1
|
Ribeiro ARS, Neuper T, Horejs-Hoeck J. The Role of STING-Mediated Activation of Dendritic Cells in Cancer Immunotherapy. Int J Nanomedicine 2024; 19:10685-10697. [PMID: 39464674 PMCID: PMC11512692 DOI: 10.2147/ijn.s477320] [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: 05/08/2024] [Accepted: 09/16/2024] [Indexed: 10/29/2024] Open
Abstract
The signaling pathway that comprises cyclic guanosine monophosphate-adenosine monophosphate (cGAMP or GMP-AMP) synthase (cGAS) and Stimulator of Interferon Genes (STING) is emerging as a druggable target for immunotherapy, with tumor-resident dendritic cells (DC) playing a critical role in mediating its effects. The STING receptor is part of the DNA-sensing cellular machinery, that can trigger the secretion of pro-inflammatory mediators, priming effector T cells and initiating specific antitumor responses. Yet, recent studies have highlighted the dual role of STING activation in the context of cancer: STING can either promote antitumor responses or enhance tumor progression. This dichotomy often depends on the cell type in which cGAS-STING signaling is induced and the activation mode, namely acute versus chronic. Of note, STING activation at the DC level appears to be particularly important for tumor eradication. This review outlines the contribution of the different conventional and plasmacytoid DC subsets and describes the mechanisms underlying STING-mediated activation of DCs in cancer. We further highlight how the STING pathway plays an intricate role in modulating the function of DCs embedded in tumor tissue. Additionally, we discuss the strategies being employed to harness STING activation for cancer treatment, such as the development of synthetic agonists and nano-based delivery systems, spotlighting the current techniques used to prompt STING engagement specifically in DCs.
Collapse
Affiliation(s)
- Ana R S Ribeiro
- Department of Biosciences and Medical Biology, Paris Lodron University of Salzburg, Salzburg, Austria
- Cancer Cluster Salzburg (CCS), Salzburg, 5020, Austria
| | - Theresa Neuper
- Department of Biosciences and Medical Biology, Paris Lodron University of Salzburg, Salzburg, Austria
- Cancer Cluster Salzburg (CCS), Salzburg, 5020, Austria
- Center for Tumor biology and Immunology (CTBI), Salzburg, 5020, Austria
| | - Jutta Horejs-Hoeck
- Department of Biosciences and Medical Biology, Paris Lodron University of Salzburg, Salzburg, Austria
- Cancer Cluster Salzburg (CCS), Salzburg, 5020, Austria
- Center for Tumor biology and Immunology (CTBI), Salzburg, 5020, Austria
| |
Collapse
|
2
|
Qian L, Zhang Z, Zhang R, Zheng X, Xiao B, Zhang X, Wu Y, Chen Y, Zhang X, Zhou P, Fu Q, Kang T, Gao Y. Activated STING-containing R-EVs from iPSC-derived MSCs promote antitumor immunity. Cancer Lett 2024; 597:217081. [PMID: 38909776 DOI: 10.1016/j.canlet.2024.217081] [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: 06/20/2024] [Accepted: 06/21/2024] [Indexed: 06/25/2024]
Abstract
We recently revealed that activated STING is secreted into RAB22A-induced extracellular vesicles (R-EVs) and promotes antitumor immunity in cancer cells. Whether mesenchymal stem cell (MSC)-derived R-EVs containing activated STING can be used as a novel antitumor immunotherapy remains unclear, as MSC-derived EVs are promising cell-free therapeutics due to their superior biocompatibility and safety, as well as low immunogenicity. Here, we report that induced pluripotent stem cell (iPSC)-derived MSCs can generate R-EVs with a size and mechanism of formation that are similar to those of R-EVs produced from cancer cells. Furthermore, these MSC-derived R-EVs containing activated STING induced IFNβ expression in recipient THP-1 monocytes and antitumor immunity in mice. Our findings reveal that the use of MSC-derived R-EVs containing activated STING is a promising cell-free strategy for antitumor immunity.
Collapse
Affiliation(s)
- Linxia Qian
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, Guangdong, China; School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, 518107, Guangdong, China
| | - Zhonghan Zhang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, Guangdong, China
| | - Ruhua Zhang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, Guangdong, China
| | - Xueping Zheng
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, Guangdong, China
| | - Beibei Xiao
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, Guangdong, China
| | - Xiaomin Zhang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, Guangdong, China
| | - Yuanzhong Wu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, Guangdong, China
| | - Yang Chen
- Departments of Endodontics, Stomatological Hospital, Southern Medical University, Guangzhou, 510000, Guangdong, China
| | - Xingding Zhang
- School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, 518107, Guangdong, China
| | - Penghui Zhou
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, Guangdong, China
| | - Qingling Fu
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, Guangdong, China.
| | - Tiebang Kang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, Guangdong, China.
| | - Ying Gao
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, Guangdong, China.
| |
Collapse
|
3
|
Bhattacharya S, Page A, Shinde P. Capecitabine loaded potato starch-chitosan nanoparticles: A novel approach for targeted therapy and improved outcomes in aggressive colon cancer. Eur J Pharm Biopharm 2024; 200:114328. [PMID: 38763329 DOI: 10.1016/j.ejpb.2024.114328] [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: 02/07/2024] [Revised: 05/08/2024] [Accepted: 05/14/2024] [Indexed: 05/21/2024]
Abstract
Aggressive colon cancer treatment poses significant challenges. This study investigates the potential of innovative carbohydrate-based nanoparticles for targeted Capecitabine (CTB) delivery. CTB nanoparticles were synthesized by conjugating CTB with potato starch and chitosan using ultrasonication, hydrolysis, and ionotropic gelation. Characterization included drug loading, rheology, Surface-Enhanced Raman Spectroscopy (SERS), Fourier-Transform Infrared Spectroscopy (FTIR), Differential Scanning Calorimetry (DSC), X-ray Diffraction (XRD), and Thermogravimetric Analysis (TGA). In vitro and in vivo antitumor activity was evaluated using HT-29 cells and N, N-dimethylhydrazine-induced Balb/c mice, respectively. Cellular assays assessed angiogenesis, migration, proliferation, and apoptosis. Nanoparticles exhibited a mean size of 245 nm, positive zeta potential (+30 mV), high loading efficacy (76 %), and sustained drug release (92 % over 100 h). CTB-loaded nanoparticles displayed superior colon histology, reduced tumour scores, and inhibited VEGD and CD31 expression compared to free CTB. Cellular assays confirmed significant antitumor effects, including reduced tube formation, migration, and proliferation, and increased apoptosis. This study demonstrates the promise of CTB-loaded potato starch-chitosan nanoparticles for aggressive colon cancer treatment. These findings highlight the potential of these nanoparticles for further evaluation in diverse cancer models.
