1
|
Huang X, Ji M, Shang X, Zhang H, Zhang X, Zhou J, Yin T. Smart on-demand drug release strategies for cancer combination therapy. J Control Release 2025; 383:113782. [PMID: 40294796 DOI: 10.1016/j.jconrel.2025.113782] [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: 01/20/2025] [Revised: 04/06/2025] [Accepted: 04/24/2025] [Indexed: 04/30/2025]
Abstract
In cancer therapy, enhancing therapeutic indices and patient compliance has been a central focus in recent drug delivery technology development. However, achieving a delicate balance between improving anti-tumor efficacy and minimizing toxicity to normal tissues remains a significant challenge. With the advent of smart on-demand drug release strategies, new opportunities have emerged. These strategies represent a promising approach to drug delivery, enabling precise control over the release of therapeutic agents in a programmed and spatiotemporal manner. Recent studies have focused on designing delivery systems capable of releasing multiple therapeutic agents sequentially, while achieving spatial resolution in vivo. Smart on-demand drug release strategies have demonstrated considerable potential in tumor combination therapy for achieving precision drug delivery and controlled release by responding to specific physiological signals or external physical stimuli in the tumor microenvironment. These strategies not only improve tumor targeting and reduce toxicity to healthy tissues but also enable sequential release in combination therapy, allowing multiple drugs to be released in a specific spatiotemporal order to enhance synergistic treatment effects. In this paper, we systematically reviewed the current research progress of smart on-demand drug release drug delivery strategies in anti-tumor combination therapy. We highlighted representative integrated drug delivery systems and discussed the challenges associated with their clinical application. Additionally, potential future research directions are proposed to further advance this promising field.
Collapse
Affiliation(s)
- Xiaolin Huang
- Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China
| | - Mengfei Ji
- Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China
| | - Xinyu Shang
- Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China
| | - Hengchuan Zhang
- Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China
| | - Xin Zhang
- Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China
| | - Jianping Zhou
- Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China.
| | - Tingjie Yin
- Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China.
| |
Collapse
|
2
|
Li X, Xu Z. Applications of Matrix Metalloproteinase-9-Related Nanomedicines in Tumors and Vascular Diseases. Pharmaceutics 2025; 17:479. [PMID: 40284474 PMCID: PMC12030376 DOI: 10.3390/pharmaceutics17040479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Revised: 03/25/2025] [Accepted: 04/03/2025] [Indexed: 04/29/2025] Open
Abstract
Matrix metalloproteinase-9 (MMP-9) is implicated in tumor progression and vascular diseases, contributing to angiogenesis, metastasis, and extracellular matrix degradation. This review comprehensively examines the relationship between MMP-9 and these pathologies, exploring the underlying molecular mechanisms and signaling pathways involved. Specifically, we discuss the contribution of MMP-9 to tumor epithelial-mesenchymal transition, angiogenesis, and metastasis, as well as its involvement in a spectrum of vascular diseases, including macrovascular, cerebrovascular, and ocular vascular diseases. This review focuses on recent advances in MMP-9-targeted nanomedicine strategies, highlighting the design and application of responsive nanoparticles for enhanced drug delivery. These nanotherapeutic strategies leverage MMP-9 overexpression to achieve targeted drug release, improved tumor penetration, and reduced systemic toxicity. We explore various nanoparticle platforms, such as liposomes and polymer nanoparticles, and discuss their mechanisms of action, including degradation, drug release, and targeting specificity. Finally, we address the challenges posed by the heterogeneity of MMP-9 expression and their implications for personalized therapies. Ultimately, this review underscores the diagnostic and therapeutic potential of MMP-9-targeted nanomedicines against tumors and vascular diseases.
Collapse
Affiliation(s)
| | - Zhuping Xu
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu 610041, China;
| |
Collapse
|
3
|
Zhang J, Gai T, Wang J, Wu Y, Zeng SM, Zhao D, Li W. Nucleic Acid-Locked Smart Carrier for Photothermal/Chemotherapy-Amplified Immunogenic Cell Death to Enhance Systemic Antitumor Efficacy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2503299. [PMID: 40184610 DOI: 10.1002/advs.202503299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 03/25/2025] [Indexed: 04/06/2025]
Abstract
Immunotherapy holds great promise in the fight against cancer; however, it often encounters poor immunogenicity with limited therapeutic efficacy. Combining multiple treatment modalities provides a trustworthy strategy for achieving a robust antitumor effect. In this study, a nucleic acid-locked smart carrier (NASC) is developed to amplify immunogenic cell death (ICD) through the synergistic integration of photothermal therapy (PTT) and chemotherapy for high-performance monotherapy. Mesoporous silica-coated gold nanorod (MSGNR) serves as the reservoir for anticancer drug doxorubicin (DOX) and is capped with a sequence-specific duplex unit containing a tumor-specific targeting AS1411 fragment, resulting in the formation of NASC. With AS1411 targeting, the NASC can specifically target and be internalized into tumor cells with high expression of nucleolin, where the duplex capping can be unlocked by the intracellularly overexpressed adenosine triphosphate. Subsequently, the released DOX synergized with MSGNR-mediated PTT following laser irradiation induces direct cell killing, which, concurrently, triggers ICD to activate antineoplastic immunity with an increased number of T lymphocytes. This triple-collaborative strategy, further combined with anti-programmed death-1 antibody (αPD-1)-mediated immune checkpoint blockade (ICB) therapy, shows a robust therapeutic efficacy in both unilateral and bilateral tumor models.
Collapse
Affiliation(s)
- Jiayang Zhang
- Department of Chemistry, Laboratory of Advanced Materials, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Fudan University, Shanghai, 200433, P. R. China
| | - Tianyu Gai
- Department of Chemistry, Laboratory of Advanced Materials, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Fudan University, Shanghai, 200433, P. R. China
| | - Jiwei Wang
- Department of Chemistry, Laboratory of Advanced Materials, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Fudan University, Shanghai, 200433, P. R. China
| | - Yucai Wu
- Department of Chemistry, Laboratory of Advanced Materials, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Fudan University, Shanghai, 200433, P. R. China
| | - Si-Ming Zeng
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan, 430072, P. R. China
| | - Dongyuan Zhao
- Department of Chemistry, Laboratory of Advanced Materials, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Fudan University, Shanghai, 200433, P. R. China
| | - Wei Li
- Department of Chemistry, Laboratory of Advanced Materials, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Fudan University, Shanghai, 200433, P. R. China
| |
Collapse
|
4
|
Linderman SW, DeRidder L, Sanjurjo L, Foote MB, Alonso MJ, Kirtane AR, Langer R, Traverso G. Enhancing immunotherapy with tumour-responsive nanomaterials. Nat Rev Clin Oncol 2025; 22:262-282. [PMID: 40050505 DOI: 10.1038/s41571-025-01000-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/05/2025] [Indexed: 03/09/2025]
Abstract
The targeted delivery of immunotherapies to tumours using tumour-responsive nanomaterials is a promising area of cancer research with the potential to address the limitations of systemic administration such as on-target off-tumour toxicities and a lack of activity owing to the immunosuppressive tumour microenvironment (TME). Attempts to address these challenges include the design and functionalization of nanomaterials capable of releasing their cargoes in response to specific TME characteristics, thus facilitating the targeted delivery of immune-checkpoint inhibitors, cytokines, mRNAs, vaccines and, potentially, chimaeric antigen receptors as well as of agents that modulate the extracellular matrix and induce immunogenic cell death. In this Review, we describe these various research efforts in the context of the dynamic properties of the TME, such as pH, reductive conditions, reactive oxygen species, hypoxia, specific enzymes, high levels of ATP and locoregional aspects, which can be leveraged to enhance the specificity and efficacy of nanomaterial-based immunotherapies. Highlighting preclinical successes and ongoing clinical trials, we evaluate the current landscape and potential of these innovative approaches. We also consider future research directions as well as the most important barriers to successful clinical translation, emphasizing the transformative potential of tumour-responsive nanomaterials in overcoming the barriers that limit the activity of traditional immunotherapies, thus improving patient outcomes.
Collapse
Affiliation(s)
- Stephen W Linderman
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Division of Hospital Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Louis DeRidder
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Harvard-MIT Division of Health Science Technology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Lucía Sanjurjo
- Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Michael B Foote
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - María José Alonso
- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela, Santiago de Compostela, Spain
- Department of Pharmacy and Pharmaceutical Technology, University of Santiago de Compostela, Santiago de Compostela, Spain
- IMDEA Nanosciences Institute, Madrid, Spain
| | - Ameya R Kirtane
- Department of Pharmaceutics, University of Minnesota, Minneapolis, MN, USA
| | - Robert Langer
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
| | - Giovanni Traverso
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Division of Gastroenterology, Hepatology and Endoscopy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
5
|
Verma M, Yadav K, Parihar R, Dutta D, Chaudhuri S, Sivakumar S. Active tumor targeting by core-shell PDMS-HA nanoparticles with sequential delivery of doxorubicin and quercetin to overcome P-glycoprotein efflux pump. NANOSCALE 2025; 17:5033-5055. [PMID: 40013710 DOI: 10.1039/d4nr03040k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/28/2025]
Abstract
The therapeutic efficacy of chemotherapy in various malignancies and solid tumors is significantly limited when used as monotherapy. This study explored a combined treatment approach for breast cancer cells involving sequential delivery of doxorubicin followed by quercetin, both delivered via polydimethylsiloxane nanoparticles decorated with hyaluronic acid. Quercetin inhibits P-glycoprotein efflux action to enhance doxorubicin activity by increasing its intracellular accumulation; hence, both synergistically suppress cancer cell growth by promoting cytotoxicity and apoptosis. Quercetin reverses multidrug resistance, induces arrest in the cell cycle, and alters the mitochondrial membrane potential. The successful delivery and internalization of these drugs into breast cancer cells were confirmed through CD44 ligand recognition, inhibiting cell viability via apoptosis (caspase-induced) and cell arrest in the G2/M phase of the cell cycle. Furthermore, in an MCF-7 (breast cancer) cell-derived xenograft tumor model using NOD/SCID mice, the core-shell PDMS-HA nanoparticle system carrying quercetin and doxorubicin resulted in approximately 65% tumor volume reduction, outperforming the loaded single drug and free drug combination. These results were supported by the TUNEL assay and proliferation index by Ki-67 immunohistochemistry staining, which show substantial cell death and tissue necrosis in the tumor sections. Histological studies of tumor tissues confirm enhanced anticancer efficacy with negligible systemic toxicity to normal organs. Overall, the PDMS-HA delivery system efficiently transports quercetin and doxorubicin to tumor cells, enhancing the antitumor effects against the MCF-7 tumor xenograft model in mice without adverse effects. This study suggests that the targeted co-delivery of phytochemicals and anti-cancer agents can synergistically overcome many barriers associated with tumor treatment.
Collapse
Affiliation(s)
- Madhu Verma
- Department of Chemical Engineering, Indian Institute of Technology Kanpur, Kanpur, Uttar Pradesh, India.
- Department of Biotechnology, National Institute of Technology Durgapur, Durgapur, West Bengal, India.
| | - Krishna Yadav
- Central Experimental Animal Facility, Indian Institute of Technology Kanpur, Kanpur, Uttar Pradesh, India
| | - Rashmi Parihar
- Central Experimental Animal Facility, Indian Institute of Technology Kanpur, Kanpur, Uttar Pradesh, India
| | - Debjani Dutta
- Department of Biotechnology, National Institute of Technology Durgapur, Durgapur, West Bengal, India.
| | - Surabhi Chaudhuri
- Department of Biotechnology, National Institute of Technology Durgapur, Durgapur, West Bengal, India.
| | - Sri Sivakumar
- Department of Chemical Engineering, Indian Institute of Technology Kanpur, Kanpur, Uttar Pradesh, India.
- Material Science Programme, Indian Institute of Technology Kanpur, Kanpur, Uttar Pradesh, India
- Centre for Environmental Science and Engineering, Center for Nanosciences, Mehta Family Centre for Engineering in Medicine, Gangwal School of Medical Sciences and Technology Indian Institute of Technology Kanpur, Kanpur, Uttar Pradesh, India
| |
Collapse
|
6
|
Zhang L, Shi J, Zhu MH, Huang Y, Lu Q, Sun P, Chen HZ, Lai X, Fang C. Liposomes-enabled cancer chemoimmunotherapy. Biomaterials 2025; 313:122801. [PMID: 39236630 DOI: 10.1016/j.biomaterials.2024.122801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 08/05/2024] [Accepted: 09/01/2024] [Indexed: 09/07/2024]
Abstract
Chemoimmunotherapy is an emerging paradigm in the clinic for treating several malignant diseases, such as non-small cell lung cancer, breast cancer, and large B-cell lymphoma. However, the efficacy of this strategy is still restricted by serious adverse events and a high therapeutic termination rate, presumably due to the lack of tumor-targeted distribution of both chemotherapeutic and immunotherapeutic agents. Targeted drug delivery has the potential to address this issue. Among the most promising nanocarriers in clinical translation, liposomes have drawn great attention in cancer chemoimmunotherapy in recent years. Liposomes-enabled cancer chemoimmunotherapy has made significant progress in clinics, with impressive therapeutic outcomes. This review summarizes the latest preclinical and clinical progress in liposome-enabled cancer chemoimmunotherapy and discusses the challenges and future directions of this field.
