1
|
Mu M, Chen B, Li H, Fan R, Yang Y, Zhou L, Han B, Zou B, Chen N, Guo G. Augmented the sensitivity of photothermal-ferroptosis therapy in triple-negative breast cancer through mitochondria-targeted nanoreactor. J Control Release 2024; 375:733-744. [PMID: 39332776 DOI: 10.1016/j.jconrel.2024.09.042] [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: 10/13/2023] [Revised: 09/16/2024] [Accepted: 09/24/2024] [Indexed: 09/29/2024]
Abstract
Ferroptosis primarily relies on reactive oxygen (ROS) production and lipid peroxide (LPO) accumulation, which opens up new opportunities for tumor therapy. However, a standalone ferroptosis process is insufficient in inhibiting tumor progression. Unlike previously reported Fe-based nanomaterials, we have engineered a novel nanoreactor named IR780/Ce@EGCG/APT, which uses metal-polyphenols network (Ce@EGCG) based on rare-earth cerium and epigallocatechin gallate (EGCG) to encapsulate IR780 and modified with the aptamer (AS1411). The intricately designed nanoreactor is specifically taken up by tumor cells, releasing Ce3+, EGCG, and IR780. On the one hand, Ce3+ triggers ROS production via a Fenton-like reaction, inducing ferroptosis in tumor cells. On the other hand, IR780 accumulates in mitochondria and disrupts mitochondrial function upon laser irradiation, leading to tumor cell apoptosis. EGCG serves as a sensitizer, simultaneously enhancing the sensitivity of tumor cells to ferroptosis and photothermal therapy. After a single dose and three times of 808 nm laser irradiation for treatment, it has been observed that the nanoreactor induces dendritic cells (DCs) maturation, facilitates cytotoxic T lymphocyte infiltration, improves immunosuppressive microenvironment, activates the systemic immune system, and generates long-term immune memory.
Collapse
Affiliation(s)
- Min Mu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, Department of Radiation Oncology and Department of Head and Neck Oncology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Bo Chen
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, Department of Radiation Oncology and Department of Head and Neck Oncology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Hui Li
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, Department of Radiation Oncology and Department of Head and Neck Oncology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Rangrang Fan
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, Department of Radiation Oncology and Department of Head and Neck Oncology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yuanli Yang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, Department of Radiation Oncology and Department of Head and Neck Oncology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Lihua Zhou
- National Institute of Measurement and Testing Technology, Chengdu 610021, Sichuan, China
| | - Bo Han
- Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, School of Pharmacy, Shihezi University, Shihezi 832002, China
| | - Bingwen Zou
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, Department of Radiation Oncology and Department of Head and Neck Oncology, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Nianyong Chen
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, Department of Radiation Oncology and Department of Head and Neck Oncology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Gang Guo
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, Department of Radiation Oncology and Department of Head and Neck Oncology, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
2
|
Zhang L, Hu H, Cai W, Chen S, Sheng P, Fu X. CaCO 3-complexed pH-responsive nanoparticles encapsulating mitoxantrone and celastrol enhance tumor chemoimmunotherapy. Int J Pharm 2024; 667:124860. [PMID: 39461678 DOI: 10.1016/j.ijpharm.2024.124860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 09/26/2024] [Accepted: 10/20/2024] [Indexed: 10/29/2024]
Abstract
Modulating the immunosuppressive tumor microenvironment (TME) while enhancing antitumor immune responses is a promising strategy. In this study, we designed an acid-sensitive nanosystem (MCCaNPs) to demonstrate effective immunotherapy against cancer through the systemic delivery of immune-stimulating chemotherapy combinations. A pH-responsive nanoplatform containing CaCO3 was prepared by the double emulsion method, and mitoxantrone (MIT) and celastrol (CEL) were simultaneously encapsulated as immunogenic cell death (ICD) inducers. Due to the acid responsiveness of CaCO3, the nanoparticles rapidly consume H+ to relieve the acidic tumor microenvironment and explosively release CEL and MIT, showing inherent immunomodulatory activity in collaborative tumor chemoimmunotherapy. MIT and CEL synergistically trigger stronger ICD by inducing tumor cells to release calreticulin (CRT), high mobility group box 1 protein (HMGB1). Following the intravenous administration of MCCaNPs, the local tumor microenvironment(TME) was reprogrammed in mice-bearing tumors. This reprogramming was characterized by a significant increase in the density of tumor-infiltrating cytotoxic T lymphocytes(CTLs), ultimately prolonging survival. Therefore, this research proposes a promising approach to trigger immunogenic cell death collaboratively, aiming to boost the tumor CTLs infiltration for anticancer immunotherapy.
Collapse
Affiliation(s)
- Liang Zhang
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang 330004, China.
| | - Huiqiang Hu
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang 330004, China.
| | - Wan Cai
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang 330004, China.
| | - Shungen Chen
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang 330004, China.
| | - Ping Sheng
- College of Life and Health Sciences, Anhui Science and Technology University, Fengyang 233100, China.
| | - Xiaomei Fu
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang 330004, China; FAN Cuisheng Studio of National Famous TCM, Nanchang 330006, China.
| |
Collapse
|
3
|
Cui C, Du M, Zhao Y, Tang J, Liu M, Min G, Chen R, Zhang Q, Sun Z, Weng H. Functional Ginger-Derived Extracellular Vesicles-Coated ZIF-8 Containing TNF-α siRNA for Ulcerative Colitis Therapy by Modulating Gut Microbiota. ACS APPLIED MATERIALS & INTERFACES 2024; 16:53460-53473. [PMID: 39303016 DOI: 10.1021/acsami.4c10562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/22/2024]
Abstract
Tumor necrosis factor-α (TNF-α) plays a causal role in the pathogenesis of ulcerative colitis (UC), and anti-TNF-α siRNA shows great promise in UC therapy. However, delivering siRNA with site-targeted stability and therapeutic efficacy is still challenging due to the complex and dynamic intestinal microenvironment. Here, based on the functional plant-derived ginger extracellular vesicles (EVs) and porous ZIF-8 nanoparticles, we propose a novel TNF-α siRNA delivery strategy (EVs@ZIF-8@siRNA) for UC targeted therapy. Ginger EVs show strong colon and macrophage targeting, as well as robust resistance to acidic degradation in the stomach. Moreover, 6-shogaol in ginger-derived EVs displays anti-inflammatory effects, which enhance the treatment efficiency by cooperation with TNF-α siRNA. In vitro experiments reveal that ZIF-8 nanoparticles have high TNF-α siRNA loading capacity and promote siRNA escape from cellular lysosomes. In vivo experiments show that the TNF-α level is reduced more significantly in colonic tissue than other nontargeted inflammation related factors, showing a good targeting of this composite nanoparticle. Furthermore, gut microbiota sequencing results demonstrate that the nanoparticles can promote intestinal barrier repair by regulating the intestinal microbial balance and restoring the intestinal health of UC mice. Therefore, the developed EVs@ZIF-8@siRNA nanoparticles may represent a novel colon-targeted oral drug, providing a promising therapeutic strategy for UC therapy.
Collapse
Affiliation(s)
- Chenyang Cui
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001 Henan, China
| | - Miao Du
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001 Henan, China
| | - Yihang Zhao
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001 Henan, China
| | - Jiaze Tang
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001 Henan, China
| | - Mengge Liu
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001 Henan, China
| | - Geng Min
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001 Henan, China
| | - Rongchen Chen
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001 Henan, China
| | - Qiang Zhang
- Department of Critical Care Medicine, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 Henan China
| | - Zhaowei Sun
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001 Henan, China
| | - Haibo Weng
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001 Henan, China
| |
Collapse
|
4
|
Zhou A, Jia J, Ji X, Cheng S, Song X, Hu J, Zhao Y, Yu L, Wang J, Wang F. Reshaped Local and Systemic Immune Responses Triggered by a Biomimetic Multifunctional Nanoplatform Coordinating Multi-Pathways for Cancer Therapy. ACS APPLIED MATERIALS & INTERFACES 2024. [PMID: 39356986 DOI: 10.1021/acsami.4c05714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/04/2024]
Abstract
Immunotherapy has fundamentally transformed the clinical cancer treatment landscape; however, achieving intricate and multifaceted modulation of the immune systems remains challenging. Here, a multipathway coordination of immunogenic cell death (ICD), autophagy, and indoleamine 2,3-dioxygenase-1 (IDO1) was achieved by a biomimetic nano-immunomodulator assembled from a chemotherapeutic agent (doxorubicin, DOX), small interfering RNA (siRNA) molecules targeting IDO1 (siIDO1), and the zeolitic imidazolate framework-8 (ZIF-8). After being camouflaged with a macrophage membrane, the biomimetic nanosystem, named mRDZ, enriched in tumors, which allowed synergistic actions of its components within tumor cells. The chemotherapeutic intervention led to a compensatory upregulation in the expression of IDO1, consequently exerting an inhibitory effect on the reactive oxygen species (ROS) and autophagic responses triggered by DOX and ZIF-8. Precise gene silencing of IDO1 by siIDO1 alleviated its suppressive influence, thereby facilitating increased ROS production and improved autophagy, ultimately bolstering tumor immunogenicity. mRDZ exhibited strong capability to boost potent local and systemic antitumor immune responses with a feature of memory, which led to the effective suppression of the growth, lung metastasis, and recurrence of the tumor. Serving as an exemplary model for the straightforward and potent reshaping of the immune system against tumors, mRDZ offers valuable insights into the development of immunomodulatory nanomaterials for cancer therapy.
Collapse
Affiliation(s)
- Ao Zhou
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China
- Key Laboratory of Nanomedical Technology (Education Department of Fujian Province), Nanomedical Technology Research Institute, School of Pharmacy, Fujian Medical University, Fuzhou 350122, China
| | - Jingyan Jia
- Key Laboratory of Nanomedical Technology (Education Department of Fujian Province), Nanomedical Technology Research Institute, School of Pharmacy, Fujian Medical University, Fuzhou 350122, China
| | - Xueyang Ji
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China
| | - Sunying Cheng
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China
| | - Xiaoxin Song
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China
| | - Jingyan Hu
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China
| | - Yan Zhao
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China
| | - Luying Yu
- Key Laboratory of Nanomedical Technology (Education Department of Fujian Province), Nanomedical Technology Research Institute, School of Pharmacy, Fujian Medical University, Fuzhou 350122, China
| | - Jieting Wang
- Key Laboratory of Nanomedical Technology (Education Department of Fujian Province), Nanomedical Technology Research Institute, School of Pharmacy, Fujian Medical University, Fuzhou 350122, China
| | - Fang Wang
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China
| |
Collapse
|
5
|
Li Z, Chen Z, Shi K, Huang P, Zeng W, Huang Q, Peng J, Yang L, Chen H, Zhao Y, Zeng X. Polyphenol-Based Self-Assembled Nanomedicine for a Three-Pronged Approach to Reversing Tumor Immunosuppression. Adv Healthc Mater 2024:e2402127. [PMID: 39344218 DOI: 10.1002/adhm.202402127] [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: 06/10/2024] [Revised: 08/14/2024] [Indexed: 10/01/2024]
Abstract
The challenges of multi-pathway immune resistance and systemic toxicity caused by the direct injection of immune checkpoint inhibitors are critical factors that compromise the effectiveness of clinical immune checkpoint blockade therapy. In this context, natural polyphenols have been employed as the primary component to construct a targeted and acid-responsive PD-L1 antibody (αPD-L1) delivery nanoplatform. This platform incorporates garcinol, an inhibitor of the Nuclear Factor Kappa-B (NF-κB) signaling pathway, to regulate pro-tumor immune escape cytokines and regulatory T cells. Additionally, the nanoplatform has been verified to induce immunogenic cell death (ICD), which promotes the maturation of dendritic cells and enhances the activity of cytotoxic T lymphocytes. In vivo and in vitro experimental results demonstrated that the nanoplatform can boost the immune response through a PD-L1 and NF-κB blocking/ICD inducing three-pronged strategy, thereby effectively combating tumor growth and metastasis.
Collapse
Affiliation(s)
- Zimu Li
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, 637371, Singapore
| | - Zirui Chen
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Kexin Shi
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Ping Huang
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Wenfeng Zeng
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Qili Huang
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Jingwen Peng
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Li Yang
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Hongzhong Chen
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Yanli Zhao
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, 637371, Singapore
| | - Xiaowei Zeng
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| |
Collapse
|
6
|
Sam Lee J, Kim M, Jin H, Kwak M, Cho E, Kim KS, Kim DE. DNA aptamer-conjugated lipid nanoparticle for targeted PTEN mRNA delivery to prostate cancer cells. Int J Pharm 2024; 662:124519. [PMID: 39067551 DOI: 10.1016/j.ijpharm.2024.124519] [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: 04/19/2024] [Revised: 07/17/2024] [Accepted: 07/24/2024] [Indexed: 07/30/2024]
Abstract
The use of messenger RNA (mRNA) as a cancer vaccine and gene therapy requires targeted vehicle delivery to the site of disease. Here, we designed a mRNA-encapsulating lipid nanoparticle (LNP) conjugated with anti-programmed death-ligand 1 (PD-L1) DNA aptamer that delivers mRNA encoding a tumor suppressor gene, namely phosphatase and tensin homolog (PTEN), to castration-resistant prostate cancer (CRPC) cells expressing PD-L1 on the cell surface. The DNA aptamer-conjugated LNP-based mRNA delivery system (Apt-LNP[PTEN mRNA]) mediated efficient mRNA delivery and transfection in CRPC cells than LNPs without targeting ligands. Cancer-targeted PTEN mRNA delivery using Apt-LNPs achieved significantly higher PTEN expression via aptamer-mediated endocytosis in target cancer cells compared with non-targeted LNP delivery, resulting in significant downregulation of AKT phosphorylation. This enhanced PI3K/AKT pathway regulation, and in turn reduced cell migration after two days along with a 70 % decrease in cell viability, leading to effective apoptotic cell death. In a CRPC xenograft model, Apt-LNP[PTEN mRNA] led to an approximate 60 % reduction in tumor growth, which was attributable to the effective PTEN restoration and PI3K/AKT signaling pathway regulation. PTEN expression was significantly enhanced in CRPC tumor tissues, which abolished cancer cell tumorigenicity. These findings demonstrated the potential of Apt-LNPs for targeted mRNA delivery to cancer cells, thus providing a promising tool for targeted mRNA delivery to a range of cancers and tissues using a conventional LNP systems.
Collapse
Affiliation(s)
- Jong Sam Lee
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Republic of Korea
| | - Minhee Kim
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Republic of Korea
| | - Hyesoo Jin
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Republic of Korea
| | - Minseo Kwak
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Republic of Korea
| | - Eunbin Cho
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Republic of Korea
| | - Keun-Sik Kim
- Department of Biomedical Laboratory Science, Konyang University, Daejeon 35365, Republic of Korea
| | - Dong-Eun Kim
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Republic of Korea.
| |
Collapse
|
7
|
Dong Z, Yang W, Zhang Y, Wang B, Wan X, Li M, Chen Y, Zhang N. Biomimetic nanomedicine cocktail enables selective cell targeting to enhance ovarian Cancer chemo- and immunotherapy. J Control Release 2024; 373:172-188. [PMID: 38972639 DOI: 10.1016/j.jconrel.2024.07.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 07/01/2024] [Accepted: 07/04/2024] [Indexed: 07/09/2024]
Abstract
Ovarian cancer is one of the deadliest cancers, and combined chemo- and immunotherapies are potential strategies to combat it. However, the anti-cancer efficacy of the combined therapies may be limited by the non-selective co-delivery of chemotherapy and immunotherapy. Herein, a combined chemo- and immunotherapy is designed to selectively target ovarian tumor (ID8) cells and dendritic cells (DCs) using ID8 cell membrane (IM) and bacterial outer membrane vesicles (OMVs), respectively. Doxorubicin (DOX) and Ovalbumin (OVA) peptide (OVA257-264) are chosen as model chemotherapy and immunotherapy agents, respectively. A DNA nanocube capable of easily loading DOX or OVA257-264 is chosen as the carrier. Firstly, the DNA nanocube is used to load DOX or OVA257-264 to prepare cube-DOX or cube-OVA. This nanocube was then encapsulated with IM to form IM@Cube-DOX and with OMV to form OMV@Cube-OVA. IM@Cube-DOX can be selectively taken up by ID8 cells, leading to effective cell killing, while OMV@Cube-OVA targets and activates DC2.4 cells in vitro. Both IM@Cube-DOX and OMV@Cube-OVA show increased accumulation at ID8 tumors in C57BL/6 mice. Combined IM@Cube-DOX + OMV@Cube-OVA therapy demonstrates better anti-tumor efficacy than non-selective delivery methods such as OMV@(Cube-DOX + Cube-OVA) or IM@(Cube-DOX + Cube-OVA) in ID8-OVA tumor-bearing mice. In conclusion, this study demonstrates a biomimetic delivery strategy that enables selective drug delivery to tumor cells and DCs, thereby enhancing the anti-tumor efficacy of combined chemo- and immunotherapy through the selective delivery strategy.