Collapse
Affiliation(s)
- Sankha Bhattacharya
- Department of Pharmaceutics, School of Pharmacy & Technology Management, SVKM'S NMIMS Deemed-to-be University, Shirpur, Maharashtra 425405, India.
| | - Amit Page
- Department of Pharmaceutics, School of Pharmacy & Technology Management, SVKM'S NMIMS Deemed-to-be University, Shirpur, Maharashtra 425405, India
| | - Prafull Shinde
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, NMIMS Deemed-to-be-University, India
| |
Collapse
|
4
|
An Y, Zhu J, Xie Q, Feng J, Gong Y, Fan Q, Cao J, Huang Z, Shi W, Lin Q, Wu L, Yang C, Ji T. Tumor Exosomal ENPP1 Hydrolyzes cGAMP to Inhibit cGAS-STING Signaling. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2308131. [PMID: 38498770 PMCID: PMC11132070 DOI: 10.1002/advs.202308131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Indexed: 03/20/2024]
Abstract
To evade immune surveillance, tumor cells express ectonucleotide pyrophosphatase phosphodiesterase 1 (ENPP1) on the surface of their membrane, which degrades extracellular cyclic GMP-AMP (cGAMP), thereby inhibiting the cyclic GMP-AMP synthase (cGAS) stimulator of interferon gene (STING) DNA-sensing pathway. To fully understand this tumor stealth mechanism, it is essential to determine whether other forms of ENPP1 with hydrolytic cGAMP activity also are present in the tumor microenvironment to regulate this innate immune pathway. Herein, it is reported that various tumor-derived exosomes carry ENPP1, and can hydrolyze synthetic 2'3'-cGAMP and endogenous 2'3'-cGAMP produced by cells to inhibit cGAS-STING pathway in immune cells. Moreover, tumor exosomal ENPP1 also can hydrolyze 2'3'-cGAMP bound to LL-37 (an effective transporter of 2'3'-cGAMP) to inhibit STING signaling. Furthermore, high expression of ENPP1 in exosomes is observed isolated from human breast and lung cancer tissue, and tumor exosomal ENPP1 inhibited the immune infiltration of CD8+ T cells and CD4+ T cells. The results elucidate the essential function of tumor exosomal ENPP1 in the cGAS-STING pathway, furthering understanding of the crosstalk between the tumor cells and immune system.
Collapse
Affiliation(s)
- Yu An
- Department of PathologyShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200011P. R. China
| | - Jinchao Zhu
- Department of PathologyShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200011P. R. China
| | - Qihui Xie
- State Key Laboratory of Oral & Maxillofacial Reconstruction and RegenerationKey Laboratory of Oral Biomedicine Ministry of EducationHubei Key Laboratory of StomatologySchool & Hospital of StomatologyWuhan UniversityWuhan430070P. R. China
| | - Jianzhou Feng
- Institute of Molecular MedicineRenji HospitalShanghai Jiao Tong University School of MedicineShanghai200127P. R. China
| | - Yanli Gong
- Institute of Molecular MedicineRenji HospitalShanghai Jiao Tong University School of MedicineShanghai200127P. R. China
| | - Qian Fan
- Institute of Molecular MedicineRenji HospitalShanghai Jiao Tong University School of MedicineShanghai200127P. R. China
| | - Jiao Cao
- Institute of Molecular MedicineRenji HospitalShanghai Jiao Tong University School of MedicineShanghai200127P. R. China
| | - Zhi Huang
- Institute of Molecular MedicineRenji HospitalShanghai Jiao Tong University School of MedicineShanghai200127P. R. China
| | - Weixiong Shi
- Institute of Molecular MedicineRenji HospitalShanghai Jiao Tong University School of MedicineShanghai200127P. R. China
| | - Qingyuan Lin
- Department of PathologyShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200011P. R. China
| | - Lingling Wu
- Institute of Molecular MedicineRenji HospitalShanghai Jiao Tong University School of MedicineShanghai200127P. R. China
| | - Chaoyong Yang
- Institute of Molecular MedicineRenji HospitalShanghai Jiao Tong University School of MedicineShanghai200127P. R. China
- The MOE Key Laboratory of Spectrochemical Analysis and InstrumentationState Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical EngineeringXiamen UniversityXiamen361005P. R. China
| | - Tianhai Ji
- Department of PathologyShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200011P. R. China
| |
Collapse
|
5
|
Heras-Murillo I, Adán-Barrientos I, Galán M, Wculek SK, Sancho D. Dendritic cells as orchestrators of anticancer immunity and immunotherapy. Nat Rev Clin Oncol 2024; 21:257-277. [PMID: 38326563 DOI: 10.1038/s41571-024-00859-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/09/2024] [Indexed: 02/09/2024]
Abstract
Dendritic cells (DCs) are a heterogeneous group of antigen-presenting innate immune cells that regulate adaptive immunity, including against cancer. Therefore, understanding the precise activities of DCs in tumours and patients with cancer is important. The classification of DC subsets has historically been based on ontogeny; however, single-cell analyses are now additionally revealing a diversity of functional states of DCs in cancer. DCs can promote the activation of potent antitumour T cells and immune responses via numerous mechanisms, although they can also be hijacked by tumour-mediated factors to contribute to immune tolerance and cancer progression. Consequently, DC activities are often key determinants of the efficacy of immunotherapies, including immune-checkpoint inhibitors. Potentiating the antitumour functions of DCs or using them as tools to orchestrate short-term and long-term anticancer immunity has immense but as-yet underexploited therapeutic potential. In this Review, we outline the nature and emerging complexity of DC states as well as their functions in regulating adaptive immunity across different cancer types. We also describe how DCs are required for the success of current immunotherapies and explore the inherent potential of targeting DCs for cancer therapy. We focus on novel insights on DCs derived from patients with different cancers, single-cell studies of DCs and their relevance to therapeutic strategies.