Collapse
Affiliation(s)
- Lele Zhang
- Hongqiao International Institute of Medicine, Tongren Hospital and State Key Laboratory of Systems Medicine for Cancer, Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jiangpei Shi
- Hongqiao International Institute of Medicine, Tongren Hospital and State Key Laboratory of Systems Medicine for Cancer, Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Mao-Hua Zhu
- Hongqiao International Institute of Medicine, Tongren Hospital and State Key Laboratory of Systems Medicine for Cancer, Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yanhu Huang
- Hongqiao International Institute of Medicine, Tongren Hospital and State Key Laboratory of Systems Medicine for Cancer, Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Qin Lu
- Hongqiao International Institute of Medicine, Tongren Hospital and State Key Laboratory of Systems Medicine for Cancer, Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Peng Sun
- Department of General Surgery, Tongren Hospital, SJTU-SM, Shanghai, 200336, China
| | - Hong-Zhuan Chen
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Biomedical Research, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Xing Lai
- Hongqiao International Institute of Medicine, Tongren Hospital and State Key Laboratory of Systems Medicine for Cancer, Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Chao Fang
- Hongqiao International Institute of Medicine, Tongren Hospital and State Key Laboratory of Systems Medicine for Cancer, Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China; Key Laboratory of Basic Pharmacology of Ministry of Education & Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, 563003, China.
| |
Collapse
|
7
|
Hou H, Liu X, Liu J, Wang Y. Carbohydrate polymer-based nanoparticles with cell membrane camouflage for cancer therapy: A review. Int J Biol Macromol 2025; 289:138620. [PMID: 39674458 DOI: 10.1016/j.ijbiomac.2024.138620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 11/21/2024] [Accepted: 12/08/2024] [Indexed: 12/16/2024]
Abstract
Recent developments in biomimetic nanoparticles, specifically carbohydrate polymer-coated cell membrane nanoparticles, have demonstrated considerable promise in treating cancer. These systems improve drug delivery by imitating natural cell actions, enhancing biocompatibility, and decreasing immune clearance. Conventional drug delivery methods frequently face challenges with non-specific dispersal and immune detection, which can hinder their efficiency and safety. These biomimetic nanoparticles improve target specificity, retention times, and therapeutic efficiency by using biological components like chitosan, hyaluronic acid, and alginate. Chitosan-based nanoparticles, which come from polysaccharides found in nature, have self-assembly abilities that make them better drug carriers. Hyaluronic acid helps target tissues more effectively, especially in cancer environments where there are high levels of hyaluronic acid receptors. Alginate-based systems also enhance drug delivery by being biocompatible and degradable, making them ideal choices for advanced therapeutic uses. Moreover, these particles hold potential for overcoming resistance to multiple drugs and boosting the body's immune reaction to tumors through precise delivery and decreased side effects of chemotherapy drugs. This review delves into the possibilities of using carbohydrate polymer-functionalized nanoparticles and their impact on enhancing the efficacy of cancer treatment.
Collapse
Affiliation(s)
- Haijia Hou
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Xuejian Liu
- Department of Pulmonary and Critical Care Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jun Liu
- Department of Thoracic Surgery, Shengjing Hospital of China Medical University, Shenyang, China.
| | - Yudong Wang
- Department of Thoracic Surgery, Shengjing Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
8
|
Yang Z, Xu C, Lee JX, Lum GZ. Magnetic Miniature Soft Robot with Reprogrammable Drug-Dispensing Functionalities: Toward Advanced Targeted Combination Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2408750. [PMID: 39246210 DOI: 10.1002/adma.202408750] [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: 06/19/2024] [Revised: 08/26/2024] [Indexed: 09/10/2024]
Abstract
Miniature robots are untethered actuators, which have great prospects to transform targeted drug delivery because they can potentially deliver high concentrations of medicine to the disease site(s) with minimal complications. However, existing miniature robots cannot perform advanced targeted combination therapy; majority of them can at most transport one type of drug, while those that can carry multiple drugs are unable to change their drug-dispensing sequence and dosage. Furthermore, the latter robots cannot transport more than three types of drugs, selectively dispense their drugs, maintain their mobility, or release their drugs at multiple sites. Here, a millimeter-scale soft robot is proposed, which can be actuated by alternating magnetic fields to dispense four types of drugs with reprogrammable drug-dispensing sequence and dosage (dispensing rates: 0.0992-0.231 µL h-1). This robot has six degrees-of-freedom motions, and it can deliver its drugs to multiple desired sites by rolling and two-anchor crawling across unstructured environments with negligible drug leakage. Such dexterity is highly desirable and unprecedented for miniature robots with drug-dispensing capabilities. The soft robot therefore has great potential to enable advanced targeted combination therapy, where four types of drugs must be delivered to various disease sites, each with a specific sequence and dosage of drugs.
Collapse
Affiliation(s)
- Zilin Yang
- School of Mechanical and Aerospace Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore, 639798, Singapore
| | - Changyu Xu
- School of Mechanical and Aerospace Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore, 639798, Singapore
| | - Jia Xin Lee
- Rehabilitation Research Institute of Singapore, Nanyang Technological University, 11 Mandalay Road, Singapore, 308232, Singapore
| | - Guo Zhan Lum
- School of Mechanical and Aerospace Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore, 639798, Singapore
- Rehabilitation Research Institute of Singapore, Nanyang Technological University, 11 Mandalay Road, Singapore, 308232, Singapore
| |
Collapse
|
9
|
Ashkarran AA, Lin Z, Rana J, Bumpers H, Sempere L, Mahmoudi M. Impact of Nanomedicine in Women's Metastatic Breast Cancer. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2301385. [PMID: 37269217 PMCID: PMC10693652 DOI: 10.1002/smll.202301385] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 05/16/2023] [Indexed: 06/04/2023]
Abstract
Metastatic breast cancer is responsible for 90% of mortalities among women suffering from various types of breast cancers. Traditional cancer treatments such as chemotherapy and radiation therapy can cause significant side effects and may not be effective in many cases. However, recent advances in nanomedicine have shown great promise in the treatment of metastatic breast cancer. For example, nanomedicine demonstrated robust capacity in detection of metastatic cancers at early stages (i.e., before the metastatic cells leave the initial tumor site), which gives clinicians a timely option to change their treatment process (for example, instead of endocrine therapy they may use chemotherapy). Here recent advances in nanomedicine technology in the identification and treatment of metastatic breast cancers are reviewed.
Collapse
Affiliation(s)
- Ali Akbar Ashkarran
- Department of Radiology and Precision Health Program, Michigan State University, East Lansing, MI, 48824, USA
| | - Zijin Lin
- Department of Radiology and Precision Health Program, Michigan State University, East Lansing, MI, 48824, USA
| | - Jatin Rana
- Division of Hematology and Oncology, Michigan State University, East Lansing, MI, 48824, USA
| | - Harvey Bumpers
- Department of Surgery, Michigan State University, East Lansing, MI, 48824, USA
| | - Lorenzo Sempere
- Department of Radiology and Precision Health Program, Michigan State University, East Lansing, MI, 48824, USA
| | - Morteza Mahmoudi
- Department of Radiology and Precision Health Program, Michigan State University, East Lansing, MI, 48824, USA
- Connors Center for Women's Health & Gender Biology, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| |
Collapse
|
10
|
Zou J, Xing X, Teng C, Zhao Q, He W, Wu X, Xia Y. Cocrystal@protein-anchoring nanococktail for combinatorially treating multidrug-resistant cancer. Acta Pharm Sin B 2024; 14:4509-4525. [PMID: 39525582 PMCID: PMC11544293 DOI: 10.1016/j.apsb.2024.08.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 06/17/2024] [Accepted: 07/10/2024] [Indexed: 11/16/2024] Open
Abstract
Multidrug resistance (MDR), the major mechanism by which various cancers develop specific resistance to therapeutic agents, has set up enormous obstacles to many forms of tumor chemotherapy. Traditional cocktail therapy administration, based on the combination of multiple drugs for anti-MDR chemotherapy, often suffers from inconsistent in vivo pharmacokinetic behaviors that cannot act synchronously on the lesions, leading to limited pharmacodynamic outcomes. Despite the emergence of nanomedicines, which has improved chemotherapeutic drugs' bioavailability and therapeutic effect on clinical application, these monotherapy-based nano-formulations still show poor progression in overcoming MDR. Herein, a "one stone and three birds" nanococktail integrated by a cocrystal@protein-anchoring strategy was purposed for triple-payload delivery, which paclitaxel-disulfiram cocrystal-like nanorods (NRs) were anchored with the basic protein drug Cytochrome c (Cyt C), followed by hyaluronic-acid modification. In particular, NRs were utilized as carrier-like particles to synchronously deliver biomacromolecule Cyt C into tumor cells and then promote cell apoptosis. Of note, on A549/Taxol drug-resistant tumor-bearing mice, the system with extraordinarily high encapsulation efficiency demonstrated prolonged in vivo circulation and increased tumor-targeting accumulation, significantly reversing tumor drug resistance and improving therapeutic efficacy. Our mechanistic study indicated that the system induced the apoptosis of Taxol-resistant tumor cells through the signal axis P-glycoprotein/Cyt C/caspase 3. Collectively, this nanococktail strategy offers a promising approach to improve the sensitivity of tumor cells to chemotherapeutic drugs and strengthen intractable drug-resistant oncotherapy.
Collapse
Affiliation(s)
- Jiahui Zou
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Xuyang Xing
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Chao Teng
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Qingling Zhao
- School of Computer Science and Engineering, Nanjing University of Science and Technology, Nanjing 210094, China
| | - Wei He
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Xuri Wu
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China
| | - Yuanzheng Xia
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Bioactive Natural Product Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| |
Collapse
|
11
|
Paul V J, Sharma P, Shanavas A. Self-Assembled Nanobiomaterials for Combination Immunotherapy. ACS APPLIED BIO MATERIALS 2024; 7:4962-4974. [PMID: 38116786 DOI: 10.1021/acsabm.3c00826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2023]
Abstract
Nanotechnological interventions for cancer immunotherapy are a rapidly evolving paradigm with immense potential. Self-assembled nanobiomaterials present safer alternatives to their nondegradable counterparts and pose better functionalities in terms of controlled drug delivery and phototherapy to activate immunogenic cell death. In this Review, we discuss several classes of self-assembled nanobiomaterials based on polymers, lipids, peptides, hydrogel, metal organic frameworks, and covalent-organic frameworks with the ability to activate systemic immune response and convert a "cold" immunosuppressive tumor mass to a "hot" antitumor immune cell rich microenvironment. The unique aspects of these materials are underpinned, and their mechanisms of combinatorial immunotherapeutic action are discussed. Future challenges associated with their clinical translation are also highlighted.
Collapse
Affiliation(s)
- Johns Paul V
- Inorganic & Organic Nanomedicine (ION) Lab, Chemical Biology Unit, Institute of Nano Science and Technology, Sector-81, Knowledge City, Sahibzada Ajit Singh Nagar, Punjab 140306, India
| | - Priyanka Sharma
- Inorganic & Organic Nanomedicine (ION) Lab, Chemical Biology Unit, Institute of Nano Science and Technology, Sector-81, Knowledge City, Sahibzada Ajit Singh Nagar, Punjab 140306, India
| | - Asifkhan Shanavas
- Inorganic & Organic Nanomedicine (ION) Lab, Chemical Biology Unit, Institute of Nano Science and Technology, Sector-81, Knowledge City, Sahibzada Ajit Singh Nagar, Punjab 140306, India
| |
Collapse
|
12
|
Hayward D, Goddard ZR, Cominetti MMD, Searcey M, Beekman AM. Light-activated azobenzene peptide inhibitor of the PD-1/PD-L1 interaction. Chem Commun (Camb) 2024; 60:8228-8231. [PMID: 39007209 PMCID: PMC11293026 DOI: 10.1039/d4cc01249f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 07/05/2024] [Indexed: 07/16/2024]
Abstract
Inhibiting the PD-1/PD-L1 protein-protein interaction is a key immunotherapy for cancer. Antibodies dominate the clinical space but are costly, with limited applicability and immune side effects. We developed a photo-controlled azobenzene peptide that selectively inhibits the PD-1/PD-L1 interaction when in the cis isomer only. Activity is demonstrated in in vitro and cellular assays.
Collapse
Affiliation(s)
- Deanne Hayward
- School of Pharmacy, University of East Anglia, Norwich Research Park, Norwich, Norfolk, NR47TJ, UK.
| | - Zoë R Goddard
- School of Pharmacy, University of East Anglia, Norwich Research Park, Norwich, Norfolk, NR47TJ, UK.
| | - Marco M D Cominetti
- School of Pharmacy, University of East Anglia, Norwich Research Park, Norwich, Norfolk, NR47TJ, UK.
| | - Mark Searcey
- School of Pharmacy, University of East Anglia, Norwich Research Park, Norwich, Norfolk, NR47TJ, UK.
| | - Andrew M Beekman
- School of Pharmacy, University of East Anglia, Norwich Research Park, Norwich, Norfolk, NR47TJ, UK.
| |
Collapse
|
13
|
Pan Y, Yu L, Liu L, Zhang J, Liang S, Parshad B, Lai J, Ma LM, Wang Z, Rao L. Genetically engineered nanomodulators elicit potent immunity against cancer stem cells by checkpoint blockade and hypoxia relief. Bioact Mater 2024; 38:31-44. [PMID: 38699238 PMCID: PMC11061653 DOI: 10.1016/j.bioactmat.2024.04.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 04/08/2024] [Accepted: 04/08/2024] [Indexed: 05/05/2024] Open
Abstract
Rapid development of checkpoint inhibitors has provided significant breakthroughs for cancer stem cell (CSC) therapy, while the therapeutic efficacy is restricted by hypoxia-mediated tumor immune evasion, especially hypoxia-induced CD47 overexpression in CSCs. Herein, we developed a genetically engineered CSC membrane-coated hollow manganese dioxide (hMnO2@gCMs) to elicit robust antitumor immunity by blocking CD47 and alleviating hypoxia to ultimately achieve the eradication of CSCs. The hMnO2 core effectively alleviated tumor hypoxia by inducing decomposition of tumor endogenous H2O2, thus suppressing the CSCs and reducing the expression of CD47. Cooperating with hypoxia relief-induced downregulation of CD47, the overexpressed SIRPα on gCM shell efficiently blocked the CD47-SIRPα "don't eat me" pathway, synergistically eliciting robust antitumor-mediated immune responses. In a B16F10-CSC bearing melanoma mouse model, the hMnO2@gCMs showed an enhanced therapeutic effect in eradicating CSCs and inhibiting tumor growth. Our work presents a simple, safe, and robust platform for CSC eradication and cancer immunotherapy.