Collapse
Affiliation(s)
- Zhuolin Dong
- Department of Pharmaceutics and Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, Henan, PR China
| | - Wenhui Yang
- Department of Pharmacy, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, PR China
| | - Yuzhen Zhang
- Department of Pharmaceutics and Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, Henan, PR China
| | - Baojin Wang
- Henan International Joint Laboratory of Ovarian Malignancies, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, PR China
| | - Xiangling Wan
- Department of Pharmaceutics and Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, Henan, PR China
| | - Mengru Li
- Department of Pharmaceutics and Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, Henan, PR China
| | - Yibing Chen
- Genetic and Prenatal Diagnosis Center, Department of Gynecology and Obstetrics, First Affiliated Hospital, Zhengzhou University, Zhengzhou 450052, Henan, PR China.
| | - Nan Zhang
- Department of Pharmaceutics and Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, Henan, PR China.
| |
Collapse
|
8
|
Luobin L, Wanxin H, Yingxin G, Qinzhou Z, Zefeng L, Danyang W, Huaqin L. Nanomedicine-induced programmed cell death in cancer therapy: mechanisms and perspectives. Cell Death Discov 2024; 10:386. [PMID: 39209834 PMCID: PMC11362291 DOI: 10.1038/s41420-024-02121-0] [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: 05/16/2024] [Revised: 07/20/2024] [Accepted: 07/30/2024] [Indexed: 09/04/2024] Open
Abstract
The balance of programmed cell death (PCD) mechanisms, including apoptosis, autophagy, necroptosis and others, is pivotal in cancer progression and treatment. Dysregulation of these pathways results in uncontrolled cell growth and resistance to conventional therapies. Nanomedicine offers a promising solution in oncology through targeted drug delivery enabling precise targeting of cancer cells while preserving healthy tissues. This approach reduces the side effects of traditional chemotherapy and enhances treatment efficacy by engaging PCD pathways. We details each PCD pathway, their mechanisms, and innovative nanomedicine strategies to activate these pathways, thereby enhancing therapeutic specificity and minimizing harm to healthy tissues. The precision of nanotechnology in targeting PCD pathways promises significant improvements in cancer treatment outcomes. This synergy between nanotechnology and targeted PCD activation could lead to more effective and less toxic cancer therapies, heralding a new era in cancer treatment.
Collapse
Affiliation(s)
- Lin Luobin
- School of Health Sciences, Guangzhou Xinhua University, 19 Huamei Road, Tianhe District, Guangzhou, 510520, China
- School of Life Sciences and Biopharmaceuticals, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - He Wanxin
- School of Health Sciences, Guangzhou Xinhua University, 19 Huamei Road, Tianhe District, Guangzhou, 510520, China
| | - Guo Yingxin
- School of Health Sciences, Guangzhou Xinhua University, 19 Huamei Road, Tianhe District, Guangzhou, 510520, China
| | - Zheng Qinzhou
- School of Life Sciences and Biopharmaceuticals, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Liang Zefeng
- School of Life Sciences and Biopharmaceuticals, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Wu Danyang
- School of Health Sciences, Guangzhou Xinhua University, 19 Huamei Road, Tianhe District, Guangzhou, 510520, China
| | - Li Huaqin
- School of Health Sciences, Guangzhou Xinhua University, 19 Huamei Road, Tianhe District, Guangzhou, 510520, China.
| |
Collapse
|
9
|
Zhou Z, Mai Y, Zhang G, Wang Y, Sun P, Jing Z, Li Z, Xu Y, Han B, Liu J. Emerging role of immunogenic cell death in cancer immunotherapy: Advancing next-generation CAR-T cell immunotherapy by combination. Cancer Lett 2024; 598:217079. [PMID: 38936505 DOI: 10.1016/j.canlet.2024.217079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 06/11/2024] [Accepted: 06/18/2024] [Indexed: 06/29/2024]
Abstract
Immunogenic cell death (ICD) is a stress-driven form of regulated cell death (RCD) in which dying tumor cells' specific signaling pathways are activated to release damage-associated molecular patterns (DAMPs), leading to the robust anti-tumor immune response as well as a reversal of the tumor immune microenvironment from "cold" to "hot". Chimeric antigen receptor (CAR)-T cell therapy, as a landmark in anti-tumor immunotherapy, plays a formidable role in hematologic malignancies but falls short in solid tumors. The Gordian knot of CAR-T cells for solid tumors includes but is not limited to, tumor antigen heterogeneity or absence, physical and immune barriers of tumors. The combination of ICD induction therapy and CAR-T cell immunotherapy is expected to promote the intensive use of CAR-T cell in solid tumors. In this review, we summarize the characteristics of ICD, stress-responsive mechanism, and the synergistic effect of various ICD-based therapies with CAR-T cells to effectively improve anti-tumor capacity.
Collapse
Affiliation(s)
- Zhaokai Zhou
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Yumiao Mai
- Department of Pediatrics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Ge Zhang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Henan Province Key Laboratory of Cardiac Injury and Repair, Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, Henan, 450052, China
| | - Yingjie Wang
- Department of Pediatrics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Pan Sun
- Department of Pediatrics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Zhaohe Jing
- Department of Pediatrics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Zhengrui Li
- Department of Oral and Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yudi Xu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Bo Han
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Jian Liu
- Department of Pediatrics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China.
| |
Collapse
|
10
|
Yu H, Huang Y, Nong Z, Lin X, Tang K, Cai Z, Huang K, Yu T, Lan H, Zhang Q, Wang Q, Yang L, Zhu J, Wu L, Luo H. In-Situ Grown Nanocrystal TiO 2 on 2D Ti 3C 2 Nanosheets with Anti-Tumor Activity from Photo-Sonodynamic Treatment and Immunology. Int J Nanomedicine 2024; 19:7963-7981. [PMID: 39130689 PMCID: PMC11316479 DOI: 10.2147/ijn.s457112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 07/06/2024] [Indexed: 08/13/2024] Open
Abstract
Introduction Traditional cancer treatment strategies often have severe toxic side effects and poor therapeutic efficacy. To address the long-standing problems related to overcoming the complexity of tumors, we develop a novel nanozyme based on the in situ oxidation of 2D Ti3C2 structure to perform simultaneous phototherapy and sonodynamic therapy on tumors. Ti3C2 nanozymes exhibit multi-enzyme activity, including intrinsic peroxidase (POD) activities, which can react with H2O2 in the tumor microenvironment. This new material can construct Ti3C2/TiO2 heterostructures in vivo. Methods Photothermal (PTT), sonodynamic (SDT) effects, and photoacoustic (PA) image-guided synergy therapy can be achieved. Finally, anticancer immune responses occur with this nanozyme. In vivo experiments revealed that the Ti3C2/TiO2 heterostructure inhibited tumor growth. Results Complementarily, our results showed that the Ti3C2/TiO2 heterostructure enhanced the immunogenic activity of tumors by recruiting cytotoxic T cells, thereby enhancing the tumor ablation effect. Mechanistic studies consistently indicated that Reactive Oxygen Species (ROS) regulates apoptosis of HCC cells by modulating NRF2/OSGIN1 signaling both in vitro and in vivo. As a result, Ti3C2 nanozyme effectively inhibited tumor through its synergistic ability to modulate ROS and enhance immune infiltration of cytotoxic T cells in the tumor microenvironment. Discussion These findings open up new avenues for enhancing 2D Ti3C2 nanosheets and suggest a new way to develop more effective sonosensitizers for the treatment of cancer.
Collapse
Affiliation(s)
- Hailing Yu
- Guangdong Provincial Engineering Research Center of Molecular Imaging, Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, Guangdong, People’s Republic of China
| | - Yongquan Huang
- Department of Ultrasound, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong, People’s Republic of China
| | - Zhisheng Nong
- School of Materials Science and Engineering, Shenyang Aerospace University, Shenyang, Liaoning, People’s Republic of China
| | - Xi Lin
- Guangdong Provincial Engineering Research Center of Molecular Imaging, Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, Guangdong, People’s Republic of China
| | - Kexin Tang
- Guangdong Provincial Engineering Research Center of Molecular Imaging, Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, Guangdong, People’s Republic of China
| | - Zeyu Cai
- Guangdong Provincial Engineering Research Center of Molecular Imaging, Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, Guangdong, People’s Republic of China
| | - Kaichen Huang
- Department of Clinical laboratory, The Third People’s Hospital of Zhuhai, Zhuhai, Guangdong, People’s Republic of China
| | - Ting Yu
- Guangdong Provincial Engineering Research Center of Molecular Imaging, Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, Guangdong, People’s Republic of China
| | - Huimin Lan
- Guangdong Provincial Engineering Research Center of Molecular Imaging, Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, Guangdong, People’s Republic of China
| | - Qianqian Zhang
- Guangdong Provincial Engineering Research Center of Molecular Imaging, Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, Guangdong, People’s Republic of China
| | - Qiang Wang
- The Green Aerotechnics Research Institute of Chongqing Jiaotong University, Chongqing, People’s Republic of China
| | - Lei Yang
- Center for Composite Materials and Structures, Harbin Institute of Technology, Harbin, Heilongjiang, People’s Republic of China
| | - Jingchuan Zhu
- School of Materials Science and Engineering, Harbin Institute of Technology, Harbin, Heilongjiang, People’s Republic of China
| | - Lili Wu
- Key Laboratory for Photonic and Electronic Bandgap Materials, Ministry of Education, School of Physics and Electronic Engineering, Harbin Normal University, Harbin, Heilongjiang, People’s Republic of China
| | - Hui Luo
- Guangdong Provincial Engineering Research Center of Molecular Imaging, Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, Guangdong, People’s Republic of China
| |
Collapse
|
11
|
Wong KY, Wong MS, Liu J. Aptamer-functionalized liposomes for drug delivery. Biomed J 2024; 47:100685. [PMID: 38081386 PMCID: PMC11340590 DOI: 10.1016/j.bj.2023.100685] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 10/21/2023] [Accepted: 12/05/2023] [Indexed: 07/26/2024] Open
Abstract
Among the various targeting ligands for drug delivery, aptamers have attracted much interest in recent years because of their smaller size compared to antibodies, ease of modification, and better batch-to-batch consistency. In addition, aptamers can be selected to target both known and even unknown cell surface biomarkers. For drug loading, liposomes are the most successful vehicle and many FDA-approved formulations are based on liposomes. In this paper, aptamer-functionalized liposomes for targeted drug delivery are reviewed. We begin with the description of related aptamers selection, followed by methods to conjugate aptamers to liposomes and the fate of such conjugates in vivo. Then a few examples of applications are reviewed. In addition to intravenous injection for systemic delivery and hoping to achieve accumulation at target sites, for certain applications, it is also possible to have aptamer/liposome conjugates applied directly at the target tissue such as intratumor injection and dropping on the surface of the eye by adhering to the cornea. While previous reviews have focused on cancer therapy, the current review mainly covers other applications in the last four years. Finally, this article discusses potential issues of aptamer targeting and some future research opportunities.
Collapse
Affiliation(s)
- Ka-Ying Wong
- Department of Chemistry, Waterloo Institute for Nanotechnology, University of Waterloo, Waterloo, Canada; Centre for Eye and Vision Research (CEVR), Pak Shek Kok, Shatin, Hong Kong.
| | - Man-Sau Wong
- Centre for Eye and Vision Research (CEVR), Pak Shek Kok, Shatin, Hong Kong; Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong; Research Center for Chinese Medicine Innovation, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong.
| | - Juewen Liu
- Department of Chemistry, Waterloo Institute for Nanotechnology, University of Waterloo, Waterloo, Canada; Centre for Eye and Vision Research (CEVR), Pak Shek Kok, Shatin, Hong Kong.
| |
Collapse
|
12
|
Yan B, Li Y, He S. Aptamer-mediated therapeutic strategies provide a potential approach for cancer. Int Immunopharmacol 2024; 136:112356. [PMID: 38820957 DOI: 10.1016/j.intimp.2024.112356] [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: 04/04/2024] [Revised: 05/21/2024] [Accepted: 05/24/2024] [Indexed: 06/02/2024]
Abstract
The treatment of tumors still faces considerable challenges. While conventional treatments such as surgery, chemotherapy, and radiation therapy provide some curative effects, their side effects and limitations highlight the importance of finding more precise treatment strategies. Aptamers have become an important target molecule in the field of drug delivery systems due to their good affinity and targeting, and they have gradually become an important link from basic research to clinical application. In this paper, we discussed the latest progress of aptamer-mediated nanodrugs, as well as aptamer-mediated photodynamic therapy, photothermal therapy, and immunotherapy strategies for tumor treatment, and explored the possibility of aptamer-mediated therapy for accurate tumor treatment. The purpose of this review is to provide novel insights for treating tumors with aptamer-mediated therapies by summarizing these innovative strategies, thereby ultimately enhancing the therapeutic efficacy for cancer patients.
Collapse
Affiliation(s)
- Bingshuo Yan
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, People's Republic of China
| | - Yuting Li
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, People's Republic of China
| | - Shiming He
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, People's Republic of China.
| |
Collapse
|
13
|
Jin B, Yang G, Guo Z, Chen Z, Liu Y, Li S, Chen H, Fang Y, Deng Y, He N. Cell-SELEX and application research of a DNA aptamer against esophageal squamous cell carcinoma (ESCC) cell line TE-1. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2024; 16:4683-4690. [PMID: 38958106 DOI: 10.1039/d4ay00895b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2024]
Abstract
Esophageal cancer is a common cancer with high morbidity and mortality that severely threatens the safety and quality of human life. The strong metastatic nature of esophageal cancer enables it to metastasize more quickly and covertly, making it difficult for current diagnostic and treatment methods to achieve efficient early screening, as well as timely and effective treatment. As a promising solution, nucleic acid aptamers, a kind of special single-stranded DNA or RNA oligonucleotide selected by the Systematic Evolution of Ligands by Exponential Enrichment (SELEX) technology, can specifically bind with different molecular targets. In this paper, random DNA single-stranded oligonucleotides were used as the initial library. Using TE-1 cells and HEEC cells as targets, specific binding sequences were selected by 15 rounds of the cell-SELEX method, and the aptamer sequence that binds to TE-1 cells with the most specificity was obtained and named Te4. The Te4 aptamer was further validated for binding specificity, binding affinity, type of target, in vitro cytotoxicity when conjugated with DOX(Te4-DOX), and in vivo distribution. Results of in vitro validation showed that Te4 has outstanding binding specificity with a Kd value of 51.16 ± 5.52 nM, and the target type of Te4 was preliminarily identified as a membrane protein. Furthermore, the cytotoxicity experiment showed that Te4-DOX has specific cytotoxicity towards cultured TE-1 cells. Finally, the results of the in vivo distribution experiment showed that the Te4 aptamer is able to specifically target tumor regions in nude mice, showing great potential to be applied in future diagnosis and targeted therapy of esophageal cancer.