Collapse
Affiliation(s)
- Ignacio Heras-Murillo
- Immunobiology Laboratory, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Irene Adán-Barrientos
- Immunobiology Laboratory, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Miguel Galán
- Immunobiology Laboratory, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Stefanie K Wculek
- Innate Immune Biology Laboratory, Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain.
| | - David Sancho
- Immunobiology Laboratory, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain.
| |
Collapse
|
6
|
Nguyen DC, Song K, Jokonya S, Yazdani O, Sellers DL, Wang Y, Zakaria ABM, Pun SH, Stayton PS. Mannosylated STING Agonist Drugamers for Dendritic Cell-Mediated Cancer Immunotherapy. ACS CENTRAL SCIENCE 2024; 10:666-675. [PMID: 38559305 PMCID: PMC10979423 DOI: 10.1021/acscentsci.3c01310] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 01/22/2024] [Accepted: 02/06/2024] [Indexed: 04/04/2024]
Abstract
The Stimulator of Interferon Genes (STING) pathway is a promising target for cancer immunotherapy. Despite recent advances, therapies targeting the STING pathway are often limited by routes of administration, suboptimal STING activation, or off-target toxicity. Here, we report a dendritic cell (DC)-targeted polymeric prodrug platform (polySTING) that is designed to optimize intracellular delivery of a diamidobenzimidazole (diABZI) small-molecule STING agonist while minimizing off-target toxicity after parenteral administration. PolySTING incorporates mannose targeting ligands as a comonomer, which facilitates its uptake in CD206+/mannose receptor+ professional antigen-presenting cells (APCs) in the tumor microenvironment (TME). The STING agonist is conjugated through a cathepsin B-cleavable valine-alanine (VA) linker for selective intracellular drug release after receptor-mediated endocytosis. When administered intravenously in tumor-bearing mice, polySTING selectively targeted CD206+/mannose receptor+ APCs in the TME, resulting in increased cross-presenting CD8+ DCs, infiltrating CD8+ T cells in the TME as well as maturation across multiple DC subtypes in the tumor-draining lymph node (TDLN). Systemic administration of polySTING slowed tumor growth in a B16-F10 murine melanoma model as well as a 4T1 murine breast cancer model with an acceptable safety profile. Thus, we demonstrate that polySTING delivers STING agonists to professional APCs after systemic administration, generating efficacious DC-driven antitumor immunity with minimal side effects. This new polymeric prodrug platform may offer new opportunities for combining efficient targeted STING agonist delivery with other selective tumor therapeutic strategies.
Collapse
Affiliation(s)
- Dinh Chuong Nguyen
- Molecular
Engineering & Sciences Institute, University
of Washington, Seattle, Washington 98195, United States
| | - Kefan Song
- Department
of Bioengineering, University of Washington, Seattle, Washington 98195, United States
| | - Simbarashe Jokonya
- Department
of Bioengineering, University of Washington, Seattle, Washington 98195, United States
| | - Omeed Yazdani
- Department
of Bioengineering, University of Washington, Seattle, Washington 98195, United States
| | - Drew L. Sellers
- Department
of Bioengineering, University of Washington, Seattle, Washington 98195, United States
| | - Yonghui Wang
- Department
of Bioengineering, University of Washington, Seattle, Washington 98195, United States
| | - ABM Zakaria
- Department
of Bioengineering, University of Washington, Seattle, Washington 98195, United States
| | - Suzie H. Pun
- Molecular
Engineering & Sciences Institute, University
of Washington, Seattle, Washington 98195, United States
- Department
of Bioengineering, University of Washington, Seattle, Washington 98195, United States
| | - Patrick S. Stayton
- Molecular
Engineering & Sciences Institute, University
of Washington, Seattle, Washington 98195, United States
- Department
of Bioengineering, University of Washington, Seattle, Washington 98195, United States
| |
Collapse
|
7
|
Wang L, Yu Z, Zhang J, Guo J. Nanoformulations of chemotherapeutic activators of the cGAS-STING pathway in tumor chemoimmunotherapy. Drug Discov Today 2024; 29:103892. [PMID: 38272174 DOI: 10.1016/j.drudis.2024.103892] [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: 10/31/2023] [Revised: 01/08/2024] [Accepted: 01/16/2024] [Indexed: 01/27/2024]
Abstract
Chemotherapeutic drugs to activate the cyclic guanosine monophosphate-adenosine monophosphate synthase (cGAS)-stimulator of interferon genes (STING) pathway have been exploited for tumor chemoimmunotherapy. The clinical translation of chemotherapeutic cGAS-STING activators is hindered by the lack of safe, efficient, and specific delivery strategies. Nanodrug delivery systems (NDDS) designed for reducing toxic effects and improving transport effectiveness potentiate in vivo delivery of chemotherapeutic cGAS-STING activators. cGAS-STING monotherapy often encounters tumor resistance without providing satisfactory therapeutic benefit; therefore combination therapy is desirable. This review describes NDDS strategies for surmounting delivery obstacles of chemotherapeutic cGAS-STING activators and highlights combinatorial regimens, which utilize therapeutics that work by different mechanisms, for optimal therapy.
Collapse
Affiliation(s)
- Lingzhi Wang
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Zhuo Yu
- Department of Hepatopathy, Shuguang Hospital, Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jihong Zhang
- Hematology Laboratory, Shengjing Hospital of China Medical University, Shenyang 110022, China.
| | - Jianfeng Guo
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China.
| |
Collapse
|
8
|
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
|
9
|
Wang Y, Liu Y, Zhang J, Peng Q, Wang X, Xiao X, Shi K. Nanomaterial-mediated modulation of the cGAS-STING signaling pathway for enhanced cancer immunotherapy. Acta Biomater 2024; 176:51-76. [PMID: 38237711 DOI: 10.1016/j.actbio.2024.01.008] [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: 10/30/2023] [Revised: 12/30/2023] [Accepted: 01/09/2024] [Indexed: 01/27/2024]
Abstract
Despite the current promise of immunotherapy, many cancer patients still suffer from challenges such as poor immune response rates, resulting in unsatisfactory clinical efficacy of existing therapies. There is an urgent need to combine emerging biomedical discoveries and innovations in traditional therapies. Modulation of the cGAS-STING signalling pathway represents an important innate immunotherapy pathway that serves as a crucial DNA sensing mechanism in innate immunity and viral defense. It has attracted increasing attention as an emerging target for cancer therapy. The recent advancements in nanotechnology have led to the significant utilization of nanomaterials in cancer immunotherapy, owing to their exceptional physicochemical properties such as large specific surface area and efficient permeability. Given the rapid development of cancer immunotherapy driven by the cGAS-STING activation, this study reviews the latest research progress in employing nanomaterials to modulate this signaling pathway. Based on the introduction of the main activation mechanisms of cGAS-STING pathway, this review focuses on nanomaterials that mediate the agonists involved and effectively activate this signaling pathway. In addition, combination nanotherapeutics based on the activation of the cGAS-STING signaling pathway are also discussed, including emerging strategies combining nanoformulated agonists with chemotherapy, radiotherapy as well as other immunomodulation in tumor targeting therapy. STATEMENT OF SIGNIFICANCE: Given the rapid development of cancer immunotherapy driven by the cGAS / STING activation, this study reviews the latest research advances in the use of nanomaterials to modulate this signaling pathway. Based on the introduction of key cGAS-STING components and their activation mechanisms, this review focuses on nanomaterials that can mediate the corresponding agonists and effectively activate this signaling pathway. In addition, combination nanotherapies based on the activation of the cGAS-STING signaling pathway are also discussed, including emerging strategies combining nanoformulated agonists with chemotherapy, radiotherapy as well as immunomodulation in cancer therapy,.
