51
|
Zhou J, Ventura CJ, Fang RH, Zhang L. Nanodelivery of STING agonists against cancer and infectious diseases. Mol Aspects Med 2022; 83:101007. [PMID: 34353637 PMCID: PMC8792206 DOI: 10.1016/j.mam.2021.101007] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 07/27/2021] [Accepted: 07/28/2021] [Indexed: 02/03/2023]
Abstract
Vaccination is a modality that has been widely explored for the treatment of various diseases. To increase the potency of vaccine formulations, immunostimulatory adjuvants have been regularly exploited, and the stimulator of interferon genes (STING) signaling pathway has recently emerged as a remarkable therapeutic target. STING is an endogenous protein on the endoplasmic reticulum that is a downstream sensor to cytosolic DNA. Upon activation, STING initiates a series of intracellular signaling cascades that ultimately generate potent type I interferon-mediated immune responses. Both natural and synthetic agonists have been used to stimulate the STING pathway, but they are usually administered locally due to low bioavailability, instability, and difficulty in bypassing the plasma membrane. With excellent pharmacokinetic profiles and versatility, nanocarriers can address many of these challenges and broaden the application of STING vaccines. Along these lines, STING-inducing nanovaccines are being developed to address a wide range of diseases. In this review, we discuss the recent advances in STING nanovaccines for anticancer, antiviral, and antibacterial applications.
Collapse
Affiliation(s)
- Jiarong Zhou
- Department of NanoEngineering, Chemical Engineering Program, Moores Cancer Center, University of California San Diego, La Jolla, CA, 92093, USA
| | - Christian J Ventura
- Department of NanoEngineering, Chemical Engineering Program, Moores Cancer Center, University of California San Diego, La Jolla, CA, 92093, USA
| | - Ronnie H Fang
- Department of NanoEngineering, Chemical Engineering Program, Moores Cancer Center, University of California San Diego, La Jolla, CA, 92093, USA.
| | - Liangfang Zhang
- Department of NanoEngineering, Chemical Engineering Program, Moores Cancer Center, University of California San Diego, La Jolla, CA, 92093, USA.
| |
Collapse
|
52
|
Chen H, Zhang Y, Li L, Guo R, Shi X, Cao X. Effective CpG Delivery Using Zwitterion-Functionalized Dendrimer-Entrapped Gold Nanoparticles to Promote T Cell-Mediated Immunotherapy of Cancer Cells. BIOSENSORS 2022; 12:71. [PMID: 35200332 PMCID: PMC8869692 DOI: 10.3390/bios12020071] [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] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/23/2022] [Accepted: 01/24/2022] [Indexed: 11/24/2022]
Abstract
Recently, cell-based immunotherapy has become one of the most promising ways to completely eliminate cancer. The major challenge is to effectively promote a proper immune response to kill the cancer cells by activated T cells. This study investigated the effect of T cell-mediated immunotherapy trigged by Au DENPs-MPC (zwitterion 2-methacryloyloxyethyl phosphorylcholine (MPC)-functionalized dendrimer-entrapped gold nanoparticles) loading oli-godeoxynucleotides (ODN) of unmethylated cytosine guanine dinucleotide (CPG). Here, we first synthesized Au DENPs-MPC, evaluated their capability to compress and transfect CpG-ODN to bone marrow dendritic cells (BMDCs), and investigated the potential to use T cells stimulated by matured BMDCs to inhibit the growth of tumor cells. The developed Au DENPs-MPC could apparently reduce the toxicity of Au DENPs, and enhanced transfer CpG-ODN to the BMDCs for the maturation as demonstrated by the 44.41-48.53% increase in different surface maturation markers. The transwell experiments certificated that ex vivo activated T cells display excellent anti-tumor ability, which could effectively inhibit the growth of tumor cells. These results suggest that Au DENPs-MPC can deliver CpG-ODN efficiently to enhance the antigen presentation ability of BMDCs to activate T cells, indicating that T cells-based immunotherapy mediated by Au DENPs-MPC loaded with CpG-ODN may become the most promising treatment of cancer.
Collapse
Affiliation(s)
| | | | | | | | - Xiangyang Shi
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, China; (H.C.); (Y.Z.); (L.L.); (R.G.)
| | - Xueyan Cao
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, China; (H.C.); (Y.Z.); (L.L.); (R.G.)
| |
Collapse
|
53
|
Ding X, Sun X, Cai H, Wu L, Liu Y, Zhao Y, Zhou D, Yu G, Zhou X. Engineering Macrophages via Nanotechnology and Genetic Manipulation for Cancer Therapy. Front Oncol 2022; 11:786913. [PMID: 35070992 PMCID: PMC8770285 DOI: 10.3389/fonc.2021.786913] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 12/13/2021] [Indexed: 12/14/2022] Open
Abstract
Macrophages play critical roles in tumor progression. In the tumor microenvironment, macrophages display highly diverse phenotypes and may perform antitumorigenic or protumorigenic functions in a context-dependent manner. Recent studies have shown that macrophages can be engineered to transport drug nanoparticles (NPs) to tumor sites in a targeted manner, thereby exerting significant anticancer effects. In addition, macrophages engineered to express chimeric antigen receptors (CARs) were shown to actively migrate to tumor sites and eliminate tumor cells through phagocytosis. Importantly, after reaching tumor sites, these engineered macrophages can significantly change the otherwise immune-suppressive tumor microenvironment and thereby enhance T cell-mediated anticancer immune responses. In this review, we first introduce the multifaceted activities of macrophages and the principles of nanotechnology in cancer therapy and then elaborate on macrophage engineering via nanotechnology or genetic approaches and discuss the effects, mechanisms, and limitations of such engineered macrophages, with a focus on using live macrophages as carriers to actively deliver NP drugs to tumor sites. Several new directions in macrophage engineering are reviewed, such as transporting NP drugs through macrophage cell membranes or extracellular vesicles, reprogramming tumor-associated macrophages (TAMs) by nanotechnology, and engineering macrophages with CARs. Finally, we discuss the possibility of combining engineered macrophages and other treatments to improve outcomes in cancer therapy.
Collapse
Affiliation(s)
- Xiaoling Ding
- Department of Immunology, Nantong University, School of Medicine, Nantong, China.,Department of Gastroenterology, The Affiliated Hospital of Nantong University, Nantong, China
| | - Xinchen Sun
- Department of Immunology, Nantong University, School of Medicine, Nantong, China.,Department of Clinical Laboratory, Taizhou Peoples' Hospital, Taizhou, China
| | - Huihui Cai
- Department of Immunology, Nantong University, School of Medicine, Nantong, China.,Department of Clinical Laboratory, The Sixth Nantong People's Hospital, Nantong, China
| | - Lei Wu
- Department of Immunology, Nantong University, School of Medicine, Nantong, China
| | - Ying Liu
- Department of Immunology, Nantong University, School of Medicine, Nantong, China
| | - Yu Zhao
- Department of Immunology, Southeast University, School of Medicine, Nanjing, China
| | - Dingjingyu Zhou
- Krieger School of Arts & Sciences, Johns Hopkins University, Baltimore, MD, United States
| | - Guiping Yu
- Department of Cardiothoracic Surgery, The Affiliated Jiangyin Hospital of Nantong University, Jiangyin, China
| | - Xiaorong Zhou
- Department of Immunology, Nantong University, School of Medicine, Nantong, China
| |
Collapse
|
54
|
Mazayen ZM, Ghoneim AM, Elbatanony RS, Basalious EB, Bendas ER. Pharmaceutical nanotechnology: from the bench to the market. FUTURE JOURNAL OF PHARMACEUTICAL SCIENCES 2022; 8:12. [PMID: 35071609 PMCID: PMC8760885 DOI: 10.1186/s43094-022-00400-0] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 01/01/2022] [Indexed: 12/24/2022] Open
Abstract
Background Nanotechnology is considered a new and rapidly emerging area in the pharmaceutical and medicinal field. Nanoparticles, as drug delivery systems, impart several advantages concerning improved efficacy as well as reduced adverse drug reactions. Main body Different types of nanosystems have been fabricated including carbon nanotubes, paramagnetic nanoparticles, dendrimers, nanoemulsions, etc. Physicochemical properties of the starting materials and the selected method of preparation play a significant aspect in determining the shape and characteristics of the developed nanoparticles. Dispersion of preformed polymers, coacervation, polymerization, nano-spray drying and supercritical fluid technology are among the most extensively used techniques for the preparation of nanocarriers. Particle size, surface charge, surface hydrophobicity and drug release are the main factors affecting nanoparticles physical stability and biological performance of the incorporated drug. In clinical practice, many nanodrugs have been used for both diagnostic and therapeutic applications and are being investigated for various indications in clinical trials. Nanoparticles are used for the cure of kidney diseases, tuberculosis, skin conditions, Alzheimer’s disease, different types of cancer as well as preparation of COVID-19 vaccines. Conclusion In this review, we will confer the advantages, types, methods of preparation, characterization methods and some of the applications of nano-systems.
Collapse
|
55
|
Rui Y, Wilson DR, Tzeng SY, Yamagata HM, Sudhakar D, Conge M, Berlinicke CA, Zack DJ, Tuesca A, Green JJ. High-throughput and high-content bioassay enables tuning of polyester nanoparticles for cellular uptake, endosomal escape, and systemic in vivo delivery of mRNA. SCIENCE ADVANCES 2022; 8:eabk2855. [PMID: 34985952 PMCID: PMC8730632 DOI: 10.1126/sciadv.abk2855] [Citation(s) in RCA: 90] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 11/05/2021] [Indexed: 05/08/2023]
Abstract
Nanoparticle-based mRNA therapeutics hold great promise, but cellular internalization and endosomal escape remain key barriers for cytosolic delivery. We developed a dual nanoparticle uptake and endosomal disruption assay using high-throughput and high-content image-based screening. Using a genetically encoded Galectin 8 fluorescent fusion protein sensor, endosomal disruption could be detected via sensor clustering on damaged endosomal membranes. Simultaneously, nucleic acid endocytosis was quantified using fluorescently tagged mRNA. We used an array of biodegradable poly(beta-amino ester)s as well as Lipofectamine and PEI to demonstrate that this assay has higher predictive capacity for mRNA delivery compared to conventional polymer and nanoparticle physiochemical characteristics. Top nanoparticle formulations enabled safe and efficacious mRNA expression in multiple tissues following intravenous injection, demonstrating that the in vitro screening method is also predictive of in vivo performance. Efficacious nonviral systemic delivery of mRNA with biodegradable particles opens up new avenues for genetic medicine and human health.
Collapse
Affiliation(s)
- Yuan Rui
- Department of Biomedical Engineering, Institute for NanoBioTechnology, and the Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - David R. Wilson
- Department of Biomedical Engineering, Institute for NanoBioTechnology, and the Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Stephany Y. Tzeng
- Department of Biomedical Engineering, Institute for NanoBioTechnology, and the Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Hannah M. Yamagata
- Department of Biomedical Engineering, Institute for NanoBioTechnology, and the Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Deepti Sudhakar
- Department of Biomedical Engineering, Institute for NanoBioTechnology, and the Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Marranne Conge
- Department of Biomedical Engineering, Institute for NanoBioTechnology, and the Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Biology, Berea College, Berea, KY, USA
| | - Cynthia A. Berlinicke
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Donald J. Zack
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Departments of Neuroscience, Molecular Biology and Genetics, and Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Anthony Tuesca
- AstraZeneca, Dosage Form and Design Development, BioPharmaceutical Development, BioPharmaceuticals R&D, Gaithersburg, MD, USA
| | - Jordan J. Green
- Department of Biomedical Engineering, Institute for NanoBioTechnology, and the Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neurosurgery and Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Departments of Materials Science and Engineering, and Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
- Bloomberg~Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
56
|
Zhan M, Yu X, Zhao W, Peng Y, Peng S, Li J, Lu L. Extracellular matrix-degrading STING nanoagonists for mild NIR-II photothermal-augmented chemodynamic-immunotherapy. J Nanobiotechnology 2022; 20:23. [PMID: 34991618 PMCID: PMC8740364 DOI: 10.1186/s12951-021-01226-3] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 12/26/2021] [Indexed: 12/14/2022] Open
Abstract
Regulation of stimulator of interferon genes (STING) pathway using agonists can boost antitumor immunity for cancer treatment, while the rapid plasma clearance, limited membrane permeability, and inefficient cytosolic transport of STING agonists greatly compromise their therapeutic efficacy. In this study, we describe an extracellular matrix (ECM)-degrading nanoagonist (dNAc) with second near-infrared (NIR-II) light controlled activation of intracellular STING pathway for mild photothermal-augmented chemodynamic-immunotherapy of breast cancer. The dNAc consists of a thermal-responsive liposome inside loading with ferrous sulfide (FeS2) nanoparticles as both NIR-II photothermal converters and Fenton catalysts, 2′3′-cyclic guanosine monophosphate-adenosine monophosphate (cGAMP) as the STING agonist, and an ECM-degrading enzyme (bromelain) on the liposome surface. Mild heat generated by dNAc upon NIR-II photoirradiation improves Fenton reaction efficacy to kill tumor cells and cause immunogenic cell death (ICD). Meanwhile, the generated heat triggers a controlled release of cGAMP from thermal-responsive liposomes to active STING pathway. The mild photothermal activation of STING pathway combined with ICD promotes anti-tumor immune responses, which leads to improved infiltration of effector T cells into tumor tissues after bromelain-mediated ECM degradation. As a result, after treatment with dNAc upon NIR-II photoactivation, both primary and distant tumors in a murine mouse model are inhibited and the liver and lung metastasis are effectively suppressed. This work presents a photoactivatable system for STING pathway and combinational immunotherapy with improved therapeutic outcome. ![]()
Collapse
Affiliation(s)
- Meixiao Zhan
- Zhuhai Institute of Translational Medicine, Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai, 519000, Guangdong, China
| | - Xiangrong Yu
- Zhuhai Institute of Translational Medicine, Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai, 519000, Guangdong, China
| | - Wei Zhao
- Zhuhai Institute of Translational Medicine, Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai, 519000, Guangdong, China
| | - Yongjun Peng
- Zhuhai Institute of Translational Medicine, Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai, 519000, Guangdong, China
| | - Shaojun Peng
- Zhuhai Institute of Translational Medicine, Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai, 519000, Guangdong, China.