Collapse
Affiliation(s)
- Baijiang Jin
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China.
| | - Gaojian Yang
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China.
| | - Zhukang Guo
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China.
- Department of Clinical Laboratory, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, China.
| | - Zhu Chen
- School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China.
- Department of Clinical Laboratory, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, China.
| | - Yuan Liu
- School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China.
- Institute for Future Sciences, University of South China, Changsha, Hunan 410000, China
| | - Song Li
- School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China.
- Institute for Future Sciences, University of South China, Changsha, Hunan 410000, China
| | - Hui Chen
- School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China.
- Institute for Future Sciences, University of South China, Changsha, Hunan 410000, China
| | - Yile Fang
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China.
- Department of Clinical Laboratory, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, China.
| | - Yan Deng
- School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China.
- Institute for Future Sciences, University of South China, Changsha, Hunan 410000, China
| | - Nongyue He
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China.
- Hunan Key Laboratory of Biomedical Nanomaterials and Devices, Hunan University of Technology, Zhuzhou 412007, Hunan, China.
| |
Collapse
|
14
|
Xie F, Qiu J, Sun C, Feng L, Jun Y, Luo C, Guo X, Zhang B, Zhou Y, Wang Y, Zhang L, Wang Q. Development of a Specific Aptamer-Modified Nano-System to Treat Esophageal Squamous Cell Carcinoma. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2309084. [PMID: 38704694 PMCID: PMC11267304 DOI: 10.1002/advs.202309084] [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: 11/24/2023] [Revised: 03/23/2024] [Indexed: 05/07/2024]
Abstract
Esophageal squamous cell carcinoma (ESCC) is a prevalent gastrointestinal cancer characterized by high mortality and an unfavorable prognosis. While combination therapies involving surgery, chemotherapy, and radiation therapy are advancing, targeted therapy for ESCC remains underdeveloped. As a result, the overall five-year survival rate for ESCC is still below 20%. Herein, ESCC-specific DNA aptamers and an innovative aptamer-modified nano-system is introduced for targeted drug and gene delivery to effectively inhibit ESCC. The EA1 ssDNA aptamer, which binds robustly to ESCC cells with high specificity and affinity, is identified using cell-based systematic evolution of ligands by exponential enrichment (cell-SELEX). An EA1-modified nano-system is developed using a natural egg yolk lipid nanovector (EA1-EYLNs-PTX/siEFNA1) that concurrently loads paclitaxel (PTX) and a small interfering RNA of Ephrin A1 (EFNA1). This combination counters ESCC's proliferation, migration, invasion, and lung metastasis. Notably, EFNA1 is overexpressed in ESCC tumors with lung metastasis and has an inverse correlation with ESCC patient prognosis. The EA1-EYLNs-PTX/siEFNA1 nano-system offers effective drug delivery and tumor targeting, resulting in significantly improved therapeutic efficacy against ESCC tumors. These insights suggest that aptamer-modified nano-systems can deliver drugs and genes with superior tumor-targeting, potentially revolutionizing targeted therapy in ESCC.
Collapse
Affiliation(s)
- Fei Xie
- The Comprehensive Cancer Center, Department of Central Laboratory, The Affiliated Huaian No.1 People's HospitalNanjing Medical UniversityHuai'anJiangsu223300China
| | - Jinrong Qiu
- The Comprehensive Cancer Center, Department of Central Laboratory, The Affiliated Huaian No.1 People's HospitalNanjing Medical UniversityHuai'anJiangsu223300China
| | - Congyong Sun
- The Comprehensive Cancer Center, Department of Central Laboratory, The Affiliated Huaian No.1 People's HospitalNanjing Medical UniversityHuai'anJiangsu223300China
| | - Lulu Feng
- The Comprehensive Cancer Center, Department of Central Laboratory, The Affiliated Huaian No.1 People's HospitalNanjing Medical UniversityHuai'anJiangsu223300China
| | - Yali Jun
- The Comprehensive Cancer Center, Department of Central Laboratory, The Affiliated Huaian No.1 People's HospitalNanjing Medical UniversityHuai'anJiangsu223300China
- The Comprehensive Cancer Center, Department of Clinical Oncology, The Affiliated Huaian No.1 People's HospitalNanjing Medical UniversityHuai'anJiangsu223300China
| | - Chao Luo
- The Comprehensive Cancer Center, Department of Central Laboratory, The Affiliated Huaian No.1 People's HospitalNanjing Medical UniversityHuai'anJiangsu223300China
| | - Xiamei Guo
- The Comprehensive Cancer Center, Department of Central Laboratory, The Affiliated Huaian No.1 People's HospitalNanjing Medical UniversityHuai'anJiangsu223300China
| | - Bowei Zhang
- The Comprehensive Cancer Center, Department of Central Laboratory, The Affiliated Huaian No.1 People's HospitalNanjing Medical UniversityHuai'anJiangsu223300China
| | - Yu Zhou
- The Comprehensive Cancer Center, Department of Clinical Oncology, The Affiliated Huaian No.1 People's HospitalNanjing Medical UniversityHuai'anJiangsu223300China
| | - Yuting Wang
- The Comprehensive Cancer Center, Department of Central Laboratory, The Affiliated Huaian No.1 People's HospitalNanjing Medical UniversityHuai'anJiangsu223300China
| | - Li Zhang
- The Comprehensive Cancer Center, Department of Central Laboratory, The Affiliated Huaian No.1 People's HospitalNanjing Medical UniversityHuai'anJiangsu223300China
| | - Qilong Wang
- The Comprehensive Cancer Center, Department of Central Laboratory, The Affiliated Huaian No.1 People's HospitalNanjing Medical UniversityHuai'anJiangsu223300China
| |
Collapse
|
15
|
Zhang J, Sang X, Yuan Y, Shen J, Fang Y, Qin M, Zheng H, Zhu Z. 4-Deoxy- ε-Pyrromycinone: A Promising Drug/Lead Compound to Treat Tumors. Drug Des Devel Ther 2024; 18:2367-2379. [PMID: 38911033 PMCID: PMC11193465 DOI: 10.2147/dddt.s461594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 06/10/2024] [Indexed: 06/25/2024] Open
Abstract
Background Anthraquinone drugs are widely used in the treatment of tumors. However, multidrug resistance and severe cardiac toxicity limit its use, which have led to the discovery of new analogues. In this paper, 4-Deoxy-ε-pyrromycinone (4-Deo), belonging to anthraquinone compounds, was first been studied with the anti-tumor effects and the safety in vitro and in vivo as a new anti-tumor drug or lead compound. Methods The quantitative analysis of 4-Deo was established by UV methodology. The anti-cancer effect of 4-Deo in vitro was evaluated by cytotoxicity experiments of H22, HepG2 and Caco2, and the anti-cancer mechanism was explored by cell apoptosis and cycle. The tumor-bearing mouse model was established by subcutaneous inoculation of H22 cells to evaluate the anti-tumor effect of 4-Deo in vivo. The safety of 4-Deo was verified by the in vitro safety experiments of healthy cells and the in vivo safety experiments of H22 tumor-bearing mice. Tumor tissue sections were labeled with CRT, HMGB1, IL-6 and CD115 to explore the preliminary anti-cancer mechanism by immunohistochemistry. Results In vitro experiments demonstrated that 4-Deo could inhibit the growth of H22 by inducing cell necrosis and blocking cells in S phase, and 4-Deo has less damage to healthy cells. In vivo experiments showed that 4-Deo increased the positive area of CRT and HMGB1, which may inhibit tumor growth by triggering immunogenic cell death (ICD). In addition, 4-Deo reduced the positive area of CSF1R, and the anti-tumor effect may be achieved by blocking the transformation of tumor-associated macrophages (TAMs) to M2 phenotype. Conclusion In summary, this paper demonstrated the promise of 4-Deo for cancer treatment in vitro and in vivo. This paper lays the foundation for the study of 4-Deo, which is beneficial for the further development anti-tumor drugs based on the lead compound of 4-Deo.
Collapse
Affiliation(s)
- Jiping Zhang
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, 311402, People’s Republic of China
| | - Xianan Sang
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, 311402, People’s Republic of China
| | - Yichao Yuan
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, 311402, People’s Republic of China
| | - Jiawei Shen
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, 311402, People’s Republic of China
| | - Yuanyuan Fang
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, 311402, People’s Republic of China
| | - Minjing Qin
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, 311402, People’s Republic of China
| | - Hangsheng Zheng
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, 311402, People’s Republic of China
| | - Zhihong Zhu
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, 311402, People’s Republic of China
| |
Collapse
|
16
|
Moon Y, Cho H, Kim K. Nano-Delivery of Immunogenic Cell Death Inducers and Immune Checkpoint Blockade Agents: Single-Nanostructure Strategies for Enhancing Immunotherapy. Pharmaceutics 2024; 16:795. [PMID: 38931916 PMCID: PMC11207855 DOI: 10.3390/pharmaceutics16060795] [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: 05/20/2024] [Revised: 06/08/2024] [Accepted: 06/09/2024] [Indexed: 06/28/2024] Open
Abstract
Cancer immunotherapy has revolutionized oncology by harnessing the patient's immune system to target and eliminate cancer cells. However, immune checkpoint blockades (ICBs) face limitations such as low response rates, particularly in immunologically 'cold' tumors. Enhancing tumor immunogenicity through immunogenic cell death (ICD) inducers and advanced drug delivery systems represents a promising solution. This review discusses the development and application of various nanocarriers, including polymeric nanoparticles, liposomes, peptide-based nanoparticles, and inorganic nanoparticles, designed to deliver ICD inducers and ICBs effectively. These nanocarriers improve therapeutic outcomes by converting cold tumors into hot tumors, thus enhancing immune responses and reducing systemic toxicity. By focusing on single-nanoparticle systems that co-deliver both ICD inducers and ICBs, this review highlights their potential in achieving higher drug concentrations at tumor sites, improving pharmacokinetics and pharmacodynamics, and facilitating clinical translation. Future research should aim to optimize these nanocarrier systems for better in vivo performance and clinical applications, ultimately advancing cancer immunotherapy.
Collapse
Affiliation(s)
- Yujeong Moon
- Department of Bioengineering, Korea University, Seoul 02841, Republic of Korea;
- Medicinal Materials Research Center, Biomedical Research Division, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Hanhee Cho
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul 03760, Republic of Korea;
| | - Kwangmeyung Kim
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul 03760, Republic of Korea;
| |
Collapse
|
17
|
Xu Y, Bai Z, Lan T, Fu C, Cheng P. CD44 and its implication in neoplastic diseases. MedComm (Beijing) 2024; 5:e554. [PMID: 38783892 PMCID: PMC11112461 DOI: 10.1002/mco2.554] [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: 08/01/2023] [Revised: 03/20/2024] [Accepted: 04/01/2024] [Indexed: 05/25/2024] Open
Abstract
CD44, a nonkinase single span transmembrane glycoprotein, is a major cell surface receptor for many other extracellular matrix components as well as classic markers of cancer stem cells and immune cells. Through alternative splicing of CD44 gene, CD44 is divided into two isoforms, the standard isoform of CD44 (CD44s) and the variant isoform of CD44 (CD44v). Different isoforms of CD44 participate in regulating various signaling pathways, modulating cancer proliferation, invasion, metastasis, and drug resistance, with its aberrant expression and dysregulation contributing to tumor initiation and progression. However, CD44s and CD44v play overlapping or contradictory roles in tumor initiation and progression, which is not fully understood. Herein, we discuss the present understanding of the functional and structural roles of CD44 in the pathogenic mechanism of multiple cancers. The regulation functions of CD44 in cancers-associated signaling pathways is summarized. Moreover, we provide an overview of the anticancer therapeutic strategies that targeting CD44 and preclinical and clinical trials evaluating the pharmacokinetics, efficacy, and drug-related toxicity about CD44-targeted therapies. This review provides up-to-date information about the roles of CD44 in neoplastic diseases, which may open new perspectives in the field of cancer treatment through targeting CD44.
Collapse
Affiliation(s)
- Yiming Xu
- Department of BiotherapyLaboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan UniversityChengduSichuanChina
| | - Ziyi Bai
- Department of BiotherapyLaboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan UniversityChengduSichuanChina
| | - Tianxia Lan
- Department of BiotherapyLaboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan UniversityChengduSichuanChina
| | - Chenying Fu
- Laboratory of Aging and Geriatric Medicine, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan UniversityChengduSichuanChina
| | - Ping Cheng
- Department of Biotherapy, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan UniversityChengduChina
| |
Collapse
|
18
|
Salman DM, Mohammad TAM. siRNA-based therapy for gastric adenocarcinoma: what's next step? Pathol Res Pract 2024; 258:155328. [PMID: 38744002 DOI: 10.1016/j.prp.2024.155328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 04/17/2024] [Accepted: 04/22/2024] [Indexed: 05/16/2024]
Abstract
Gastric cancer continues to have a high death rate despite advancements in their diagnosis and treatment. Novel treatment techniques are thus desperately needed. This is where double-stranded RNA molecules known as small interfering RNA (siRNA), which may selectively target the mRNA of disease-causing genes, may find use in medicine. For siRNAs to function properly in the human body, they must be shielded from deterioration. Furthermore, in order to maintain organ function, they must only target the tumor and spare normal tissue. siRNAs have been designed using clever delivery mechanisms including polymers and lipids to achieve these objectives. Although siRNA protection is not hard to acquire, it is still challenging to target cancer cells with them. Here, we first discuss the basic characteristics of gastric cancer before describing the properties of siRNA and typical delivery methods created specifically for gastric tumors. Lastly, we provide a succinct overview of research using siRNAs to treat gastric tumors.
Collapse
Affiliation(s)
- Dyar Mudhafar Salman
- Department of Clinical Pharmacy, College of Pharmacy, Hawler Medical University, Erbil, Kurdistan Region, Iraq; Faculty of Pharmacy, Tishk International University, Erbil, Kurdistan Region, Iraq
| | - Talar Ahmad Merza Mohammad
- Department of Clinical Pharmacy, College of Pharmacy, Hawler Medical University, Erbil, Kurdistan Region, Iraq; Pharmacy department, School of Medicine, University of Kurdistan Hewlêr (UKH), Erbil, Kurdistan Region, Iraq.
| |
Collapse
|
19
|
Choi Y, Seok SH, Yoon HY, Ryu JH, Kwon IC. Advancing cancer immunotherapy through siRNA-based gene silencing for immune checkpoint blockade. Adv Drug Deliv Rev 2024; 209:115306. [PMID: 38626859 DOI: 10.1016/j.addr.2024.115306] [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: 12/07/2023] [Revised: 04/01/2024] [Accepted: 04/09/2024] [Indexed: 05/23/2024]
Abstract
Cancer immunotherapy represents a revolutionary strategy, leveraging the patient's immune system to inhibit tumor growth and alleviate the immunosuppressive effects of the tumor microenvironment (TME). The recent emergence of immune checkpoint blockade (ICB) therapies, particularly following the first approval of cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) inhibitors like ipilimumab, has led to significant growth in cancer immunotherapy. The extensive explorations on diverse immune checkpoint antibodies have broadened the therapeutic scope for various malignancies. However, the clinical response to these antibody-based ICB therapies remains limited, with less than 15% responsiveness and notable adverse effects in some patients. This review introduces the emerging strategies to overcome current limitations of antibody-based ICB therapies, mainly focusing on the development of small interfering ribonucleic acid (siRNA)-based ICB therapies and innovative delivery systems. We firstly highlight the diverse target immune checkpoint genes for siRNA-based ICB therapies, incorporating silencing of multiple genes to boost anti-tumor immune responses. Subsequently, we discuss improvements in siRNA delivery systems, enhanced by various nanocarriers, aimed at overcoming siRNA's clinical challenges such as vulnerability to enzymatic degradation, inadequate pharmacokinetics, and possible unintended target interactions. Additionally, the review presents various combination therapies that integrate chemotherapy, phototherapy, stimulatory checkpoints, ICB antibodies, and cancer vaccines. The important point is that when used in combination with siRNA-based ICB therapy, the synergistic effect of traditional therapies is strengthened, improving host immune surveillance and therapeutic outcomes. Conclusively, we discuss the insights into innovative and effective cancer immunotherapeutic strategies based on RNA interference (RNAi) technology utilizing siRNA and nanocarriers as a novel approach in ICB cancer immunotherapy.