Collapse
Affiliation(s)
- Yaxin Wang
- College of Pharmacy, Nankai University, Tianjin 300350, PR China
| | - Yunmeng Liu
- College of Pharmacy, Nankai University, Tianjin 300350, PR China
| | - Jincheng Zhang
- College of Pharmacy, Nankai University, Tianjin 300350, PR China
| | - Qikai Peng
- College of Pharmacy, Nankai University, Tianjin 300350, PR China
| | - Xingdong Wang
- College of Pharmacy, Nankai University, Tianjin 300350, PR China
| | - Xiyue Xiao
- College of Pharmacy, Nankai University, Tianjin 300350, PR China
| | - Kai Shi
- College of Pharmacy, Nankai University, Tianjin 300350, PR China.
| |
Collapse
|
10
|
Liu X, Cheng Y, Mu Y, Zhang Z, Tian D, Liu Y, Hu X, Wen T. Diverse drug delivery systems for the enhancement of cancer immunotherapy: an overview. Front Immunol 2024; 15:1328145. [PMID: 38298192 PMCID: PMC10828056 DOI: 10.3389/fimmu.2024.1328145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 01/03/2024] [Indexed: 02/02/2024] Open
Abstract
Despite the clear benefits demonstrated by immunotherapy, there is still an inevitable off-target effect resulting in serious adverse immune reactions. In recent years, the research and development of Drug Delivery System (DDS) has received increased prominence. In decades of development, DDS has demonstrated the ability to deliver drugs in a precisely targeted manner to mitigate side effects and has the advantages of flexible control of drug release, improved pharmacokinetics, and drug distribution. Therefore, we consider that combining cancer immunotherapy with DDS can enhance the anti-tumor ability. In this paper, we provide an overview of the latest drug delivery strategies in cancer immunotherapy and briefly introduce the characteristics of DDS based on nano-carriers (liposomes, polymer nano-micelles, mesoporous silica, extracellular vesicles, etc.) and coupling technology (ADCs, PDCs and targeted protein degradation). Our aim is to show readers a variety of drug delivery platforms under different immune mechanisms, and analyze their advantages and limitations, to provide more superior and accurate targeting strategies for cancer immunotherapy.
Collapse
Affiliation(s)
- Xu Liu
- Department of Respiratory and Infectious Disease of Geriatrics, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yang Cheng
- Department of Respiratory and Infectious Disease of Geriatrics, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yao Mu
- Department of Respiratory and Infectious Disease of Geriatrics, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | | | - Dan Tian
- Department of Thoracic Surgery, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Yunpeng Liu
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, Liaoning, China
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, Liaoning, China
- Liaoning Province Clinical Research Center for Cancer, The First Hospital of China Medical University, Shenyang, Liaoning, China
- Clinical Cancer Treatment and Research Center of Shenyang, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xuejun Hu
- Department of Respiratory and Infectious Disease of Geriatrics, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Ti Wen
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, Liaoning, China
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, Liaoning, China
- Liaoning Province Clinical Research Center for Cancer, The First Hospital of China Medical University, Shenyang, Liaoning, China
- Clinical Cancer Treatment and Research Center of Shenyang, The First Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
11
|
Huang C, Shao N, Huang Y, Chen J, Wang D, Hu G, Zhang H, Luo L, Xiao Z. Overcoming challenges in the delivery of STING agonists for cancer immunotherapy: A comprehensive review of strategies and future perspectives. Mater Today Bio 2023; 23:100839. [PMID: 38024837 PMCID: PMC10630661 DOI: 10.1016/j.mtbio.2023.100839] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 10/12/2023] [Accepted: 10/19/2023] [Indexed: 12/01/2023] Open
Abstract
STING (Stimulator of Interferon Genes) agonists have emerged as promising agents in the field of cancer immunotherapy, owing to their excellent capacity to activate the innate immune response and combat tumor-induced immunosuppression. This review provides a comprehensive exploration of the strategies employed to develop effective formulations for STING agonists, with particular emphasis on versatile nano-delivery systems. The recent advancements in delivery systems based on lipids, natural/synthetic polymers, and proteins for STING agonists are summarized. The preparation methodologies of nanoprecipitation, self-assembly, and hydrogel, along with their advantages and disadvantages, are also discussed. Furthermore, the challenges and opportunities in developing next-generation STING agonist delivery systems are elaborated. This review aims to serve as a reference for researchers in designing novel and effective STING agonist delivery systems for cancer immunotherapy.
Collapse
Affiliation(s)
- Cuiqing Huang
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, The First Affiliated Hospital of Jinan University, Guangzhou, 510632, China
- Department of Ultrasound, Guangdong Women and Children Hospital, Guangzhou, 511400, China
| | - Ni Shao
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, The First Affiliated Hospital of Jinan University, Guangzhou, 510632, China
| | - Yanyu Huang
- Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, CA, 95817, USA
| | - Jifeng Chen
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, The First Affiliated Hospital of Jinan University, Guangzhou, 510632, China
| | - Duo Wang
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, The First Affiliated Hospital of Jinan University, Guangzhou, 510632, China
| | - Genwen Hu
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, The First Affiliated Hospital of Jinan University, Guangzhou, 510632, China
- Department of Radiology, Shenzhen People's Hospital, The Second Clinical Medical College of Jinan University, Shenzhen, 518020, China
| | - Hong Zhang
- Department of Interventional Vascular Surgery, The Sixth Affiliated Hospital of Jinan University, Dongguan, 523560, China
| | - Liangping Luo
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, The First Affiliated Hospital of Jinan University, Guangzhou, 510632, China
| | - Zeyu Xiao
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, The First Affiliated Hospital of Jinan University, Guangzhou, 510632, China
| |
Collapse
|
12
|
Zhou S, Cheng F, Zhang Y, Su T, Zhu G. Engineering and Delivery of cGAS-STING Immunomodulators for the Immunotherapy of Cancer and Autoimmune Diseases. Acc Chem Res 2023; 56:2933-2943. [PMID: 37802125 PMCID: PMC10882213 DOI: 10.1021/acs.accounts.3c00394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/08/2023]
Abstract