| | - Jingchao Li
- Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai, 201620, China.
| | - Ligong Lu
- Zhuhai Institute of Translational Medicine, Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai, 519000, Guangdong, China.
| |
Collapse
|
57
|
Zhang BD, Wu JJ, Li WH, Hu HG, Zhao L, He PY, Zhao YF, Li YM. STING and TLR7/8 agonists-based nanovaccines for synergistic antitumor immune activation. NANO RESEARCH 2022; 15:6328-6339. [PMID: 35464625 PMCID: PMC9014842 DOI: 10.1007/s12274-022-4282-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 02/28/2022] [Accepted: 03/01/2022] [Indexed: 05/07/2023]
Abstract
UNLABELLED Immunostimulatory therapies based on pattern recognition receptors (PRRs) have emerged as an effective approach in the fight against cancer, with the ability to recruit tumor-specific lymphocytes in a low-immunogenicity tumor environment. The agonist cyclic dinucleotides (CDNs) of the stimulator of interferon gene (STING) are a group of very promising anticancer molecules that increase tumor immunogenicity by activating innate immunity. However, the tumor immune efficacy of CDNs is limited by several factors, including relatively narrow cytokine production, inefficient delivery to STING, and rapid clearance. In addition, a single adjuvant molecule is unable to elicit a broad cytokine response and thus cannot further amplify the anticancer effect. To address this problem, two or more agonist molecules are often used together to synergistically enhance immune efficacy. In this work, we found that a combination of the STING agonist CDGSF and the Toll-like receptor 7/8 (TLR7/8) agonist 522 produced a broader cytokine response. Subsequently, we developed multicomponent nanovaccines (MCNVs) consisting of a PC7A polymer as a nanocarrier encapsulating the antigen OVA and adjuvant molecules. These MCNVs activate bone marrow-derived dendritic cells (BMDCs) to produce multiple proinflammatory factors that promote antigen cross-presentation to stimulate specific antitumor T-cell responses. In in vivo experiments, we observed that MCNVs triggered a strong T-cell response in tumor-infiltrating lymphocytes, resulting in significant tumor regression and, notably, a 100% survival rate in mice through 25 days without other partnering therapies. These data suggest that our nanovaccines have great potential to advance cancer immunotherapy with increased durability and potency. ELECTRONIC SUPPLEMENTARY MATERIAL Supplementary material (synthesis of CDGSF, 522, PC7A and OVA; preparation of MCNVs; representative gating strategies for flow cytometry) is available in the online version of this article at 10.1007/s12274-022-4282-x.
Collapse
Affiliation(s)
- Bo-Dou Zhang
- Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Department of Chemistry, Center for Synthetic and Systems Biology, Tsinghua University, Beijing, 100084 China
| | - Jun-Jun Wu
- Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Department of Chemistry, Center for Synthetic and Systems Biology, Tsinghua University, Beijing, 100084 China
| | - Wen-Hao Li
- Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Department of Chemistry, Center for Synthetic and Systems Biology, Tsinghua University, Beijing, 100084 China
| | - Hong-Guo Hu
- Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Department of Chemistry, Center for Synthetic and Systems Biology, Tsinghua University, Beijing, 100084 China
| | - Lang Zhao
- Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Department of Chemistry, Center for Synthetic and Systems Biology, Tsinghua University, Beijing, 100084 China
| | - Pei-Yang He
- Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Department of Chemistry, Center for Synthetic and Systems Biology, Tsinghua University, Beijing, 100084 China
| | - Yu-Fen Zhao
- Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Department of Chemistry, Center for Synthetic and Systems Biology, Tsinghua University, Beijing, 100084 China
- Institute of Drug Discovery Technology, Ningbo University, Ningbo, 315201 China
| | - Yan-Mei Li
- Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Department of Chemistry, Center for Synthetic and Systems Biology, Tsinghua University, Beijing, 100084 China
- Beijing Institute for Brain Disorders, Beijing, 100069 China
| |
Collapse
|
58
|
Li K, Zhang Z, Mei Y, Li M, Yang Q, WU Q, Yang H, HE LIANGCAN, Liu S. Targeting innate immune system by nanoparticles for cancer immunotherapy. J Mater Chem B 2022; 10:1709-1733. [DOI: 10.1039/d1tb02818a] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Various cancer therapies have advanced remarkably over the past decade. Unlike the direct therapeutic targeting of tumor cells, cancer immunotherapy is a new strategy that boosts the host's immune system...
Collapse
|
59
|
Tao X, He H, Peng J, Xu R, Fu J, Hu Y, Li L, Yang X, Feng X, Zhang C, Zhang L, Yu X, Shen A, Huang K, Fu Q. Overexpression of PDE4D in mouse liver is sufficient to trigger NAFLD and hypertension in a CD36-TGF-β1 pathway: therapeutic role of roflumilast. Pharmacol Res 2022; 175:106004. [PMID: 34826603 DOI: 10.1016/j.phrs.2021.106004] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Revised: 11/14/2021] [Accepted: 11/22/2021] [Indexed: 12/18/2022]
Abstract
Emerging evidence has shown that nonalcoholic fatty liver disease (NAFLD) may be both a consequence and a cause of hypertension. Recent studies have demonstrated that phosphodiesterase 4 (PDE4)-cAMP signaling represents a pathway relevant to the pathophysiology of metabolic disorders. This study aims to investigate the impact and the underlying mechanism of PDE4 in the pathogenesis of NAFLD and its associated hypertension. Here we demonstrated that high-fat-diet (HFD) fed mice developed NAFLD and hypertension, with an associated increase in hepatic PDE4D expression, which can be prevented and even reversed by PDE4 inhibitor roflumilast. Furthermore, we demonstrated that hepatic overexpression of PDE4D drove significant hepatic steatosis and elevation of blood pressure. Mechanistically, PDE4D activated fatty acid translocase CD36 signaling which facilitates hepatic lipid deposition, resulting in TGF-β1 production by hepatocytes and excessive TGF-β1 signaling in vessels and consequent hypertension. Specific silencing of TGF-β1 in hepatocytes by siRNA using poly (β-amino ester) nanoparticles significantly normalized hepatic PDE4D overexpression-activated TGF-β1 signaling in vessels and hypertension. Together, the conclusions indicated that PDE4D plays an important role in the pathogenesis of NAFLD and associated hypertension via activation of CD36-TGF-β1 signaling in the liver. PDE4 inhibitor such as roflumilast, which is clinically approved for chronic obstructive pulmonary disease (COPD) treatment, has the potential to be used as a preventive or therapeutic drug against NAFLD and associated hypertension in the future.
Collapse
Affiliation(s)
- Xiang Tao
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Clinical Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Haiqing He
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Jiangtong Peng
- Clinical Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Rui Xu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Key Laboratory for Drug Target Research and Pharmacodynamic Evaluation of Hubei Province, Wuhan, China
| | - Jing Fu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Key Laboratory for Drug Target Research and Pharmacodynamic Evaluation of Hubei Province, Wuhan, China
| | - Yuting Hu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Key Laboratory for Drug Target Research and Pharmacodynamic Evaluation of Hubei Province, Wuhan, China
| | - Li Li
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Key Laboratory for Drug Target Research and Pharmacodynamic Evaluation of Hubei Province, Wuhan, China
| | - Xiaoyan Yang
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Key Laboratory for Drug Target Research and Pharmacodynamic Evaluation of Hubei Province, Wuhan, China
| | - Xiuling Feng
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chao Zhang
- Institute of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lingmin Zhang
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Xiyong Yu
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Ao Shen
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China.
| | - Kai Huang
- Clinical Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Qin Fu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Key Laboratory for Drug Target Research and Pharmacodynamic Evaluation of Hubei Province, Wuhan, China.
| |
Collapse
|
60
|
Harnessing the combined potential of cancer immunotherapy and nanomedicine: A new paradigm in cancer treatment. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2021; 40:102492. [PMID: 34775062 DOI: 10.1016/j.nano.2021.102492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Revised: 10/16/2021] [Accepted: 10/29/2021] [Indexed: 11/21/2022]
Abstract
Cancer immunotherapy has recently emerged as a rising star due to its ability to activate patients' immune systems to fight tumors and prevent relapse. Conversely, the interest in cancer nanomedicine has seemingly waned due to its lackluster clinical translation. Despite being hailed as a game-changer in oncology, cancer immunotherapy still faces numerous challenges. Combining both entities together has thus been one among several solutions proposed to circumvent these challenges. This solution has since gained traction and has also led to a renaissance of cancer nanomedicine. While most combinations are currently experimental at best, some have progressed on to clinical trials. This review thus seeks to examine the advantages and disadvantages of integrating both modalities as a cancer treatment. The opportunities, challenges and future directions of this emerging field will also be explored with the hope that such a combination will lead to a paradigm shift in cancer treatments.
Collapse
|
61
|
Considerations for the delivery of STING ligands in cancer immunotherapy. J Control Release 2021; 339:235-247. [PMID: 34592386 DOI: 10.1016/j.jconrel.2021.09.033] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 09/23/2021] [Accepted: 09/24/2021] [Indexed: 12/23/2022]
Abstract
Several studies have shown the importance of the cGAS-STING pathway in antigen-presenting cells for anti-cancer immunity. Cyclic GMP-AMP (cGAMP) - STING ligand is a negatively charged dinucleotide prone to degradation by hydrolases. Once administered in its soluble form, high doses are needed which in turn may cause side effects such as T cell apoptosis. Moreover, due to its negative charge, transfection of cGAMP into negatively-charged membrane cells is hampered. In order to achieve successful transfection and protection from enzymatic degradation there is a need for a suitable carrier for cGAMP. In this review, we therefore describe currently reported carriers for cGAMP, and correlate their characteristics to the effect they cause. To achieve targeted delivery to the tumor microenvironment, the route of administration and physicochemical parameters of the particles (containing a carrier and cGAMP) such as size and charge need to be determined. Therefore, the choice of the particle formulation and its impact on the preclinical outcome will be discussed.
Collapse
|
62
|
STING Signaling and Skin Cancers. Cancers (Basel) 2021; 13:cancers13225603. [PMID: 34830754 PMCID: PMC8615888 DOI: 10.3390/cancers13225603] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 11/03/2021] [Accepted: 11/08/2021] [Indexed: 12/15/2022] Open
Abstract
Recent developments in immunotherapy against malignancies overcome the disadvantages of traditional systemic treatments; however, this immune checkpoint treatment is not perfect and cannot obtain a satisfactory clinical outcome in all cases. Therefore, an additional therapeutic option for malignancy is needed in oncology. Stimulator of interferon genes (STING) has recently been highlighted as a strong type I interferon driver and shows anti-tumor immunity against various malignancies. STING-targeted anti-tumor immunotherapy is expected to enhance the anti-tumor effects and clinical outcomes of immunotherapy against malignancies. In this review, we focus on recent advancements in the knowledge gained from research on STING signaling in skin cancers. In addition to the limitations of STING-targeted immunotherapy, we also discuss the clinical application of STING agonists in the treatment of skin cancer.