Collapse
Affiliation(s)
- Yuanwei Pan
- The Research and Application Center of Precision Medicine, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, 450014, China
- Institute of Biomedical Health Technology and Engineering, Shenzhen Bay Laboratory, Shenzhen, 518132, China
| | - Ling Yu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
- Department of Critical Care Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, 510120, China
| | - Lujie Liu
- Institute of Biomedical Health Technology and Engineering, Shenzhen Bay Laboratory, Shenzhen, 518132, China
- Medical Research Center, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Jing Zhang
- Institute of Biomedical Health Technology and Engineering, Shenzhen Bay Laboratory, Shenzhen, 518132, China
| | - Shuang Liang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Badri Parshad
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02129, USA
| | - Jialin Lai
- Institute of Biomedical Health Technology and Engineering, Shenzhen Bay Laboratory, Shenzhen, 518132, China
| | - Li-Min Ma
- Medical Research Center, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Zhaohui Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Lang Rao
- The Research and Application Center of Precision Medicine, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, 450014, China
- Institute of Biomedical Health Technology and Engineering, Shenzhen Bay Laboratory, Shenzhen, 518132, China
| |
Collapse
|
14
|
Kong C, Sun J, Hu X, Li G, Wu S. A tumor targeted nano micelle carrying astragaloside IV for combination treatment of bladder cancer. Sci Rep 2024; 14:17704. [PMID: 39085255 PMCID: PMC11291986 DOI: 10.1038/s41598-024-66010-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 06/26/2024] [Indexed: 08/02/2024] Open
Abstract
Immune checkpoint inhibitors (ICIs) are effective agents for tumor immunotherapy. However, their clinical effectiveness is unsatisfactory due to off-target effects and a suppressive immune microenvironment. This study developed a nanodrug delivery system for bladder cancer (BCa) using PCL-MPEG and PCL-PEG-CHO to synthesize internal hydrophobic and external hydrophilic micelles (PP) that encapsulated water-insoluble astragaloside IV (PPA). The aldehyde group on the surface of PPA reacted with the amino group of aPD-L1, allowing the decoration of this antibody on the surface of the micelles. The resultingPPA@aPD-L1effectively piggybacked astragaloside IV and aPD-L1 antibody. These findings suggest that PPA@aPD-L1 is relatively stable in circulation and efficiently binds to BCa cells with the aid of aPD-L1. Additionally, this strategy prolongs the drug's retention time in tumors. Compared to PBS, PP, and PPA with PPA + aPD-L1 groups, PPA@aPD-L1significantly prolonged the survival of mice with BCa and reduced tumor volume. Mechanistic studies showed that PPA inhibited the NF-κB and STAT3 signaling pathways in tumor cells. Additionally, PPA@aPD-L1increased IFN-γ and decreased IL-10 expression in bladder tumors, affecting the number and type of intratumorally infiltrating T cells. Our study presents a simple and effective drug delivery system that combines herbal monomers with ICIs. It has demonstrated a potent ability to suppress tumor growth and holds potential for future applications.
Collapse
Affiliation(s)
- Chenfan Kong
- Institute of Urology, The affiliated Luohu Hospital of Shenzhen University, Shenzhen University, Shenzhen, 518000, China
- Science and Education Department, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, 518033, China
- Department of Oncology, Shenzhen Baoan People's Hospital, Shenzhen, 518101, China
| | - Jianrong Sun
- Department of Oncology, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, 518033, China
| | - Xinzi Hu
- Department of Oncology, Shenzhen Baoan People's Hospital, Shenzhen, 518101, China
| | - Guangzhi Li
- Institute of Urology, The affiliated Luohu Hospital of Shenzhen University, Shenzhen University, Shenzhen, 518000, China.
| | - Song Wu
- Department of Urology, The Affiliated Shenzhen Hospital of Shanghai University of Traditional Chinese Medicine, Shenzhen, 518009, China.
| |
Collapse
|
15
|
Zhang T, Ping W, Suo M, Pan Y, Huang R, Chen J, Lyu M, Zhang N, Ning S, Tang BZ. Stimuli-Responsive Hydrogels Potentiating Photothermal Therapy against Cancer Stem Cell-Induced Breast Cancer Metastasis. ACS NANO 2024. [PMID: 39046933 DOI: 10.1021/acsnano.4c04067] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/27/2024]
Abstract
The self-renewal and differentiation properties of cancer stem cells (CSCs) result in chemoresistance in breast cancer. Even though numerous drugs have been developed to target CSCs, they have suffered from inefficient delivery and accumulation at the focal site. Here, a thermoresponsive hydrogel is developed by coencapsulating aggregation-induced emission (AIE)-active photothermal agent and thioridazine (THZ), demonstrating a controllable delivery system triggered by the AIE agent to augment THZ-mediated CSC ablation. Upon near-infrared laser stimuli, the photothermal effect from the AIE agent induces hydrogel deformation for burst drug release. The precise in situ tumor administration of the hydrogel accelerates drug diffusion and accumulation in deep breast cancer lesions. Thus, THZ can invade tumors and provoke massive CSC apoptosis via dopamine receptor blockage and oxidative stress induction. Consequently, effective CSC inhibition and significant suppression of tumor recurrence and metastasis are demonstrated in mice with breast cancer. We believe that this intelligent hydrogel-based delivery system represents a promising treatment strategy for metastatic breast cancer with clinical potential.
Collapse
Affiliation(s)
- Tianfu Zhang
- Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, School of Biomedical Engineering, Guangzhou Medical University, Guangdong 511436, China
| | - Wei Ping
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong, University of Science and Technology, Wuhan, Hubei 430030, China
| | - Meng Suo
- Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, School of Biomedical Engineering, Guangzhou Medical University, Guangdong 511436, China
| | - You Pan
- Department of Breast Surgery, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530000, China
| | - Rong Huang
- Department of Breast Surgery, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530000, China
| | - Jingqi Chen
- Department of Breast Surgery, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530000, China
| | - Meng Lyu
- Department of Gastrointestinal Surgery and Department of Geriatrics, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020, China
| | - Ni Zhang
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong, University of Science and Technology, Wuhan, Hubei 430030, China
| | - Shipeng Ning
- Department of Breast Surgery, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530000, China
| | - Ben Zhong Tang
- School of Science and Engineering, Shenzhen Institute of Aggregate Science and Technology, The Chinese University of Hong Kong, Shenzhen (CUHK-Shenzhen), Guangdong 518172, China
| |
Collapse
|
16
|
Cheng Z, Fobian SF, Gurrieri E, Amin M, D'Agostino VG, Falahati M, Zalba S, Debets R, Garrido MJ, Saeed M, Seynhaeve ALB, Balcioglu HE, Ten Hagen TLM. Lipid-based nanosystems: the next generation of cancer immune therapy. J Hematol Oncol 2024; 17:53. [PMID: 39030582 PMCID: PMC11265205 DOI: 10.1186/s13045-024-01574-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 07/11/2024] [Indexed: 07/21/2024] Open
Abstract
Immunotherapy has become an important part of the oncotherapy arsenal. Its applicability in various cancer types is impressive, as well as its use of endogenous mechanisms to achieve desired ends. However, off-target or on-target-off-tumor toxicity, limited activity, lack of control in combination treatments and, especially for solid tumors, low local accumulation, have collectively limited clinical use thereof. These limitations are partially alleviated by delivery systems. Lipid-based nanoparticles (NPs) have emerged as revolutionary carriers due to favorable physicochemical characteristics, with specific applications and strengths particularly useful in immunotherapeutic agent delivery. The aim of this review is to highlight the challenges faced by immunotherapy and how lipid-based NPs have been, and may be further utilized to address such challenges. We discuss recent fundamental and clinical applications of NPs in a range of areas and provide a detailed discussion of the main obstacles in immune checkpoint inhibition therapies, adoptive cellular therapies, and cytokine therapies. We highlight how lipid-based nanosystems could address these through either delivery, direct modulation of the immune system, or targeting of the immunosuppressive tumor microenvironment. We explore advanced and emerging liposomal and lipid nanoparticle (LNP) systems for nucleic acid delivery, intrinsic and extrinsic stimulus-responsive formulations, and biomimetic lipid-based nanosystems in immunotherapy. Finally, we discuss the key challenges relating to the clinical use of lipid-based NP immunotherapies, suggesting future research directions for the near term to realize the potential of these innovative lipid-based nanosystems, as they become the crucial steppingstone towards the necessary enhancement of the efficacy of immunotherapy.
Collapse
Affiliation(s)
- Ziyun Cheng
- Precision Medicine in Oncology (PrMiO), Department of Pathology, Erasmus MC Cancer Institute, Erasmus Medical Center, Rotterdam, The Netherlands
- Nanomedicine Innovation Center Erasmus (NICE), Erasmus Medical Center, Rotterdam, The Netherlands
| | - Seth-Frerich Fobian
- Precision Medicine in Oncology (PrMiO), Department of Pathology, Erasmus MC Cancer Institute, Erasmus Medical Center, Rotterdam, The Netherlands
- Nanomedicine Innovation Center Erasmus (NICE), Erasmus Medical Center, Rotterdam, The Netherlands
| | - Elena Gurrieri
- Laboratory of Biotechnology and Nanomedicine, Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Mohamadreza Amin
- Precision Medicine in Oncology (PrMiO), Department of Pathology, Erasmus MC Cancer Institute, Erasmus Medical Center, Rotterdam, The Netherlands
- Nanomedicine Innovation Center Erasmus (NICE), Erasmus Medical Center, Rotterdam, The Netherlands
| | - Vito Giuseppe D'Agostino
- Laboratory of Biotechnology and Nanomedicine, Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Mojtaba Falahati
- Precision Medicine in Oncology (PrMiO), Department of Pathology, Erasmus MC Cancer Institute, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Sara Zalba
- Department of Pharmaceutical Sciences, School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Navarra Institute for Health Research, Pamplona, Spain
| | - Reno Debets
- Laboratory of Tumor Immunology, Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus Medical Center, Rotterdam, The Netherlands
| | - María J Garrido
- Department of Pharmaceutical Sciences, School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Navarra Institute for Health Research, Pamplona, Spain
| | - Mesha Saeed
- Precision Medicine in Oncology (PrMiO), Department of Pathology, Erasmus MC Cancer Institute, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Ann L B Seynhaeve
- Precision Medicine in Oncology (PrMiO), Department of Pathology, Erasmus MC Cancer Institute, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Hayri E Balcioglu
- Laboratory of Tumor Immunology, Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus Medical Center, Rotterdam, The Netherlands.
| | - Timo L M Ten Hagen
- Precision Medicine in Oncology (PrMiO), Department of Pathology, Erasmus MC Cancer Institute, Erasmus Medical Center, Rotterdam, The Netherlands.
- Nanomedicine Innovation Center Erasmus (NICE), Erasmus Medical Center, Rotterdam, The Netherlands.
| |
Collapse
|
17
|
Sun J, Gong J, Gong L, Zhu C, Li-Yang L, Wang J, Yang Y, Zhang S, Liu S, Fu JJ, Xu P. High Manganese Content of Lipid NanoMn (LNM) by Microfluidic Technology for Enhancing Anti-Tumor Immunity. Pharmaceutics 2024; 16:556. [PMID: 38675217 PMCID: PMC11054818 DOI: 10.3390/pharmaceutics16040556] [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: 02/18/2024] [Revised: 03/30/2024] [Accepted: 04/05/2024] [Indexed: 04/28/2024] Open
Abstract
Immunotherapy is a clinically effective method for treating tumors. Manganese can activate the cGAS-STING signaling pathway and induce an anti-tumor immune response. However, its efficacy is hindered by non-specific distribution and low uptake rates. In this study, we employed microfluidic technology to design and develop an innovative preparation process, resulting in the creation of a novel manganese lipid nanoparticle (LNM). The lipid manganese nanoparticle produced in this process boasts a high manganese payload, excellent stability, the capacity for large-scale production, and high batch repeatability. LNM has effectively demonstrated the ability to activate the cGAS-STING signaling pathway, induce the production of pro-inflammatory cytokines, and inhibit tumor development. Notably, LNM does not require combination chemotherapy drugs or other immune activators. Therefore, LNM presents a safe, straightforward, and efficient strategy for anti-tumor immune activation, with the potential for scalable production.
Collapse
Affiliation(s)
- Jiawei Sun
- Department of Pharmaceutics, College of Pharmacy, Inner Mongolia Medical University, Hohhot 010110, China; (J.S.); (S.L.)
| | - Jingjing Gong
- Beijing Key Laboratory of Tumor Systems Biology, Institute of Systems Biomedicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China; (J.G.); (L.G.); (C.Z.); (L.L.-Y.); (J.W.); (Y.Y.); (S.Z.)
| | - Lidong Gong
- Beijing Key Laboratory of Tumor Systems Biology, Institute of Systems Biomedicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China; (J.G.); (L.G.); (C.Z.); (L.L.-Y.); (J.W.); (Y.Y.); (S.Z.)
| | - Chuanda Zhu
- Beijing Key Laboratory of Tumor Systems Biology, Institute of Systems Biomedicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China; (J.G.); (L.G.); (C.Z.); (L.L.-Y.); (J.W.); (Y.Y.); (S.Z.)
| | - Longhao Li-Yang
- Beijing Key Laboratory of Tumor Systems Biology, Institute of Systems Biomedicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China; (J.G.); (L.G.); (C.Z.); (L.L.-Y.); (J.W.); (Y.Y.); (S.Z.)
| | - Jingya Wang
- Beijing Key Laboratory of Tumor Systems Biology, Institute of Systems Biomedicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China; (J.G.); (L.G.); (C.Z.); (L.L.-Y.); (J.W.); (Y.Y.); (S.Z.)
| | - Yuanyuan Yang
- Beijing Key Laboratory of Tumor Systems Biology, Institute of Systems Biomedicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China; (J.G.); (L.G.); (C.Z.); (L.L.-Y.); (J.W.); (Y.Y.); (S.Z.)
| | - Shiming Zhang
- Beijing Key Laboratory of Tumor Systems Biology, Institute of Systems Biomedicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China; (J.G.); (L.G.); (C.Z.); (L.L.-Y.); (J.W.); (Y.Y.); (S.Z.)
| | - Silu Liu
- Department of Pharmaceutics, College of Pharmacy, Inner Mongolia Medical University, Hohhot 010110, China; (J.S.); (S.L.)
| | - Ji-Jun Fu
- School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Pengcheng Xu
- Department of Pharmaceutics, College of Pharmacy, Inner Mongolia Medical University, Hohhot 010110, China; (J.S.); (S.L.)
| |
Collapse
|
18
|
Sun S, Yang Q, Jiang D, Zhang Y. Nanobiotechnology augmented cancer stem cell guided management of cancer: liquid-biopsy, imaging, and treatment. J Nanobiotechnology 2024; 22:176. [PMID: 38609981 PMCID: PMC11015566 DOI: 10.1186/s12951-024-02432-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Accepted: 03/20/2024] [Indexed: 04/14/2024] Open
Abstract
Cancer stem cells (CSCs) represent both a key driving force and therapeutic target of tumoral carcinogenesis, tumor evolution, progression, and recurrence. CSC-guided tumor diagnosis, treatment, and surveillance are strategically significant in improving cancer patients' overall survival. Due to the heterogeneity and plasticity of CSCs, high sensitivity, specificity, and outstanding targeting are demanded for CSC detection and targeting. Nanobiotechnologies, including biosensors, nano-probes, contrast enhancers, and drug delivery systems, share identical features required. Implementing these techniques may facilitate the overall performance of CSC detection and targeting. In this review, we focus on some of the most recent advances in how nanobiotechnologies leverage the characteristics of CSC to optimize cancer diagnosis and treatment in liquid biopsy, clinical imaging, and CSC-guided nano-treatment. Specifically, how nanobiotechnologies leverage the attributes of CSC to maximize the detection of circulating tumor DNA, circulating tumor cells, and exosomes, to improve positron emission computed tomography and magnetic resonance imaging, and to enhance the therapeutic effects of cytotoxic therapy, photodynamic therapy, immunotherapy therapy, and radioimmunotherapy are reviewed.