The cyclic GMP-AMP synthase-stimulator interferon gene (cGAS-STING) pathway is an emerging therapeutic target for the prophylaxis and therapy of a variety of diseases, ranging from cancer, infectious diseases, to autoimmune disorders. As a cytosolic double stranded DNA (dsDNA) sensor, cGAS can bind with relatively long dsDNA, resulting in conformational change and activation of cGAS. Activated cGAS catalyzes the conversion of adenosine triphosphate (ATP) and guanosine triphosphate (GTP) into cGAMP, a cyclic dinucleotide (CDN). CDNs, including 2'3'-cGAMP, stimulate adapter protein STING on the endoplasmic membrane, triggering interferon regulatory factor 3 (IRF3) phosphorylation and nuclear factor kappa B (NF-κB) activation. This results in antitumor and antiviral type I interferon (IFN-I) responses. Moreover, cGAS-STING overactivation and the resulting IFN-I responses have been associated with a number of inflammatory and autoimmune diseases. This makes cGAS-STING appealing immunomodulatory targets for the prophylaxis and therapy of various related diseases. However, drug development of CDNs and CDN derivatives is challenged by their limited biostability, difficult formulation, poor pharmacokinetics, and inefficient tissue accumulation and cytosolic delivery. Though recent synthetic small molecular CDN- or non-CDN-based STING agonists have been reported with promising preclinical therapeutic efficacy, their therapeutic efficacy and safety remain to be fully evaluated preclinically and clinically. Therefore, it is highly desirable and clinically significant to advance drug development for cGAS-STING activation by innovative approaches, such as drug delivery systems and drug development for pharmacological immunomodulation of cGAS. In this Account, we summarize our recent research in the engineering and delivery of immunostimulatory or immunoregulatory modulators for cGAS and STING for the immunotherapy of cancer and autoimmune diseases. To improve the delivery efficiency of CDNs, we developed ionizable and pH-responsive polymeric nanocarriers to load STING agonists, aiming to improve the cellular uptake and facilitate the endosomal escape to induce efficient STING activation. We also codelivered STING agonists with complementary immunostimulatants in nanoparticle-in-hydrogel composites to synergetically elicit potent innate and adaptive antitumor responses that eradicate local and distant large tumors. Further, taking advantage of the simplicity of manufacturing and the established nucleic acid delivery system, we developed oligonucleotide-based cGAS agonists as immunostimulant immunotherapeutics as well as adjuvants for peptide antigens for cancer immunotherapy. To suppress the overly strong proinflammatory responses associated with cGAS-STING overactivation in some of the autoimmune disorders, we devised nanomedicine-in-hydrogel (NiH) that codelivers a cGAS inhibitor and cell-free DNA (cfDNA)-scavenging cationic nanoparticles (cNPs) for systemic immunosuppression in rheumatoid arthritis (RA) therapy. Lastly, we discussed current drug development by targeting cGAS-STING for cancer, infectious diseases, and autoimmune diseases, as well as the potential opportunities for utilizing cGAS-STING pathway for versatile applications in disease treatment.
Collapse
Affiliation(s)
- Shurong Zhou
- Department of Pharmaceutical Sciences, College of Pharmacy; Biointerfaces Institute. University of Michigan. Ann Arbor, Michigan 48109, United States
| | - Furong Cheng
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200032, China
| | - Yu Zhang
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 31002, China
| | - Ting Su
- Department of Pharmaceutics, School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia 23298, United States
| | - Guizhi Zhu
- Department of Pharmaceutical Sciences, College of Pharmacy; Biointerfaces Institute. University of Michigan. Ann Arbor, Michigan 48109, United States
| |
Collapse
|
13
|
Liu Q, Zou J, Chen Z, He W, Wu W. Current research trends of nanomedicines. Acta Pharm Sin B 2023; 13:4391-4416. [PMID: 37969727 PMCID: PMC10638504 DOI: 10.1016/j.apsb.2023.05.018] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 04/25/2023] [Accepted: 05/05/2023] [Indexed: 11/17/2023] Open
Abstract
Owing to the inherent shortcomings of traditional therapeutic drugs in terms of inadequate therapeutic efficacy and toxicity in clinical treatment, nanomedicine designs have received widespread attention with significantly improved efficacy and reduced non-target side effects. Nanomedicines hold tremendous theranostic potential for treating, monitoring, diagnosing, and controlling various diseases and are attracting an unfathomable amount of input of research resources. Against the backdrop of an exponentially growing number of publications, it is imperative to help the audience get a panorama image of the research activities in the field of nanomedicines. Herein, this review elaborates on the development trends of nanomedicines, emerging nanocarriers, in vivo fate and safety of nanomedicines, and their extensive applications. Moreover, the potential challenges and the obstacles hindering the clinical translation of nanomedicines are also discussed. The elaboration on various aspects of the research trends of nanomedicines may help enlighten the readers and set the route for future endeavors.
Collapse
Affiliation(s)
- Qiuyue Liu
- Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai 200443, China
- Key Laboratory of Smart Drug Delivery of MOE, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Jiahui Zou
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Zhongjian Chen
- Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai 200443, China
| | - Wei He
- Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai 200443, China
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Wei Wu
- Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai 200443, China
- Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, China
- Key Laboratory of Smart Drug Delivery of MOE, School of Pharmacy, Fudan University, Shanghai 201203, China
- Fudan Zhangjiang Institute, Shanghai 201203, China
| |
Collapse
|
14
|
Go EJ, Yang H, Park W, Lee SJ, Han JH, Kong SJ, Lee WS, Han DK, Chon HJ, Kim C. Systemic Delivery of a STING Agonist-Loaded Positively Charged Liposome Selectively Targets Tumor Immune Microenvironment and Suppresses Tumor Angiogenesis. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2300544. [PMID: 37381624 DOI: 10.1002/smll.202300544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 06/13/2023] [Indexed: 06/30/2023]
Abstract
Although stimulator of interferon genes (STING) agonists has shown great promise in preclinical studies, the clinical development of STING agonist therapy is challenged by its limited systemic delivery. Here, positively charged fusogenic liposomes loaded with a STING agonist (PoSTING) are designed for systemic delivery and to preferentially target the tumor microenvironment. When PoSTING is administered intravenously, it selectively targets not only tumor cells but also immune and tumor endothelial cells (ECs). In particular, delivery of STING agonists to tumor ECs normalizes abnormal tumor vasculatures, induces intratumoral STING activation, and elicits robust anti-tumor T cell immunity within the tumor microenvironment. Therefore, PoSTING can be used as a systemic delivery platform to overcome the limitations of using STING agonists in clinical trials.