Collapse
|
63
|
Yang Y, Liu Q, Shi X, Zheng Q, Chen L, Sun Y. Advances in plant-derived natural products for antitumor immunotherapy. Arch Pharm Res 2021; 44:987-1011. [PMID: 34751930 DOI: 10.1007/s12272-021-01355-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Accepted: 10/29/2021] [Indexed: 12/28/2022]
Abstract
In recent years, immunotherapy has emerged as a novel antitumor strategy in addition to traditional surgery, radiotherapy and chemotherapy. It uniquely focuses on immune cells and immunomodulators in the tumor microenvironment and helps eliminate tumors at the root by rebuilding the immune system. Despite remarkable breakthroughs, cancer immunotherapy still faces many challenges: lack of predictable and prognostic biomarkers, adverse side effects, acquired treatment resistance, high costs, etc. Therefore, more efficacious and efficient, safer and cheaper antitumor immunomodulatory drugs have become an urgent requirement. For decades, plant-derived natural products obtained from land and sea have provided the most important source for the development of antitumor drugs. Currently, more attention is being paid to the discovery of potential cancer immunotherapy modulators from plant-derived natural products, such as polysaccharides, phenols, terpenoids, quinones and alkaloids. Some of these agents have outstanding advantages of multitargeting and low side effects and low cost compared to conventional immunotherapeutic agents. We intend to summarize the progress of comprehensive research on these plant-derived natural products and their derivatives and discuss their possible mechanisms in regulating the immune system and their efficacy as monotherapies or in combination with regular chemotherapeutic agents.
Collapse
Affiliation(s)
- Yi Yang
- Fujian Provincial Key Laboratory of Medical Instrument and Pharmaceutical Technology, College of Biological Science and Technology, Fuzhou University, Fuzhou, 350108, China
| | - Qinying Liu
- Fujian Provincial Key Laboratory of Tumor Biotherapy, Fujian Cancer Hospital, Fujian Medical University Cancer Hospital, Fuzhou, 350014, China
| | - Xianai Shi
- Fujian Provincial Key Laboratory of Medical Instrument and Pharmaceutical Technology, College of Biological Science and Technology, Fuzhou University, Fuzhou, 350108, China
| | - Qiuhong Zheng
- Fujian Provincial Key Laboratory of Tumor Biotherapy, Fujian Cancer Hospital, Fujian Medical University Cancer Hospital, Fuzhou, 350014, China
| | - Li Chen
- Fujian Provincial Key Laboratory of Medical Instrument and Pharmaceutical Technology, College of Biological Science and Technology, Fuzhou University, Fuzhou, 350108, China.
| | - Yang Sun
- Fujian Provincial Key Laboratory of Tumor Biotherapy, Fujian Cancer Hospital, Fujian Medical University Cancer Hospital, Fuzhou, 350014, China.
- Department of Gyn-Surgical Oncology, Fujian Cancer Hospital, Fujian Medical University Cancer Hospital, Fuzhou, 350014, China.
| |
Collapse
|
64
|
Recent advances in immunotherapy, immunoadjuvant, and nanomaterial-based combination immunotherapy. Coord Chem Rev 2021. [DOI: 10.1016/j.ccr.2021.214009] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
65
|
Lakshmanan VK, Jindal S, Packirisamy G, Ojha S, Lian S, Kaushik A, Alzarooni AIMA, Metwally YAF, Thyagarajan SP, Do Jung Y, Chouaib S. Nanomedicine-based cancer immunotherapy: recent trends and future perspectives. Cancer Gene Ther 2021; 28:911-923. [PMID: 33558704 DOI: 10.1038/s41417-021-00299-4] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Accepted: 01/20/2021] [Indexed: 01/30/2023]
Abstract
The combination of cancer immunotherapy with efficient functionalized nanosystems has emerged as a beneficial treatment strategy and its use has increased rapidly. The roles of stimuli-responsive nanosystems and nanomedicine-based cancer immunotherapy, a subsidiary discipline in the field of immunology, are pivotal. The present era is witnessing rapid advancements in the use of nanomedicine as a platform for investigating novel therapeutic applications and modern intelligent healthcare management strategies. The development of cancer nanomedicine has posthaste ratified the outcomes of immunotherapy to the subsequent stage in the current era of medical research. This review focuses on key findings with respect to the effectiveness of nanomedicine-based cancer immunotherapies and their applications, which include i) immune checkpoint inhibitors and nanomedicine, ii) CRISPR-Cas nanoparticles (NPs) in cancer immunotherapy, iii) combination cancer immunotherapy with core-shell nanoparticles, iv) biomimetic NPs for cancer immunotherapy, and v) CAR-T cells and cancer nanoimmunotherapy. By evaluating the state-of-the-art tools and taking the challenges involved into consideration, various aspects of the proposed nano-enabled therapeutic approaches have been discussed in this review.
Collapse
Affiliation(s)
- Vinoth-Kumar Lakshmanan
- Centre for Preclinical and Translational Medical Research (CPTMR), Central Research Facility (CRF), Faculty of Clinical Research, Sri Ramachandra Institute of Higher Education and Research, Porur, Chennai, Tamil Nadu, India. .,Thumbay Research Institute for Precision Medicine, Gulf Medical University, Ajman, United Arab Emirates. .,Department of Biomedical Sciences, College of Medicine, Gulf Medical University, Ajman, United Arab Emirates.
| | - Shlok Jindal
- Department of Biotechnology, Indian Institute of Technology (IIT) Roorkee, Roorkee, Uttarakhand, India
| | - Gopinath Packirisamy
- Department of Biotechnology, Indian Institute of Technology (IIT) Roorkee, Roorkee, Uttarakhand, India. .,Centre for Nanotechnology, Indian Institute of Technology (IIT) Roorkee, Roorkee, Uttarakhand, India.
| | - Shreesh Ojha
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, Abu Dhabi, United Arab Emirates
| | - Sen Lian
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Ajeet Kaushik
- NanoBio Tech Laboratory, Health System Engineering, Department of Natural Sciences, Division of Sciences, Arts & Mathematics, Florida Polytechnic University, Lakeland, FL, USA
| | | | - Yasser Abdelraouf Farahat Metwally
- Department of Urology, H.H. Sheikh Khalifa General Hospital, Al Salama, Opp. Ministry of Community Development, Umm Al Quwain, United Arab Emirates
| | - Sadras Panchatcharam Thyagarajan
- Centre for Preclinical and Translational Medical Research (CPTMR), Central Research Facility (CRF), Faculty of Clinical Research, Sri Ramachandra Institute of Higher Education and Research, Porur, Chennai, Tamil Nadu, India
| | - Young Do Jung
- Department of Biochemistry, Chonnam National University Medical School, Gwangju, South Korea
| | - Salem Chouaib
- Thumbay Research Institute for Precision Medicine, Gulf Medical University, Ajman, United Arab Emirates. .,INSERM UMR1186, Integrative Tumor Immunology and Genetic Oncology, Gustave Roussy, Equipe Labellisée par la Ligue Contre le Cancer, EPHE, Faculté de Médecine, Université Paris-Sud, Université Paris-Saclay, Villejuif, France.
| |
Collapse
|
66
|
Thangam R, Patel KD, Kang H, Paulmurugan R. Advances in Engineered Polymer Nanoparticle Tracking Platforms towards Cancer Immunotherapy-Current Status and Future Perspectives. Vaccines (Basel) 2021; 9:vaccines9080935. [PMID: 34452059 PMCID: PMC8402739 DOI: 10.3390/vaccines9080935] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 08/03/2021] [Accepted: 08/13/2021] [Indexed: 12/17/2022] Open
Abstract
Engineering polymeric nanoparticles for their shape, size, surface chemistry, and functionalization using various targeting molecules has shown improved biomedical applications for nanoparticles. Polymeric nanoparticles have created tremendous therapeutic platforms, particularly applications related to chemo- and immunotherapies in cancer. Recently advancements in immunotherapies have broadened this field in immunology and biomedical engineering, where "immunoengineering" creates solutions to target translational science. In this regard, the nanoengineering field has offered the various techniques necessary to manufacture and assemble multifunctional polymeric nanomaterial systems. These include nanoparticles functionalized using antibodies, small molecule ligands, targeted peptides, proteins, and other novel agents that trigger and encourage biological systems to accept the engineered materials as immune enhancers or as vaccines to elevate therapeutic functions. Strategies to engineer polymeric nanoparticles with therapeutic and targeting molecules can provide solutions for developing immune vaccines via maintaining the receptor storage in T- and B cells. Furthermore, cancer immunotherapy using polymeric nanomaterials can serve as a gold standard approach for treating primary and metastasized tumors. The current status of the limited availability of immuno-therapeutic drugs highlights the importance of polymeric nanomaterial platforms to improve the outcomes via delivering anticancer agents at localized sites, thereby enhancing the host immune response in cancer therapy. This review mainly focuses on the potential scientific enhancements and recent developments in cancer immunotherapies by explicitly discussing the role of polymeric nanocarriers as nano-vaccines. We also briefly discuss the role of multifunctional nanomaterials for their therapeutic impacts on translational clinical applications.
Collapse
Affiliation(s)
- Ramar Thangam
- Department of Materials Science and Engineering, Korea University, Seoul 02841, Korea; (K.D.P.); (H.K.)
- Institute for High Technology Materials and Devices, Korea University, Seoul 02841, Korea
- Correspondence: (R.T.); (R.P.)
| | - Kapil D. Patel
- Department of Materials Science and Engineering, Korea University, Seoul 02841, Korea; (K.D.P.); (H.K.)
| | - Heemin Kang
- Department of Materials Science and Engineering, Korea University, Seoul 02841, Korea; (K.D.P.); (H.K.)
- Institute for High Technology Materials and Devices, Korea University, Seoul 02841, Korea
- Department of Biomicrosystem Technology, Korea University, Seoul 02841, Korea
| | - Ramasamy Paulmurugan
- Department of Radiology, Molecular Imaging Program at Stanford, School of Medicine, Stanford University, Palo Alto, CA 94304, USA
- Department of Radiology, Canary Center at Stanford for Cancer Early Detection, School of Medicine, Stanford University, Palo Alto, CA 94304, USA
- Correspondence: (R.T.); (R.P.)
| |
Collapse
|
67
|
Yan H, Chen W. The Promise and Challenges of Cyclic Dinucleotides as Molecular Adjuvants for Vaccine Development. Vaccines (Basel) 2021; 9:917. [PMID: 34452042 PMCID: PMC8402453 DOI: 10.3390/vaccines9080917] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 08/05/2021] [Accepted: 08/10/2021] [Indexed: 12/14/2022] Open
Abstract
Cyclic dinucleotides (CDNs), originally discovered as bacterial second messengers, play critical roles in bacterial signal transduction, cellular processes, biofilm formation, and virulence. The finding that CDNs can trigger the innate immune response in eukaryotic cells through the stimulator of interferon genes (STING) signalling pathway has prompted the extensive research and development of CDNs as potential immunostimulators and novel molecular adjuvants for induction of systemic and mucosal innate and adaptive immune responses. In this review, we summarize the chemical structure, biosynthesis regulation, and the role of CDNs in enhancing the crosstalk between host innate and adaptive immune responses. We also discuss the strategies to improve the efficient delivery of CDNs and the recent advance and future challenges in the development of CDNs as potential adjuvants in prophylactic vaccines against infectious diseases and in therapeutic vaccines against cancers.
Collapse
Affiliation(s)
- Hongbin Yan
- Department of Chemistry, Brock University, St. Catharines, ON L2S 3A1, Canada
| | - Wangxue Chen
- Human Health and Therapeutics Research Centre, National Research Council Canada, Ottawa, ON K1A 0R6, Canada
- Department of Biological Sciences, Brock University, St. Catharines, ON L2S 3A1, Canada
| |
Collapse
|
68
|
Sadiq IZ, Abubakar FS, Dan-Iya BI. Role of nanoparticles in tackling COVID-19 pandemic: a bio-nanomedical approach. JOURNAL OF TAIBAH UNIVERSITY FOR SCIENCE 2021. [DOI: 10.1080/16583655.2021.1944488] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Idris Zubairu Sadiq
- Department of Biochemistry, Faculty of life sciences, Ahmadu Bello University, Zaria, Nigeria
- African Center of Excellence in Neglected Tropical Diseases and Forensic Biotechnology, Ahmadu Bello University, Zaria, Nigeria
| | - Fatima Sadiq Abubakar
- Department of Biochemistry, Faculty of life sciences, Ahmadu Bello University, Zaria, Nigeria
- African Center of Excellence in Neglected Tropical Diseases and Forensic Biotechnology, Ahmadu Bello University, Zaria, Nigeria
- National Agricultural Extension and Liaison Services, Ahmadu Bello University, Zaria, Nigeria
| | - Bilal Ibrahim Dan-Iya
- Pharmacy Technician Departments, College of Health Sciences and Technology, Kano, Nigeria
- Department of Biochemistry, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Darul Ehsan, Malaysia
| |
Collapse
|
69
|
Zhou Q, Zhou Y, Li T, Ge Z. Nanoparticle-Mediated STING Agonist Delivery for Enhanced Cancer Immunotherapy. Macromol Biosci 2021; 21:e2100133. [PMID: 34117839 DOI: 10.1002/mabi.202100133] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 05/17/2021] [Indexed: 12/12/2022]
Abstract
Stimulator of interferon genes (STING) are located in the endoplasmic reticulum of cells, which have been demonstrated to show considerable potentials to achieve efficient antitumor immunity by inducing various pro-inflammatory cytokines and chemokines, such as type I interferons. A variety of STING agonists have been prepared for STING activation, and many of them have been promoted to preclinical trials or clinical applications for the immunotherapy of cancers. However, the intrinsic disadvantages of the small molecule STING agonists can limit the in vivo application and final therapeutic efficacy due to low bioavailability of targeting tissues. Moreover, a cascade of physiological barriers for in vivo STING activation also limit the accumulation of STING agonists in targeting tissues. Drug delivery systems play an important role to improve the STING activation efficiency. In recent years, a variety of nanoparticle-mediated STING agonist delivery systems have been engineered and exploited to address the challenges related to the in vivo STING activation, including liposomes, polymeric micelles, polymersomes, and so on. In this review article, the progresses concerning STING agonists and related delivery systems in recent years will be summarized and discussed.