Collapse
Affiliation(s)
- Si Sun
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Qiang Yang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Dawei Jiang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China.
- Key Laboratory of Biological Targeted Therapy, the Ministry of Education, Wuhan, 430022, China.
| | - Yuan Zhang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
19
|
Ibrahim AA, Nsairat H, Al-Sulaibi M, El-Tanani M, Jaber AM, Lafi Z, Barakat R, Abuarqoub DA, Mahmoud IS, Obare SO, Aljabali AAA, Alkilany AM, Alshaer W. Doxorubicin conjugates: a practical approach for its cardiotoxicity alleviation. Expert Opin Drug Deliv 2024; 21:399-422. [PMID: 38623735 DOI: 10.1080/17425247.2024.2343882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 02/29/2024] [Indexed: 04/17/2024]
Abstract
INTRODUCTION Doxorubicin (DOX) emerges as a cornerstone in the arsenal of potent chemotherapeutic agents. Yet, the clinical deployment of DOX is tarnished by its proclivity to induce severe cardiotoxic effects, culminating in heart failure and other consequential morbidities. In response, a panoply of strategies has undergone rigorous exploration over recent decades, all aimed at attenuating DOX's cardiotoxic impact. The advent of encapsulating DOX within lipidic or polymeric nanocarriers has yielded a dual triumph, augmenting DOX's therapeutic efficacy while mitigating its deleterious side effects. AREAS COVERED Recent strides have spotlighted the emergence of DOX conjugates as particularly auspicious avenues for ameliorating DOX-induced cardiotoxicity. These conjugates entail the fusion of DOX through physical or chemical bonds with diminutive natural or synthetic moieties, polymers, biomolecules, and nanoparticles. This spectrum encompasses interventions that impinge upon DOX's cardiotoxic mechanism, modulate cellular uptake and localization, confer antioxidative properties, or refine cellular targeting. EXPERT OPINION The endorsement of DOX conjugates as a compelling stratagem to mitigate DOX-induced cardiotoxicity resounds from this exegesis, amplifying safety margins and the therapeutic profile of this venerated chemotherapeutic agent. Within this ambit, DOX conjugates stand as a beacon of promise in the perpetual pursuit of refining chemotherapy-induced cardiac compromise.
Collapse
Affiliation(s)
- Abed Alqader Ibrahim
- Department of Nanoscience, Joint School of Nanoscience and Nanoengineering, University of North Carolina at Greensboro, Greensboro, NC, USA
| | - Hamdi Nsairat
- Pharmacological and Diagnostic Research Center, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman, Jordan
| | - Mazen Al-Sulaibi
- Pharmacological and Diagnostic Research Center, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman, Jordan
| | - Mohamed El-Tanani
- Pharmacological and Diagnostic Research Center, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman, Jordan
- College of Pharmacy, Ras Al Khaimah Medical and Health Sciences University, Ras Al Khaimah, United Arab Emirates
| | - Areej M Jaber
- Pharmacological and Diagnostic Research Center, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman, Jordan
| | - Zainab Lafi
- Pharmacological and Diagnostic Research Center, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman, Jordan
| | - Rahmeh Barakat
- Pharmacological and Diagnostic Research Center, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman, Jordan
| | - Duaa Azmi Abuarqoub
- Department of Pharmacology and Biomedical Sciences, Faculty of Pharmacy and Medical Sciences, University of Petra, Amman, Jordan
- Cell Therapy Center, The University of Jordan, Amman, Jordan
| | - Ismail Sami Mahmoud
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, The Hashemite University, Zarqa, Jordan
| | - Sherine O Obare
- Department of Nanoscience, Joint School of Nanoscience and Nanoengineering, University of North Carolina at Greensboro, Greensboro, NC, USA
- Department of Nanoengineering, Joint School of Nanoscience and Nanoengineering, North Carolina Agricultural and Technical State University, Greensboro, NC, USA
| | - Alaa A A Aljabali
- Faculty of Pharmacy, Department of Pharmaceutical Sciences, Yarmouk University, Irbid, Jordan
| | | | - Walhan Alshaer
- Cell Therapy Center, The University of Jordan, Amman, Jordan
| |
Collapse
|
20
|
Guo F, Du Y, Wang Y, Wang M, Wang L, Yu N, Luo S, Wu F, Yang G. Targeted drug delivery systems for matrix metalloproteinase-responsive anoparticles in tumor cells: A review. Int J Biol Macromol 2024; 257:128658. [PMID: 38065446 DOI: 10.1016/j.ijbiomac.2023.128658] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 12/04/2023] [Accepted: 12/05/2023] [Indexed: 01/26/2024]
Abstract
Nanodrug delivery systems based on tumor microenvironment responses have shown excellent performance in tumor-targeted therapy, given their unique targeting and drug-release characteristics. Matrix metalloproteinases (MMPs) have been widely explored owing to their high specificity and expression in various tumor microenvironments. The design of an enzyme-sensitive nanodelivery system using MMPs as targeted receptors could markedly improve the performance of drug targeting. The current review focuses on the development and application of MMP-responsive drug carriers, and summarizes the classification of single- and multi-target nanocarriers based on their MMP responsiveness. The potential applications and challenges of this nanodrug delivery system are discussed to provide a reference for designing high-performance nanodrug delivery systems.
Collapse
Affiliation(s)
- Fangyuan Guo
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China; Research Institute of Pharmaceutical Particle Technology, Zhejiang University of Technology, Hangzhou 310014, China
| | - Yinzhou Du
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China
| | - Yujia Wang
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China
| | - Mengqi Wang
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China
| | - Lianyi Wang
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China
| | - Nan Yu
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China
| | - Shuai Luo
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China
| | - Fang Wu
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China
| | - Gensheng Yang
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China; Research Institute of Pharmaceutical Particle Technology, Zhejiang University of Technology, Hangzhou 310014, China.
| |
Collapse
|
21
|
Lu Y, Chen L, Wu Z, Zhou P, Dai J, Li J, Wen Q, Fan Y, Zeng F, Chen Y, Fu S. Self-driven bioactive hybrids co-deliver doxorubicin and indocyanine green nanoparticles for chemo/photothermal therapy of breast cancer. Biomed Pharmacother 2023; 169:115846. [PMID: 37944443 DOI: 10.1016/j.biopha.2023.115846] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Revised: 10/27/2023] [Accepted: 11/05/2023] [Indexed: 11/12/2023] Open
Abstract
Breast cancer is characterized by insidious onset, rapid progression, easy recurrence, and metastasis. Conventional monotherapies are usually ineffective due to insufficient drug delivery. Therefore, the combination of multimodal therapy with tumor microenvironment (TME)-responsive nanoplatforms is increasingly being considered for the targeted treatment of breast cancer. We synthesized bioactive hybrid nanoparticles for synergistic chemotherapy and photothermal therapy. Briefly, doxorubicin (DOX) and indocyanine green (ICG)-loaded nanoparticles (DI) of average particle size 113.58 ± 2.14 nm were synthesized, and their surface were modified with polydopamine (PDA) and attached to the anaerobic probiotic Bifidobacterium infantis (Bif). The bioactive Bif@DIP hybrid showed good photothermal conversion efficiency of about 38.04%. In addition, the self-driving ability of Bif allowed targeted delivery of the PDA-coated DI nanoparticles (DIP) to the hypoxic regions of the tumor. The low pH and high GSH levels in the TME stimulated the controlled release of DOX and ICG from the Bif@DIP hybrid, which then triggered apoptosis of tumor cells and induced immunogenic cell death (ICD), resulting in effective and sustained anti-tumor effect with minimum systemic toxicity. Thus, the self-driven Bif@DIP hybrid is a promising nanodrug for the targeted chemotherapy and photothermal therapy against solid cancers.
Collapse
Affiliation(s)
- Yun Lu
- Department of Oncology, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, PR China
| | - Lan Chen
- Department of Oncology, the Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan 646000, PR China
| | - Zhouxue Wu
- Department of Oncology, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, PR China
| | - Ping Zhou
- Department of Radiology, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, PR China
| | - Jie Dai
- Department of Oncology, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, PR China
| | - Jianmei Li
- Department of Oncology, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, PR China
| | - Qian Wen
- Department of Oncology, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, PR China
| | - Yu Fan
- Department of Oncology, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, PR China
| | - Fancai Zeng
- Laboratory of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan 646000, PR China
| | - Yue Chen
- Department of Nuclear Medicine, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, PR China; Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou, Sichuan 646000, PR China
| | - Shaozhi Fu
- Department of Oncology, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, PR China; Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou, Sichuan 646000, PR China.
| |
Collapse
|
22
|
Lv L, Shi Y, Deng Z, Xu J, Ye Z, He J, Chen G, Yu X, Wu J, Huang X, Li G. A polymeric nanocarrier that eradicates breast cancer stem cells and delivers chemotherapeutic drugs. Biomater Res 2023; 27:133. [PMID: 38102651 PMCID: PMC10722842 DOI: 10.1186/s40824-023-00465-9] [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: 09/19/2023] [Accepted: 11/20/2023] [Indexed: 12/17/2023] Open
Abstract
BACKGROUND Drug nanocarriers can markedly reduce the toxicities and side effects of encapsulated chemotherapeutic drugs in the clinic. However, these drug nanocarriers have little effect on eradicating breast cancer stem cells (BCSCs). Although compounds that can inhibit BCSCs have been reported, these compounds are difficult to use as carriers for the widespread delivery of conventional chemotherapeutic drugs. METHODS Herein, we synthesize a polymeric nanocarrier, hyaluronic acid-block-poly (curcumin-dithiodipropionic acid) (HA-b-PCDA), and explore the use of HA-b-PCDA to simultaneously deliver chemotherapeutic drugs and eradicate BCSCs. RESULTS Based on molecular docking and molecular dynamics studies, HA-b-PCDA delivers 35 clinical chemotherapeutic drugs. To further verify the drug deliver ability of HA-b-PCDA, doxorubicin, paclitaxel, docetaxel, gemcitabine and camptothecin are employed as model drugs to prepare nanoparticles. These drug-loaded HA-b-PCDA nanoparticles significantly inhibit the proliferation and stemness of BCSC-enriched 4T1 mammospheres. Moreover, doxorubicin-loaded HA-b-PCDA nanoparticles efficiently inhibit tumor growth and eradicate approximately 95% of BCSCs fraction in vivo. Finally, HA-b-PCDA eradicates BCSCs by activating Hippo and inhibiting the JAK2/STAT3 pathway. CONCLUSION HA-b-PCDA is a polymeric nanocarrier that eradicates BCSCs and potentially delivers numerous clinical chemotherapeutic drugs.
Collapse
Affiliation(s)
- Li Lv
- Department of Pharmacy, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510120, China
| | - Yonghui Shi
- Department of Pharmacy, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510120, China
| | - Zhicheng Deng
- Shenshan Medical Center, Memorial Hospital of Sun Yat-Sen University, Shanwei, Guangdong, 516600, China
| | - Jiajia Xu
- School of Pharmacy, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Zicong Ye
- Department of Pharmacy, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510120, China
| | - Jianxiong He
- Department of Pharmacy, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510120, China
| | - Guanghui Chen
- Department of Pharmacy, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510120, China
| | - Xiaoxia Yu
- Department of Pharmacy, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510120, China
| | - Junyan Wu
- Department of Pharmacy, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510120, China.
| | - Xingzhen Huang
- School of Pharmacy, Guangxi Medical University, Nanning, Guangxi, 530021, China.
| | - Guocheng Li
- Department of Pharmacy, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510120, China.
- Shenshan Medical Center, Memorial Hospital of Sun Yat-Sen University, Shanwei, Guangdong, 516600, China.
| |
Collapse
|
23
|
Youness RA, Mohamed AH, Efthimiadou EK, Mekky RY, Braoudaki M, Fahmy SA. A Snapshot of Photoresponsive Liposomes in Cancer Chemotherapy and Immunotherapy: Opportunities and Challenges. ACS OMEGA 2023; 8:44424-44436. [PMID: 38046305 PMCID: PMC10688172 DOI: 10.1021/acsomega.3c04134] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Accepted: 08/18/2023] [Indexed: 12/05/2023]
Abstract
To provide precise medical regimens, photonics technologies have been involved in the field of nanomedicine. Phototriggered liposomes have been cast as promising nanosystems that achieve controlled release of payloads in several pathological conditions such as cancer, autoimmune, and infectious diseases. In contrast to the conventional liposomes, this photoresponsive element greatly improves therapeutic efficacy and reduces the adverse effects of gene/drug therapy during treatment. Recently, cancer immunotherpay has been one of the hot topics in the field of oncology due to the great success and therapeutic benefits that were well-recognized by the patients. However, several side effects have been encountered due to the unmonitored augmentation of the immune system. This Review highlights the most recent advancements in the development of photoresponsive liposome nanosystems in the field of oncology, with a specific emphasis on challenges and opportunities in the field of cancer immunotherapy.
Collapse
Affiliation(s)
- Rana A. Youness
- Biology
and Biochemistry Department, Faculty of Biotechnology, German International University (GIU), New Administrative Capital, Cairo 4824201, Egrypt
- Biology
and Biochemistry Department, Molecular Genetics Research Team (MGRT),
School of Life and Medical Sciences, University
of Hertfordshire Hosted by Global Academic Foundation, Cairo 11835, Egypt
| | - Adham H. Mohamed
- Department
of Chemistry, Faculty of Science, Cairo
University, Giza 12613, Egypt
| | - Eleni K. Efthimiadou
- Inorganic
Chemistry Laboratory, Department of Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis, Zografou 157 71, Greece
| | - Radwa Y. Mekky
- Department
of Pharmacology and Toxicology, Faculty of Pharmacy, October University for Modern Sciences and Arts (MSA University), Cairo 12622, Egypt
| | - Maria Braoudaki
- Clinical,
Pharmaceutical, and Biological Science Department, School of Life
and Medical Sciences, University of Hertfordshire, Hatfield AL10 9AB, U.K.
| | - Sherif Ashraf Fahmy
- Chemistry
Department, School of Life and Medical Sciences, University of Hertfordshire Hosted by Global Academic Foundation, Cairo 11835, Egypt
| |
Collapse
|
24
|
Cheng L, Yu J, Hao T, Wang W, Wei M, Li G. Advances in Polymeric Micelles: Responsive and Targeting Approaches for Cancer Immunotherapy in the Tumor Microenvironment. Pharmaceutics 2023; 15:2622. [PMID: 38004600 PMCID: PMC10675796 DOI: 10.3390/pharmaceutics15112622] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 11/01/2023] [Accepted: 11/09/2023] [Indexed: 11/26/2023] Open
Abstract
In recent years, to treat a diverse array of cancer forms, considerable advancements have been achieved in the field of cancer immunotherapies. However, these therapies encounter multiple challenges in clinical practice, such as high immune-mediated toxicity, insufficient accumulation in cancer tissues, and undesired off-target reactions. To tackle these limitations and enhance bioavailability, polymer micelles present potential solutions by enabling precise drug delivery to the target site, thus amplifying the effectiveness of immunotherapy. This review article offers an extensive survey of recent progress in cancer immunotherapy strategies utilizing micelles. These strategies include responsive and remodeling approaches to the tumor microenvironment (TME), modulation of immunosuppressive cells within the TME, enhancement of immune checkpoint inhibitors, utilization of cancer vaccine platforms, modulation of antigen presentation, manipulation of engineered T cells, and targeting other components of the TME. Subsequently, we delve into the present state and constraints linked to the clinical utilization of polymeric micelles. Collectively, polymer micelles demonstrate excellent prospects in tumor immunotherapy by effectively addressing the challenges associated with conventional cancer immunotherapies.