Collapse
Affiliation(s)
- Eun-Jin Go
- Medical Oncology, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, Gyeonggi, 13496, Republic of Korea
- Laboratory of Translational Immuno-Oncology, CHA University School of Medicine, Seongnam, Gyeonggi, 13496, Republic of Korea
| | - Hannah Yang
- Medical Oncology, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, Gyeonggi, 13496, Republic of Korea
- Laboratory of Translational Immuno-Oncology, CHA University School of Medicine, Seongnam, Gyeonggi, 13496, Republic of Korea
| | - Wooram Park
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Seoburo 2066, Suwon, Gyeonggi, 16419, Republic of Korea
| | - Seung Joon Lee
- Medical Oncology, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, Gyeonggi, 13496, Republic of Korea
- Laboratory of Translational Immuno-Oncology, CHA University School of Medicine, Seongnam, Gyeonggi, 13496, Republic of Korea
| | - Jun-Hyeok Han
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Seoburo 2066, Suwon, Gyeonggi, 16419, Republic of Korea
| | - So Jung Kong
- Medical Oncology, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, Gyeonggi, 13496, Republic of Korea
- Laboratory of Translational Immuno-Oncology, CHA University School of Medicine, Seongnam, Gyeonggi, 13496, Republic of Korea
| | - Won Suk Lee
- Medical Oncology, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, Gyeonggi, 13496, Republic of Korea
- Laboratory of Translational Immuno-Oncology, CHA University School of Medicine, Seongnam, Gyeonggi, 13496, Republic of Korea
| | - Dong Keun Han
- Department of Biomedical Science, CHA University, Seongnam, Gyeonggi, 13496, Republic of Korea
| | - Hong Jae Chon
- Medical Oncology, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, Gyeonggi, 13496, Republic of Korea
- Laboratory of Translational Immuno-Oncology, CHA University School of Medicine, Seongnam, Gyeonggi, 13496, Republic of Korea
| | - Chan Kim
- Medical Oncology, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, Gyeonggi, 13496, Republic of Korea
- Laboratory of Translational Immuno-Oncology, CHA University School of Medicine, Seongnam, Gyeonggi, 13496, Republic of Korea
| |
Collapse
|
15
|
Hao Y, Ji Z, Zhou H, Wu D, Gu Z, Wang D, ten Dijke P. Lipid-based nanoparticles as drug delivery systems for cancer immunotherapy. MedComm (Beijing) 2023; 4:e339. [PMID: 37560754 PMCID: PMC10407046 DOI: 10.1002/mco2.339] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Revised: 06/26/2023] [Accepted: 07/04/2023] [Indexed: 08/11/2023] Open
Abstract
Immune checkpoint inhibitors (ICIs) have shown remarkable success in cancer treatment. However, in cancer patients without sufficient antitumor immunity, numerous data indicate that blocking the negative signals elicited by immune checkpoints is ineffective. Drugs that stimulate immune activation-related pathways are emerging as another route for improving immunotherapy. In addition, the development of nanotechnology presents a promising platform for tissue and cell type-specific delivery and improved uptake of immunomodulatory agents, ultimately leading to enhanced cancer immunotherapy and reduced side effects. In this review, we summarize and discuss the latest developments in nanoparticles (NPs) for cancer immuno-oncology therapy with a focus on lipid-based NPs (lipid-NPs), including the characteristics and advantages of various types. Using the agonists targeting stimulation of the interferon genes (STING) transmembrane protein as an exemplar, we review the potential of various lipid-NPs to augment STING agonist therapy. Furthermore, we present recent findings and underlying mechanisms on how STING pathway activation fosters antitumor immunity and regulates the tumor microenvironment and provide a summary of the distinct STING agonists in preclinical studies and clinical trials. Ultimately, we conduct a critical assessment of the obstacles and future directions in the utilization of lipid-NPs to enhance cancer immunotherapy.
Collapse
Affiliation(s)
- Yang Hao
- Department of Laboratory AnimalsCollege of Animal SciencesJilin UniversityChangchunChina
- Department of Basic MedicineChangzhi Medical CollegeChangzhiChina
- Department of Cell and Chemical Biology and Oncode InstituteLeiden University Medical CenterLeidenThe Netherlands
| | - Zhonghao Ji
- Department of Laboratory AnimalsCollege of Animal SciencesJilin UniversityChangchunChina
- Department of Basic MedicineChangzhi Medical CollegeChangzhiChina
| | - Hengzong Zhou
- Department of Laboratory AnimalsCollege of Animal SciencesJilin UniversityChangchunChina
| | - Dongrun Wu
- Departure of Philosophy, Faculty of HumanitiesLeiden UniversityLeidenThe Netherlands
| | - Zili Gu
- Department of RadiologyLeiden University Medical CenterLeidenThe Netherlands
| | - Dongxu Wang
- Department of Laboratory AnimalsCollege of Animal SciencesJilin UniversityChangchunChina
| | - Peter ten Dijke
- Department of Cell and Chemical Biology and Oncode InstituteLeiden University Medical CenterLeidenThe Netherlands
| |
Collapse
|
16
|
Liu Y, Fei Y, Wang X, Yang B, Li M, Luo Z. Biomaterial-enabled therapeutic modulation of cGAS-STING signaling for enhancing antitumor immunity. Mol Ther 2023; 31:1938-1959. [PMID: 37002605 PMCID: PMC10362396 DOI: 10.1016/j.ymthe.2023.03.026] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 03/07/2023] [Accepted: 03/23/2023] [Indexed: 04/03/2023] Open
Abstract
cGAS-STING signaling is a central component in the therapeutic action of most existing cancer therapies. The accumulated knowledge of tumor immunoregulatory network in recent years has spurred the development of cGAS-STING agonists for tumor treatment as an effective immunotherapeutic strategy. However, the clinical translation of these agonists is thus far unsatisfactory because of the low immunostimulatory efficacy and unrestricted side effects under clinically relevant conditions. Interestingly, the rational integration of biomaterial technology offers a promising approach to overcome these limitations for more effective and safer cGAS-STING-mediated tumor therapy. Herein, we first outline the cGAS-STING signaling axis and generally discuss its association with tumors. We then symmetrically summarize the recent progress in those biomaterial-based cGAS-STING agonism strategies to generate robust antitumor immunity, categorized by the chemical nature of those cGAS-STING stimulants and carrier substrates. Finally, a perspective is provided to discuss the existing challenges and potential opportunities in cGAS-STING modulation for tumor therapy.
Collapse
Affiliation(s)
- Yingqi Liu
- School of Life Science, Chongqing University, Chongqing 400044, P. R. China
| | - Yang Fei
- School of Life Science, Chongqing University, Chongqing 400044, P. R. China
| | - Xuan Wang
- School of Life Science, Chongqing University, Chongqing 400044, P. R. China
| | - Bingbing Yang
- School of Life Science, Chongqing University, Chongqing 400044, P. R. China
| | - Menghuan Li
- School of Life Science, Chongqing University, Chongqing 400044, P. R. China.
| | - Zhong Luo
- School of Life Science, Chongqing University, Chongqing 400044, P. R. China.
| |
Collapse
|
17
|
Hu Y, Zhang W, Chu X, Wang A, He Z, Si CL, Hu W. Dendritic cell-targeting polymer nanoparticle-based immunotherapy for cancer: A review. Int J Pharm 2023; 635:122703. [PMID: 36758880 DOI: 10.1016/j.ijpharm.2023.122703] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 02/01/2023] [Accepted: 02/04/2023] [Indexed: 02/10/2023]
Abstract
Cancer immunity is dependent on dynamic interactions between T cells and dendritic cells (DCs). Polymer-based nanoparticles target DC receptors to improve anticancer immune responses. In this paper, DC surface receptors and their specific coupling natural ligands and antibodies are reviewed and compared. Moreover, reaction mechanisms are described, and the synergistic effects of immune adjuvants are demonstrated. Also, extracellular-targeting antigen-delivery strategies and intracellular stimulus responses are reviewed to promote the rational design of polymer delivery systems.