Collapse
Affiliation(s)
- Qinghao Zhou
- CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, Anhui, 230026, China
| | - Yu Zhou
- CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, Anhui, 230026, China
| | - Taiyuan Li
- CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, Anhui, 230026, China
| | - Zhishen Ge
- CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, Anhui, 230026, China
| |
Collapse
|
70
|
Gao M, Xie Y, Lei K, Zhao Y, Kurum A, Van Herck S, Guo Y, Hu X, Tang L. A Manganese Phosphate Nanocluster Activates the cGAS‐STING Pathway for Enhanced Cancer Immunotherapy. ADVANCED THERAPEUTICS 2021. [DOI: 10.1002/adtp.202100065] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Min Gao
- Institute of Bioengineering École Polytechnique Fédérale de Lausanne (EPFL) Lausanne 1015 Switzerland
| | - Yu‐Qing Xie
- Institute of Bioengineering École Polytechnique Fédérale de Lausanne (EPFL) Lausanne 1015 Switzerland
| | - Kewen Lei
- Institute of Materials Science and Engineering École Polytechnique Fédérale de Lausanne (EPFL) Lausanne 1015 Switzerland
| | - Yu Zhao
- Institute of Bioengineering École Polytechnique Fédérale de Lausanne (EPFL) Lausanne 1015 Switzerland
- Institute of Materials Science and Engineering École Polytechnique Fédérale de Lausanne (EPFL) Lausanne 1015 Switzerland
| | - Armand Kurum
- Institute of Materials Science and Engineering École Polytechnique Fédérale de Lausanne (EPFL) Lausanne 1015 Switzerland
| | - Simon Van Herck
- Institute of Bioengineering École Polytechnique Fédérale de Lausanne (EPFL) Lausanne 1015 Switzerland
| | - Yugang Guo
- Institute of Bioengineering École Polytechnique Fédérale de Lausanne (EPFL) Lausanne 1015 Switzerland
- Institute of Materials Science and Engineering École Polytechnique Fédérale de Lausanne (EPFL) Lausanne 1015 Switzerland
| | - Xiaomeng Hu
- Institute of Bioengineering École Polytechnique Fédérale de Lausanne (EPFL) Lausanne 1015 Switzerland
| | - Li Tang
- Institute of Bioengineering École Polytechnique Fédérale de Lausanne (EPFL) Lausanne 1015 Switzerland
- Institute of Materials Science and Engineering École Polytechnique Fédérale de Lausanne (EPFL) Lausanne 1015 Switzerland
| |
Collapse
|
71
|
Liu L, Kshirsagar P, Christiansen J, Gautam SK, Aithal A, Gulati M, Kumar S, Solheim JC, Batra SK, Jain M, Wannemuehler MJ, Narasimhan B. Polyanhydride nanoparticles stabilize pancreatic cancer antigen MUC4β. J Biomed Mater Res A 2021; 109:893-902. [PMID: 32776461 PMCID: PMC8100985 DOI: 10.1002/jbm.a.37080] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 07/24/2020] [Accepted: 07/28/2020] [Indexed: 02/06/2023]
Abstract
Pancreatic cancer (PC) is one of the most lethal malignancies and represents an increasing and challenging threat, especially with an aging population. The identification of immunogenic PC-specific upregulated antigens and an enhanced understanding of the immunosuppressive tumor microenvironment have provided opportunities to enable the immune system to recognize cancer cells. Due to its differential upregulation and functional role in PC, the transmembrane mucin MUC4 is an attractive target for immunotherapy. In the current study we characterized the antigen stability, antigenicity and release kinetics of a MUC4β-nanovaccine to guide further optimization and, in vivo evaluation. Amphiphilic polyanhydride copolymers based on 20 mol % 1,8-bis(p-carboxyphenoxy)-3,6-dioxaoctane and 80 mol % 1,6-bis(p-carboxyphenoxy)hexane were used to synthesize nanoparticles. Structurally stable MUC4β protein was released from the particles in a sustained manner and characterized by gel electrophoresis and fluorescence spectroscopy. Modest levels of protein degradation were observed upon release. The released protein was also analyzed by MUC4β-specific monoclonal antibodies using ELISA and showed no significant loss of epitope availability. Further, mice immunized with multiple formulations of combination vaccines containing MUC4β-loaded nanoparticles generated MUC4β-specific antibody responses. These results indicate that polyanhydride nanoparticles are viable MUC4β vaccine carriers, laying the foundation for evaluation of this platform for PC immunotherapy.
Collapse
Affiliation(s)
- Luman Liu
- Department of Chemical and Biological Engineering, Iowa State University, Ames, Iowa
| | - Prakash Kshirsagar
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska
| | - John Christiansen
- Department of Veterinary Microbiology and Preventative Medicine, Iowa State University, Ames, Iowa
| | - Shailendra K. Gautam
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska
| | - Abhijit Aithal
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska
| | - Mansi Gulati
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska
| | - Sushil Kumar
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska
| | - Joyce C. Solheim
- Nanovaccine Institute, Iowa State University, Ames, Iowa
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska
| | - Surinder K. Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska
- Nanovaccine Institute, Iowa State University, Ames, Iowa
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska
| | - Maneesh Jain
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska
- Nanovaccine Institute, Iowa State University, Ames, Iowa
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska
| | - Michael J. Wannemuehler
- Department of Veterinary Microbiology and Preventative Medicine, Iowa State University, Ames, Iowa
- Nanovaccine Institute, Iowa State University, Ames, Iowa
| | - Balaji Narasimhan
- Department of Chemical and Biological Engineering, Iowa State University, Ames, Iowa
- Nanovaccine Institute, Iowa State University, Ames, Iowa
| |
Collapse
|
72
|
|
73
|
An D, Fu J, Xie Z, Xing C, Zhang B, Wang B, Qiu M. Progress in the therapeutic applications of polymer-decorated black phosphorus and black phosphorus analog nanomaterials in biomedicine. J Mater Chem B 2021; 8:7076-7120. [PMID: 32648567 DOI: 10.1039/d0tb00824a] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Wonderful black phosphorus (BP) and some BP analogs (BPAs) have been increasingly studied for their biomedical applications owing to their fascinating properties and biodegradability, but opportunities and challenges have always coexisted in their study. Poor stability upon exposure to the natural environment is the major obstacle hampering their in vivo applications. BP/polymer and BPAs/polymer nanocomposites can not only efficiently prevent their oxidation and aggregation but also exhibit "biological activity" due to synergistic effects. In this review, we briefly describe the synthesis methods and stability strategies of BP/polymer and BPAs/polymer. Then, advances pertaining to their exciting therapeutic applications in various fields are systematically introduced, such as cancer therapy (phototherapy, drug delivery, and synergistic immunotherapy), bone regeneration, and neurogenesis. Some challenges for future clinical trials and possible directions for further study are finally discussed.
Collapse
Affiliation(s)
- Dong An
- International Collaborative Laboratory of 2D Materials for Optoelectronics Science and Technology of Ministry of Education, Institute of Microscale Optoelectronics, Shenzhen University, Shenzhen 518060, China. and Key Laboratory of Marine Chemistry Theory and Technology (Ocean University of China), Ministry of Education, Qingdao, 266100, P. R. China.
| | - Jianye Fu
- International Collaborative Laboratory of 2D Materials for Optoelectronics Science and Technology of Ministry of Education, Institute of Microscale Optoelectronics, Shenzhen University, Shenzhen 518060, China. and Key Laboratory of Marine Chemistry Theory and Technology (Ocean University of China), Ministry of Education, Qingdao, 266100, P. R. China.
| | - Zhongjian Xie
- Shenzhen International Institute for Biomedical Research, Shenzhen 518116, P. R. China
| | - Chenyang Xing
- International Collaborative Laboratory of 2D Materials for Optoelectronics Science and Technology of Ministry of Education, Institute of Microscale Optoelectronics, Shenzhen University, Shenzhen 518060, China.
| | - Bin Zhang
- International Collaborative Laboratory of 2D Materials for Optoelectronics Science and Technology of Ministry of Education, Institute of Microscale Optoelectronics, Shenzhen University, Shenzhen 518060, China.
| | - Bing Wang
- International Collaborative Laboratory of 2D Materials for Optoelectronics Science and Technology of Ministry of Education, Institute of Microscale Optoelectronics, Shenzhen University, Shenzhen 518060, China.
| | - Meng Qiu
- Key Laboratory of Marine Chemistry Theory and Technology (Ocean University of China), Ministry of Education, Qingdao, 266100, P. R. China.
| |
Collapse
|
74
|
Dosta P, Cryer AM, Prado M, Dion MZ, Ferber S, Kalash S, Artzi N. Delivery of Stimulator of Interferon Genes (STING) Agonist Using Polypeptide‐Modified Dendrimer Nanoparticles in the Treatment of Melanoma. ADVANCED NANOBIOMED RESEARCH 2021. [DOI: 10.1002/anbr.202100006] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Affiliation(s)
- Pere Dosta
- Institute for Medical Engineering and Science Massachusetts Institute of Technology Cambridge MA 02139 USA
- Department of Medicine Division of Engineering in Medicine Brigham and Women's Hospital Harvard Medical School Boston MA 02115 USA
| | - Alexander M. Cryer
- Institute for Medical Engineering and Science Massachusetts Institute of Technology Cambridge MA 02139 USA
- Department of Medicine Division of Engineering in Medicine Brigham and Women's Hospital Harvard Medical School Boston MA 02115 USA
| | - Michaela Prado
- Institute for Medical Engineering and Science Massachusetts Institute of Technology Cambridge MA 02139 USA
- Department of Medicine Division of Engineering in Medicine Brigham and Women's Hospital Harvard Medical School Boston MA 02115 USA
| | - Michelle Z. Dion
- Institute for Medical Engineering and Science Massachusetts Institute of Technology Cambridge MA 02139 USA
- Department of Medicine Division of Engineering in Medicine Brigham and Women's Hospital Harvard Medical School Boston MA 02115 USA
| | - Shiran Ferber
- Institute for Medical Engineering and Science Massachusetts Institute of Technology Cambridge MA 02139 USA
- Department of Medicine Division of Engineering in Medicine Brigham and Women's Hospital Harvard Medical School Boston MA 02115 USA
| | - Santhosh Kalash
- Institute for Medical Engineering and Science Massachusetts Institute of Technology Cambridge MA 02139 USA
- Department of Medicine Division of Engineering in Medicine Brigham and Women's Hospital Harvard Medical School Boston MA 02115 USA
| | - Natalie Artzi
- Institute for Medical Engineering and Science Massachusetts Institute of Technology Cambridge MA 02139 USA
- Department of Medicine Division of Engineering in Medicine Brigham and Women's Hospital Harvard Medical School Boston MA 02115 USA
| |
Collapse
|
75
|
Huang X, Pan J, Xu F, Shao B, Wang Y, Guo X, Zhou S. Bacteria-Based Cancer Immunotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:2003572. [PMID: 33854892 PMCID: PMC8025040 DOI: 10.1002/advs.202003572] [Citation(s) in RCA: 156] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Revised: 11/03/2020] [Indexed: 05/24/2023]
Abstract
In the past decade, bacteria-based cancer immunotherapy has attracted much attention in the academic circle due to its unique mechanism and abundant applications in triggering the host anti-tumor immunity. One advantage of bacteria lies in their capability in targeting tumors and preferentially colonizing the core area of the tumor. Because bacteria are abundant in pathogen-associated molecular patterns that can effectively activate the immune cells even in the tumor immunosuppressive microenvironment, they are capable of enhancing the specific immune recognition and elimination of tumor cells. More attractively, during the rapid development of synthetic biology, using gene technology to enable bacteria to be an efficient producer of immunotherapeutic agents has led to many creative immunotherapy paradigms. The combination of bacteria and nanomaterials also displays infinite imagination in the multifunctional endowment for cancer immunotherapy. The current progress report summarizes the recent advances in bacteria-based cancer immunotherapy with specific foci on the applications of naive bacteria-, engineered bacteria-, and bacterial components-based cancer immunotherapy, and at the same time discusses future directions in this field of research based on the present developments.