Collapse
Affiliation(s)
- Youngjin Choi
- Medicinal Materials Research Center, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Su Hyun Seok
- Medicinal Materials Research Center, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Hong Yeol Yoon
- Medicinal Materials Research Center, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea; Division of Bio-Medical Science &Technology, KIST School, University of Science and Technology, Seoul 02792, Republic of Korea
| | - Ju Hee Ryu
- Medicinal Materials Research Center, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea.
| | - Ick Chan Kwon
- Medicinal Materials Research Center, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea; KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea.
| |
Collapse
|
20
|
Gatto MS, Johnson MP, Najahi-Missaoui W. Targeted Liposomal Drug Delivery: Overview of the Current Applications and Challenges. Life (Basel) 2024; 14:672. [PMID: 38929656 PMCID: PMC11204409 DOI: 10.3390/life14060672] [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: 03/30/2024] [Revised: 05/14/2024] [Accepted: 05/20/2024] [Indexed: 06/28/2024] Open
Abstract
In drug development, it is not uncommon that an active substance exhibits efficacy in vitro but lacks the ability to specifically reach its target in vivo. As a result, targeted drug delivery has become a primary focus in the pharmaceutical sciences. Since the approval of Doxil® in 1995, liposomes have emerged as a leading nanoparticle in targeted drug delivery. Their low immunogenicity, high versatility, and well-documented efficacy have led to their clinical use against a wide variety of diseases. That being said, every disease is accompanied by a unique set of physiological conditions, and each liposomal product must be formulated with this consideration. There are a multitude of different targeting techniques for liposomes that can be employed depending on the application. Passive techniques such as PEGylation or the enhanced permeation and retention effect can improve general pharmacokinetics, while active techniques such as conjugating targeting molecules to the liposome surface may bring even further specificity. This review aims to summarize the current strategies for targeted liposomes in the treatment of diseases.
Collapse
Affiliation(s)
| | | | - Wided Najahi-Missaoui
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, GA 30602, USA; (M.S.G.); (M.P.J.)
| |
Collapse
|
21
|
Lang X, Wang X, Han M, Guo Y. Nanoparticle-Mediated Synergistic Chemoimmunotherapy for Cancer Treatment. Int J Nanomedicine 2024; 19:4533-4568. [PMID: 38799699 PMCID: PMC11127654 DOI: 10.2147/ijn.s455213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Accepted: 05/07/2024] [Indexed: 05/29/2024] Open
Abstract
Until now, there has been a lack of effective strategies for cancer treatment. Immunotherapy has high potential in treating several cancers but its efficacy is limited as a monotherapy. Chemoimmunotherapy (CIT) holds promise to be widely used in cancer treatment. Therefore, identifying their involvement and potential synergy in CIT approaches is decisive. Nano-based drug delivery systems (NDDSs) are ideal delivery systems because they can simultaneously target immune cells and cancer cells, promoting drug accumulation, and reducing the toxicity of the drug. In this review, we first introduce five current immunotherapies, including immune checkpoint blocking (ICB), adoptive cell transfer therapy (ACT), cancer vaccines, oncolytic virus therapy (OVT) and cytokine therapy. Subsequently, the immunomodulatory effects of chemotherapy by inducing immunogenic cell death (ICD), promoting tumor killer cell infiltration, down-regulating immunosuppressive cells, and inhibiting immune checkpoints have been described. Finally, the NDDSs-mediated collaborative drug delivery systems have been introduced in detail, and the development of NDDSs-mediated CIT nanoparticles has been prospected.
Collapse
Affiliation(s)
- Xiaoxue Lang
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, People’s Republic of China
| | - Xiangtao Wang
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, People’s Republic of China
| | - Meihua Han
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, People’s Republic of China
| | - Yifei Guo
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, People’s Republic of China
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, People’s Republic of China
- Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine Prescription, Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing, People’s Republic of China
| |
Collapse
|
22
|
Hu M, Bao J, Zhang Y, Wang L, Zhang Y, Zhang J, Tang J, Zou Q. Supramolecular Nanoparticles of Histone and Hyaluronic Acid for Co-Delivery of siRNA and Photosensitizer In Vitro. Int J Mol Sci 2024; 25:5424. [PMID: 38791462 PMCID: PMC11121309 DOI: 10.3390/ijms25105424] [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: 04/15/2024] [Revised: 05/07/2024] [Accepted: 05/14/2024] [Indexed: 05/26/2024] Open
Abstract
Small interfering RNA (siRNA) has significant potential as a treatment for cancer by targeting specific genes or molecular pathways involved in cancer development and progression. The addition of siRNA to other therapeutic strategies, like photodynamic therapy (PDT), can enhance the anticancer effects, providing synergistic benefits. Nevertheless, the effective delivery of siRNA into target cells remains an obstacle in cancer therapy. Herein, supramolecular nanoparticles were fabricated via the co-assembly of natural histone and hyaluronic acid for the co-delivery of HMGB1-siRNA and the photosensitizer chlorin e6 (Ce6) into the MCF-7 cell. The produced siRNA-Ce6 nanoparticles (siRNA-Ce6 NPs) have a spherical morphology and exhibit uniform distribution. In vitro experiments demonstrate that the siRNA-Ce6 NPs display good biocompatibility, enhanced cellular uptake, and improved cytotoxicity. These outcomes indicate that the nanoparticles constructed by the co-assembly of histone and hyaluronic acid hold enormous promise as a means of siRNA and photosensitizer co-delivery towards synergetic therapy.
Collapse
Affiliation(s)
- Minxing Hu
- Research and Industrialization of New Drug Release Technology Joint Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Jianwei Bao
- Research and Industrialization of New Drug Release Technology Joint Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Yuanmei Zhang
- Research and Industrialization of New Drug Release Technology Joint Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Lele Wang
- Research and Industrialization of New Drug Release Technology Joint Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Ya Zhang
- Research and Industrialization of New Drug Release Technology Joint Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Jiaxin Zhang
- Research and Industrialization of New Drug Release Technology Joint Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Jihui Tang
- Research and Industrialization of New Drug Release Technology Joint Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Qianli Zou
- Research and Industrialization of New Drug Release Technology Joint Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei 230032, China
- Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei 230000, China
| |
Collapse
|
23
|
Nele V, Campani V, Alia Moosavian S, De Rosa G. Lipid nanoparticles for RNA delivery: Self-assembling vs driven-assembling strategies. Adv Drug Deliv Rev 2024; 208:115291. [PMID: 38514018 DOI: 10.1016/j.addr.2024.115291] [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: 12/22/2023] [Revised: 02/20/2024] [Accepted: 03/14/2024] [Indexed: 03/23/2024]
Abstract
Among non-viral vectors, lipid nanovectors are considered the gold standard for the delivery of RNA therapeutics. The success of lipid nanoparticles for RNA delivery, with three products approved for human use, has stimulated further investigation into RNA therapeutics for different pathologies. This requires decoding the pathological intracellular processes and tailoring the delivery system to the target tissue and cells. The complexity of the lipid nanovectors morphology originates from the assembling of the lipidic components, which can be elicited by various methods able to drive the formation of nanoparticles with the desired organization. In other cases, pre-formed nanoparticles can be mixed with RNA to induce self-assembly and structural reorganization into RNA-loaded nanoparticles. In this review, the most relevant lipid nanovectors and their potentialities for RNA delivery are described on the basis of the assembling mechanism and of the particle architecture.
Collapse
Affiliation(s)
- Valeria Nele
- Department of Pharmacy, University of Naples Federico II, Via D. Montesano, 49 80131 Naples, Italy
| | - Virginia Campani
- Department of Pharmacy, University of Naples Federico II, Via D. Montesano, 49 80131 Naples, Italy
| | - Seyedeh Alia Moosavian
- Department of Pharmacy, University of Naples Federico II, Via D. Montesano, 49 80131 Naples, Italy
| | - Giuseppe De Rosa
- Department of Pharmacy, University of Naples Federico II, Via D. Montesano, 49 80131 Naples, Italy.
| |
Collapse
|
24
|
Wang L, He S, Liu R, Xue Y, Quan Y, Shi R, Yang X, Lin Q, Sun X, Zhang Z, Zhang L. A pH/ROS dual-responsive system for effective chemoimmunotherapy against melanoma via remodeling tumor immune microenvironment. Acta Pharm Sin B 2024; 14:2263-2280. [PMID: 38799639 PMCID: PMC11119573 DOI: 10.1016/j.apsb.2023.12.001] [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: 09/24/2023] [Revised: 12/05/2023] [Accepted: 12/06/2023] [Indexed: 05/29/2024] Open
Abstract
Chemotherapeutics can induce immunogenic cell death (ICD) in tumor cells, offering new possibilities for cancer therapy. However, the efficiency of the immune response generated is insufficient due to the inhibitory nature of the tumor microenvironment (TME). Here, we developed a pH/reactive oxygen species (ROS) dual-response system to enhance chemoimmunotherapy for melanoma. The system productively accumulated in tumors by specific binding of phenylboronic acid (PBA) to sialic acids (SA). The nanoparticles (NPs) rapidly swelled and released quercetin (QUE) and doxorubicin (DOX) upon the stimulation of tumor microenvironment (TME). The in vitro and in vivo results consistently demonstrated that the NPs improved anti-tumor efficacy and prolonged survival of mice, significantly enhancing the effects of the combination. Our study revealed DOX was an ICD inducer, stimulating immune responses and promoting maturation of dendritic cells (DCs). Additionally, QUE served as a TME regulator by inhibiting the cyclooxygenase-2 (COX2)-prostaglandin E2 (PGE2) axis, which influenced various immune cells, including increasing cytotoxic T cells (CLTs) infiltration, promoting M1 macrophage polarization, and reducing regulatory T cells (Tregs) infiltration. The combination synergistically facilitated chemoimmunotherapy efficacy by remodeling the immunosuppressive microenvironment. This work presents a promising strategy to increase anti-tumor efficiency of chemotherapeutic agents.
Collapse
Affiliation(s)
- Leilei Wang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Shanshan He
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Rong Liu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Yuan Xue
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Yuan Quan
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Rongying Shi
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Xueying Yang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Qing Lin
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Xun Sun
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Zhirong Zhang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Ling Zhang
- Med-X Center for Materials, College of Polymer Science and Engineering, Sichuan University, Chengdu 610065, China
| |
Collapse
|
25
|
Cho H, Kim K. Multi-functional nanomedicines for combinational cancer immunotherapy that transform cold tumors to hot tumors. Expert Opin Drug Deliv 2024; 21:627-638. [PMID: 38682272 DOI: 10.1080/17425247.2024.2348656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 04/24/2024] [Indexed: 05/01/2024]
Abstract
INTRODUCTION Currently, cancer immunotherapy is widely used as a groundbreaking method that can completely cure advanced cancers. However, this new immunotherapy has the challenge of low patient response, which is often due to many patients' tumors having an immunosuppressive environment, known as cold tumors. AREAS COVERED This review aims to introduce various nanomedicine-derived combinational cancer immunotherapy that can transform cold tumor into hot tumors. Initially, we discuss new technologies for combinational immunotherapy based on multifunctional nanomedicines that can deliver combinational immunogenic cell death (ICD) inducers, immune checkpoint blockades (ICBs) and immune modulators (IMs) to targeted tumor tissues at the same time. Ultimately, we highlight how multifunctional nanomedicines for combinational cancer immunotherapy can be used to transform cold tumor into hot tumors against advanced cancers. EXPERT OPINION Nanomedicine-derived combinational cancer immunotherapy for delivering multiple ICD inducers, ICBs, and IMs at the same time is recognized as a new potential technology that can activate tumor immunity and simultaneously increase the therapeutic efficacy of immune cells that can transform effectively the cold tumors into hot tumors. Finally, nanomedicine-derived combinational cancer immunotherapy can solve the serious problems of low therapeutic efficacy that occurs when treating single drug or simple combinational drugs in cancer immunotherapy.
Collapse
Affiliation(s)
- Hanhee Cho
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Woman's University, Seoul, Republic of Korea
| | | |
Collapse
|
26
|
Camorani S, Caliendo A, Morrone E, Agnello L, Martini M, Cantile M, Cerrone M, Zannetti A, La Deda M, Fedele M, Ricciardi L, Cerchia L. Bispecific aptamer-decorated and light-triggered nanoparticles targeting tumor and stromal cells in breast cancer derived organoids: implications for precision phototherapies. J Exp Clin Cancer Res 2024; 43:92. [PMID: 38532439 PMCID: PMC10964525 DOI: 10.1186/s13046-024-03014-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 03/17/2024] [Indexed: 03/28/2024] Open
Abstract
BACKGROUND Based on the established role of cancer-stroma cross-talk in tumor growth, progression and chemoresistance, targeting interactions between tumor cells and their stroma provides new therapeutic approaches. Dual-targeted nanotherapeutics selectively acting on both tumor and stromal cells may overcome the limits of tumor cell-targeting single-ligand nanomedicine due to the complexity of the tumor microenvironment. METHODS Gold-core/silica-shell nanoparticles embedding a water-soluble iridium(III) complex as photosensitizer and luminescent probe (Iren-AuSiO2_COOH) were efficiently decorated with amino-terminated EGFR (CL4) and PDGFRβ (Gint4.T) aptamers (Iren-AuSiO2_Aptamer). The targeting specificity, and the synergistic photodynamic and photothermal effects of either single- and dual-aptamer-decorated nanoparticles have been assessed by confocal microscopy and cell viability assays, respectively, on different human cell types including mesenchymal subtype triple-negative breast cancer (MES-TNBC) MDA-MB-231 and BT-549 cell lines (both EGFR and PDGFRβ positive), luminal/HER2-positive breast cancer BT-474 and epidermoid carcinoma A431 cells (only EGFR positive) and adipose-derived mesenchymal stromal/stem cells (MSCs) (only PDGFRβ positive). Cells lacking expression of both receptors were used as negative controls. To take into account the tumor-stroma interplay, fluorescence imaging and cytotoxicity were evaluated in preclinical three-dimensional (3D) stroma-rich breast cancer models. RESULTS We show efficient capability of Iren-AuSiO2_Aptamer nanoplatforms to selectively enter into target cells, and kill them, through EGFR and/or PDGFRβ recognition. Importantly, by targeting EGFR+ tumor/PDGFRβ+ stromal cells in the entire tumor bulk, the dual-aptamer-engineered nanoparticles resulted more effective than unconjugated or single-aptamer-conjugated nanoparticles in either 3D spheroids cocultures of tumor cells and MSCs, and in breast cancer organoids derived from pathologically and molecularly well-characterized tumors. CONCLUSIONS Our study proposes smart, novel and safe multifunctional nanoplatforms simultaneously addressing cancer-stroma within the tumor microenvironment, which are: (i) actively delivered to the targeted cells through highly specific aptamers; (ii) localized by means of their luminescence, and (iii) activated via minimally invasive light, launching efficient tumor death, thus providing innovative precision therapeutics. Given the unique features, the proposed dual targeted nanoformulations may open a new door to precision cancer treatment.