Collapse
Affiliation(s)
- Lichun Cheng
- Department of Pharmacy, The Second Hospital of Dalian Medical University, Dalian 116027, China; (L.C.); (T.H.); (W.W.)
- School of Pharmacy, China Medical University, Shenyang 110122, China;
| | - Jiankun Yu
- School of Pharmacy, China Medical University, Shenyang 110122, China;
| | - Tangna Hao
- Department of Pharmacy, The Second Hospital of Dalian Medical University, Dalian 116027, China; (L.C.); (T.H.); (W.W.)
| | - Wenshuo Wang
- Department of Pharmacy, The Second Hospital of Dalian Medical University, Dalian 116027, China; (L.C.); (T.H.); (W.W.)
| | - Minjie Wei
- School of Pharmacy, China Medical University, Shenyang 110122, China;
| | - Guiru Li
- Department of Pharmacy, The Second Hospital of Dalian Medical University, Dalian 116027, China; (L.C.); (T.H.); (W.W.)
| |
Collapse
|
25
|
Doustmihan A, Fathi M, Mazloomi M, Salemi A, Hamblin MR, Jahanban-Esfahlan R. Molecular targets, therapeutic agents and multitasking nanoparticles to deal with cancer stem cells: A narrative review. J Control Release 2023; 363:57-83. [PMID: 37739017 DOI: 10.1016/j.jconrel.2023.09.029] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Revised: 09/08/2023] [Accepted: 09/17/2023] [Indexed: 09/24/2023]
Abstract
There is increasing evidence that malignant tumors are initiated and maintained by a sub-population of tumor cells that have similar biological properties to normal adult stem cells. This very small population of Cancer Stem Cells (CSC) comprises tumor initiating cells responsible for cancer recurrence, drug resistance and metastasis. Conventional treatments such as chemotherapy, radiotherapy and surgery, in addition to being potentially toxic and non-specific, may paradoxically increase the population, spread and survival of CSCs. Next-generation sequencing and omics technologies are increasing our understanding of the pathways and factors involved in the development of CSCs, and can help to discover new therapeutic targets against CSCs. In addition, recent advances in nanomedicine have provided hope for the development of optimal specific therapies to eradicate CSCs. Moreover, the use of artificial intelligence and nano-informatics can elucidate new drug targets, and help to design drugs and nanoparticles (NPs) to deal with CSCs. In this review, we first summarize the properties of CSCs and describe the signaling pathways and molecular characteristics responsible for the emergence and survival of CSCs. Also, the location of CSCs within the tumor and the effect of host factors on the creation and maintenance of CSCs are discussed. Newly discovered molecular targets involved in cancer stemness and some novel therapeutic compounds to combat CSCs are highlighted. The optimum properties of anti-CSC NPs, including blood circulation and stability, tumor accumulation and penetration, cellular internalization, drug release, endosomal escape, and aptamers designed for specific targeting of CSCs are covered. Finally, some recent smart NPs designed for therapeutic and theranostic purposes to overcome CSCs are discussed.
Collapse
Affiliation(s)
- Abolfazl Doustmihan
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Marziyeh Fathi
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - MirAhmad Mazloomi
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Aysan Salemi
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran; Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Michael R Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein, 2028, South Africa.
| | - Rana Jahanban-Esfahlan
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
26
|
Gao S, Liu M, Liu D, Kong X, Fang Y, Li Y, Wu H, Ji J, Yang X, Zhai G. Biomimetic biomineralization nanoplatform-mediated differentiation therapy and phototherapy for cancer stem cell inhibition and antitumor immunity activation. Asian J Pharm Sci 2023; 18:100851. [PMID: 37915760 PMCID: PMC10616143 DOI: 10.1016/j.ajps.2023.100851] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 08/24/2023] [Accepted: 09/11/2023] [Indexed: 11/03/2023] Open
Abstract
Growing evidence suggests that the presence of cancer stem cells (CSCs) is a major challenge in current tumor treatments, especially the transition from non-CSCs to differentiation of CSCs for evading conventional therapies and driving metastasis. Here we propose a therapeutic strategy of synergistic differentiation therapy and phototherapy to induce differentiation of CSCs into mature tumor cells by differentiation inducers and synergistic elimination of them and normal cancer cells through phototherapy. In this work, we synthesized a biomimetic nanoplatform loaded with IR-780 and all-trans retinoic acid (ATRA) via biomineralization. This method can integrate aluminum ions into small-sized protein carriers to form nanoclusters, which undergo responsive degradation under acidic conditions and facilitate deep tumor penetration. With the help of CSC differentiation induced by ATRA, IR-780 inhibited the self-renewal of CSCs and cancer progression by generating hyperthermia and reactive oxygen species in a synergistic manner. Furthermore, ATRA can boost immunogenic cell death induced by phototherapy, thereby strongly causing a systemic anti-tumor immune response and efficiently eliminating CSCs and tumor cells. Taken together, this dual strategy represents a new paradigm of targeted eradication of CSCs and tumors by inducing CSC differentiation, improving photothermal therapy/photodynamic therapy and enhancing antitumor immunity.
Collapse
Affiliation(s)
- Shan Gao
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Meng Liu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Dongzhu Liu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Xinru Kong
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Yuelin Fang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Yingying Li
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Hang Wu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Jianbo Ji
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Xiaoye Yang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Guangxi Zhai
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| |
Collapse
|
27
|
Wang Y, Cui Y, Dai T, Yue Y. Reduction-responsive supramolecular hybridized paclitaxel nanoparticles for tumor treatment. Front Bioeng Biotechnol 2023; 11:1257788. [PMID: 37724094 PMCID: PMC10505395 DOI: 10.3389/fbioe.2023.1257788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 08/21/2023] [Indexed: 09/20/2023] Open
Abstract
Powerful chemotherapeutics have been used to combat tumor cells, but serious adverse effects and poor therapeutic efficiency restrict their clinical performance. Herein, we developed reduction-responsive supramolecular hybridized paclitaxel nanoparticles (PTX@HOMNs) for improved tumor treatment. The nanocarrier is composed of F127 and strengthened by a disulfide bond linked organosilica network, which ensures the desirable stability during blood circulation and controlled drug release at tumor sites. The as-prepared PTX@HOMNs could effectively accumulate at tumor regions. After entering tumor cells, PTX@HOMNs can respond to intracellular glutathione, and trigger active drug release for chemotherapy. As a result, PTX@HOMNs exhibited potent antitumor activity against ovarian tumors in vitro and in vivo. Our work provides a deep insight into constructing simple and controlled drug delivery nanoplatforms for improved tumor treatment.
Collapse
Affiliation(s)
| | | | | | - Ying Yue
- Department of Gynecological Oncology, The First Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
28
|
Liu H, Liu M, Zhao Y, Mo R. Nanomedicine strategies to counteract cancer stemness and chemoresistance. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2023; 4:630-656. [PMID: 37720349 PMCID: PMC10501898 DOI: 10.37349/etat.2023.00157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 05/07/2023] [Indexed: 09/19/2023] Open
Abstract
Cancer stem-like cells (CSCs) identified by self-renewal ability and tumor-initiating potential are responsible for tumor recurrence and metastasis in many cancers. Conventional chemotherapy fails to eradicate CSCs that hold a state of dormancy and possess multi-drug resistance. Spurred by the progress of nanotechnology for drug delivery and biomedical applications, nanomedicine has been increasingly developed to tackle stemness-associated chemotherapeutic resistance for cancer therapy. This review focuses on advances in nanomedicine-mediated therapeutic strategies to overcome chemoresistance by specifically targeting CSCs, the combination of chemotherapeutics with chemopotentiators, and programmable controlled drug release. Perspectives from materials and formulations at the nano-scales are specifically surveyed. Future opportunities and challenges are also discussed.
Collapse
Affiliation(s)
- Huayu Liu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, Jiangsu, China
| | - Mingqi Liu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, Jiangsu, China
| | - Yanan Zhao
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, Jiangsu, China
| | - Ran Mo
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, Jiangsu, China
| |
Collapse
|
29
|
He JJ, Li QQ, Zhao C, Zhou J, Wu J, Zhang HB, Zhao YQ, Zhang HH, Lei TY, Zhao XY, You Z, Song QB, Xu B. Advancement and Applications of Nanotherapy for Cancer Immune Microenvironment. Curr Med Sci 2023; 43:631-646. [PMID: 37558863 DOI: 10.1007/s11596-023-2763-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 04/27/2023] [Indexed: 08/11/2023]
Abstract
Cancer treatment has evolved rapidly due to major advances in tumor immunity research. However, due to the complexity, heterogeneity, and immunosuppressive microenvironment of tumors, the overall efficacy of immunotherapy is only 20%. In recent years, nanoparticles have attracted more attention in the field of cancer immunotherapy because of their remarkable advantages in biocompatibility, precise targeting, and controlled drug delivery. However, the clinical application of nanomedicine also faces many problems concerning biological safety, and the synergistic mechanism of nano-drugs with immunity remains to be elucidated. Our study summarizes the functional characteristics and regulatory mechanisms of nanoparticles in the cancer immune microenvironment and how nanoparticles activate and long-term stimulate innate immunity and adaptive immunity. Finally, the current problems and future development trends regarding the application of nanoparticles are fully discussed and prospected to promote the transformation and application of nanomedicine used in cancer treatment.
Collapse
Affiliation(s)
- Jun-Ju He
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Qing-Qing Li
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Chen Zhao
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Jin Zhou
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Jie Wu
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Hui-Bo Zhang
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Ya-Qi Zhao
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Hao-Han Zhang
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Tian-Yu Lei
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Xin-Yi Zhao
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Zuo You
- Department of Traditional Chinese Medicine, Xianfeng County People's Hospital, Enshi, 445000, China
| | - Qi-Bin Song
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Bin Xu
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|
30
|
Xiong Y, Yong Z, Xu C, Deng Q, Wang Q, Li S, Wang C, Zhang Z, Yang X, Li Z. Hyperbaric Oxygen Activates Enzyme-Driven Cascade Reactions for Cooperative Cancer Therapy and Cancer Stem Cells Elimination. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2301278. [PMID: 37114827 PMCID: PMC10375084 DOI: 10.1002/advs.202301278] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 04/10/2023] [Indexed: 06/19/2023]
Abstract
Tumor starvation induced by intratumor glucose depletion emerges as a promising strategy for anticancer therapy. However, its antitumor potencies are severely compromised by intrinsic tumor hypoxia, low delivery efficiencies, and undesired off-target toxicity. Herein, a multifunctional cascade bioreactor (HCG), based on the self-assembly of pH-responsive hydroxyethyl starch prodrugs, copper ions, and glucose oxidase (GOD), is engineered, empowered by hyperbaric oxygen (HBO) for efficient cooperative therapy against aggressive breast cancers. Once internalized by tumor cells, HCG undergoes disassembly and releases cargoes in response to acidic tumor microenvironment. Subsequently, HBO activates GOD-catalyzed oxidation of glucose to H2 O2 and gluconic acid by ameliorating tumor hypoxia, fueling copper-catalyzed •OH generation and pH-responsive drug release. Meanwhile, HBO degrades dense tumor extracellular matrix, promoting tumor accumulation and penetration of HCG. Moreover, along with the consumption of glucose and the redox reaction of copper ions, the antioxidant capacity of tumor cells is markedly reduced, collectively boosting oxidative stress. As a result, the combination of HCG and HBO can not only remarkably suppress the growth of orthotopic breast tumors but also restrain pulmonary metastases by inhibiting cancer stem cells. Considering the clinical accessibility of HBO, this combined strategy holds significant translational potentials for GOD-based therapies.