Collapse
Affiliation(s)
- Xuehui Huang
- Key Laboratory of Advanced Technologies of MaterialsMinistry of EducationSchool of Materials Science and EngineeringSouthwest Jiaotong UniversityChengdu610031China
| | - Jingmei Pan
- Key Laboratory of Advanced Technologies of MaterialsMinistry of EducationSchool of Materials Science and EngineeringSouthwest Jiaotong UniversityChengdu610031China
| | - Funeng Xu
- Key Laboratory of Advanced Technologies of MaterialsMinistry of EducationSchool of Materials Science and EngineeringSouthwest Jiaotong UniversityChengdu610031China
| | - Binfen Shao
- School of Life Science and EngineeringSouthwest Jiaotong UniversityChengdu610031China
| | - Yi Wang
- School of Life Science and EngineeringSouthwest Jiaotong UniversityChengdu610031China
| | - Xing Guo
- Key Laboratory of Advanced Technologies of MaterialsMinistry of EducationSchool of Materials Science and EngineeringSouthwest Jiaotong UniversityChengdu610031China
| | - Shaobing Zhou
- Key Laboratory of Advanced Technologies of MaterialsMinistry of EducationSchool of Materials Science and EngineeringSouthwest Jiaotong UniversityChengdu610031China
| |
Collapse
|
76
|
Perni S, Preedy EC, Prokopovich P. Amplify antimicrobial photo dynamic therapy efficacy with poly-beta-amino esters (PBAEs). Sci Rep 2021; 11:7275. [PMID: 33790379 PMCID: PMC8012660 DOI: 10.1038/s41598-021-86773-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 03/17/2021] [Indexed: 01/03/2023] Open
Abstract
Light-activated antimicrobial agents (photosensitisers) are promising alternatives to antibiotics for the treatment of skin infections and wounds through antimicrobial photo dynamic therapy (aPDT); utilisation of this technique is still restricted by general low efficacy requiring long exposure time (in the order of tens of minutes) that make the treatment very resource intensive. We report for the first time the possibility of harvesting the cell penetrating properties of poly-beta-amino esters (PBAEs) in combination with toluidine blue O (TBO) to shorten aPDT exposure time. Candidates capable of inactivation rates 30 times quicker than pure TBO were discovered and further improvements through PBAE backbone optimisation could be foreseen. Efficacy of the complexes was PBAE-dependent on a combination of TBO uptake and a newly discovered and unexpected role of PBAEs on reactive species production. Chemometric approach of partial least square regression was employed to assess the critical PBAE properties involved in this newly observed phenomenon in order to elicit a possible mechanism. The superior antimicrobial performance of this new approach benefits from the use of well established, low-cost and safe dye (TBO) coupled with inexpensive, widely tested and biodegradable polymers also known to be safe. Moreover, no adverse cytotoxic effects of the PBAEs adjuvated TBO delivery have been observed on a skin cells in vitro model demonstrating the safety profile of this new technology.
Collapse
Affiliation(s)
- Stefano Perni
- Cardiff School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Redwood Building, King Edward VII Avenue, Cardiff, CF10 3NB, UK
| | - Emily C Preedy
- Cardiff School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Redwood Building, King Edward VII Avenue, Cardiff, CF10 3NB, UK
| | - Polina Prokopovich
- Cardiff School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Redwood Building, King Edward VII Avenue, Cardiff, CF10 3NB, UK.
| |
Collapse
|
77
|
Muluh TA, Chen Z, Li Y, Xiong K, Jin J, Fu S, Wu J. Enhancing Cancer Immunotherapy Treatment Goals by Using Nanoparticle Delivery System. Int J Nanomedicine 2021; 16:2389-2404. [PMID: 33790556 PMCID: PMC8007559 DOI: 10.2147/ijn.s295300] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 02/14/2021] [Indexed: 12/14/2022] Open
Abstract
Recently, there has been an incredible increase in research about the abnormal growth of cells (neoplasm), focusing on the management, treatment and preventing reoccurrence. It has been understood that the natural defense system, composed of a variety of immune defensive cells, does not just limit its function in eliminating neoplastic cells, but also controls the growth and spread of tumor cells of different kinds to other parts of the body. Cancer immunotherapy, is a cancer treatment plan that educates the body’s defensive system to forestall, control, and eliminate tumor cells. The effectiveness of immunotherapy is achieved, to its highest efficacy, by the use of nanoparticles (NPs) for precise and timely delivery of immunotherapies to specific targeted neoplasms, with less or no harm to the healthy cells. Immunotherapies have been affirmed in clinical trials as a cancer regimen for various types of cancers, the side effects resulting from imprecise and non-targeted conveyance is well managed with the use of nanoparticles. Nonetheless, we will concentrate on enhancing cancer immunotherapy approaches by the use of nanoparticles for the productivity of antitumor immunity. Nanoparticles will be presented and utilized as an objective immunotherapy delivery system for high exactness and are thus a promising methodology for cancer treatment.
Collapse
Affiliation(s)
- Tobias Achu Muluh
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, People's Republic of China
| | - Zhuo Chen
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, People's Republic of China
| | - Yi Li
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, People's Republic of China
| | - Kang Xiong
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, People's Republic of China
| | - Jing Jin
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, People's Republic of China
| | - ShaoZhi Fu
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, People's Republic of China.,Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou, 646000, Sichuan, People's Republic of China.,Department of Oncology, Academician (Expert) Workstation of Sichuan Province, Luzhou, Sichuan, 646000, People's Republic of China
| | - JingBo Wu
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, People's Republic of China.,Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou, 646000, Sichuan, People's Republic of China.,Department of Oncology, Academician (Expert) Workstation of Sichuan Province, Luzhou, Sichuan, 646000, People's Republic of China
| |
Collapse
|
78
|
Effective delivery of STING agonist using exosomes suppresses tumor growth and enhances antitumor immunity. J Biol Chem 2021; 296:100523. [PMID: 33711340 PMCID: PMC8042450 DOI: 10.1016/j.jbc.2021.100523] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 03/01/2021] [Accepted: 03/05/2021] [Indexed: 12/20/2022] Open
Abstract
The Stimulator of Interferon Genes (STING) pathway is implicated in the innate immune response and is important in both oncogenesis and cancer treatment. Specifically, activation of the cytosolic DNA sensor STING in antigen-presenting cells (APCs) induces a type I interferon response and cytokine production that facilitates antitumor immune therapy. However, use of STING agonists (STINGa) as a cancer therapeutic has been limited by unfavorable pharmacological properties and targeting inefficiency due to rapid clearance and limited uptake into the cytosol. Exosomes, a class of extracellular vesicles shed by all cells are under consideration for their use as effective carriers of drugs owing to their innate ability to be taken up by cells and their biocompatibility for optimal drug biodistribution. Therefore, we engineered exosomes to deliver the STING agonist cyclic GMP-AMP (iExoSTINGa), to exploit their favorable pharmacokinetics and pharmacodynamics. Selective targeting of the STING pathway in APCs with iExoSTINGa was associated with superior potency compared with STINGa alone in suppressing B16F10 tumor growth. Moreover, iExoSTINGa showed superior uptake of STINGa into dendritic cells compared with STINGa alone, which led to increased accumulation of activated CD8+ T-cells and an antitumor immune response. Our study highlights the potential of exosomes in general, and iExoSTINGa specifically, in enhancing cancer therapy outcomes.
Collapse
|
79
|
Kyriakides TR, Raj A, Tseng TH, Xiao H, Nguyen R, Mohammed FS, Halder S, Xu M, Wu MJ, Bao S, Sheu WC. Biocompatibility of nanomaterials and their immunological properties. Biomed Mater 2021; 16:10.1088/1748-605X/abe5fa. [PMID: 33578402 PMCID: PMC8357854 DOI: 10.1088/1748-605x/abe5fa] [Citation(s) in RCA: 89] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 02/12/2021] [Indexed: 12/16/2022]
Abstract
Nanomaterials (NMs) have revolutionized multiple aspects of medicine by enabling novel sensing, diagnostic, and therapeutic approaches. Advancements in processing and fabrication have also allowed significant expansion in the applications of the major classes of NMs based on polymer, metal/metal oxide, carbon, liposome, or multi-scale macro-nano bulk materials. Concomitantly, concerns regarding the nanotoxicity and overall biocompatibility of NMs have been raised. These involve putative negative effects on both patients and those subjected to occupational exposure during manufacturing. In this review, we describe the current state of testing of NMs including those that are in clinical use, in clinical trials, or under development. We also discuss the cellular and molecular interactions that dictate their toxicity and biocompatibility. Specifically, we focus on the reciprocal interactions between NMs and host proteins, lipids, and sugars and how these induce responses in immune and other cell types leading to topical and/or systemic effects.
Collapse
Affiliation(s)
- Themis R Kyriakides
- Department of Biomedical Engineering, Yale University, New Haven, CT 06405, United States of America
- Department of Pathology, Yale University, New Haven, CT 06405, United States of America
- Vascular Biology and Therapeutics Program, Yale University, New Haven, CT 06405, United States of America
| | - Arindam Raj
- Department of Mechanical Engineering and Materials Science, Yale University, New Haven, CT 06405, United States of America
| | - Tiffany H Tseng
- Department of Pathology, Yale University, New Haven, CT 06405, United States of America
- Vascular Biology and Therapeutics Program, Yale University, New Haven, CT 06405, United States of America
| | - Hugh Xiao
- Department of Biomedical Engineering, Yale University, New Haven, CT 06405, United States of America
| | - Ryan Nguyen
- Department of Biomedical Engineering, Yale University, New Haven, CT 06405, United States of America
| | - Farrah S Mohammed
- Department of Biomedical Engineering, Yale University, New Haven, CT 06405, United States of America
| | - Saiti Halder
- Department of Biomedical Engineering, Yale University, New Haven, CT 06405, United States of America
| | - Mengqing Xu
- Department of Biomedical Engineering, Yale University, New Haven, CT 06405, United States of America
- Vascular Biology and Therapeutics Program, Yale University, New Haven, CT 06405, United States of America
| | - Michelle J Wu
- Department of Biomedical Engineering, Yale University, New Haven, CT 06405, United States of America
| | - Shuozhen Bao
- Department of Biomedical Engineering, Yale University, New Haven, CT 06405, United States of America
- Vascular Biology and Therapeutics Program, Yale University, New Haven, CT 06405, United States of America
| | - Wendy C Sheu
- Department of Biomedical Engineering, Yale University, New Haven, CT 06405, United States of America
| |
Collapse
|
80
|
Pu F, Chen F, Liu J, Zhang Z, Shao Z. Immune Regulation of the cGAS-STING Signaling Pathway in the Tumor Microenvironment and Its Clinical Application. Onco Targets Ther 2021; 14:1501-1516. [PMID: 33688199 PMCID: PMC7935450 DOI: 10.2147/ott.s298958] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Accepted: 02/19/2021] [Indexed: 12/25/2022] Open
Abstract
As a DNA receptor in the cytoplasm, cyclic GMP-AMP synthase (cGAS) contributes to the recognition of abnormal DNA in the cytoplasm and contributes to the stimulator of interferon genes (STING) signaling pathway. cGAS could mediate the expression of interferon-related genes, inflammatory-related factors, and downstream chemokines, thus initiating the immune response. The STING protein is a key effector downstream of the DNA receptor pathway. It is widely expressed across cell types such as immune cells, tumor cells, and stromal cells and plays a role in signal transduction for cytoplasmic DNA sensing and immunity. STING agonists, as novel agonists, are used in preclinical research and in the treatment of various tumors via clinical trials and have displayed attractive application prospects. Studying the cGAS-STING signaling pathway will deepen our understanding of tumor immunity and provide a basis for the research and development of antitumor drugs.