Collapse
Affiliation(s)
- Yeye Hu
- Institute of Translational Medicine, School of Medicine, Yangzhou University, Yangzhou 225009, China; Tianjin Key Laboratory of Pulp & Paper, Tianjin University of Science & Technology, Tianjin 300457, China
| | - Wei Zhang
- School of Life Sciences, Huaiyin Normal University, Huaian 223300, China
| | - Xiaozhong Chu
- School of Chemistry & Chemical Engineering, Huaiyin Normal University, Huaian 223300, China
| | - Aoran Wang
- School of Chemistry & Chemical Engineering, Huaiyin Normal University, Huaian 223300, China
| | - Ziliang He
- School of Life Sciences, Huaiyin Normal University, Huaian 223300, China
| | - Chuan-Ling Si
- Tianjin Key Laboratory of Pulp & Paper, Tianjin University of Science & Technology, Tianjin 300457, China.
| | - Weicheng Hu
- Institute of Translational Medicine, School of Medicine, Yangzhou University, Yangzhou 225009, China; Affiliated Hospital of Yangzhou University, Yangzhou 225009, China; Jiangsu Key Laboratory of Experimental & Translational Non-Coding RNA Research, School of Medicine, Yangzhou University, Yangzhou 225009, China.
| |
Collapse
|
18
|
Fang H, Wu Y, Chen L, Cao Z, Deng Z, Zhao R, Zhang L, Yang Y, Liu Z, Chen Q. Regulating the Obesity-Related Tumor Microenvironment to Improve Cancer Immunotherapy. ACS NANO 2023; 17:4748-4763. [PMID: 36809912 DOI: 10.1021/acsnano.2c11159] [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: 06/18/2023]
Abstract
Obesity usually induces systemic metabolic disturbances, including in the tumor microenvironment (TME). This is because adaptive metabolism related to obesity in the TME with a low level of prolyl hydroxylase-3 (PHD3) depletes the major fatty acid fuels of CD8+ T cells and leads to the poor infiltration and unsatisfactory function of CD8+ T cells. Herein, we discovered that obesity could aggravate the immunosuppressive TME and weaken CD8+ T cell-mediated tumor cell killing. We have thus developed gene therapy to relieve the obesity-related TME to promote cancer immunotherapy. An efficient gene carrier was prepared by modifying polyethylenimine with p-methylbenzenesulfonyl (abbreviated as PEI-Tos) together with hyaluronic acid (HA) shielding, achieving excellent gene transfection in tumors after intravenous administration. HA/PEI-Tos/pDNA (HPD) containing the plasmid encoding PHD3 (pPHD3) can effectively upregulate the expression of PHD3 in tumor tissues, revising the immunosuppressive TME and significantly increasing the infiltration of CD8+ T cells, thereby improving the responsiveness of immune checkpoint antibody-mediated immunotherapy. Efficient therapeutic efficacy was achieved using HPD together with αPD-1 in colorectal tumor and melanoma-bearing obese mice. This work provides an effective strategy to improve immunotherapy of tumors in obese mice, which may provide a useful reference for the immunotherapy of obesity-related cancer in the clinic.
Collapse
Affiliation(s)
- Huapan Fang
- Institute of Functional Nano and Soft Materials, Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou 215123, China
- College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Yicheng Wu
- Institute of Functional Nano and Soft Materials, Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou 215123, China
| | - Linfu Chen
- Institute of Functional Nano and Soft Materials, Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou 215123, China
| | - Zhiqin Cao
- Institute of Functional Nano and Soft Materials, Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou 215123, China
| | - Zheng Deng
- Institute of Functional Nano and Soft Materials, Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou 215123, China
| | - Rui Zhao
- Institute of Functional Nano and Soft Materials, Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou 215123, China
| | - Lin Zhang
- Institute of Functional Nano and Soft Materials, Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou 215123, China
| | - Yang Yang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
| | - Zhuang Liu
- Institute of Functional Nano and Soft Materials, Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou 215123, China
| | - Qian Chen
- Institute of Functional Nano and Soft Materials, Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou 215123, China
| |
Collapse
|
19
|
Li Y, Li X, Yi J, Cao Y, Qin Z, Zhong Z, Yang W. Nanoparticle-Mediated STING Activation for Cancer Immunotherapy. Adv Healthc Mater 2023:e2300260. [PMID: 36905358 DOI: 10.1002/adhm.202300260] [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: 01/25/2023] [Revised: 03/02/2023] [Indexed: 03/12/2023]
Abstract
As the first line of host defense against pathogenic infections, innate immunity plays a key role in antitumor immunotherapy. The cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) (cGAS-STING) pathway has attracted much attention because of the secretion of various proinflammatory cytokines and chemokines. Many STING agonists have been identified and applied into preclinical or clinical trials for cancer immunotherapy. However, the fast excretion, low bioavailability, nonspecificity, and adverse effects of the small molecule STING agonists limit their therapeutic efficacy and in vivo application. Nanodelivery systems with appropriate size, charge, and surface modification are capable of addressing these dilemmas. In this review, the mechanism of the cGAS-STING pathway is discussed and the STING agonists, focusing on nanoparticle-mediated STING therapy and combined therapy for cancers, are summarized. Finally, the future direction and challenges of nano-STING therapy are expounded, emphasizing the pivotal scientific problems and technical bottlenecks and hoping to provide general guidance for its clinical application.