Collapse
Affiliation(s)
- Simona Camorani
- Institute of Experimental Endocrinology and Oncology "Gaetano Salvatore", National Research Council, 80131, Naples, Italy
| | - Alessandra Caliendo
- Institute of Experimental Endocrinology and Oncology "Gaetano Salvatore", National Research Council, 80131, Naples, Italy
| | - Elena Morrone
- CNR-NANOTEC Institute of Nanotechnology, National Research Council, Rende, CS, Italy
- Department of Chemistry and Chemical Technologies, University of Calabria, Rende, CS, Italy
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Perugia, Italy
| | - Lisa Agnello
- Institute of Experimental Endocrinology and Oncology "Gaetano Salvatore", National Research Council, 80131, Naples, Italy
| | - Matteo Martini
- Institute of Light and Matter, UMR 5306, Claude Bernard University Lyon 1, Villeurbanne, France
| | - Monica Cantile
- Institutional Biobank-Scientific Directorate, National Cancer Institute INT-IRCCS Fondazione G. Pascale, 80131, Naples, Italy
| | - Margherita Cerrone
- Pathology Unit, National Cancer Institute INT-IRCCS Fondazione G. Pascale, 80131, Naples, Italy
| | - Antonella Zannetti
- Institute of Biostructures and Bioimaging, National Research Council, 80145, Naples, Italy
| | - Massimo La Deda
- CNR-NANOTEC Institute of Nanotechnology, National Research Council, Rende, CS, Italy
- Department of Chemistry and Chemical Technologies, University of Calabria, Rende, CS, Italy
| | - Monica Fedele
- Institute of Experimental Endocrinology and Oncology "Gaetano Salvatore", National Research Council, 80131, Naples, Italy
| | - Loredana Ricciardi
- CNR-NANOTEC Institute of Nanotechnology, National Research Council, Rende, CS, Italy.
| | - Laura Cerchia
- Institute of Experimental Endocrinology and Oncology "Gaetano Salvatore", National Research Council, 80131, Naples, Italy.
| |
Collapse
|
27
|
Hajimolaali M, Dorkoosh FA, Antimisiaris SG. Review of recent preclinical and clinical research on ligand-targeted liposomes as delivery systems in triple negative breast cancer therapy. J Liposome Res 2024:1-26. [PMID: 38520185 DOI: 10.1080/08982104.2024.2325963] [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: 11/03/2023] [Accepted: 02/27/2024] [Indexed: 03/25/2024]
Abstract
Triple-negative breast Cancer (TNBC) is one of the deadliest types, making up about 20% of all breast cancers. Chemotherapy is the traditional manner of progressed TNBC treatment; however, it has a short-term result with a high reversibility pace. The lack of targeted treatment limited and person-dependent treatment options for those suffering from TNBC cautions to be the worst type of cancer among breast cancer patients. Consequently, appropriate treatment for this disease is considered a major clinical challenge. Therefore, various treatment methods have been developed to treat TNBC, among which chemotherapy is the most common and well-known approach recently studied. Although effective methods are chemotherapies, they are often accompanied by critical limitations, especially the lack of specific functionality. These methods lead to systematic toxicity and, ultimately, the expansion of multidrug-resistant (MDR) cancer cells. Therefore, finding novel and efficient techniques to enhance the targeting of TNBC treatment is an essential requirement. Liposomes have demonstrated that they are an effective method for drug delivery; however, among a large number of liposome-based drug delivery systems annually developed, a small number have just received authorization for clinical application. The new approaches to using liposomes target their structure with various ligands to increase therapeutic efficiency and diminish undesired side effects on various body tissues. The current study describes the most recent strategies and research associated with functionalizing the liposomes' structure with different ligands as targeted drug carriers in treating TNBCs in preclinical and clinical stages.
Collapse
Affiliation(s)
- Mohammad Hajimolaali
- Department of Pharmacy, Laboratory of Pharmaceutical Technology, University of Patras, Patras, Greece
| | - Farid Abedin Dorkoosh
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
- Medical Biomaterial Research Center (MBRC), Tehran University of Medical Sciences, Tehran, Iran
| | - Sophia G Antimisiaris
- Department of Pharmacy, Laboratory of Pharmaceutical Technology, University of Patras, Patras, Greece
- Institute of Chemical Engineering, Foundation for Research and Technology Hellas, FORTH/ICEHT, Patras, Greece
| |
Collapse
|
28
|
Ashby G, Keng KE, Hayden CC, Stachowiak JC. A live cell imaging-based assay for tracking particle uptake by clathrin-mediated endocytosis. Methods Enzymol 2024; 700:413-454. [PMID: 38971609 DOI: 10.1016/bs.mie.2024.02.010] [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] [Indexed: 07/08/2024]
Abstract
A popular strategy for therapeutic delivery to cells and tissues is to encapsulate therapeutics inside particles that cells internalize via endocytosis. The efficacy of particle uptake by endocytosis is often studied in bulk using flow cytometry and Western blot analysis and confirmed using confocal microscopy. However, these techniques do not reveal the detailed dynamics of particle internalization and how the inherent heterogeneity of many types of particles may impact their endocytic uptake. Toward addressing these gaps, here we present a live-cell imaging-based method that utilizes total internal reflection fluorescence microscopy to track the uptake of a large ensemble of individual particles in parallel, as they interact with the cellular endocytic machinery. To analyze the resulting data, we employ an open-source tracking algorithm in combination with custom data filters. This analysis reveals the dynamic interactions between particles and endocytic structures, which determine the probability of particle uptake. In particular, our approach can be used to examine how variations in the physical properties of particles (size, targeting, rigidity), as well as heterogeneity within the particle population, impact endocytic uptake. These data impact the design of particles toward more selective and efficient delivery of therapeutics to cells.
Collapse
Affiliation(s)
- Grant Ashby
- Department of Biomedical Engineering, The University of Texas at Austin
| | - Kayla E Keng
- Department of Biomedical Engineering, The University of Texas at Austin
| | - Carl C Hayden
- Department of Biomedical Engineering, The University of Texas at Austin
| | - Jeanne C Stachowiak
- Department of Biomedical Engineering, The University of Texas at Austin; Department of Chemical Engineering, The University of Texas at Austin.
| |
Collapse
|
29
|
Gong X, Zheng C, Cai Y, Zhang W, Zhu B, Rong R, Kong Y, Zhang Y, Wang J, Li Y, Zhang P. Adenosine-modulating synthetic high-density lipoprotein for chemoimmunotherapy of triple-negative breast cancer. J Control Release 2024; 367:637-648. [PMID: 38295994 DOI: 10.1016/j.jconrel.2024.01.064] [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: 10/19/2023] [Revised: 01/05/2024] [Accepted: 01/28/2024] [Indexed: 02/13/2024]
Abstract
Adenosine (ADO) is a common chemotherapy-associated immune checkpoint that hinders anti-tumor immunity-mediated efficacy of chemotherapy. Herein, we created a synthetic high-density lipoprotein (sHDL) by co-assembly of a doxorubicin (DOX)-apolipoprotein A1 mimetic peptide conjugate, PSB-603 (an A2BR inhibitor), phospholipid, and cholesterol oleate with a microfluidic-based method. The obtained DP-sHDL showed a self-promoted drug delivery to cancer cells via remodeling tumor microenvironment. DP-sHDL could trigger the release of ATP from cancer cells and inhibit its conversion into ADO. Consequently, DP-sHDL, while increasing immunogenic cell death, reduced intratumoral ADO levels by 58%. This treatment improved both the density and activity of CD8+ T cells as well as NK cells and relieved the immunosuppressive microenvironment, and led to a substantial inhibition of 4T1 tumor growth, thereby extending the survival of mice. The efficacy of DP-sHDL could be further improved when used in combination with immune checkpoint blockade therapy. We envision that this platform provides a simple yet promising strategy to enhance anti-tumor response of chemotherapy by relieving treatment-associated immunosuppression.
Collapse
Affiliation(s)
- Xiang Gong
- National Advanced Medical Engineering Research Center, China State Institute of Pharmaceutical Industry, Shanghai 201203, China; State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Chao Zheng
- National Advanced Medical Engineering Research Center, China State Institute of Pharmaceutical Industry, Shanghai 201203, China; State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Ying Cai
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wen Zhang
- National Advanced Medical Engineering Research Center, China State Institute of Pharmaceutical Industry, Shanghai 201203, China; State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Binyu Zhu
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Rong Rong
- Yantai Key Laboratory of Nanomedicine & Advanced Preparations, Yantai Institute of Materia Medica, Yantai 264000, China
| | - Ying Kong
- Yantai Key Laboratory of Nanomedicine & Advanced Preparations, Yantai Institute of Materia Medica, Yantai 264000, China
| | - Yuan Zhang
- Department of Pulmonary and Critical Care Medicine, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
| | - Jian Wang
- National Advanced Medical Engineering Research Center, China State Institute of Pharmaceutical Industry, Shanghai 201203, China.
| | - Yaping Li
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; Yantai Key Laboratory of Nanomedicine & Advanced Preparations, Yantai Institute of Materia Medica, Yantai 264000, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Pengcheng Zhang
- School of Biomedical Engineering & State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University, Shanghai 201210, China.
| |
Collapse
|
30
|
Li Y, Liu W, Xu H, Zhou Y, Xie W, Guo Y, Liao Z, Jiang X, Liu J, Ren C. Aptamers combined with immune checkpoints for cancer detection and targeted therapy: A review. Int J Biol Macromol 2024; 262:130032. [PMID: 38342267 DOI: 10.1016/j.ijbiomac.2024.130032] [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: 08/03/2023] [Revised: 02/02/2024] [Accepted: 02/05/2024] [Indexed: 02/13/2024]
Abstract
In recent years, remarkable strides have been made in the field of immunotherapy, which has emerged as a standard treatment for many cancers. As a kind of immunotherapy drug, monoclonal antibodies employed in immune checkpoint therapy have proven beneficial for patients with diverse cancer types. However, owing to the extensive heterogeneity of clinical responses and the complexity and variability of the immune system and tumor microenvironment (TME), accurately predicting its efficacy remains a challenge. Recent advances in aptamers provide a promising approach for monitoring alterations within the immune system and TME, thereby facilitating targeted immunotherapy, particularly focused on immune checkpoint blockade, with enhanced antitumor efficiency. Aptamers have been widely used in tumor cell detection, biosensors, drug discovery, and biomarker screening due to their high specificity and high affinity with their targets. This review aims to comprehensively examine the research status and progress of aptamers in cancer diagnosis and immunotherapy, with a specific emphasis on those related to immune checkpoints. Additionally, we will discuss the future research directions and potential therapeutic targets for aptamer-based immune checkpoint therapy, aiming to provide a theoretical basis for targeting immunotherapy molecules and blocking tumor immune escape.
Collapse
Affiliation(s)
- Yihan Li
- NHC Key Laboratory of Carcinogenesis, Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China
| | - Weidong Liu
- NHC Key Laboratory of Carcinogenesis, Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China
| | - Hongjuan Xu
- NHC Key Laboratory of Biological Nanotechnology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Yao Zhou
- NHC Key Laboratory of Carcinogenesis, Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China
| | - Wen Xie
- NHC Key Laboratory of Carcinogenesis, Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China
| | - Youwei Guo
- NHC Key Laboratory of Carcinogenesis, Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China
| | - Ziling Liao
- NHC Key Laboratory of Carcinogenesis, Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China
| | - Xingjun Jiang
- NHC Key Laboratory of Carcinogenesis, Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Jie Liu
- Department of Critical care medicine, Hainan Hospital of Chinese PLA General Hosptial; project supported by Hainan Province Clinical Medical Center, China.
| | - Caiping Ren
- NHC Key Laboratory of Carcinogenesis, Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China; Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, China.
| |
Collapse
|
31
|
Xu L, Cao Y, Xu Y, Li R, Xu X. Redox-Responsive Polymeric Nanoparticle for Nucleic Acid Delivery and Cancer Therapy: Progress, Opportunities, and Challenges. Macromol Biosci 2024; 24:e2300238. [PMID: 37573033 DOI: 10.1002/mabi.202300238] [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: 05/25/2023] [Revised: 07/25/2023] [Indexed: 08/14/2023]
Abstract
Cancer development and progression of cancer are closely associated with the activation of oncogenes and loss of tumor suppressor genes. Nucleic acid drugs (e.g., siRNA, mRNA, and DNA) are widely used for cancer therapy due to their specific ability to regulate the expression of any cancer-associated genes. However, nucleic acid drugs are negatively charged biomacromolecules that are susceptible to serum nucleases and cannot cross cell membrane. Therefore, specific delivery tools are required to facilitate the intracellular delivery of nucleic acid drugs. In the past few decades, a variety of nanoparticles (NPs) are designed and developed for nucleic acid delivery and cancer therapy. In particular, the polymeric NPs in response to the abnormal redox status in cancer cells have garnered much more attention as their potential in redox-triggered nanostructure dissociation and rapid intracellular release of nucleic acid drugs. In this review, the important genes or signaling pathways regulating the abnormal redox status in cancer cells are briefly introduced and the recent development of redox-responsive NPs for nucleic acid delivery and cancer therapy is systemically summarized. The future development of NPs-mediated nucleic acid delivery and their challenges in clinical translation are also discussed.
Collapse
Affiliation(s)
- Lei Xu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, P. R. China
- Guangzhou Key Laboratory of Medical Nanomaterials, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, P. R. China
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan, 528200, P. R. China
| | - Yuan Cao
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, P. R. China
- Guangzhou Key Laboratory of Medical Nanomaterials, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, P. R. China
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan, 528200, P. R. China
| | - Ya Xu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, P. R. China
- Guangzhou Key Laboratory of Medical Nanomaterials, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, P. R. China
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan, 528200, P. R. China
| | - Rong Li
- The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, P. R. China
| | - Xiaoding Xu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, P. R. China
- Guangzhou Key Laboratory of Medical Nanomaterials, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, P. R. China
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan, 528200, P. R. China
- The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, P. R. China
| |
Collapse
|
32
|
Paun RA, Jurchuk S, Tabrizian M. A landscape of recent advances in lipid nanoparticles and their translational potential for the treatment of solid tumors. Bioeng Transl Med 2024; 9:e10601. [PMID: 38435821 PMCID: PMC10905562 DOI: 10.1002/btm2.10601] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 08/30/2023] [Accepted: 09/05/2023] [Indexed: 03/05/2024] Open
Abstract
Lipid nanoparticles (LNPs) are biocompatible drug delivery systems that have found numerous applications in medicine. Their versatile nature enables the encapsulation and targeting of various types of medically relevant molecular cargo, including oligonucleotides, proteins, and small molecules for the treatment of diseases, such as cancer. Cancers that form solid tumors are particularly relevant for LNP-based therapeutics due to the enhanced permeation and retention effect that allows nanoparticles to accumulate within the tumor tissue. Additionally, LNPs can be formulated for both locoregional and systemic delivery depending on the tumor type and stage. To date, LNPs have been used extensively in the clinic to reduce systemic toxicity and improve outcomes in cancer patients by encapsulating chemotherapeutic drugs. Next-generation lipid nanoparticles are currently being developed to expand their use in gene therapy and immunotherapy, as well as to enable the co-encapsulation of multiple drugs in a single system. Other developments include the design of targeted LNPs to specific cells and tissues, and triggerable release systems to control cargo delivery at the tumor site. This review paper highlights recent developments in LNP drug delivery formulations and focuses on the treatment of solid tumors, while also discussing some of their current translational limitations and potential opportunities in the field.