Collapse
Affiliation(s)
- Feifei Pu
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Fengxia Chen
- Department of Radiation and Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, People's Republic of China
| | - Jianxiang Liu
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Zhicai Zhang
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Zengwu Shao
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| |
Collapse
|
81
|
Fryer AL, Abdullah A, Taylor JM, Crack PJ. The Complexity of the cGAS-STING Pathway in CNS Pathologies. Front Neurosci 2021; 15:621501. [PMID: 33633536 PMCID: PMC7900568 DOI: 10.3389/fnins.2021.621501] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 01/19/2021] [Indexed: 12/21/2022] Open
Abstract
Neuroinflammation driven by type-I interferons in the CNS is well established to exacerbate the progression of many CNS pathologies both acute and chronic. The role of adaptor protein Stimulator of Interferon Genes (STING) is increasingly appreciated to instigate type-I IFN-mediated neuroinflammation. As an upstream regulator of type-I IFNs, STING modulation presents a novel therapeutic opportunity to mediate inflammation in the CNS. This review will detail the current knowledge of protective and detrimental STING activity in acute and chronic CNS pathologies and the current therapeutic avenues being explored.
Collapse
Affiliation(s)
- Amelia L Fryer
- Neuropharmacology Laboratory, Department of Pharmacology and Therapeutics, University of Melbourne, Melbourne, VIC, Australia
| | - Amar Abdullah
- Neuropharmacology Laboratory, Department of Pharmacology and Therapeutics, University of Melbourne, Melbourne, VIC, Australia
| | - Juliet M Taylor
- Neuropharmacology Laboratory, Department of Pharmacology and Therapeutics, University of Melbourne, Melbourne, VIC, Australia
| | - Peter J Crack
- Neuropharmacology Laboratory, Department of Pharmacology and Therapeutics, University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
82
|
Mitchell MJ, Billingsley MM, Haley RM, Wechsler ME, Peppas NA, Langer R. Engineering precision nanoparticles for drug delivery. Nat Rev Drug Discov 2021; 20:101-124. [PMID: 33277608 PMCID: PMC7717100 DOI: 10.1038/s41573-020-0090-8] [Citation(s) in RCA: 3736] [Impact Index Per Article: 934.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/16/2020] [Indexed: 12/12/2022]
Abstract
In recent years, the development of nanoparticles has expanded into a broad range of clinical applications. Nanoparticles have been developed to overcome the limitations of free therapeutics and navigate biological barriers - systemic, microenvironmental and cellular - that are heterogeneous across patient populations and diseases. Overcoming this patient heterogeneity has also been accomplished through precision therapeutics, in which personalized interventions have enhanced therapeutic efficacy. However, nanoparticle development continues to focus on optimizing delivery platforms with a one-size-fits-all solution. As lipid-based, polymeric and inorganic nanoparticles are engineered in increasingly specified ways, they can begin to be optimized for drug delivery in a more personalized manner, entering the era of precision medicine. In this Review, we discuss advanced nanoparticle designs utilized in both non-personalized and precision applications that could be applied to improve precision therapies. We focus on advances in nanoparticle design that overcome heterogeneous barriers to delivery, arguing that intelligent nanoparticle design can improve efficacy in general delivery applications while enabling tailored designs for precision applications, thereby ultimately improving patient outcome overall.
Collapse
Affiliation(s)
- Michael J Mitchell
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA.
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | | | - Rebecca M Haley
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Marissa E Wechsler
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Nicholas A Peppas
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA.
- Department of Chemical Engineering, The University of Texas at Austin, Austin, TX, USA.
- Department of Pediatrics, The University of Texas at Austin, Austin, TX, USA.
- Department of Surgery and Perioperative Care, The University of Texas at Austin, Austin, TX, USA.
- Department of Molecular Pharmaceutics and Drug Delivery, The University of Texas at Austin, Austin, TX, USA.
| | - Robert Langer
- Department of Chemical Engineering and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
83
|
Khot S, Rawal SU, Patel MM. Dissolvable-soluble or biodegradable polymers. DRUG DELIVERY DEVICES AND THERAPEUTIC SYSTEMS 2021:367-394. [DOI: 10.1016/b978-0-12-819838-4.00024-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
84
|
Nguyen DC, Shae D, Pagendarm HM, Becker KW, Wehbe M, Kilchrist KV, Pastora LE, Palmer CR, Seber P, Christov PP, Duvall CL, Wilson JT. Amphiphilic Polyelectrolyte Graft Copolymers Enhance the Activity of Cyclic Dinucleotide STING Agonists. Adv Healthc Mater 2021; 10:e2001056. [PMID: 33225632 PMCID: PMC7856189 DOI: 10.1002/adhm.202001056] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 10/08/2020] [Indexed: 11/10/2022]
Abstract
Cyclic dinucleotide (CDN) agonists of stimulator of interferon genes (STING) hold great therapeutic potential, but their activity is hindered by poor drug-like properties that restrict cytosolic bioavailability. Here, this challenge is addressed through the synthesis and evaluation of a novel series of PEGMA-co-DEAEMA-co-BMA copolymers with pH-responsive, membrane-destabilizing activity to enhance intracellular delivery of the CDN, cGAMP. Copolymers are synthesized with PEGMA of two different molecular weights (300 and 950 Da) and over a range of PEG mass fraction and polymer molecular weight, and relationships between copolymer structure, self-assembly, endosomal escape, and cGAMP activity are elucidated. A subset of polymers that self-assembled into 50-800 nm nanoparticles is identified, which can be loaded with cGAMP via a simple mixing strategy, resulting in significantly enhanced immunostimulatory activity. Increased cGAMP activity is found to be highly correlated with the capacity of carriers to enhance intracellular CDN uptake and to promote endosomal destabilization, findings that establish efficient cytosolic delivery as a criterion for CDN carriers. Additionally, it is demonstrated that a lead CDN carrier formulation can enhance STING activation in vivo in a model of intratumoral immunotherapy. Collectively, these investigations demonstrate the utility of PEGMA-co-DEAEMA-co-BMA copolymers as carriers for CDNs and potentially other cytosolically-acting drug cargo.
Collapse
Affiliation(s)
- D. Chuong Nguyen
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN 37235, USA
- The SyBBURE Searle Undergraduate Research Program, Vanderbilt University, Nashville, TN, 37235, USA
| | - Daniel Shae
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Hayden M. Pagendarm
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Kyle W. Becker
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Mohamed Wehbe
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Kameron V. Kilchrist
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Lucinda E. Pastora
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Christian R. Palmer
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Pedro Seber
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Plamen P. Christov
- Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN, 37232
| | - Craig L. Duvall
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - John T. Wilson
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN 37235, USA
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN, 37232
- Vanderbilt Institute of Nanoscale Science and Engineering, Vanderbilt University, Nashville, TN 37232
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| |
Collapse
|
85
|
Liu Y, Lu X, Qin N, Qiao Y, Xing S, Liu W, Feng F, Liu Z, Sun H. STING, a promising target for small molecular immune modulator: A review. Eur J Med Chem 2020; 211:113113. [PMID: 33360799 DOI: 10.1016/j.ejmech.2020.113113] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Revised: 12/13/2020] [Accepted: 12/14/2020] [Indexed: 12/19/2022]
Abstract
Stimulator of interferon genes (STING) plays a crucial role in human innate immune system, which is gradually concerned following the emerging immunotherapy. Activated STING induces the production of type I interferons (IFNs) and proinflammatory cytokines through STING-TBK1-IRF3/NF-κB pathway, which could be applied into the treatment of infection, inflammation, and tumorigenesis. Here, we provided a detailed summary of STING from its structure, function and regulation. Especially, we illustrated the canonical or noncanonical cyclic dinucleotides (CDNs) and synthetic small molecules for STING activation or inhibition and their efficacy in related diseases. Importantly, we particularly emphasized the discovery, development and modification of STING agonist or antagonist, attempting to enlighten reader's mind for enriching small molecular modulator of STING. In addition, we summarized biological evaluation methods for the assessment of small molecules activity.
Collapse
Affiliation(s)
- Yijun Liu
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, People's Republic of China
| | - Xin Lu
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, People's Republic of China
| | - Nan Qin
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing 211198, People's Republic of China
| | - Yuting Qiao
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, People's Republic of China
| | - Shuaishuai Xing
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, People's Republic of China
| | - Wenyuan Liu
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, People's Republic of China
| | - Feng Feng
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing 211198, People's Republic of China; Jiangsu Food and Pharmaceuticals Science College, Institute of Food and Pharmaceuticals Research, 223005, People's Republic of China
| | - Zongliang Liu
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, 264005, PR China.
| | - Haopeng Sun
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, People's Republic of China.
| |
Collapse
|
86
|
Iqbal S, Qu Y, Dong Z, Zhao J, Rauf Khan A, Rehman S, Zhao Z. Poly (β‐amino esters) based potential drug delivery and targeting polymer; an overview and perspectives (review). Eur Polym J 2020. [DOI: 10.1016/j.eurpolymj.2020.110097] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
87
|
Neshat SY, Tzeng SY, Green JJ. Gene delivery for immunoengineering. Curr Opin Biotechnol 2020; 66:1-10. [PMID: 32554325 PMCID: PMC7313888 DOI: 10.1016/j.copbio.2020.05.008] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 05/04/2020] [Accepted: 05/25/2020] [Indexed: 12/14/2022]
Abstract
A growing number of gene delivery strategies are being employed for immunoengineering in applications ranging from infectious disease prevention to cancer therapy. Viral vectors tend to have high gene transfer capability but may be hampered by complications related to their intrinsic immunogenicity. Non-viral methods of gene delivery, including polymeric, lipid-based, and inorganic nanoparticles as well as physical delivery techniques, have also been widely investigated. By using either ex vivo engineering of immune cells that are subsequently adoptively transferred or in vivo transfection of cells for in situ genetic programming, researchers have developed different approaches to precisely modulate immune responses. In addition to expressing a gene of interest through intracellular delivery of plasmid DNA and mRNA, researchers are also delivering oligonucleotides to knock down gene expression and immunostimulatory nucleic acids to tune immune activity. Many of these biotechnologies are now in clinical trials and have high potential to impact medicine.
Collapse
Affiliation(s)
- Sarah Y Neshat
- Department of Biomedical Engineering and the Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Stephany Y Tzeng
- Department of Biomedical Engineering and the Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA.
| | - Jordan J Green
- Department of Biomedical Engineering and the Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD 21218, USA; Departments of Materials Science and Engineering and Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA; Departments of Oncology, Ophthalmology, and Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Bloomberg∼Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA.
| |
Collapse
|
88
|
Chattopadhyay S, Hu CMJ. Nanomedicinal delivery of stimulator of interferon genes agonists: recent advances in virus vaccination. Nanomedicine (Lond) 2020; 15:2883-2894. [PMID: 33252301 DOI: 10.2217/nnm-2020-0269] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The discovery of stimulator of interferon genes (STING) and their agonists as primary components that link antiviral innate and adaptive immunity has motivated growing research on STING agonist-mediated immunotherapy and vaccine development. To overcome the delivery challenge in shuttling highly polar STING agonists, typically in the form of cyclic dinucleotides, to target cells and to STING proteins in cellular cytosol, numerous nanoformulation strategies have been implemented for effective STING activation. While many STING-activating nanoparticles are developed to enhance anticancer immunotherapy, their adoption as vaccine adjuvant has vastly propelled antiviral vaccination efforts against challenging public health threats, including HIV, influenza and coronaviruses. In light of the COVID-19 pandemic that has thrusted vaccine development into the public spotlight, this review highlights advances in nanomedicinal STING agonist delivery with an emphasis on their applications in antiviral vaccination.
Collapse
Affiliation(s)
- Saborni Chattopadhyay
- Taiwan International Graduate Program in Molecular Medicine, National Yang-Ming University & Academic Sinica, Taipei 11529, Taiwan.,Institute of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan
| | - Che-Ming Jack Hu
- Taiwan International Graduate Program in Molecular Medicine, National Yang-Ming University & Academic Sinica, Taipei 11529, Taiwan.,Institute of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan.,Center of Applied Nanomedicine, National Cheng Kung University, Tainan 701, Taiwan
| |
Collapse
|
89
|
Li J, Burgess DJ. Nanomedicine-based drug delivery towards tumor biological and immunological microenvironment. Acta Pharm Sin B 2020; 10:2110-2124. [PMID: 33304781 PMCID: PMC7714990 DOI: 10.1016/j.apsb.2020.05.008] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 05/15/2020] [Accepted: 05/21/2020] [Indexed: 12/17/2022] Open
Abstract
The complex tumor microenvironment is a most important factor in cancer development. The biological microenvironment is composed of a variety of barriers including the extracellular matrix and associated cells such as endothelia cells, pericytes, and cancer-associated fibroblasts. Different strategies can be utilized to enhance nanoparticle-based drug delivery and distribution into tumor tissues addressing the extracellular matrix or cellular components. In addition to the biological microenvironment, the immunological conditions around the tumor tissue can be very complicated and cancer cells have various ways of evading immune surveillance. Nanoparticle drug delivery systems can enhance cancer immunotherapy by tuning the immunological response and memory of various immune cells such as T cells, B cells, macrophages, and dendritic cells. In this review, the main components in the tumor biological and immunological environment are discussed. The focus is on recent advances in nanoparticle-based drug delivery systems towards targets within the tumor microenvironment to improve cancer chemotherapy and immunotherapy.