Collapse
Affiliation(s)
- Yuxuan Xiong
- National Engineering Research Center for NanomedicineCollege of Life Science and TechnologyHuazhong University of Science and TechnologyWuhan430074P. R. China
| | - Zhengtao Yong
- National Engineering Research Center for NanomedicineCollege of Life Science and TechnologyHuazhong University of Science and TechnologyWuhan430074P. R. China
| | - Chen Xu
- National Engineering Research Center for NanomedicineCollege of Life Science and TechnologyHuazhong University of Science and TechnologyWuhan430074P. R. China
| | - Qingyuan Deng
- National Engineering Research Center for NanomedicineCollege of Life Science and TechnologyHuazhong University of Science and TechnologyWuhan430074P. R. China
| | - Qiang Wang
- National Engineering Research Center for NanomedicineCollege of Life Science and TechnologyHuazhong University of Science and TechnologyWuhan430074P. R. China
| | - Shiyou Li
- National Engineering Research Center for NanomedicineCollege of Life Science and TechnologyHuazhong University of Science and TechnologyWuhan430074P. R. China
| | - Chong Wang
- National Engineering Research Center for NanomedicineCollege of Life Science and TechnologyHuazhong University of Science and TechnologyWuhan430074P. R. China
| | - Zhijie Zhang
- National Engineering Research Center for NanomedicineCollege of Life Science and TechnologyHuazhong University of Science and TechnologyWuhan430074P. R. China
| | - Xiangliang Yang
- National Engineering Research Center for NanomedicineCollege of Life Science and TechnologyHuazhong University of Science and TechnologyWuhan430074P. R. China
- Key Laboratory of Molecular Biophysics of Ministry of EducationCollege of Life Science and TechnologyHuazhong University of Science and TechnologyWuhan430074P. R. China
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia MedicalHuazhong University of Science and TechnologyWuhan430074P. R. China
- Hubei Engineering Research Center for Biomaterials and Medical Protective MaterialsHuazhong University of Science and TechnologyWuhan430074P. R. China
- Hubei Bioinformatics and Molecular Imaging Key LaboratoryCollege of Life Science and TechnologyHuazhong University of Science and TechnologyWuhan430074P. R. China
- GBA Research Innovation Institute for NanotechnologyGuangdong510530P. R. China
| | - Zifu Li
- National Engineering Research Center for NanomedicineCollege of Life Science and TechnologyHuazhong University of Science and TechnologyWuhan430074P. R. China
- Key Laboratory of Molecular Biophysics of Ministry of EducationCollege of Life Science and TechnologyHuazhong University of Science and TechnologyWuhan430074P. R. China
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia MedicalHuazhong University of Science and TechnologyWuhan430074P. R. China
- Hubei Engineering Research Center for Biomaterials and Medical Protective MaterialsHuazhong University of Science and TechnologyWuhan430074P. R. China
- Hubei Bioinformatics and Molecular Imaging Key LaboratoryCollege of Life Science and TechnologyHuazhong University of Science and TechnologyWuhan430074P. R. China
| |
Collapse
|
31
|
Yue M, Guo T, Nie DY, Zhu YX, Lin M. Advances of nanotechnology applied to cancer stem cells. World J Stem Cells 2023; 15:514-529. [PMID: 37424953 PMCID: PMC10324502 DOI: 10.4252/wjsc.v15.i6.514] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 03/01/2023] [Accepted: 04/18/2023] [Indexed: 06/26/2023] Open
Abstract
Cancer stem cells (CSCs) are a small proportion of the cells that exist in cancer tissues. They are considered to be the culprit of tumor genesis, development, drug resistance, metastasis and recurrence because of their self-renewal, proliferation, and differentiation potential. The elimination of CSCs is thus the key to cure cancer, and targeting CSCs provides a new method for tumor treatment. Due to the advantages of controlled sustained release, targeting and high biocompatibility, a variety of nanomaterials are used in the diagnosis and treatments targeting CSCs and promote the recognition and removal of tumor cells and CSCs. This article mainly reviews the research progress of nanotechnology in sorting CSCs and nanodrug delivery systems targeting CSCs. Furthermore, we identify the problems and future research directions of nanotechnology in CSC therapy. We hope that this review will provide guidance for the design of nanotechnology as a drug carrier so that it can be used in clinic for cancer therapy as soon as possible.
Collapse
Affiliation(s)
- Miao Yue
- Clinical Laboratory, Nanjing University of Chinese Medicine, Taizhou 225300, Jiangsu Province, China
| | - Ting Guo
- Taizhou School of Clinical Medicine, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou 225300, Jiangsu Province, China
| | - Deng-Yun Nie
- Clinical Laboratory, Nanjing University of Chinese Medicine, Taizhou 225300, Jiangsu Province, China
| | - Yin-Xing Zhu
- Taizhou School of Clinical Medicine, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou 225300, Jiangsu Province, China
| | - Mei Lin
- Taizhou School of Clinical Medicine, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou 225300, Jiangsu Province, China.
| |
Collapse
|
32
|
Singh AK, Malviya R, Prajapati B, Singh S, Yadav D, Kumar A. Nanotechnology-Aided Advancement in Combating the Cancer Metastasis. Pharmaceuticals (Basel) 2023; 16:899. [PMID: 37375846 PMCID: PMC10304141 DOI: 10.3390/ph16060899] [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: 04/27/2023] [Revised: 05/28/2023] [Accepted: 06/14/2023] [Indexed: 06/29/2023] Open
Abstract
Modern medicine has been working to find a cure for cancer for almost a century, but thus far, they have not been very successful. Although cancer treatment has come a long way, more work has to be carried out to boost specificity and reduce systemic toxicity. The diagnostic industry is on the cusp of a technological revolution, and early diagnosis is essential for improving prognostic outlook and patient quality of life. In recent years, nanotechnology's use has expanded, demonstrating its efficacy in enhancing fields such as cancer treatment, radiation therapy, diagnostics, and imaging. Applications for nanomaterials are diverse, ranging from enhanced radiation adjuvants to more sensitive early detection instruments. Cancer, particularly when it has spread beyond the original site of cancer, is notoriously tough to combat. Many people die from metastatic cancer, which is why it remains a huge issue. Cancer cells go through a sequence of events known as the "metastatic cascade" throughout metastasis, which may be used to build anti-metastatic therapeutic techniques. Conventional treatments and diagnostics for metastasis have their drawbacks and hurdles that must be overcome. In this contribution, we explore in-depth the potential benefits that nanotechnology-aided methods might offer to the detection and treatment of metastatic illness, either alone or in conjunction with currently available conventional procedures. Anti-metastatic drugs, which can prevent or slow the spread of cancer throughout the body, can be more precisely targeted and developed with the help of nanotechnology. Furthermore, we talk about how nanotechnology is being applied to the treatment of patients with cancer metastases.
Collapse
Affiliation(s)
- Arun Kumar Singh
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida 203201, India; (A.K.S.); (D.Y.)
| | - Rishabha Malviya
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida 203201, India; (A.K.S.); (D.Y.)
| | - Bhupendra Prajapati
- Shree S. K. Patel College of Pharmaceutical Education and Research, Ganpat University, Kherva 384012, India
| | - Sudarshan Singh
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Deepika Yadav
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida 203201, India; (A.K.S.); (D.Y.)
| | - Arvind Kumar
- Chandigarh Engineering College, Jhanjeri, Mohali 140307, India;
| |
Collapse
|
33
|
Li L, Ni R, Zheng D, Chen L. Eradicating the tumor "seeds": nanomedicines-based therapies against cancer stem cells. Daru 2023; 31:83-94. [PMID: 36971921 PMCID: PMC10238364 DOI: 10.1007/s40199-023-00456-0] [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: 08/17/2022] [Accepted: 03/05/2023] [Indexed: 03/29/2023] Open
Abstract
OBJECTIVES Cancer stem cells (CSCs), a small subpopulation of cells with high tumorigenesis and strong intrinsic drug resistance, exhibit self-renewal and differentiation abilities. CSCs play a crucial role in tumor progression, drug resistance, recurrence and metastasis,and conventional therapy is not enough to eradicate them. Therefore, developing novel therapies targeting CSCs to increase drug sensitivity and preventing relapse is essential. The objective of this review is to present nanotherapies that target and eradicate the tumor "seeds". EVIDENCE ACQUISITION Evidence was collected and sorted from the literature ranging from 2000 to 2022, using appropriate keywords and key phrases as search terms within scientific databases such as Web of Science, PubMed and Google Scholar. RESULTS Nanoparticle drug delivery systems have been successfully applied to gain longer circulation time, more precise targeting capability and better stability during cancer treatment. Nanotechnology-based strategies that have been used to target CSCs, include (1) encapsulating small molecular drugs and genes by nanotechnology, (2) targeting CSC signaling pathways, (3) utilizing nanocarriers targeting for specific markers of CSCs, (4) improving photothermal/ photodynamic therapy (PTT/PDT), 5)targeting the metabolism of CSCs and 6) enhancing nanomedicine-aided immunotherapy. CONCLUSION This review summarizes the biological hallmarks and markers of CSCs, and the nanotechnology-based therapies to kill them. Nanoparticle drug delivery systems are appropriate means for delivering drugs to tumors through enhanced permeability and retention (EPR) effect. Furthermore, surface modification with special ligands or antibodies improves the recognition and uptake of tumor cells or CSCs. It is expected that this review can offer insights into features of CSCs and the exploration of targeting nanodrug delivery systems.
Collapse
Affiliation(s)
- Lin Li
- Department of Pharmacy, Women and Children's Hospital of Chongqing Medical University, Chongqing Health Center for Women and Children, 401147, Chongqing, China
| | - Rui Ni
- Department of Pharmacy, Daping Hospital, Army Medical University, 400042, Chongqing, China
| | - Dan Zheng
- Department of Pharmacy, Women and Children's Hospital of Chongqing Medical University, Chongqing Health Center for Women and Children, 401147, Chongqing, China
| | - Lin Chen
- Department of Pharmacy, Women and Children's Hospital of Chongqing Medical University, Chongqing Health Center for Women and Children, 401147, Chongqing, China.
| |
Collapse
|
34
|
Ma GL, Lin WF. Immune checkpoint inhibition mediated with liposomal nanomedicine for cancer therapy. Mil Med Res 2023; 10:20. [PMID: 37106400 PMCID: PMC10142459 DOI: 10.1186/s40779-023-00455-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 04/08/2023] [Indexed: 04/29/2023] Open
Abstract
Immune checkpoint blockade (ICB) therapy for cancer has achieved great success both in clinical results and on the market. At the same time, success drives more attention from scientists to improve it. However, only a small portion of patients are responsive to this therapy, and it comes with a unique spectrum of side effects termed immune-related adverse events (irAEs). The use of nanotechnology could improve ICBs' delivery to the tumor, assist them in penetrating deeper into tumor tissues and alleviate their irAEs. Liposomal nanomedicine has been investigated and used for decades, and is well-recognized as the most successful nano-drug delivery system. The successful combination of ICB with liposomal nanomedicine could help improve the efficacy of ICB therapy. In this review, we highlighted recent studies using liposomal nanomedicine (including new emerging exosomes and their inspired nano-vesicles) in associating ICB therapy.
Collapse
Affiliation(s)
- Guang-Long Ma
- Faculty of Medicine, Centre for Cancer Immunology, University of Southampton, Southampton, SO16 6YD UK
| | - Wei-Feng Lin
- Department of Molecular Chemistry and Materials Science, Weizmann Institute of Science, 76100 Rehovot, Israel
- Key Laboratory of Bio-Inspired Smart Interfacial Science and Technology of Ministry of Education, School of Chemistry, Beihang University, Beijing, 100191 China
| |
Collapse
|
35
|
Zhang H, Wang R, Wu C, Feng W, Zhong Q, Chen X, Wang T, Mao C. Diffusion-mediated carving of interior topologies of all-natural protein nanoparticles to tailor sustained drug release for effective breast cancer therapy. Biomaterials 2023; 295:122027. [PMID: 36805237 DOI: 10.1016/j.biomaterials.2023.122027] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 01/01/2023] [Accepted: 01/25/2023] [Indexed: 01/30/2023]
Abstract
Proteins are promising base materials for developing drug carriers with efficient blood circulation due to low possibilities of clearance by macrophages. However, such natural biopolymers have highly sophisticated molecular structures, preventing them from being assembled into nano-platforms with manipulable payload release profiles. Here, we report the self-assembly of two natural proteins (milk casein and rice protein) into protein nanoparticles (NPs, ∼150 nm) with tailorable release profiles. Diffusion of plant-derived paclitaxel (PTX)-containing eugenol into the hydrophobic cores of the NPs and subsequent dialysis to remove eugenol from the cores lead to the carving of the NP interiors. With the increase in the mass ratios of casein and rice protein, this process generates all-natural NPs with PTX loaded in their full cavities, semi-full cavities, or solid cores. These NPs can be efficiently uptaken by breast cancer cells and could kill the cancer cells efficiently. PTX in these NPs demonstrates increasingly sustained in vivo release profiles from full cavities, semi-full cavities, to solid cores, gradually extending its pharmacokinetic profiles in blood plasma to favor drug accumulation in breast tumor models. Consequently, the NPs with solid cores completely inhibit tumor growth in vivo, more effectively than those with full and semi-full cavities. Our work opens up a new avenue to the use of diffusion-mediated nanoscale carving in producing biomaterials with controllable interior topologies relevant to drug release profiles.
Collapse
Affiliation(s)
- Hao Zhang
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Jiangnan University, Wuxi 21422, China; National Engineering Research Center of Cereal Fermentation and Food Biomanufacturing, Jiangnan University, Wuxi 21422, China; Jiangsu Provincial Research Center for Bioactive Product Processing Technology, Jiangnan University, Wuxi 21422, China; School of Food Science and Technology, Jiangnan University, Wuxi 21422, China
| | - Ren Wang
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Jiangnan University, Wuxi 21422, China; National Engineering Research Center of Cereal Fermentation and Food Biomanufacturing, Jiangnan University, Wuxi 21422, China; Jiangsu Provincial Research Center for Bioactive Product Processing Technology, Jiangnan University, Wuxi 21422, China; School of Food Science and Technology, Jiangnan University, Wuxi 21422, China
| | - Chao Wu
- National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, China
| | - Wei Feng
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Jiangnan University, Wuxi 21422, China; National Engineering Research Center of Cereal Fermentation and Food Biomanufacturing, Jiangnan University, Wuxi 21422, China; Jiangsu Provincial Research Center for Bioactive Product Processing Technology, Jiangnan University, Wuxi 21422, China; School of Food Science and Technology, Jiangnan University, Wuxi 21422, China
| | - Qixin Zhong
- Department of Food Science, The University of Tennessee, Knoxville, TN 37996, USA
| | - Xianfu Chen
- State Key Laboratory of Materials-Oriented Chemical Engineering and College of Chemical Engineering, Nanjing Tech University, Nanjing 210009, China
| | - Tao Wang
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Jiangnan University, Wuxi 21422, China; National Engineering Research Center of Cereal Fermentation and Food Biomanufacturing, Jiangnan University, Wuxi 21422, China; Jiangsu Provincial Research Center for Bioactive Product Processing Technology, Jiangnan University, Wuxi 21422, China; School of Food Science and Technology, Jiangnan University, Wuxi 21422, China.
| | - Chuanbin Mao
- School of Materials Science and Engineering, Zhejiang University, Hangzhou, China; Department of Chemistry and Biochemistry and Stephenson Life Science Research Center, University of Oklahoma, Norman, OK 73019, USA.
| |
Collapse
|
36
|
Zhao Z, Wang D, Li Y. Versatile biomimetic nanomedicine for treating cancer and inflammation disease. MEDICAL REVIEW (2021) 2023; 3:123-151. [PMID: 37724085 PMCID: PMC10471090 DOI: 10.1515/mr-2022-0046] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 02/11/2023] [Indexed: 09/20/2023]
Abstract
Nanosized drug delivery systems (NDDSs) have emerged as a powerful tool to optimize drug delivery in complex diseases, including cancer and inflammation. However, the therapeutic effect of NDDSs is still far from satisfactory due to their poor circulation time, low delivery efficiency, and innate toxicity. Fortunately, biomimetic approaches offer new opportunities to develop nanomedicine, which is derived from a variety of native biomolecules including cells, exosomes, bacteria, and so on. Since inheriting the superior biocompatibility and versatile functions of natural materials, biomimetic nanomedicine can mimic biological processes, prolong blood circulation, and lower immunogenicity, serving as a desired platform for precise drug delivery for treating cancer and inflammatory disease. In this review, we outline recent advances in biomimetic NDDSs, which consist of two concepts: biomimetic exterior camouflage and bioidentical molecule construction. We summarize engineering strategies that further functionalized current biomimetic NDDSs. A series of functional biomimetic NDDSs created by our group are introduced. We conclude with an outlook on remaining challenges and possible directions for biomimetic NDDSs. We hope that better technologies can be inspired and invented to advance drug delivery systems for cancer and inflammation therapy.