Collapse
Affiliation(s)
- Radu A. Paun
- Department of Biomedical Engineering, Faculty of Medicine and Health SciencesMcGill UniversityMontrealQuebecCanada
| | - Sarah Jurchuk
- Department of Biomedical Engineering, Faculty of Medicine and Health SciencesMcGill UniversityMontrealQuebecCanada
| | - Maryam Tabrizian
- Department of Biomedical Engineering, Faculty of Medicine and Health SciencesMcGill UniversityMontrealQuebecCanada
- Faculty of Dentistry and Oral Health SciencesMcGill UniversityMontrealQuebecCanada
| |
Collapse
|
33
|
Ou Y, Wang M, Xu Q, Sun B, Jia Y. Small molecule agents for triple negative breast cancer: Current status and future prospects. Transl Oncol 2024; 41:101893. [PMID: 38290250 PMCID: PMC10840364 DOI: 10.1016/j.tranon.2024.101893] [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: 11/16/2023] [Revised: 01/20/2024] [Accepted: 01/23/2024] [Indexed: 02/01/2024] Open
Abstract
Triple-negative breast cancer (TNBC) is a subtype of breast cancer with poor prognosis. The number of cases increased by 2.26 million in 2020, making it the most commonly diagnosed cancer type in the world. TNBCs lack hormone receptor (HR) and human epidermal growth factor 2 (HER2), which limits treatment options. Currently, paclitaxel-based drugs combined with other chemotherapeutics remain the main treatment for TNBC. There is currently no consensus on the best therapeutic regimen for TNBC. However, there have been successful clinical trials exploring large-molecule monoclonal antibodies, small-molecule targeted drugs, and novel antibody-drug conjugate (ADC). Although monoclonal antibodies have produced clinical success, their large molecular weight can limit therapeutic benefits. It is worth noting that in the past 30 years, the FDA has approved small molecule drugs for HER2-positive breast cancers. The lack of effective targets and the occurrence of drug resistance pose significant challenges in the treatment of TNBC. To improve the prognosis of TNBC, it is crucial to search for effective targets and to overcome drug resistance. This review examines the clinical efficacy, adverse effects, resistance mechanisms, and potential solutions of targeted small molecule drugs in both monotherapies and combination therapies. New therapeutic targets, including nuclear export protein 1 (XPO1) and hedgehog (Hh), are emerging as potential options for researchers and become integrated into clinical trials for TNBC. Additionally, there is growing interest in the potential of targeted protein degradation chimeras (PROTACs), degraders of rogue proteins, as a future therapy direction. This review provides potentially valuable insights with clinical implications.
Collapse
Affiliation(s)
- Yan Ou
- The First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Mengchao Wang
- The First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Qian Xu
- The First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Binxu Sun
- The First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Yingjie Jia
- The First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China.
| |
Collapse
|
34
|
Gong Y, Zhang C, Li H, Yu X, Li Y, Liu Z, He R. Ferroptosis-Related lncRNA to Predict the Clinical Outcomes and Molecular Characteristics of Kidney Renal Papillary Cell Carcinoma. Curr Issues Mol Biol 2024; 46:1886-1903. [PMID: 38534739 DOI: 10.3390/cimb46030123] [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: 11/14/2023] [Revised: 12/21/2023] [Accepted: 02/27/2024] [Indexed: 03/28/2024] Open
Abstract
Kidney renal papillary cell carcinoma (KIRP) is a highly heterogeneous type of kidney cancer, resulting in limited effective prognostic targets for KIRP patients. Long non-coding RNAs (lncRNAs) have emerged as crucial regulators in the regulation of ferroptosis and iron metabolism, making them potential targets for the treatment and prognosis of KIRP. In this study, we constructed a ferroptosis-related lncRNA risk score model (FRM) based on the TCGA-KIRP dataset, which represents a novel subtype of KIRP not previously reported. The model demonstrated promising diagnostic accuracy and holds potential for clinical translation. We observed significant differences in metabolic activities, immune microenvironment, mutation landscape, ferroptosis sensitivity, and drug sensitivity between different risk groups. The high-risk groups exhibit significantly higher fractions of cancer-associated fibroblasts (CAFs), hematopoietic stem cells (HSC), and pericytes. Drugs (IC50) analysis provided a range of medication options based on different FRM typing. Additionally, we employed single-cell transcriptomics to further analyze the impact of immune invasion on the occurrence and development of KIRP. Overall, we have developed an accurate prognostic model based on the expression patterns of ferroptosis-related lncRNAs for KIRP. This model has the potential to contribute to the evaluation of patient prognosis, molecular characteristics, and treatment modalities, and can be further translated into clinical applications.
Collapse
Affiliation(s)
- Yubo Gong
- School of Life Science and Technology, Wuhan Polytechnic University, Wuhan 430023, China
| | - Chenchen Zhang
- School of Life Science and Technology, Wuhan Polytechnic University, Wuhan 430023, China
| | - Hao Li
- School of Life Science and Technology, Wuhan Polytechnic University, Wuhan 430023, China
| | - Xiaojie Yu
- School of Life Science and Technology, Wuhan Polytechnic University, Wuhan 430023, China
| | - Yuejia Li
- School of Life Science and Technology, Wuhan Polytechnic University, Wuhan 430023, China
| | - Zhiguo Liu
- School of Life Science and Technology, Wuhan Polytechnic University, Wuhan 430023, China
| | - Ruyi He
- School of Life Science and Technology, Wuhan Polytechnic University, Wuhan 430023, China
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan 430062, China
| |
Collapse
|
35
|
Sesarman A, Luput L, Rauca VF, Patras L, Licarete E, Meszaros MS, Dume BR, Negrea G, Toma VA, Muntean D, Porfire A, Banciu M. Targeting of M2 macrophages with IL-13-functionalized liposomal prednisolone inhibits melanoma angiogenesis in vivo. J Liposome Res 2024:1-12. [PMID: 38379249 DOI: 10.1080/08982104.2024.2315452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 01/31/2024] [Indexed: 02/22/2024]
Abstract
The intricate cooperation between cancer cells and nontumor stromal cells within melanoma microenvironment (MME) enables tumor progression and metastasis. We previously demonstrated that the interplay between tumor-associated macrophages (TAMs) and melanoma cells can be disrupted by using long-circulating liposomes (LCLs) encapsulating prednisolone phosphate (PLP) (LCL-PLP) that inhibited tumor angiogenesis coordinated by TAMs. In this study, our goal was to improve LCL specificity for protumor macrophages (M2-like (i.e., TAMs) macrophages) and to induce a more precise accumulation at tumor site by loading PLP into IL-13-conjugated liposomes (IL-13-LCL-PLP), since IL-13 receptor is overexpressed in this type of macrophages. The IL-13-LCL-PLP liposomal formulation was obtained by covalent attachment of thiolated IL-13 to maleimide-functionalized LCL-PLP. C57BL/6 mice bearing B16.F10 s.c melanoma tumors were used to investigate the antitumor action of LCL-PLP and IL-13-LCL-PLP. Our results showed that IL-13-LCL-PLP formulation remained stable in biological fluids after 24h and it was preferentially taken up by M2 polarized macrophages. IL-13-LCL-PLP induced strong tumor growth inhibition compared to nonfunctionalized LCL-PLP at the same dose, by altering TAMs-mediated angiogenesis and oxidative stress, limiting resistance to apoptosis and invasive features in MME. These findings suggest IL-13-LCL-PLP might become a promising delivery platform for chemotherapeutic agents in melanoma.
Collapse
Affiliation(s)
- Alina Sesarman
- Department of Molecular Biology and Biotechnology, and Center of Systems Biology, Biodiversity and Bioresources, Faculty of Biology and Geology, Babes-Bolyai University, Cluj-Napoca, Romania
| | - Lavinia Luput
- Department of Molecular Biology and Biotechnology, and Center of Systems Biology, Biodiversity and Bioresources, Faculty of Biology and Geology, Babes-Bolyai University, Cluj-Napoca, Romania
| | - Valentin-Florian Rauca
- Department of Molecular Biology and Biotechnology, and Center of Systems Biology, Biodiversity and Bioresources, Faculty of Biology and Geology, Babes-Bolyai University, Cluj-Napoca, Romania
- Department of Dermatology and Allergology, School of Medicine, Technical University of Munich, Munich, Germany
| | - Laura Patras
- Department of Molecular Biology and Biotechnology, and Center of Systems Biology, Biodiversity and Bioresources, Faculty of Biology and Geology, Babes-Bolyai University, Cluj-Napoca, Romania
| | - Emilia Licarete
- Department of Molecular Biology and Biotechnology, and Center of Systems Biology, Biodiversity and Bioresources, Faculty of Biology and Geology, Babes-Bolyai University, Cluj-Napoca, Romania
- Molecular Biology Centre, Institute for Interdisciplinary Research in Bio-Nano-Sciences of Babes-Bolyai University, Cluj-Napoca, Romania
| | - Marta-Szilvia Meszaros
- Department of Molecular Biology and Biotechnology, and Center of Systems Biology, Biodiversity and Bioresources, Faculty of Biology and Geology, Babes-Bolyai University, Cluj-Napoca, Romania
| | - Bogdan Razvan Dume
- Doctoral School in Integrative Biology, Faculty of Biology and Geology, "Babes-Bolyai" University, Cluj-Napoca, Romania
| | - Giorgiana Negrea
- Doctoral School in Integrative Biology, Faculty of Biology and Geology, "Babes-Bolyai" University, Cluj-Napoca, Romania
| | - Vlad-Alexandru Toma
- Department of Molecular Biology and Biotechnology, and Center of Systems Biology, Biodiversity and Bioresources, Faculty of Biology and Geology, Babes-Bolyai University, Cluj-Napoca, Romania
- Department of Experimental Biology and Biochemistry, nstitute of Biological Research, branch of NIRDBS Bucharest, Cluj-Napoca, Romania
| | - Dana Muntean
- Department of Pharmaceutical Technology and Biopharmaceutics, Faculty of Pharmacy, University of Medicine and Pharmacy "Iuliu Hatieganu", Cluj-Napoca, Romania
| | - Alina Porfire
- Department of Pharmaceutical Technology and Biopharmaceutics, Faculty of Pharmacy, University of Medicine and Pharmacy "Iuliu Hatieganu", Cluj-Napoca, Romania
| | - Manuela Banciu
- Department of Molecular Biology and Biotechnology, and Center of Systems Biology, Biodiversity and Bioresources, Faculty of Biology and Geology, Babes-Bolyai University, Cluj-Napoca, Romania
| |
Collapse
|
36
|
Sanati M, Afshari AR, Ahmadi SS, Kesharwani P, Sahebkar A. Advances in liposome-based delivery of RNA therapeutics for cancer treatment. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2024; 204:177-218. [PMID: 38458738 DOI: 10.1016/bs.pmbts.2023.12.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/10/2024]
Abstract
Liposomal drug delivery systems stand as versatile therapeutic platforms for precisely targeting related elements in cancerous tissues owing to their intrinsic passive and acquired active targeting capabilities and exceptional compatibility with physiologic environments. When the capacity of liposomes as nanocarriers is combined with the revolutionary potential of RNA therapies in affecting undruggable targets, the outcome would be promising drug candidates as game-changers in the cancer treatment arena. However, optimizing liposome composition, physicochemical properties, and surface chemistry is paramount to maximizing their pharmacokinetic and pharmacodynamic attributes. This review highlighted the potential of liposomes as nanovehicles for RNA therapeutics through a literature review and looked at the most recent preclinical and clinical advancements in utilizing liposomal RNA therapeutics for cancer management. Notably, the discovery of novel targets, advancements in liposome engineering, and organizing well-planned clinical trials would help uncover the incredible potential of these nanotherapeutics in cancer patients.
Collapse
Affiliation(s)
- Mehdi Sanati
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Birjand University of Medical Sciences, Birjand, Iran; Experimental and Animal Study Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Amir R Afshari
- Natural Products and Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, Iran; Department of Physiology and Pharmacology, Faculty of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Seyed Sajad Ahmadi
- Department of Ophthalmology, Khatam-Ol-Anbia Hospital, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
37
|
Peng C, Xu Y, Wu J, Wu D, Zhou L, Xia X. TME-Related Biomimetic Strategies Against Cancer. Int J Nanomedicine 2024; 19:109-135. [PMID: 38192633 PMCID: PMC10773252 DOI: 10.2147/ijn.s441135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 12/21/2023] [Indexed: 01/10/2024] Open
Abstract
The tumor microenvironment (TME) plays an important role in various stages of tumor generation, metastasis, and evasion of immune monitoring and treatment. TME targeted therapy is based on TME components, related pathways or active molecules as therapeutic targets. Therefore, TME targeted therapy based on environmental differences between TME and normal cells has been widely studied. Biomimetic nanocarriers with low clearance, low immunogenicity, and high targeting have enormous potential in tumor treatment. This review introduces the composition and characteristics of TME, including cancer‑associated fibroblasts (CAFs), extracellular matrix (ECM), tumor blood vessels, non-tumor cells, and the latest research progress of biomimetic nanoparticles (NPs) based on TME. It also discusses the opportunities and challenges of clinical transformation of biomimetic nanoparticles.
Collapse
Affiliation(s)
- Cheng Peng
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, People’s Republic of China
| | - Yilin Xu
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, People’s Republic of China
| | - Jing Wu
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, People’s Republic of China
| | - Donghai Wu
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, People’s Republic of China
| | - Lili Zhou
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, People’s Republic of China
| | - Xinhua Xia
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, People’s Republic of China
| |
Collapse
|
38
|
Xi Y, Chen L, Tang J, Yu B, Shen W, Niu X. Amplifying "eat me signal" by immunogenic cell death for potentiating cancer immunotherapy. Immunol Rev 2024; 321:94-114. [PMID: 37550950 DOI: 10.1111/imr.13251] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 07/06/2023] [Accepted: 07/15/2023] [Indexed: 08/09/2023]
Abstract
Immunogenic cell death (ICD) is a unique mode of cell death, which can release immunogenic damage-associated molecular patterns (DAMPs) and tumor-associated antigens to trigger long-term protective antitumor immune responses. Thus, amplifying "eat me signal" during tumor ICD cascade is critical for cancer immunotherapy. Some therapies (radiotherapy, photodynamic therapy (PDT), photothermal therapy (PTT), etc.) and inducers (chemotherapeutic agents, etc.) have enabled to initiate and/or facilitate ICD and activate antitumor immune responses. Recently, nanostructure-based drug delivery systems have been synthesized for inducing ICD through combining treatment of chemotherapeutic agents, photosensitizers for PDT, photothermal transformation agents for PTT, radiosensitizers for radiotherapy, etc., which can release loaded agents at an appropriate dosage in the designated place at the appropriate time, contributing to higher efficiency and lower toxicity. Also, immunotherapeutic agents in combination with nanostructure-based drug delivery systems can produce synergetic antitumor effects, thus potentiating immunotherapy. Overall, our review outlines the emerging ICD inducers, and nanostructure drug delivery systems loading diverse agents to evoke ICD through chemoradiotherapy, PDT, and PTT or combining immunotherapeutic agents. Moreover, we discuss the prospects and challenges of harnessing ICD induction-based immunotherapy, and highlight the significance of multidisciplinary and interprofessional collaboration to promote the optimal translation of this treatment strategy.
Collapse
Affiliation(s)
- Yong Xi
- Department of Thoracic Surgery, Ningbo Medical Center Lihuili Hospital, Ningbo University, Ningbo, China
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Lijie Chen
- School of Medicine, Xiamen University, Xiamen, China
- China Medical University, Shenyang, China
| | - Jian Tang
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Bentong Yu
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Weiyu Shen
- Department of Thoracic Surgery, Ningbo Medical Center Lihuili Hospital, Ningbo University, Ningbo, China
| | - Xing Niu
- China Medical University, Shenyang, China
| |
Collapse
|
39
|
Zou J. Site-specific delivery of cisplatin and paclitaxel mediated by liposomes: A promising approach in cancer chemotherapy. ENVIRONMENTAL RESEARCH 2023; 238:117111. [PMID: 37734579 DOI: 10.1016/j.envres.2023.117111] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 08/25/2023] [Accepted: 09/09/2023] [Indexed: 09/23/2023]
Abstract
The site-specific delivery of drugs, especially anti-cancer drugs has been an interesting field for researchers and the reason is low accumulation of cytotoxic drugs in cancer cells. Although combination cancer therapy has been beneficial in providing cancer drug sensitivity, targeted delivery of drugs appears to be more efficient. One of the safe, biocompatible and efficient nano-scale delivery systems in anti-cancer drug delivery is liposomes. Their particle size is small and they have other properties such as adjustable physico-chemical properties, ease of functionalization and high entrapment efficiency. Cisplatin is a chemotherapy drug with clinical approval in patients, but its accumulation in cancer cells is low due to lack of targeted delivery and repeated administration results in resistance development. Gene and drug co-administration along with cisplatin/paclitaxel have resulted in increased sensitivity in tumor cells, but there is still space for more progress in cancer therapy. The delivery of cisplatin/paclitaxel by liposomes increases accumulation of drug in tumor cells and impairs activity of efflux pumps in promoting cytotoxicity. Moreover, phototherapy along with cisplatin/paclitaxel delivery can increase potential in tumor suppression. Smart nanoparticles including pH-sensitive nanoparticles provide site-specific delivery of cisplatin/paclitaxel. The functionalization of liposomes can be performed by ligands to increase targetability towards tumor cells in mediating site-specific delivery of cisplatin/paclitaxel. Finally, liposomes can mediate co-delivery of cisplatin/paclitaxel with drugs or genes in potentiating tumor suppression. Since drug resistance has caused therapy failure in cancer patients, and cisplatin/paclitaxel are among popular chemotherapy drugs, delivery of these drugs mediates targeted suppression of cancers and prevents development of drug resistance. Because of biocompatibility and safety of liposomes, they are currently used in clinical trials for treatment of cancer patients. In future, the optimal dose of using liposomes and optimal concentration of loading cisplatin/paclitaxel on liposomal nanocarriers in clinical trials should be determined.