Collapse
Affiliation(s)
- Jin Li
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT 06269-3092, USA
| | - Diane J. Burgess
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT 06269-3092, USA
| |
Collapse
|
90
|
Shofolawe-Bakare OT, Stokes LD, Hossain M, Smith AE, Werfel TA. Immunostimulatory biomaterials to boost tumor immunogenicity. Biomater Sci 2020; 8:5516-5537. [PMID: 33049007 PMCID: PMC7837217 DOI: 10.1039/d0bm01183e] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cancer immunotherapy is exhibiting great promise as a new therapeutic modality for cancer treatment. However, immunotherapies are limited by the inability of some tumors to provoke an immune response. These tumors with a 'cold' immunological phenotype are characterized by low numbers of tumor-infiltrating lymphocytes, high numbers of immunosuppressive leukocytes (e.g. regulatory T cells, tumor-associated macrophages), and high production of immune-dampening signals (e.g. IL-10, TGF-β, IDO-1). Strategies to boost the aptitude of tumors to initiate an immune response (i.e. boost tumor immunogenicity) will turn 'cold' tumors 'hot' and augment the anti-tumor efficacy of current immunotherapies. Approaches to boost tumor immunogenicity already show promise; however, multifaceted delivery and immunobiology challenges exist. For instance, systemic delivery of many immune-stimulating agents causes off-target toxicity and/or the development of autoimmunity, limiting the administrable dose below the threshold needed to achieve efficacy. Moreover, once administered in vivo, molecules such as the nucleic acid-based agonists for many pattern recognition receptors are either rapidly cleared or degraded, and don't efficiently traffic to the intracellular compartments where the receptors are located. Thus, these nucleic acid-based drugs are ineffective without a delivery system. Biomaterials-based approaches aim to enhance current strategies to boost tumor immunogenicity, enable novel strategies, and spare dose-limiting toxicities. Here, we review recent progress to improve cancer immunotherapies by boosting immunogenicity within tumors using immunostimulatory biomaterials.
Collapse
|
91
|
Motedayen Aval L, Pease JE, Sharma R, Pinato DJ. Challenges and Opportunities in the Clinical Development of STING Agonists for Cancer Immunotherapy. J Clin Med 2020; 9:E3323. [PMID: 33081170 PMCID: PMC7602874 DOI: 10.3390/jcm9103323] [Citation(s) in RCA: 145] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 10/09/2020] [Accepted: 10/12/2020] [Indexed: 02/06/2023] Open
Abstract
Immune checkpoint inhibitors (ICI) have revolutionised cancer therapy. However, they have been effective in only a small subset of patients and a principal mechanism underlying immune-refractoriness is a 'cold' tumour microenvironment, that is, lack of a T-cell-rich, spontaneously inflamed phenotype. As such, there is a demand to develop strategies to transform the tumour milieu of non-responsive patients to one supporting T-cell-based inflammation. The cyclic guanosine monophosphate-adenosine monophosphate synthase-stimulator of interferon genes (cGAS-STING) pathway is a fundamental regulator of innate immune sensing of cancer, with potential to enhance tumour rejection through the induction of a pro-inflammatory response dominated by Type I interferons. Recognition of these positive immune-modulatory properties has rapidly elevated the STING pathway as a putative target for immunotherapy, leading to a myriad of preclinical and clinical studies assessing natural and synthetic cyclic dinucleotides and non-nucleotidyl STING agonists. Despite pre-clinical evidence of efficacy, clinical translation has resulted into disappointingly modest efficacy. Poor pharmacokinetic and physiochemical properties of cyclic dinucleotides are key barriers to the development of STING agonists, most of which require intra-tumoral dosing. Development of systemically administered non-nucleotidyl STING agonists, or conjugation with liposomes, polymers and hydrogels may overcome pharmacokinetic limitations and improve drug delivery. In this review, we summarise the body of evidence supporting a synergistic role of STING agonists with currently approved ICI therapies and discuss whether, despite the numerous obstacles encountered to date, the clinical development of STING agonist as novel anti-cancer therapeutics may still hold the promise of broadening the reach of cancer immunotherapy.
Collapse
Affiliation(s)
- Leila Motedayen Aval
- Department of Surgery & Cancer, Imperial College London, Hammersmith Hospital, Du Cane Road, London W120HS, UK; (L.M.A.); (R.S.)
| | - James E. Pease
- Inflammation, Repair & Development, National Heart & Lung Institute, Imperial College London, London SW7 2AZ, UK;
| | - Rohini Sharma
- Department of Surgery & Cancer, Imperial College London, Hammersmith Hospital, Du Cane Road, London W120HS, UK; (L.M.A.); (R.S.)
| | - David J. Pinato
- Department of Surgery & Cancer, Imperial College London, Hammersmith Hospital, Du Cane Road, London W120HS, UK; (L.M.A.); (R.S.)
| |
Collapse
|
92
|
Karlsson J, Rhodes KR, Green JJ, Tzeng SY. Poly(beta-amino ester)s as gene delivery vehicles: challenges and opportunities. Expert Opin Drug Deliv 2020; 17:1395-1410. [PMID: 32700581 PMCID: PMC7658038 DOI: 10.1080/17425247.2020.1796628] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 07/13/2020] [Indexed: 12/19/2022]
Abstract
INTRODUCTION Gene delivery technologies are being developed for an increasing number of biomedical applications, with delivery vehicles including viruses and non-viral materials. Among biomaterials used for non-viral gene delivery, poly(beta-amino ester)s (PBAEs), a class of synthetic, biodegradable polymers, have risen as a leading gene delivery vehicle that has been used for multiple applications in vitro and in vivo. AREAS COVERED This review summarizes the key properties of PBAEs and their development, including a discussion of the advantages and disadvantages of PBAEs for gene delivery applications. The use of PBAEs to improve the properties of other drug delivery vehicles is also summarized. EXPERT OPINION PBAEs are designed to have multiple characteristics that are ideal for gene delivery, including their reversible positive charge, which promotes binding to nucleic acids as well as imparting high buffering capacity, and their rapid degradability under mild conditions. Simultaneously, some of their properties also lead to nanoparticle instability and low transfection efficiency in physiological environments. The ease with which PBAEs can be chemically modified as well as non-covalently blended with other materials, however, allows them to be customized specifically to overcome delivery barriers for varied applications.
Collapse
Affiliation(s)
- Johan Karlsson
- Department of Biomedical Engineering and the Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Kelly R. Rhodes
- Department of Biomedical Engineering and the Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Jordan J. Green
- Department of Biomedical Engineering and the Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD 21218, USA
- Departments of Materials Science and Engineering and Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Departments of Oncology, Ophthalmology, and Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
- Bloomberg~Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Stephany Y. Tzeng
- Department of Biomedical Engineering and the Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| |
Collapse
|
93
|
Park KS, Xu C, Sun X, Louttit C, Moon JJ. Improving STING Agonist Delivery for Cancer Immunotherapy Using Biodegradable Mesoporous Silica Nanoparticles. ADVANCED THERAPEUTICS 2020; 3:2000130. [PMID: 34295963 PMCID: PMC8294168 DOI: 10.1002/adtp.202000130] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Indexed: 12/19/2022]
Abstract
Stimulator of interferon genes (STING) activation by intratumoral STING agonist treatment has been recently shown to eradicate tumors in preclinical models of cancer immunotherapy, generating intense research interest and leading to multiple clinical trials. However, there are many challenges associated with STING agonist-based cancer immunotherapy, including low cellular uptake of STING agonists. Here, biodegradable mesoporous silica nanoparticles (bMSN) with an average size of 80 nm are developed for efficient cellular delivery of STING agonists. STING agonists delivered via bMSN potently activate innate and adaptive immune cells, leading to strong antitumor efficacy and prolonged animal survival in murine models of melanoma. Delivery of immunotherapeutic agents via biodegradable bMSN is a promising approach for improving cancer immunotherapy.
Collapse
Affiliation(s)
- Kyung Soo Park
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA, Biointerfaces Institute, University of Michigan, Ann Arbor MI 48109, USA
| | - Cheng Xu
- Biointerfaces Institute, University of Michigan, Ann Arbor MI 48109, USA, Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor MI 48109, USA
| | - Xiaoqi Sun
- Biointerfaces Institute, University of Michigan, Ann Arbor MI 48109, USA, Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor MI 48109, USA
| | - Cameron Louttit
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA, Biointerfaces Institute, University of Michigan, Ann Arbor MI 48109, USA
| | - James J Moon
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA, Biointerfaces Institute, University of Michigan, Ann Arbor MI 48109, USA, Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor MI 48109, USA
| |
Collapse
|
94
|
Bockamp E, Rosigkeit S, Siegl D, Schuppan D. Nano-Enhanced Cancer Immunotherapy: Immunology Encounters Nanotechnology. Cells 2020; 9:E2102. [PMID: 32942725 PMCID: PMC7565449 DOI: 10.3390/cells9092102] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 09/07/2020] [Accepted: 09/11/2020] [Indexed: 02/06/2023] Open
Abstract
Cancer immunotherapy utilizes the immune system to fight cancer and has already moved from the laboratory to clinical application. However, and despite excellent therapeutic outcomes in some hematological and solid cancers, the regular clinical use of cancer immunotherapies reveals major limitations. These include the lack of effective immune therapy options for some cancer types, unresponsiveness to treatment by many patients, evolving therapy resistance, the inaccessible and immunosuppressive nature of the tumor microenvironment (TME), and the risk of potentially life-threatening immune toxicities. Given the potential of nanotechnology to deliver, enhance, and fine-tune cancer immunotherapeutic agents, the combination of cancer immunotherapy with nanotechnology can overcome some of these limitations. In this review, we summarize innovative reports and novel strategies that successfully combine nanotechnology and cancer immunotherapy. We also provide insight into how nanoparticular combination therapies can be used to improve therapy responsiveness, to reduce unwanted toxicity, and to overcome adverse effects of the TME.
Collapse
Affiliation(s)
- Ernesto Bockamp
- Institute of Translational Immunology, University Medical Center, Johannes Gutenberg University Mainz, 55131 Mainz, Germany; (E.B.); (S.R.); (D.S.)
- Research Center for Immunotherapy, University Medical Center, Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| | - Sebastian Rosigkeit
- Institute of Translational Immunology, University Medical Center, Johannes Gutenberg University Mainz, 55131 Mainz, Germany; (E.B.); (S.R.); (D.S.)
- Research Center for Immunotherapy, University Medical Center, Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| | - Dominik Siegl
- Institute of Translational Immunology, University Medical Center, Johannes Gutenberg University Mainz, 55131 Mainz, Germany; (E.B.); (S.R.); (D.S.)
- Research Center for Immunotherapy, University Medical Center, Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| | - Detlef Schuppan
- Institute of Translational Immunology, University Medical Center, Johannes Gutenberg University Mainz, 55131 Mainz, Germany; (E.B.); (S.R.); (D.S.)
- Research Center for Immunotherapy, University Medical Center, Johannes Gutenberg University Mainz, 55131 Mainz, Germany
- Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| |
Collapse
|
95
|
Abstract
Personalized cancer vaccines (PCVs) are reinvigorating vaccine strategies in cancer immunotherapy. In contrast to adoptive T-cell therapy and checkpoint blockade, the PCV strategy modulates the innate and adaptive immune systems with broader activation to redeploy antitumor immunity with individualized tumor-specific antigens (neoantigens). Following a sequential scheme of tumor biopsy, mutation analysis, and epitope prediction, the administration of neoantigens with synthetic long peptide (SLP) or mRNA formulations dramatically improves the population and activity of antigen-specific CD4+ and CD8+ T cells. Despite the promising prospect of PCVs, there is still great potential for optimizing prevaccination procedures and vaccine potency. In particular, the arduous development of tumor-associated antigen (TAA)-based vaccines provides valuable experience and rational principles for augmenting vaccine potency which is expected to advance PCV through the design of adjuvants, delivery systems, and immunosuppressive tumor microenvironment (TME) reversion since current personalized vaccination simply admixes antigens with adjuvants. Considering the broader application of TAA-based vaccine design, these two strategies complement each other and can lead to both personalized and universal therapeutic methods. Chemical strategies provide vast opportunities for (1) exploring novel adjuvants, including synthetic molecules and materials with optimizable activity, (2) constructing efficient and precise delivery systems to avoid systemic diffusion, improve biosafety, target secondary lymphoid organs, and enhance antigen presentation, and (3) combining bioengineering methods to innovate improvements in conventional vaccination, "smartly" re-educate the TME, and modulate antitumor immunity. As chemical strategies have proven versatility, reliability, and universality in the design of T cell- and B cell-based antitumor vaccines, the union of such numerous chemical methods in vaccine construction is expected to provide new vigor and vitality in cancer treatment.