Collapse
Affiliation(s)
- Zhiwen Zhao
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Dangge Wang
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
- Yantai Key Laboratory of Nanomedicine & Advanced Preparations, Yantai Institute of Materia Medica, Yantai, China
| | - Yaping Li
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, China
| |
Collapse
|
37
|
Awad RM, Breckpot K. Novel technologies for applying immune checkpoint blockers. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2023; 382:1-101. [PMID: 38225100 DOI: 10.1016/bs.ircmb.2023.03.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/03/2023]
Abstract
Cancer cells develop several ways to subdue the immune system among others via upregulation of inhibitory immune checkpoint (ICP) proteins. These ICPs paralyze immune effector cells and thereby enable unfettered tumor growth. Monoclonal antibodies (mAbs) that block ICPs can prevent immune exhaustion. Due to their outstanding effects, mAbs revolutionized the field of cancer immunotherapy. However, current ICP therapy regimens suffer from issues related to systemic administration of mAbs, including the onset of immune related adverse events, poor pharmacokinetics, limited tumor accessibility and immunogenicity. These drawbacks and new insights on spatiality prompted the exploration of novel administration routes for mAbs for instance peritumoral delivery. Moreover, novel ICP drug classes that are adept to novel delivery technologies were developed to circumvent the drawbacks of mAbs. We therefore review the state-of-the-art and novel delivery strategies of ICP drugs.
Collapse
Affiliation(s)
- Robin Maximilian Awad
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Karine Breckpot
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium.
| |
Collapse
|
38
|
Sun F, Hou H, Li Y, Tang W, Wang J, Lu L, Fu J, Liu Z, Gao D, Zhao F, Gao X, Ling P, Wang F, Tan H. Glycol-Split Heparin-Linked Prodrug Nanoparticles Target the Mitochondrion Apparatus for Cancer Metastasis Treatment. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023:e2206491. [PMID: 36965026 DOI: 10.1002/smll.202206491] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 03/02/2023] [Indexed: 06/18/2023]
Abstract
The progression and metastasis of solid tumors rely strongly on neovascularization. However, angiogenesis inhibitors alone cannot meet the needs of tumor therapy. This study prepared a new drug conjugate (PTX-GSHP-CYS-ES2, PGCE) by combining polysaccharides (heparin without anticoagulant activity, GSHP), chemotherapeutic drugs (paclitaxel, PTX), and antiangiogenic drugs (ES2). Furthermore, a tumor-targeted prodrug nanoparticle delivery system is established. The nanoparticles appear to accumulate in the mitochondrial of tumor cells and achieve ES2 and PTX release under high glutathione and acidic environment. It has been confirmed that PGCE inhibited the expression of multiple metastasis-related proteins by targeting the tumor cell mitochondrial apparatus and disrupting their structure. Furthermore, PGCE nanoparticles inhibit migration, invasion, and angiogenesis in B16F10 tumor-bearing mice and suppress tumor growth and metastasis in vitro. Further in vitro and in vivo experiments show that PGCE has strong antitumor growth and metastatic effects and exhibits efficient anti-angiogenesis properties. This multi-targeted nanoparticle system potentially enhances the antitumor and anti-metastatic effects of combination chemotherapy and antiangiogenic drugs.
Collapse
Affiliation(s)
- Feng Sun
- National Glycoengineering Research Center, Shandong University, Qingdao, Shandong, 266237, China
- NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate-Based Medicine, Shandong University, Qingdao, 266237, China
- Shandong Provincial Technology Innovation Center of Carbohydrate, Shandong University, Qingdao, 266237, China
| | - Huiwen Hou
- National Glycoengineering Research Center, Shandong University, Qingdao, Shandong, 266237, China
- NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate-Based Medicine, Shandong University, Qingdao, 266237, China
- Shandong Provincial Technology Innovation Center of Carbohydrate, Shandong University, Qingdao, 266237, China
| | - Yan Li
- National Glycoengineering Research Center, Shandong University, Qingdao, Shandong, 266237, China
- NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate-Based Medicine, Shandong University, Qingdao, 266237, China
- Shandong Provincial Technology Innovation Center of Carbohydrate, Shandong University, Qingdao, 266237, China
| | - Wen Tang
- National Glycoengineering Research Center, Shandong University, Qingdao, Shandong, 266237, China
- NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate-Based Medicine, Shandong University, Qingdao, 266237, China
- Shandong Provincial Technology Innovation Center of Carbohydrate, Shandong University, Qingdao, 266237, China
| | - Jie Wang
- National Glycoengineering Research Center, Shandong University, Qingdao, Shandong, 266237, China
- NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate-Based Medicine, Shandong University, Qingdao, 266237, China
- Shandong Provincial Technology Innovation Center of Carbohydrate, Shandong University, Qingdao, 266237, China
| | - Lu Lu
- National Glycoengineering Research Center, Shandong University, Qingdao, Shandong, 266237, China
- NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate-Based Medicine, Shandong University, Qingdao, 266237, China
- Shandong Provincial Technology Innovation Center of Carbohydrate, Shandong University, Qingdao, 266237, China
| | - Jiaai Fu
- National Glycoengineering Research Center, Shandong University, Qingdao, Shandong, 266237, China
- NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate-Based Medicine, Shandong University, Qingdao, 266237, China
- Shandong Provincial Technology Innovation Center of Carbohydrate, Shandong University, Qingdao, 266237, China
| | - Zengmei Liu
- National Glycoengineering Research Center, Shandong University, Qingdao, Shandong, 266237, China
- NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate-Based Medicine, Shandong University, Qingdao, 266237, China
- Shandong Provincial Technology Innovation Center of Carbohydrate, Shandong University, Qingdao, 266237, China
| | - Didi Gao
- National Glycoengineering Research Center, Shandong University, Qingdao, Shandong, 266237, China
- NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate-Based Medicine, Shandong University, Qingdao, 266237, China
- Shandong Provincial Technology Innovation Center of Carbohydrate, Shandong University, Qingdao, 266237, China
| | - Feiyan Zhao
- National Glycoengineering Research Center, Shandong University, Qingdao, Shandong, 266237, China
- NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate-Based Medicine, Shandong University, Qingdao, 266237, China
- Shandong Provincial Technology Innovation Center of Carbohydrate, Shandong University, Qingdao, 266237, China
| | - Xinqing Gao
- National Glycoengineering Research Center, Shandong University, Qingdao, Shandong, 266237, China
- NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate-Based Medicine, Shandong University, Qingdao, 266237, China
- Shandong Provincial Technology Innovation Center of Carbohydrate, Shandong University, Qingdao, 266237, China
| | - Peixue Ling
- National Glycoengineering Research Center, Shandong University, Qingdao, Shandong, 266237, China
- NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate-Based Medicine, Shandong University, Qingdao, 266237, China
- Shandong Provincial Technology Innovation Center of Carbohydrate, Shandong University, Qingdao, 266237, China
- School of Pharmaceutical sciences, Shandong University, Jinan, 250012, China
| | - Fengshan Wang
- National Glycoengineering Research Center, Shandong University, Qingdao, Shandong, 266237, China
- NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate-Based Medicine, Shandong University, Qingdao, 266237, China
- School of Pharmaceutical sciences, Shandong University, Jinan, 250012, China
| | - Haining Tan
- National Glycoengineering Research Center, Shandong University, Qingdao, Shandong, 266237, China
- NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate-Based Medicine, Shandong University, Qingdao, 266237, China
- Shandong Provincial Technology Innovation Center of Carbohydrate, Shandong University, Qingdao, 266237, China
- School of Pharmaceutical sciences, Shandong University, Jinan, 250012, China
| |
Collapse
|
39
|
Liu L, Pan Y, Zhao C, Huang P, Chen X, Rao L. Boosting Checkpoint Immunotherapy with Biomaterials. ACS NANO 2023; 17:3225-3258. [PMID: 36746639 DOI: 10.1021/acsnano.2c11691] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
The immune checkpoint blockade (ICB) therapy has revolutionized the field of cancer treatment, while low response rates and systemic toxicity limit its clinical outcomes. With the rapid advances in nanotechnology and materials science, various types of biomaterials have been developed to maximize therapeutic efficacy while minimizing side effects by increasing tumor antigenicity, reversing immunosuppressive microenvironment, amplifying antitumor immune response, and reducing extratumoral distribution of checkpoint inhibitors as well as enhancing their retention within target sites. In this review, we reviewed current design strategies for different types of biomaterials to augment ICB therapy effectively and then discussed present representative biomaterial-assisted immune modulation and targeted delivery of checkpoint inhibitors to boost ICB therapy. Current challenges and future development prospects for expanding the ICB with biomaterials were also summarized. We anticipate this review will be helpful for developing emerging biomaterials for ICB therapy and promoting the clinical application of ICB therapy.
Collapse
Affiliation(s)
- Lujie Liu
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen 518060, China
- Institute of Biomedical Health Technology and Engineering, Shenzhen Bay Laboratory, Shenzhen 518132, China
| | - Yuanwei Pan
- Institute of Biomedical Health Technology and Engineering, Shenzhen Bay Laboratory, Shenzhen 518132, China
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 119074
| | - Chenchen Zhao
- Institute of Biomedical Health Technology and Engineering, Shenzhen Bay Laboratory, Shenzhen 518132, China
| | - Peng Huang
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen 518060, China
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 119074
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), Singapore 138673
| | - Lang Rao
- Institute of Biomedical Health Technology and Engineering, Shenzhen Bay Laboratory, Shenzhen 518132, China
| |
Collapse
|
40
|
Zhao P, Li H, Bu W. A Forward Vision for Chemodynamic Therapy: Issues and Opportunities. Angew Chem Int Ed Engl 2023; 62:e202210415. [PMID: 36650984 DOI: 10.1002/anie.202210415] [Citation(s) in RCA: 135] [Impact Index Per Article: 67.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Indexed: 01/19/2023]
Abstract
Since the insight to fuse Fenton chemistry and nanomedicine into cancer therapy, great signs of progress have been made in the field of chemodynamic therapy (CDT). However, the exact mechanism of CDT is obscured by the unique tumor chemical environment and inevitable nanoparticle-cell interactions, thus impeding further development. In this Scientific Perspective, the significance of CDT is clarified, the complex mechanism is deconstructed into primitive chemical and biological interactions, and the mechanism research directions based on the chemical kinetics and biological signaling pathways are discussed in detail. Moreover, beneficial outlooks are presented to enlighten the evolution of next-generation CDT. Hopefully, this Scientific Perspective can inspire new ideas and advances for CDT and provide a reference for breaking down the interdisciplinary barriers in the field of nanomedicine.
Collapse
Affiliation(s)
- Peiran Zhao
- Department of Materials Science and State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200433, P.R. China
| | - Huiyan Li
- Department of Materials Science and State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200433, P.R. China
| | - Wenbo Bu
- Department of Materials Science and State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200433, P.R. China
| |
Collapse
|
41
|
Zhao P, Li H, Bu W. A Forward Vision for Chemodynamic Therapy: Issues and Opportunities. Angew Chem Int Ed Engl 2023. [DOI: 10.1002/ange.202210415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Affiliation(s)
- Peiran Zhao
- Department of Materials Science and State Key Laboratory of Molecular Engineering of Polymers Fudan University Shanghai 200433 P.R. China
| | - Huiyan Li
- Department of Materials Science and State Key Laboratory of Molecular Engineering of Polymers Fudan University Shanghai 200433 P.R. China
| | - Wenbo Bu
- Department of Materials Science and State Key Laboratory of Molecular Engineering of Polymers Fudan University Shanghai 200433 P.R. China
| |
Collapse
|
42
|
Jia W, Zhou L, Li L, Zhou P, Shen Z. Nano-Based Drug Delivery of Polyphenolic Compounds for Cancer Treatment: Progress, Opportunities, and Challenges. Pharmaceuticals (Basel) 2023; 16:ph16010101. [PMID: 36678599 PMCID: PMC9865384 DOI: 10.3390/ph16010101] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 01/04/2023] [Accepted: 01/05/2023] [Indexed: 01/12/2023] Open
Abstract
Polyphenols and their derivates, a kind of natural product distributed in herb plants, vegetables, and fruits, are the most abundant antioxidants in the human diet and have been found to display cancer-preventative effects in several epidemiological studies. The scientific community has also validated the anti-cancer bioactivities and low toxicities of polyphenolic compounds, including flavones, tannins, phenolic acids, and anthocyanins, through in vitro and in vivo studies. However, the low stability, weak targeting ability, poor solubility, and low bioavailability of pure polyphenolic agents have significantly impaired their treatment efficacy. Nowadays, nano-based technology has been applied to surmount these restrictions and maximize the treatment efficacy of polyphenols. In this review, we summarize the advantages and related mechanisms of polyphenols in cancer treatment. Moreover, aiming at the poor solubility and low bioavailability of pure polyphenols in vivo, the advantages of nano-based delivery systems and recent research developments are highlighted. Herein, particular emphasis is mainly placed on the most widely used nanomaterials in the delivery of natural products, including liposomes, micelles, and nanogels. Finally, we present an overview and the challenges of future implementations of nano-based delivery systems of polyphenolic compounds in the cancer therapeutic field.