Collapse
Affiliation(s)
- Jianyong Zou
- Department of Thoracic Surgery, The first Affiliated Hospital of Sun Yat-Sen University, 510080, Guangzhou, PR China.
| |
Collapse
|
40
|
Xu G, Jiang Y, Li Y, Ge J, Xu X, Chen D, Wu J. A novel immunogenic cell death-related genes signature for predicting prognosis, immune landscape and immunotherapy effect in hepatocellular carcinoma. J Cancer Res Clin Oncol 2023; 149:16261-16277. [PMID: 37698679 DOI: 10.1007/s00432-023-05370-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 08/29/2023] [Indexed: 09/13/2023]
Abstract
OBJECTIVE Immunogenic cell death (ICD) has emerged as a promising strategy to activate the adaptive immune response, modulate the tumor microenvironment (TME) and enhance the efficacy of immune therapy. However, the relationship between ICD and TME reprogramming in hepatocellular carcinoma (HCC) remains poorly understood. METHODS Transcriptional profiles and clinical spectrum of 486 HCC patients were obtained from TCGA and GEO databases. We utilized consensus clustering analysis to construct two distinct molecular subtypes and established an ICD-based scoring system (named ICD score) via WGCNA and LASSO Cox regression to predict the prognosis of the HCC cohort. Then we employed CIBERSORT and ESTIMATE methods to analyze the immune landscape of ICD score in HCC. Subsequently, the immunophenoscore (IPS) and tumor immune dysfunction and rejection (TIDE) analyses were performed to determine whether the ICD score could influence the immune therapeutic effect. Based on the ICD scoring system, a novel nomogram was generated to provide a numerical probability of HCC patients' overall survival (OS). RESULTS We identified two independent ICD clusters (cluster A/B), and cluster B possessed a worse prognosis and higher immune cell infiltration. Using ICD scoring system, the HCC patients were divided into high- and low-ICD-score groups. Through integrative analyses, the high-ICD cohort owned advanced TNM stage, high pathologic grade and increased suppressive immune cell enrichment. We developed a nomogram containing the ICD score, demonstrating a high predictive accuracy with a C-index of 0.703. We further discovered that PSMD2 and PSMD14 could serve as ICD-associated prognostic biomarkers and therapeutic targets in HCC. CONCLUSION The ICD score exhibits a high degree of reliability for predicting prognosis and may provide valuable guidance for the selection of immunotherapy for HCC patients. This novel scoring system enables the estimation of clinical immunotherapy response for HCC patients, offering new opportunities for personalized immunotherapy.
Collapse
Affiliation(s)
- Guangming Xu
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang Province, Hangzhou, 310003, China
| | - Yifan Jiang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang Province, Hangzhou, 310003, China
| | - Yu Li
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang Province, Hangzhou, 310003, China
| | - Jiangzhen Ge
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang Province, Hangzhou, 310003, China
| | - Xiaofeng Xu
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang Province, Hangzhou, 310003, China
| | - Diyu Chen
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang Province, Hangzhou, 310003, China.
- Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Zhejiang Province, Hangzhou, 310003, China.
| | - Jian Wu
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang Province, Hangzhou, 310003, China.
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Zhejiang Province, Hangzhou, 310003, China.
- Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment For Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Zhejiang Province, Hangzhou, 310003, China.
- Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Zhejiang Province, Hangzhou, 310003, China.
| |
Collapse
|
41
|
Wang R, Li N, Zhang T, Sun Y, He X, Lu X, Chu L, Sun K. Tumor microenvironment-responsive micelles assembled from a prodrug of mitoxantrone and 1-methyl tryptophan for enhanced chemo-immunotherapy. Drug Deliv 2023; 30:2182254. [PMID: 36840464 PMCID: PMC9970211 DOI: 10.1080/10717544.2023.2182254] [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] [Indexed: 02/26/2023] Open
Abstract
Mitoxantrone (MX) can induce the immunogenic-cell death (ICD) of tumor cells and activate anti-tumor immune responses. However, it can also cause high expression of indole amine 2, 3-dioxygenase (IDO) during ICD, leading to T-cell apoptosis and a weakened immune response. An IDO inhibitor, 1-methyl tryptophan (1-MT), can inhibit the activity of IDO caused by MX, resulting in enhanced chemo-immunotherapy. Here, MX-1-MT was connected by ester bond which could be broken in an acidic tumor microenvironment. MX-1-MT was combined with polyethylene glycol (PEG) via a disulfide bond which could be reduced by glutathione overexpressed in tumors, thereby accelerating drug release at target sites. Folic acid-modified distearoyl phosphoethanolamine-polyethylene glycol (DSPE-PEG-FA) was introduced to form targeting micelles. The micelles were of uniform particle size, high stability, and high responsiveness. They could be taken-up by drug-resistant MCF-7/ADR cells, displayed high targeting ability, and induced enhanced cytotoxicity and ICD. Due to 1-MT addition, micelles could inhibit IDO. In vivo studies demonstrated that micelles could accumulate in the tumor tissues of nude mice, resulting in an enhanced antitumor effect and few side-effects.
Collapse
Affiliation(s)
- Ru Wang
- Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Ministry of Education, Yantai University, Yantai, Shandong Province, China
| | - Nuannuan Li
- Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Ministry of Education, Yantai University, Yantai, Shandong Province, China
| | - Tianyu Zhang
- Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Ministry of Education, Yantai University, Yantai, Shandong Province, China
| | - Yiying Sun
- Yantai Saipute Analyzing Service Co. Ltd, Yantai, Shandong Province, China
| | - Xiaoyan He
- Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Ministry of Education, Yantai University, Yantai, Shandong Province, China
| | - Xiaoyan Lu
- Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Ministry of Education, Yantai University, Yantai, Shandong Province, China
| | - Liuxiang Chu
- Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Ministry of Education, Yantai University, Yantai, Shandong Province, China
| | - Kaoxiang Sun
- Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Ministry of Education, Yantai University, Yantai, Shandong Province, China,CONTACT Kaoxiang Sun Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Ministry of Education, Yantai University, Yantai, Shandong Province, China
| |
Collapse
|
42
|
Hasan I, Roy S, Ehexige E, Wu R, Chen Y, Gao Z, Guo B, Chang C. A state-of-the-art liposome technology for glioblastoma treatment. NANOSCALE 2023; 15:18108-18138. [PMID: 37937394 DOI: 10.1039/d3nr04241c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2023]
Abstract
Glioblastoma (GBM) is a challenging problem due to the poor BBB permeability of cancer drugs, its recurrence after the treatment, and high malignancy and is difficult to treat with the currently available therapeutic strategies. Furthermore, the prognosis and survival rate of GBM are still poor after surgical removal via conventional combination therapy. Owing to the existence of the formidable blood-brain barrier (BBB) and the aggressive, infiltrating nature of GBM growth, the diagnosis and treatment of GBM are quite challenging. Recently, liposomes and their derivatives have emerged as super cargos for the delivery of both hydrophobic and hydrophilic drugs for the treatment of glioblastoma because of their advantages, such as biocompatibility, long circulation, and ease of physical and chemical modification, which facilitate the capability of targeting specific sites, circumvention of BBB transport restrictions, and amplification of the therapeutic efficacy. Herein, we provide a timely update on the burgeoning liposome-based drug delivery systems and potential challenges in these fields for the diagnosis and treatment of brain tumors. Furthermore, we focus on the most recent liposome-based drug delivery cargos, including pH-sensitive, temperature-sensitive, and biomimetic liposomes, to enhance the multimodality in imaging and therapeutics of glioblastoma. Furthermore, we highlight the future difficulties and directions for the research and clinical translation of liposome-based drug delivery. Hopefully, this review will trigger the interest of researchers to expedite the development of liposome cargos and even their clinical translation for improving the prognosis of glioblastoma.
Collapse
Affiliation(s)
- Ikram Hasan
- School of Biomedical Engineering, Medical School, Shenzhen University, Shenzhen, Guangdong, 518060, China.
| | - Shubham Roy
- Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application and School of Science, Harbin Institute of Technology, Shenzhen 518055, China.
- Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Harbin Institute of Technology, Shenzhen 518055, China
| | - Ehexige Ehexige
- School of Biomedical Engineering, Medical School, Shenzhen University, Shenzhen, Guangdong, 518060, China.
| | - Runling Wu
- School of Biomedical Engineering, Medical School, Shenzhen University, Shenzhen, Guangdong, 518060, China.
| | - Yu Chen
- Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application and School of Science, Harbin Institute of Technology, Shenzhen 518055, China.
- Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Harbin Institute of Technology, Shenzhen 518055, China
- College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen, 518060, China
| | - Zhengyuan Gao
- School of Biomedical Engineering, Medical School, Shenzhen University, Shenzhen, Guangdong, 518060, China.
| | - Bing Guo
- Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application and School of Science, Harbin Institute of Technology, Shenzhen 518055, China.
- Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Harbin Institute of Technology, Shenzhen 518055, China
| | - Chunqi Chang
- School of Biomedical Engineering, Medical School, Shenzhen University, Shenzhen, Guangdong, 518060, China.
| |
Collapse
|
43
|
Li N, Zhang T, Wang R, Sun Y, Chu L, Lu X, Sun K. Homotypic targeted nanoplatform enable efficient chemoimmunotherapy and reduced DOX cardiotoxicity in chemoresistant cancer via TGF-β1 blockade. J Control Release 2023; 361:147-160. [PMID: 37536544 DOI: 10.1016/j.jconrel.2023.07.063] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 07/18/2023] [Accepted: 07/31/2023] [Indexed: 08/05/2023]
Abstract
Doxorubicin (DOX) with broad-spectrum antitumor activity has been reported to induce effective immunogenic cell death (ICD) effect. However, the serious cardiotoxicity and chemoresistance severely restrict the widely clinical application of DOX. Herein, for the first time, a bio-inspired nanoplatform via co-assembly of DOX-conjugated polyethyleneimine (PEI-DOX), cancer cell membrane (CCM) and TGF-β1 siRNA (siTGF-β1) was rationally designed, which can not only overcome the drawbacks of DOX but also display high capability to modulate the tumor microenvironment and prevent the tumor progressing and metastasis. Experimental studies confirmed the pH-sensitivity of PEI-DOX and the homotypic-targeting and immuno-escapable ability of CCM, resulting an enhanced accumulation of DOX and siTGF-β1 in tumor sites. In addition to this, the bio-inspired nanoplatform could also improve the stability and facilitate the endosomal escape of siTGF-β1. All these effects ensured the silence efficiency of siTGF-β1 in tumor sites, which could further modulate the chemoresistant and immunosuppressive tumor microenvironment, resulting a synergistic effect with DOX to prevent tumor progressing and metastasis. Additionally, even trapped in cardiac tissues, siTGF-β1 could inhibit the production of TGF-β1 and ROS induced by DOX, resulting a reduced myocardial damage. Therefore, our newly designed bio-inspired nano-delivery system may be a promising nanoplatform with efficient chemoimmunotherapy to ameliorate DOX-induced cardiotoxicity and combat tumor growth and metastasis in chemoresistant cancer.
Collapse
Affiliation(s)
- Nuannuan Li
- Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Ministry of Education, Yantai University, Yantai, China
| | - Tianyu Zhang
- Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Ministry of Education, Yantai University, Yantai, China
| | - Ru Wang
- Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Ministry of Education, Yantai University, Yantai, China
| | - Yiying Sun
- Yantai Saipute Analyzing Service Co. Ltd, Yantai, Shandong Province, China
| | - Liuxiang Chu
- Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Ministry of Education, Yantai University, Yantai, China
| | - Xiaoyan Lu
- Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Ministry of Education, Yantai University, Yantai, China
| | - Kaoxiang Sun
- Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Ministry of Education, Yantai University, Yantai, China.
| |
Collapse
|
44
|
Pourtalebi Jahromi L, Rothammer M, Fuhrmann G. Polysaccharide hydrogel platforms as suitable carriers of liposomes and extracellular vesicles for dermal applications. Adv Drug Deliv Rev 2023; 200:115028. [PMID: 37517778 DOI: 10.1016/j.addr.2023.115028] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 06/26/2023] [Accepted: 07/27/2023] [Indexed: 08/01/2023]
Abstract
Lipid-based nanocarriers have been extensively investigated for their application in drug delivery. Particularly, liposomes are now clinically established for treating various diseases such as fungal infections. In contrast, extracellular vesicles (EVs) - small cell-derived nanoparticles involved in cellular communication - have just recently sparked interest as drug carriers but their development is still at the preclinical level. To drive this development further, the methods and technologies exploited in the context of liposome research should be applied in the domain of EVs to facilitate and accelerate their clinical translation. One of the crucial steps for EV-based therapeutics is designing them as proper dosage forms for specific applications. This review offers a comprehensive overview of state-of-the-art polysaccharide-based hydrogel platforms designed for artificial and natural vesicles with application in drug delivery to the skin. We discuss their various physicochemical and biological properties and try to create a sound basis for the optimization of EV-embedded hydrogels as versatile therapeutic avenues.
Collapse
Affiliation(s)
- Leila Pourtalebi Jahromi
- Friedrich-Alexander-Universität Erlangen-Nürnberg, Department of Biology, Pharmaceutical Biology, Staudtstr. 5, 91058 Erlangen, Germany
| | - Markus Rothammer
- Friedrich-Alexander-Universität Erlangen-Nürnberg, Department of Biology, Pharmaceutical Biology, Staudtstr. 5, 91058 Erlangen, Germany
| | - Gregor Fuhrmann
- Friedrich-Alexander-Universität Erlangen-Nürnberg, Department of Biology, Pharmaceutical Biology, Staudtstr. 5, 91058 Erlangen, Germany; FAU NeW, Nikolaus-Fiebiger-Str. 10, 91058 Erlangen, Germany.
| |
Collapse
|
45
|
Bertrand P. Aptamers Targeting the PD-1/PD-L1 Axis: A Perspective. J Med Chem 2023; 66:10878-10888. [PMID: 37561598 DOI: 10.1021/acs.jmedchem.3c00551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/12/2023]
Abstract
Aptamers have emerged in recent years as alternatives to antibodies or small molecules to interfere with the immune check points by blocking the PD-1/PD-L1 interactions and represent an interesting perspective for immuno-oncology. Aptamers are RNA or DNA nucleotides able to bind to a target with high affinity, with the target ranging from small molecules to proteins and up to cells. Aptamers are identified by the SELEX method that can be modified for specific purposes. The range of applications of aptamers covers therapy as well as new alternative assay technologies similar to ELISA. Aptamers' limited plasma stability can be managed using delivery strategies. The goal of this Perspective is to give an overview of the current development of aptamers targeting the most studied immune checkpoint modulators, PD-1 and PD-L1, and analogous strategies with aptamers for other immuno-related targets.