Collapse
Affiliation(s)
- Yongjuan Li
- Medical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, 450001, China.,The Center of Infection and Immunity, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Xinyan Li
- Medical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, 450001, China.,The Center of Infection and Immunity, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Jinmeng Yi
- Medical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, 450001, China.,The Center of Infection and Immunity, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Yongjian Cao
- School of Pharmaceutical Sciences, Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Zhihai Qin
- Medical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, 450001, China.,The Center of Infection and Immunity, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Zhiyuan Zhong
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, State Key Laboratory of Radiation Medicine and Protection, and College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Weijing Yang
- School of Pharmaceutical Sciences, Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou University, Zhengzhou, Henan, 450001, China
| |
Collapse
|
20
|
Nanotechnology for next-generation cancer immunotherapy: State of the art and future perspectives. J Control Release 2023; 356:14-25. [PMID: 36805873 DOI: 10.1016/j.jconrel.2023.02.016] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 02/10/2023] [Indexed: 02/23/2023]
Abstract
Over the past decade, immunotherapy aiming to activate an effective antitumor immune response has ushered in a new era of cancer treatment. However, the efficacy of cancer immunotherapy is limited by low response rates and high systemic toxicity. Nanotechnology is an encouraging platform for the development of next-generation cancer immunotherapy to effectively treat advanced cancer. Nanotechnology-enabled immunotherapy has remarkable advantages, ranging from the increased bioavailability and stability of immunotherapeutic agents to the enhanced activation of immune cells and favorable safety profiles. Nanotechnology-enabled immunotherapy can target solid tumors through reprogramming or stimulating immune cells (i.e., nanovaccines); modulating the immunosuppressive tumor microenvironment; or targeting tumor cells and altering their responses to immune cells to generate effective antitumor immunity. In this Oration, I introduce the advanced strategies currently being pursued by our laboratory and other groups to improve the therapeutic efficacy of cancer immunotherapy and discuss the potential challenges and future directions.
Collapse
|
21
|
Liposomal Delivery of MIW815 (ADU-S100) for Potentiated STING Activation. Pharmaceutics 2023; 15:pharmaceutics15020638. [PMID: 36839960 PMCID: PMC9966736 DOI: 10.3390/pharmaceutics15020638] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 02/03/2023] [Accepted: 02/10/2023] [Indexed: 02/16/2023] Open
Abstract
Stimulator of interferon genes (STING) agonists can improve the anticancer efficacy of immune checkpoint blockade by amplifying tumor immunogenicity. However, the clinical translation of cyclic dinucleotides (CDNs) as STING agonists is hindered by their poor drug-like properties. In this study, we investigated the design criteria for DOTAP/cholesterol liposomes for the systemic delivery of ADU-S100 and delineated the impact of key formulation factors on the loading efficiency, serum stability, and STING agonistic activity of ADU-S100. Our findings demonstrate that the cationic liposomal formulation of ADU-S100 can be optimized to greatly potentiate STING activation in antigen-presenting cells.
Collapse
|
22
|
Chen S, Peng A, Chen M, Zhan M. Nanomedicines targeting activation of STING to reshape tumor immune microenvironment and enhance immunotherapeutic efficacy. Front Oncol 2023; 12:1093240. [PMID: 36741735 PMCID: PMC9890065 DOI: 10.3389/fonc.2022.1093240] [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: 11/08/2022] [Accepted: 12/29/2022] [Indexed: 01/19/2023] Open
Abstract
Immunotherapy has greatly enhanced the effectiveness of cancer treatments, but the efficacy of many current immunotherapies is still limited by the tumor-suppressive immune microenvironment. Multiple studies have shown that activating the stimulation of IFN genes (STING) pathway and inducing innate immunity can significantly impact the tumor immune microenvironment and improve antitumor therapy. While natural or synthetic STING agonists have been identified or developed for preclinical and clinical use, small molecule agonists have limited utility due to degradation and lack of targeting. As such, the delivery and release of STING agonists into tumor tissue is a major challenge that must be addressed in order to further advance the use of STING agonists. To address this challenge, various nanomedicines have been developed. In this paper, we concisely review the antitumor immunotherapeutic mechanisms of STING agonists, highlighting the latest developments in STING agonists and the current progress of nanomedicines for activating STING. We classify the different nanomedicines according to the STING agonists they utilize in order to facilitate understanding of recent advances in this field. Finally, we also discuss the prospects and challenges of this field.
Collapse
Affiliation(s)
- Shanshan Chen
- Zhuhai Interventional Medical Center, Zhuhai Precision Medical Center, Zhuhai People’s Hospital, Zhuhai Hospital Affiliated with Jinan University, Jinan University, Zhuhai, China,Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People’s Hospital Affiliated with Jinan University, Jinan University, Zhuhai, China
| | - Anghui Peng
- Zhuhai Interventional Medical Center, Zhuhai Precision Medical Center, Zhuhai People’s Hospital, Zhuhai Hospital Affiliated with Jinan University, Jinan University, Zhuhai, China,Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People’s Hospital Affiliated with Jinan University, Jinan University, Zhuhai, China
| | - Muhe Chen
- Zhuhai Interventional Medical Center, Zhuhai Precision Medical Center, Zhuhai People’s Hospital, Zhuhai Hospital Affiliated with Jinan University, Jinan University, Zhuhai, China,Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People’s Hospital Affiliated with Jinan University, Jinan University, Zhuhai, China,*Correspondence: Muhe Chen, ; Meixiao Zhan,
| | - Meixiao Zhan
- Zhuhai Interventional Medical Center, Zhuhai Precision Medical Center, Zhuhai People’s Hospital, Zhuhai Hospital Affiliated with Jinan University, Jinan University, Zhuhai, China,Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People’s Hospital Affiliated with Jinan University, Jinan University, Zhuhai, China,*Correspondence: Muhe Chen, ; Meixiao Zhan,
| |
Collapse
|
23
|
Huang R, Ning Q, Zhao J, Zhao X, Zeng L, Yi Y, Tang S. Targeting STING for cancer immunotherapy: From mechanisms to translation. Int Immunopharmacol 2022; 113:109304. [DOI: 10.1016/j.intimp.2022.109304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 09/17/2022] [Accepted: 09/29/2022] [Indexed: 11/05/2022]
|
24
|
Nanodelivery of cGAS-STING activators for tumor immunotherapy. Trends Pharmacol Sci 2022; 43:957-972. [PMID: 36089410 DOI: 10.1016/j.tips.2022.08.006] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 08/13/2022] [Accepted: 08/16/2022] [Indexed: 12/24/2022]
Abstract
Activation of the cyclic guanosine monophosphate-adenosine monophosphate synthase (cGAS)-stimulator of interferon genes (STING) pathway has great potential to promote antitumor immunity. Development of activators for the cGAS-STING pathway (cGAS-STING activators) has profoundly revolutionized tumor immunotherapy. However, successful clinical application of cGAS-STING activators is contingent on having appropriate systems to achieve safe, effective, and specific delivery. There is an increasing emphasis on the design and application of nano drug delivery systems (NDDS) that can facilitate the delivery potential of cGAS-STING activators. In this review, we discuss barriers for translational development of cGAS-STING activators (DNA damaging drugs and STING agonists) and recent advances of NDDS for these agents in tumor immunotherapy.
Collapse
|