Collapse
Affiliation(s)
- Wenhui Jia
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China School of Basic Medical Sciences & Forensic Medicine, Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu 610041, China
| | - Li Zhou
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400016, China
| | - Lei Li
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Ping Zhou
- Department of Radiotherapy, The First Affiliated Hospital of Hainan Medical University, Haikou 571199, China
- Correspondence: (P.Z.); (Z.S.)
| | - Zhisen Shen
- Department of Otorhinolaryngology and Head and Neck Surgery, The Affiliated Lihuili Hospital, Ningbo University, Ningbo 315211, China
- Correspondence: (P.Z.); (Z.S.)
| |
Collapse
|
43
|
Wang Y, Xia L, Lin J, Gong L, Xia Y, Xu Y, Liu L, Bao J, Zhang C, Chai Y, Li H. Thioridazine combined with carboplatin results in synergistic inhibition of triple negative breast cancer by targeting cancer stem cells. Transl Oncol 2022; 26:101549. [PMID: 36191461 PMCID: PMC9530598 DOI: 10.1016/j.tranon.2022.101549] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Revised: 07/14/2022] [Accepted: 09/16/2022] [Indexed: 11/18/2022] Open
Abstract
Cancer stem cells (CSCs) in triple-negative breast cancer (TNBC) are closely related to tumorigenesis and metastasis. Thioridazine (THZ) is a usual phenothiazine antipsychotic drug that can destroy CSCs. We aimed to explore whether THZ could sensitize metastatic TNBC cells, especially the CSCs, to carboplatin (CBP) treatment. Metastatic TNBC cells, 4T1 cells, and tumor-bearing mice were treated with THZ and CBP as monotherapy or combination therapy. MTT, flow cytometry, electron microscopy, immunohistochemistry and western blotting were applied to assess the cell viability, apoptosis, mitochondrial morphology and the relevant protein levels, respectively. Tumor size and lung metastasis under different treatments as well as tumorigenesis of residual tumor cells from each group were monitored. THZ combined with CBP inhibited 4T1 tumor cell proliferation and induced apoptosis by inhibiting the PI3K-AKT-mTOR pathway and activating estrogen receptor stress. THZ also showed strong activity against breast CSCs, THZ combined with CBP significantly destroyed cancer cells, inhibited lung metastasis and relieved the tumor burden; Our data demonstrated that THZ can sensitize TNBC cells to CBP treatment and this combination therapy may provide a bright strategy for TNBC treatment by targeting both cancer cells and CSCs.
Collapse
Affiliation(s)
- Yi Wang
- Department of Oncology, The First People's Hospital of Hefei/The Third Affiliated Hospital of Anhui Medical University, Hefei 230061, Anhui, PR China
| | - Leiming Xia
- Department of Hematopathology, The Fourth Affiliated Hospital of Anhui Medical University, Hefei 230002, Anhui, PR China
| | - Jing Lin
- Department of Oncology, The First People's Hospital of Hefei/The Third Affiliated Hospital of Anhui Medical University, Hefei 230061, Anhui, PR China
| | - Li Gong
- Department of Oncology, East District of First Affiliated Hospital of Anhui Medical University, Hefei 231600, Anhui, PR China
| | - Yang Xia
- Department of Oncology, Taizhou People's Hospital/The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou 225300, Jiangsu, PR China
| | - Yang Xu
- Department of Oncology, The First People's Hospital of Hefei/The Third Affiliated Hospital of Anhui Medical University, Hefei 230061, Anhui, PR China
| | - Liu Liu
- Department of Oncology, The First People's Hospital of Hefei/The Third Affiliated Hospital of Anhui Medical University, Hefei 230061, Anhui, PR China
| | - Jian Bao
- Department of Oncology, The First People's Hospital of Hefei/The Third Affiliated Hospital of Anhui Medical University, Hefei 230061, Anhui, PR China
| | - Congshu Zhang
- Department of Oncology, The First People's Hospital of Hefei/The Third Affiliated Hospital of Anhui Medical University, Hefei 230061, Anhui, PR China
| | - Yuqing Chai
- Department of Oncology, The First People's Hospital of Hefei/The Third Affiliated Hospital of Anhui Medical University, Hefei 230061, Anhui, PR China
| | - Hongxia Li
- Department of Oncology, The First People's Hospital of Hefei/The Third Affiliated Hospital of Anhui Medical University, Hefei 230061, Anhui, PR China.
| |
Collapse
|
44
|
Wang L, Jiang W, Su Y, Zhan M, Peng S, Liu H, Lu L. Self-Splittable Transcytosis Nanoraspberry for NIR-II Photo-Immunometabolic Cancer Therapy in Deep Tumor Tissue. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2204067. [PMID: 36073839 PMCID: PMC9661837 DOI: 10.1002/advs.202204067] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 08/11/2022] [Indexed: 05/17/2023]
Abstract
Cancer photo-immunotherapy (CPIT) as an ideal strategy can rapidly release hostile signals by appropriate dosage of focal laser irradiation to unmask primary tumor immunogenicity and can activate adaptive immunity to control distant metastases. However, many factors, including disordered immunometabolism, poor penetration of photothermal agents and immuno-regulators, inadequate laser penetration into the deep tumor region, restrict the therapeutic outcomes of CPIT. Here, a second near-infrared window (NIR-II) photo-immunometabolic cancer therapy (PICT) by a programmed raspberry-structured nanoadjuvant (PRNMT ) is presented that can potentiates efficient immunogenic cell death (ICD) in deep tumor tissue and alleviates immunometabolic disorder. The PRNMT is architected through self-assembly of indoleamine 2,3-dioxygenase 1 (IDO-1) inhibitor modified small-sized CuS nanoparticles (CuS5 ) and tumor microenvironment (TME) responsive cationized polymeric matrix. The TME can trigger the splitting and surface cationization of PRNMT into small cationized CuS5 that feature high transcytosis potential and TME immunometabolic regulation. Upon NIR-II irradiation, CuS5 induce homogeneous ICD and release immunometabolic regulator in deep tumor tissues, which ameliorates IDO-1 mediated immunometabolic disorder and further suppresses regulatory T cells infiltration. PRNMT mediated PICT effectively delays the primary murine mammary carcinoma 4T1 tumor growth and inhibits the lethal pulmonary metastasis in combination with programmed cell death protein 1 (PD1) blockade.
Collapse
Affiliation(s)
- Li Wang
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and TreatmentZhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University)ZhuhaiGuangdong519000P. R. China
- Department of RadiologyThe First Affiliated Hospital of USTCUniversity of Science and Technology of ChinaHefeiAnhui230001P. R. China
| | - Wei Jiang
- Department of RadiologyThe First Affiliated Hospital of USTCUniversity of Science and Technology of ChinaHefeiAnhui230001P. R. China
| | - Yanhong Su
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and TreatmentZhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University)ZhuhaiGuangdong519000P. R. China
| | - Meixiao Zhan
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and TreatmentZhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University)ZhuhaiGuangdong519000P. R. China
| | - Shaojun Peng
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and TreatmentZhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University)ZhuhaiGuangdong519000P. R. China
| | - Hang Liu
- Department of RadiologyThe First Affiliated Hospital of USTCUniversity of Science and Technology of ChinaHefeiAnhui230001P. R. China
| | - Ligong Lu
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and TreatmentZhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University)ZhuhaiGuangdong519000P. R. China
| |
Collapse
|
45
|
Overcoming challenges to enable targeting of metastatic breast cancer tumour microenvironment with nano-therapeutics: Current status and future perspectives. OPENNANO 2022. [DOI: 10.1016/j.onano.2022.100083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
46
|
Wang Y, Minden A. Current Molecular Combination Therapies Used for the Treatment of Breast Cancer. Int J Mol Sci 2022; 23:ijms231911046. [PMID: 36232349 PMCID: PMC9569555 DOI: 10.3390/ijms231911046] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 09/13/2022] [Accepted: 09/15/2022] [Indexed: 11/23/2022] Open
Abstract
Breast cancer is the second leading cause of death for women worldwide. While monotherapy (single agent) treatments have been used for many years, they are not always effective, and many patients relapse after initial treatment. Moreover, in some patients the response to therapy becomes weaker, or resistance to monotherapy develops over time. This is especially problematic for metastatic breast cancer or triple-negative breast cancer. Recently, combination therapies (in which two or more drugs are used to target two or more pathways) have emerged as promising new treatment options. Combination therapies are often more effective than monotherapies and demonstrate lower levels of toxicity during long-term treatment. In this review, we provide a comprehensive overview of current combination therapies, including molecular-targeted therapy, hormone therapy, immunotherapy, and chemotherapy. We also describe the molecular basis of breast cancer and the various treatment options for different breast cancer subtypes. While combination therapies are promising, we also discuss some of the challenges. Despite these challenges, the use of innovative combination therapy holds great promise compared with traditional monotherapies. In addition, the use of multidisciplinary technologies (such as nanotechnology and computer technology) has the potential to optimize combination therapies even further.
Collapse
|
47
|
Hou X, Zhong D, Chen H, Gu Z, Gong Q, Ma X, Zhang H, Zhu H, Luo K. Recent advances in hyaluronic acid-based nanomedicines: Preparation and application in cancer therapy. Carbohydr Polym 2022; 292:119662. [PMID: 35725165 DOI: 10.1016/j.carbpol.2022.119662] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 05/06/2022] [Accepted: 05/23/2022] [Indexed: 12/11/2022]
|
48
|
Yu L, Jin Y, Song M, Zhao Y, Zhang H. When Natural Compounds Meet Nanotechnology: Nature-Inspired Nanomedicines for Cancer Immunotherapy. Pharmaceutics 2022; 14:1589. [PMID: 36015215 PMCID: PMC9412684 DOI: 10.3390/pharmaceutics14081589] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 07/25/2022] [Accepted: 07/28/2022] [Indexed: 02/01/2023] Open
Abstract
Recent significant strides of natural compounds in immunomodulation have highlighted their great potential against cancer. Despite many attempts being made for cancer immunotherapy, the biomedical application of natural compounds encounters a bottleneck because of their unclear mechanisms, low solubility and bioavailability, and limited efficacy. Herein, we summarize the immune regulatory mechanisms of different natural compounds at each step of the cancer-immunity cycle and highlight their anti-tumor potential and current limitations. We then propose and present various drug delivery strategies based on nanotechnology, including traditional nanoparticles (NPs)-based delivery strategies (lipid-based NPs, micelles, and polysaccharide/peptide/protein-based NPs) and novel delivery strategies (cell-derived NPs and carrier-free NPs), thus providing solutions to break through existing bottlenecks. Furthermore, representative applications of nature-inspired nanomedicines are also emphasized in detail with the advantages and disadvantages discussed. Finally, the challenges and prospects of natural compounds for cancer immunotherapy are provided, hopefully, to facilitate their far-reaching development toward clinical translation.
Collapse
Affiliation(s)
- Linna Yu
- People’s Hospital of Qianxinan Buyi and Miao Minority Autonomous Prefecture, Xingyi 562400, China;
| | - Yi Jin
- Key Laboratory of Drug Quality Control and Pharmacovigilance (Ministry of Education), State Key Laboratory of Natural Medicines, Department of Pharmaceutics, NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing 210009, China; (Y.J.); (M.S.)
| | - Mingjie Song
- Key Laboratory of Drug Quality Control and Pharmacovigilance (Ministry of Education), State Key Laboratory of Natural Medicines, Department of Pharmaceutics, NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing 210009, China; (Y.J.); (M.S.)
| | - Yu Zhao
- People’s Hospital of Qianxinan Buyi and Miao Minority Autonomous Prefecture, Xingyi 562400, China;
| | - Huaqing Zhang
- Key Laboratory of Drug Quality Control and Pharmacovigilance (Ministry of Education), State Key Laboratory of Natural Medicines, Department of Pharmaceutics, NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing 210009, China; (Y.J.); (M.S.)
| |
Collapse
|
49
|
Ahmad MZ, Alasiri AS, Alasmary MY, Abdullah MM, Ahmad J, Abdel Wahab BA, M Alqahtani SA, Pathak K, Mustafa G, Khan MA, Saikia R, Gogoi U. Emerging advances in nanomedicine for breast cancer immunotherapy: opportunities and challenges. Immunotherapy 2022; 14:957-983. [PMID: 35852105 DOI: 10.2217/imt-2021-0348] [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] [Indexed: 11/21/2022] Open
Abstract
Breast cancer is one of the most common causes of cancer-related morbidity and mortality in women worldwide. Early diagnosis and an appropriate therapeutic approach for all cancers are climacterics for a favorable prognosis. Targeting the immune system in breast cancer is already a clinical reality with notable successes, specifically with checkpoint blockade antibodies and chimeric antigen receptor T-cell therapy. However, there have been inevitable setbacks in the clinical application of cancer immunotherapy, including inadequate immune responses due to insufficient delivery of immunostimulants to immune cells and uncontrolled immune system modulation. Rapid advancements and new evidence have suggested that nanomedicine-based immunotherapy may be a viable option for treating breast cancer.
Collapse
Affiliation(s)
- Mohammad Zaki Ahmad
- Department of Pharmaceutics, College of Pharmacy, Najran University, Najran, Najran, 11001, Kingdom of Saudi Arabia
| | - Ali S Alasiri
- Department of Pharmaceutics, College of Pharmacy, Najran University, Najran, Najran, 11001, Kingdom of Saudi Arabia
| | - Mohammed Yahia Alasmary
- Medical Department, College of Medicine, Najran University, Najran, 11001, Kingdom of Saudi Arabia
| | - M M Abdullah
- Advanced Materials & Nano-Research Centre, Department of Physics, Faculty of Science & Arts, Najran University, Najran, 11001, Kingdom Saudi Arabia
| | - Javed Ahmad
- Department of Pharmaceutics, College of Pharmacy, Najran University, Najran, Najran, 11001, Kingdom of Saudi Arabia
| | - Basel A Abdel Wahab
- Department of Pharmacology, College of Pharmacy, Najran University, Najran, 11001, Kingdom of Saudi Arabia
- Department of Pharmacology, College of Medicine, Assiut University, Assiut, 71515, Egypt
| | - Saif Aboud M Alqahtani
- Internal Medicine Department, College of Medicine, King Khalid University, Abha, 61421, Kingdom of Saudi Arabia
| | - Kalyani Pathak
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, 786004, Assam, India
| | - Gulam Mustafa
- College of Pharmacy, Shaqra University, Ad-Dawadmi Riyadh, Kingdom of Saudi Arabia
| | - Mohammad Ahmad Khan
- Department of Pharmacology, School of Pharmaceutical Education & Research, Jamia Hamdard, New Delhi, 110062, India
| | - Riya Saikia
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, 786004, Assam, India
| | - Urvashee Gogoi
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, 786004, Assam, India
| |
Collapse
|
50
|
Xie X, Jiang K, Li B, Hou S, Tang H, Shao B, Ping Y, Zhang Q. A small-molecule self-assembled nanodrug for combination therapy of photothermal-differentiation-chemotherapy of breast cancer stem cells. Biomaterials 2022; 286:121598. [DOI: 10.1016/j.biomaterials.2022.121598] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 05/16/2022] [Accepted: 05/19/2022] [Indexed: 02/07/2023]
|