Collapse
Affiliation(s)
- Philippe Bertrand
- University of Poitiers, IC2MP UMR 7285 CNRS, 4 rue Michel Brunet B27, TSA 51106, 86073 Poitiers cedex 9, France
| |
Collapse
|
46
|
Subhan MA, Filipczak N, Torchilin VP. Advances with Lipid-Based Nanosystems for siRNA Delivery to Breast Cancers. Pharmaceuticals (Basel) 2023; 16:970. [PMID: 37513882 PMCID: PMC10386415 DOI: 10.3390/ph16070970] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Revised: 07/01/2023] [Accepted: 07/04/2023] [Indexed: 07/30/2023] Open
Abstract
Breast cancer is the most frequently diagnosed cancer among women. Breast cancer is also the key reason for worldwide cancer-related deaths among women. The application of small interfering RNA (siRNA)-based drugs to combat breast cancer requires effective gene silencing in tumor cells. To overcome the challenges of drug delivery to tumors, various nanosystems for siRNA delivery, including lipid-based nanoparticles that protect siRNA from degradation for delivery to cancer cells have been developed. These nanosystems have shown great potential for efficient and targeted siRNA delivery to breast cancer cells. Lipid-based nanosystems remain promising as siRNA drug delivery carriers for effective and safe cancer therapy including breast cancer. Lipid nanoparticles (LNPs) encapsulating siRNA enable efficient and specific silencing of oncogenes in breast tumors. This review discusses a variety of lipid-based nanosystems including cationic lipids, sterols, phospholipids, PEG-lipid conjugates, ionizable liposomes, exosomes for effective siRNA drug delivery to breast tumors, and the clinical translation of lipid-based siRNA nanosystems for solid tumors.
Collapse
Affiliation(s)
- Md Abdus Subhan
- Department of Chemistry, ShahJalal University of Science and Technology, Sylhet 3114, Bangladesh
- Division of Nephrology, University of Rochester Medical Center, School of Medicine and Dentistry, 601 Elmwood Ave, Box 675, Rochester, NY 14642, USA
| | - Nina Filipczak
- Center for Pharmaceutical Biotechnology and Nanomedicine, Department of Pharmaceutical Sciences, Northeastern University, Boston, MA 02115, USA
| | - Vladimir P Torchilin
- Center for Pharmaceutical Biotechnology and Nanomedicine, Department of Pharmaceutical Sciences, Northeastern University, Boston, MA 02115, USA
- Department of Chemical Engineering, Northeastern University, Boston, MA 02115, USA
| |
Collapse
|
47
|
Ding YN, Ding HY, Li H, Yang R, Huang JY, Chen H, Wang LH, Wang YJ, Hu CM, An YL, Zhang ZY, Yu WP, Tang QS, Shao GL. Photosensitive Small Extracellular Vesicles Regulate the Immune Microenvironment of Triple Negative Breast Cancer. Acta Biomater 2023:S1742-7061(23)00329-X. [PMID: 37302734 DOI: 10.1016/j.actbio.2023.06.004] [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: 02/28/2023] [Revised: 05/25/2023] [Accepted: 06/06/2023] [Indexed: 06/13/2023]
Abstract
Currently, the treatment of triple-negative breast cancer (TNBC) is limited by the special pathological characteristics of this disease. In recent years, photodynamic therapy (PDT) has created new hope for the treatment of TNBC. Moreover, PDT can induce immunogenic cell death (ICD) and improve tumor immunogenicity. However, even though PDT can improve the immunogenicity of TNBC, the inhibitory immune microenvironment of TNBC still weakens the antitumor immune response. Therefore, we used the neutral sphingomyelinase inhibitor GW4869 to inhibit the secretion of small extracellular vesicles (sEVs) by TNBC cells to improve the tumor immune microenvironment and enhance antitumor immunity. In addition, bone mesenchymal stem cell (BMSC)-derived sEVs have good biological safety and a strong drug loading capacity, which can effectively improve the efficiency of drug delivery. In this study, we first obtained primary BMSCs and sEVs, and then the photosensitizers Ce6 and GW4869 were loaded into the sEVs by electroporation to produce immunomodulatory photosensitive nanovesicles (Ce6-GW4869/sEVs). When administered to TNBC cells or orthotopic TNBC models, these photosensitive sEVs could specifically target TNBC and improve the tumor immune microenvironment. Moreover, PDT combined with GW4869-based therapy showed a potent synergistic antitumor effect mediated by direct killing of TNBC and activation of antitumor immunity. Here, we designed photosensitive sEVs that could target TNBC and regulate the tumor immune microenvironment, providing a potential approach for improving the effectiveness of TNBC treatment. STATEMENT OF SIGNIFICANCE: We designed an immunomodulatory photosensitive nanovesicle (Ce6-GW4869/sEVs) with the photosensitizer Ce6 to achieve photodynamic therapy and the neutral sphingomyelinase inhibitor GW4869 to inhibit the secretion of small extracellular vesicles (sEVs) by triple-negative breast cancer (TNBC) cells to improve the tumor immune microenvironment and enhance antitumor immunity. In this study, the immunomodulatory photosensitive nanovesicle could target TNBC cells and regulate the tumor immune microenvironment, thus providing a potential approach for improving the treatment effect in TNBC. We found that the reduction in tumor sEVs secretion induced by GW4869 improved the tumor-suppressive immune microenvironment. Moreover, similar therapeutic strategies can also be applied in other kinds of tumors, especially immunosuppressive tumors, which is of great value for the clinical translation of tumor immunotherapy.
Collapse
Affiliation(s)
- Yi-Nan Ding
- Medical School of Southeast University, Nanjing 210009, China
| | - Hui-Yan Ding
- Medical School of Southeast University, Nanjing 210009, China
| | - Han Li
- Department of tuberculosis, the Second Hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing, China
| | - Rui Yang
- Center of Reproductive Medicine, State Key Laboratory of Reproductive Medicine, Research Institute for Reproductive Health and Genetic Diseases, The Affiliated Wuxi Maternity and Child Health Care Hospital of Nanjing Medical University, Wuxi, 214002, Jiangsu, China
| | - Jia-Yan Huang
- Department of tuberculosis, the Second Hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing, China
| | - He Chen
- First people's hospital of Changzhou, Changzhou, Jiangsu, China
| | - Lu-Hong Wang
- Medical School of Southeast University, Nanjing 210009, China
| | - Yun-Juan Wang
- Medical School of Southeast University, Nanjing 210009, China
| | - Chun-Mei Hu
- Department of tuberculosis, the Second Hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yan-Li An
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, China
| | - Zhi-Yuan Zhang
- Department of Neurosurgery, Nanjing Jinling hospital, Nanjing University, Nanjing 210002, China
| | - Wei-Ping Yu
- Medical School of Southeast University, Nanjing 210009, China..
| | - Qiu-Sha Tang
- Medical School of Southeast University, Nanjing 210009, China..
| | - Guo-Liang Shao
- Department of interventional oncology, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China..
| |
Collapse
|
48
|
Wei M, Shen X, Fan X, Li J, Bai J. PD-L1 aptamer-functionalized degradable hafnium oxide nanoparticles for near infrared-II diagnostic imaging and radiosensitization. Front Bioeng Biotechnol 2023; 11:1224339. [PMID: 37351473 PMCID: PMC10282151 DOI: 10.3389/fbioe.2023.1224339] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 05/30/2023] [Indexed: 06/24/2023] Open
Abstract
Immune checkpoint blockade is now recognized as a paradigm-shifting cancer therapeutic strategy, whereas there remains difficulty in accurately predicting immunotherapy efficacy by PD-L1 expression. In addition, radiotherapy for cancer patients faces the problem of insufficient dose of radiotherapy at the tumor site while which have been not tolerated by normal tissues. In this study, we created PD-L1 aptamer-anchored spherical nucleic acids (SNAs) with a shell made of PD-L1 aptamer and indocyanine green (ICG) embedded in a mesoporous hafnium oxide nanoparticle core (Hf@ICG-Apt). Upon low pH irradiation in the tumor sites, the nano-system enabled the release of ICG in the high PD-L1 expression tumor to develop a high tumor-to-background ratio of 7.97 ± 0.76 and enhanced the ICG tumor retention to more than 48 h. Moreover, Hf@ICG-Apt improved radiation therapy (RT) when combined with radiation. Notably, Hf@ICG-Apt showed scarcely any systemic toxicity in vivo. Overall, this research offered a novel approach for applying reliable monitoring of PD-L1 expression and localization and robust RT sensitization against cancer with good biosafety.
Collapse
Affiliation(s)
- Min Wei
- Cancer Center and Department of Breast and Thyroid Surgery, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- Fujian Key Laboratory of Precision Diagnosis and Treatment in Breast Cancer, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- Xiamen Key Laboratory of Endocrine-Related Cancer Precision Medicine, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- Xiamen Research Center of Clinical Medicine in Breast and Thyroid Cancers, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- Cancer Research Center of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Xiao Shen
- Cancer Center and Department of Breast and Thyroid Surgery, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- Fujian Key Laboratory of Precision Diagnosis and Treatment in Breast Cancer, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- Xiamen Key Laboratory of Endocrine-Related Cancer Precision Medicine, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- Xiamen Research Center of Clinical Medicine in Breast and Thyroid Cancers, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- Cancer Research Center of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Xueqi Fan
- Cancer Center and Department of Breast and Thyroid Surgery, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- Fujian Key Laboratory of Precision Diagnosis and Treatment in Breast Cancer, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- Xiamen Key Laboratory of Endocrine-Related Cancer Precision Medicine, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- Xiamen Research Center of Clinical Medicine in Breast and Thyroid Cancers, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- Cancer Research Center of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Jiwei Li
- Department of Respiratory, Critical Care and Sleep Medicine, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Jingwen Bai
- Cancer Center and Department of Breast and Thyroid Surgery, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- Fujian Key Laboratory of Precision Diagnosis and Treatment in Breast Cancer, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- Xiamen Key Laboratory of Endocrine-Related Cancer Precision Medicine, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- Xiamen Research Center of Clinical Medicine in Breast and Thyroid Cancers, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- Cancer Research Center of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- Department of Oncology, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| |
Collapse
|
49
|
Zhang Y, Wu X, Zhu H, Cong Y. Development and in functional study of a bi-specific sustained release drug-loaded nano-liposomes for hepatocellular carcinoma. J Biomater Appl 2023:8853282231179313. [PMID: 37243614 DOI: 10.1177/08853282231179313] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
BACKGROUND Lenvatinib (LEN) is a first-line therapy for patients with hepatocellular carcinoma (HCC), but has a larger adverse effect profile. In this study, we developed a liposome with drug-carrying function and magnetic resonance imaging (MRI) imaging function to investigate the targeted drug-carrying function and MRI tracing ability of liposome for HCC. METHODS Magnetic nano-liposomes (MNL) with dual targeting function of epithelial cell adhesion molecule (EpCAM) and vimentin and capable of encapsulating LEN drugs were prepared. The characterization performance, drug loading efficiency and cytotoxicity of EpCAM/vimentin-LEN-MNL were tested, and the dual-targeting slow release drug loading function and MRI tracing ability were investigated in cellular and animal models. RESULTS EpCAM/vimentin-LEN-MNL has a mean particle size of 218.37 ± 5.13 nm and a mean potential of 32.86 ± 4.62 mV, and is spherical in shape and can be uniformly dispersed in solution. The encapsulation rate was 92.66 ± 0.73% and the drug loading rate was 9.35 ± 0.16%. It has low cytotoxicity, can effectively inhibit HCC cell proliferation and promote HCC cell apoptosis, and has specific targeting function and MRI tracing ability for HCC cells. CONCLUSIONS In this study, an HCC-specific dual-targeted sustained-release drug delivery liposome with dual-targeted recognition and sensitive MRI tracer was successfully prepared, which provides an important scientific basis for maximizing the multiple effects of nano-carriers in tumor diagnosis and treatment.
Collapse
Affiliation(s)
- Yufei Zhang
- Shanghai Seventh People's Hospital, Shanghai, China
| | - Xiaoxiong Wu
- Shanghai Seventh People's Hospital, Shanghai, China
| | - Hongfan Zhu
- Shanghai Seventh People's Hospital, Shanghai, China
| | - Yun Cong
- Shanghai Seventh People's Hospital, Shanghai, China
| |
Collapse
|
50
|
Liang X, Gao H, Xiao J, Han S, He J, Yuan R, Yang S, Yao C. Abrine, an IDO1 inhibitor, suppresses the immune escape and enhances the immunotherapy of anti-PD-1 antibody in hepatocellular carcinoma. Front Immunol 2023; 14:1185985. [PMID: 37334368 PMCID: PMC10272936 DOI: 10.3389/fimmu.2023.1185985] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 05/16/2023] [Indexed: 06/20/2023] Open
Abstract
Background Indoleamine-2,3-dioxygenase 1 (IDO1) is responsible for tumor immune escape by regulating T cell-associated immune responses and promoting the activation of immunosuppressive. Given the vital role of IDO1 in immune response, further investigation on the regulation of IDO1 in tumors is needed. Methods Herein, we used ELISA kit to detect the interferon-gamma (IFN-γ), Tryptophan (Trp), and kynurenic acid (Kyn) levels; western blot, Flow cytometry, and immunofluorescence assays detected the expression of the proteins; Molecular docking assay, SPR assay and Cellular Thermal Shift Assay (CETSA) were used to detect the interaction between IDO1 and Abrine; nano live label-free system was used to detect the phagocytosis activity; tumor xenografts animal experiments were used to explore the anti-tumor effect of Abrine; flow cytometry detected the immune cells changes. Results The important immune and inflammatory response cytokine interferon-gamma (IFN-γ) up-regulated the IDO1 expression in cancer cells through the methylation of 6-methyladenosine (m6A) m6A modification of RNA, metabolism of Trp into Kyn, and JAK1/STAT1 signaling pathway, which could be inhibited by IDO1 inhibitor Abrine. CD47 is IFN-γ-stimulated genes (ISGs) and prevents the phagocytosis of macrophages, leading to the cancer immune escape, and this effect could be inhibited by Abrine both in vivo and in vitro. The PD-1/PD-L1 axis is an important immune checkpoint in regulating immune response, overexpression of PD-1 or PD-L1 promotes immune suppression, while in this study Abrine could inhibit the expression of PD-L1 in cancer cells or tumor tissue. The combination treatment of Abrine and anti-PD-1 antibody has a synergistic effect on suppressing the tumor growth through up-regulating CD4+ or CD8+ T cells, down-regulating the Foxp3+ Treg cells, and inhibiting the expression of IDO1, CD47, and PD-L1. Conclusion Overall, this study reveals that Abrine as an IDO1 inhibitor has an inhibition effect on immune escape and has a synergistic effect with the anti-PD-1 antibody on the treatment of HCC.
Collapse
Affiliation(s)
- Xiaowei Liang
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, China
- Engineering Research Center in Ministry of Education for Innovative Drugs of Traditional Chinese Medicine and Zhuang Yao Medicine, Nanning, China
| | - Hongwei Gao
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, China
- Engineering Research Center in Ministry of Education for Innovative Drugs of Traditional Chinese Medicine and Zhuang Yao Medicine, Nanning, China
| | - Jian Xiao
- Engineering Research Center in Ministry of Education for Innovative Drugs of Traditional Chinese Medicine and Zhuang Yao Medicine, Nanning, China
| | - Shan Han
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, China
- Engineering Research Center in Ministry of Education for Innovative Drugs of Traditional Chinese Medicine and Zhuang Yao Medicine, Nanning, China
| | - Jia He
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, China
- Engineering Research Center in Ministry of Education for Innovative Drugs of Traditional Chinese Medicine and Zhuang Yao Medicine, Nanning, China
| | - Renyikun Yuan
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, China
- Engineering Research Center in Ministry of Education for Innovative Drugs of Traditional Chinese Medicine and Zhuang Yao Medicine, Nanning, China
| | - Shilin Yang
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, China
- Engineering Research Center in Ministry of Education for Innovative Drugs of Traditional Chinese Medicine and Zhuang Yao Medicine, Nanning, China
| | - Chun Yao
- Engineering Research Center in Ministry of Education for Innovative Drugs of Traditional Chinese Medicine and Zhuang Yao Medicine, Nanning, China
| |
Collapse
|