Collapse
Affiliation(s)
- Wen-Hao Li
- Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Department of Chemistry, Tsinghua University, 100084 Beijing, China
| | - Yan-Mei Li
- Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Department of Chemistry, Tsinghua University, 100084 Beijing, China.,Beijing Institute for Brain Disorders, 100069 Beijing, China.,Center for Synthetic and Systems Biology, Tsinghua University, 100084 Beijing, China
| |
Collapse
|
96
|
Abdou P, Wang Z, Chen Q, Chan A, Zhou DR, Gunadhi V, Gu Z. Advances in engineering local drug delivery systems for cancer immunotherapy. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2020; 12:e1632. [PMID: 32255276 PMCID: PMC7725287 DOI: 10.1002/wnan.1632] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 03/03/2020] [Accepted: 03/04/2020] [Indexed: 12/13/2022]
Abstract
Cancer immunotherapy aims to leverage the immune system to suppress the growth of tumors and to inhibit metastasis. The recent promising clinical outcomes associated with cancer immunotherapy have prompted research and development efforts towards enhancing the efficacy of immune checkpoint blockade, cancer vaccines, cytokine therapy, and adoptive T cell therapy. Advancements in biomaterials, nanomedicine, and micro-/nano-technology have facilitated the development of enhanced local delivery systems for cancer immunotherapy, which can enhance treatment efficacy while minimizing toxicity. Furthermore, locally administered cancer therapies that combine immunotherapy with chemotherapy, radiotherapy, or phototherapy have the potential to achieve synergistic antitumor effects. Herein, the latest studies on local delivery systems for cancer immunotherapy are surveyed, with an emphasis on the therapeutic benefits associated with the design of biomaterials and nanomedicines. This article is categorized under: Nanotechnology Approaches to Biology > Nanoscale Systems in Biology Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease.
Collapse
Affiliation(s)
- Peter Abdou
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA
- Jonsson Comprehensive Cancer Center, California NanoSystems Institute, and Center for Minimally Invasive Therapeutics, University of California, Los Angeles, CA 90095, USA
| | - Zejun Wang
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA
- Jonsson Comprehensive Cancer Center, California NanoSystems Institute, and Center for Minimally Invasive Therapeutics, University of California, Los Angeles, CA 90095, USA
| | - Qian Chen
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, 199 Ren’ai Road, Suzhou, 215123, Jiangsu, PR China
| | - Amanda Chan
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA
- Jonsson Comprehensive Cancer Center, California NanoSystems Institute, and Center for Minimally Invasive Therapeutics, University of California, Los Angeles, CA 90095, USA
| | - Daojia R. Zhou
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA
- Jonsson Comprehensive Cancer Center, California NanoSystems Institute, and Center for Minimally Invasive Therapeutics, University of California, Los Angeles, CA 90095, USA
| | - Vivienne Gunadhi
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA
| | - Zhen Gu
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA
- Jonsson Comprehensive Cancer Center, California NanoSystems Institute, and Center for Minimally Invasive Therapeutics, University of California, Los Angeles, CA 90095, USA
| |
Collapse
|
97
|
Siddique S, Chow JCL. Application of Nanomaterials in Biomedical Imaging and Cancer Therapy. NANOMATERIALS (BASEL, SWITZERLAND) 2020; 10:E1700. [PMID: 32872399 PMCID: PMC7559738 DOI: 10.3390/nano10091700] [Citation(s) in RCA: 186] [Impact Index Per Article: 37.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 08/24/2020] [Accepted: 08/27/2020] [Indexed: 12/11/2022]
Abstract
Nanomaterials, such as nanoparticles, nanorods, nanosphere, nanoshells, and nanostars, are very commonly used in biomedical imaging and cancer therapy. They make excellent drug carriers, imaging contrast agents, photothermal agents, photoacoustic agents, and radiation dose enhancers, among other applications. Recent advances in nanotechnology have led to the use of nanomaterials in many areas of functional imaging, cancer therapy, and synergistic combinational platforms. This review will systematically explore various applications of nanomaterials in biomedical imaging and cancer therapy. The medical imaging modalities include magnetic resonance imaging, computed tomography, positron emission tomography, single photon emission computerized tomography, optical imaging, ultrasound, and photoacoustic imaging. Various cancer therapeutic methods will also be included, including photothermal therapy, photodynamic therapy, chemotherapy, and immunotherapy. This review also covers theranostics, which use the same agent in diagnosis and therapy. This includes recent advances in multimodality imaging, image-guided therapy, and combination therapy. We found that the continuous advances of synthesis and design of novel nanomaterials will enhance the future development of medical imaging and cancer therapy. However, more resources should be available to examine side effects and cell toxicity when using nanomaterials in humans.
Collapse
Affiliation(s)
- Sarkar Siddique
- Department of Physics, Ryerson University, Toronto, ON M5B 2K3, Canada;
| | - James C. L. Chow
- Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1X6, Canada
- Department of Radiation Oncology, University of Toronto, Toronto, ON M5T 1P5, Canada
| |
Collapse
|
98
|
Zheng J, Mo J, Zhu T, Zhuo W, Yi Y, Hu S, Yin J, Zhang W, Zhou H, Liu Z. Comprehensive elaboration of the cGAS-STING signaling axis in cancer development and immunotherapy. Mol Cancer 2020; 19:133. [PMID: 32854711 PMCID: PMC7450153 DOI: 10.1186/s12943-020-01250-1] [Citation(s) in RCA: 165] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 08/17/2020] [Indexed: 02/08/2023] Open
Abstract
Cellular recognition of microbial DNA is an evolutionarily conserved mechanism by which the innate immune system detects pathogens. Cyclic GMP-AMP synthase (cGAS) and its downstream effector, stimulator of interferon genes (STING), are involved in mediating fundamental innate antimicrobial immunity by promoting the release of type I interferons (IFNs) and other inflammatory cytokines. Accumulating evidence suggests that the activation of the cGAS-STING axis is critical for antitumor immunity. The downstream cytokines regulated by cGAS-STING, especially type I IFNs, serve as bridges connecting innate immunity with adaptive immunity. Accordingly, a growing number of studies have focused on the synthesis and screening of STING pathway agonists. However, chronic STING activation may lead to a protumor phenotype in certain malignancies. Hence, the cGAS-STING signaling pathway must be orchestrated properly when STING agonists are used alone or in combination. In this review, we discuss the dichotomous roles of the cGAS-STING pathway in tumor development and the latest advances in the use of STING agonists.
Collapse
Affiliation(s)
- Juyan Zheng
- Department of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, and National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, People's Republic of China.,Institute of Clinical Pharmacology, Engineering Research Center for applied Technology of Pharmacogenomics of Ministry of Education, Central South University, Changsha, 410078, People's Republic of China
| | - Junluan Mo
- Shenzhen center for chronic disease control and Prevention, Shenzhen, 518020, People's Republic of China
| | - Tao Zhu
- Department of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, and National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, People's Republic of China.,Institute of Clinical Pharmacology, Engineering Research Center for applied Technology of Pharmacogenomics of Ministry of Education, Central South University, Changsha, 410078, People's Republic of China
| | - Wei Zhuo
- Department of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, and National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, People's Republic of China.,Institute of Clinical Pharmacology, Engineering Research Center for applied Technology of Pharmacogenomics of Ministry of Education, Central South University, Changsha, 410078, People's Republic of China
| | - Yueneng Yi
- Hunan Yineng Biological Medicine Co., Ltd, Changsha, 410205, People's Republic of China
| | - Shuo Hu
- Department of Nuclear Medicine, Key Laboratory of Biological Nanotechnology of National Health Commission, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People's Republic of China
| | - Jiye Yin
- Department of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, and National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, People's Republic of China.,Institute of Clinical Pharmacology, Engineering Research Center for applied Technology of Pharmacogenomics of Ministry of Education, Central South University, Changsha, 410078, People's Republic of China
| | - Wei Zhang
- Department of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, and National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, People's Republic of China.,Institute of Clinical Pharmacology, Engineering Research Center for applied Technology of Pharmacogenomics of Ministry of Education, Central South University, Changsha, 410078, People's Republic of China
| | - Honghao Zhou
- Department of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, and National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, People's Republic of China.,Institute of Clinical Pharmacology, Engineering Research Center for applied Technology of Pharmacogenomics of Ministry of Education, Central South University, Changsha, 410078, People's Republic of China
| | - Zhaoqian Liu
- Department of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, and National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, People's Republic of China. .,Institute of Clinical Pharmacology, Engineering Research Center for applied Technology of Pharmacogenomics of Ministry of Education, Central South University, Changsha, 410078, People's Republic of China.
| |
Collapse
|
99
|
Shae D, Baljon JJ, Wehbe M, Christov PP, Becker KW, Kumar A, Suryadevara N, Carson CS, Palmer CR, Knight FC, Joyce S, Wilson JT. Co-delivery of Peptide Neoantigens and Stimulator of Interferon Genes Agonists Enhances Response to Cancer Vaccines. ACS NANO 2020; 14:9904-9916. [PMID: 32701257 PMCID: PMC7775800 DOI: 10.1021/acsnano.0c02765] [Citation(s) in RCA: 105] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
Cancer vaccines targeting patient-specific neoantigens have emerged as a promising strategy for improving responses to immune checkpoint blockade. However, neoantigenic peptides are poorly immunogenic and inept at stimulating CD8+ T cell responses, motivating a need for new vaccine technologies that enhance their immunogenicity. The stimulator of interferon genes (STING) pathway is an endogenous mechanism by which the innate immune system generates an immunological context for priming and mobilizing neoantigen-specific T cells. Owing to this critical role in tumor immune surveillance, a synthetic cancer nanovaccine platform (nanoSTING-vax) was developed that mimics immunogenic cancer cells in its capacity to efficiently promote co-delivery of peptide antigens and the STING agonist, cGAMP. The co-loading of cGAMP and peptides into pH-responsive, endosomolytic polymersomes promoted the coordinated delivery of both cGAMP and peptide antigens to the cytosol, thereby eliciting inflammatory cytokine production, co-stimulatory marker expression, and antigen cross-presentation. Consequently, nanoSTING-vax significantly enhanced CD8+ T cell responses to a range of peptide antigens. Therapeutic immunization with nanoSTING-vax, in combination with immune checkpoint blockade, inhibited tumor growth in multiple murine tumor models, even leading to complete tumor rejection and generation of durable antitumor immune memory. Collectively, this work establishes nanoSTING-vax as a versatile platform for enhancing immune responses to neoantigen-targeted cancer vaccines.
Collapse
Affiliation(s)
- Daniel Shae
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Jessalyn J. Baljon
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Mohamed Wehbe
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Plamen P. Christov
- Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN, 37232
| | - Kyle W. Becker
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Amrendra Kumar
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN 37212, USA
| | - Naveenchandra Suryadevara
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN 37212, USA
| | - Carcia S. Carson
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Christian R. Palmer
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Frances C. Knight
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Sebastian Joyce
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN 37212, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - John T. Wilson
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN 37235, USA
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
- Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN, 37232
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| |
Collapse
|
100
|
Gogoi H, Mansouri S, Jin L. The Age of Cyclic Dinucleotide Vaccine Adjuvants. Vaccines (Basel) 2020; 8:E453. [PMID: 32823563 PMCID: PMC7563944 DOI: 10.3390/vaccines8030453] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Revised: 08/10/2020] [Accepted: 08/12/2020] [Indexed: 02/07/2023] Open
Abstract
As prophylactic vaccine adjuvants for infectious diseases, cyclic dinucleotides (CDNs) induce safe, potent, long-lasting humoral and cellular memory responses in the systemic and mucosal compartments. As therapeutic cancer vaccine adjuvants, CDNs induce potent anti-tumor immunity, including cytotoxic T cells and NK cells activation that achieve durable regression in multiple mouse models of tumors. Clinical trials are ongoing to fulfill the promise of CDNs (ClinicalTrials.gov: NCT02675439, NCT03010176, NCT03172936, and NCT03937141). However, in October 2018, the first clinical data with Merck's CDN MK-1454 showed zero activity as a monotherapy in patients with solid tumors or lymphomas (NCT03010176). Lately, the clinical trial from Aduro's CDN ADU-S100 monotherapy was also disappointing (NCT03172936). The emerging hurdle in CDN vaccine development calls for a timely re-evaluation of our understanding on CDN vaccine adjuvants. Here, we review the status of CDN vaccine adjuvant research, including their superior adjuvant activities, in vivo mode of action, and confounding factors that affect their efficacy in humans. Lastly, we discuss the strategies to overcome the hurdle and advance promising CDN adjuvants in humans.
Collapse
Affiliation(s)
| | | | - Lei Jin
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Florida, Gainesville, FL 32610, USA; (H.G.); (S.M.)
| |
Collapse
|