1
|
Hu C, Jiang Y, Chen Y, Wang Y, Wu Z, Zhang Q, Wu M. Low-Intensity Focused Ultrasound-Responsive Phase-Transitional Liposomes Loaded with STING Agonist Enhances Immune Activation for Breast Cancer Immunotherapy. Cancers (Basel) 2024; 16:3657. [PMID: 39518096 PMCID: PMC11545222 DOI: 10.3390/cancers16213657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 10/26/2024] [Accepted: 10/27/2024] [Indexed: 11/16/2024] Open
Abstract
Background: Pharmacologically targeting the STING pathway offers a novel approach to cancer immunotherapy. However, small-molecule STING agonists face challenges such as poor tumor accumulation, rapid clearance, and short-lived effects within the tumor microenvironment, thus limiting their therapeutic potential. To address the challenges of poor specificity and inadequate targeting of STING in breast cancer treatment, herein, we report the design and development of a targeted liposomal delivery system modified with the tumor-targeting peptide iRGD (iRGD-STING-PFP@liposomes). With LIFU irradiation, the liposomal system exploits acoustic cavitation, where gas nuclei form and collapse within the hydrophobic region of the liposome lipid bilayer (transient pore formation), which leads to significantly enhanced drug release. Methods: Transmission electron microscopy (TEM) was used to investigate the physicochemical properties of the targeted liposomes. Encapsulation efficiency and in vitro release were assessed using the dialysis bag method, while the effects of iRGD on liposome targeting were evaluated through laser confocal microscopy. The CCK-8 assay was used to investigate the toxicity and cell growth effects of this system on 4T1 breast cancer cells and HUVEC vascular endothelial cells. A subcutaneous breast cancer tumor model was established to evaluate the tumor-killing effects and therapeutic mechanism of the newly developed liposomes. Results: The liposome carrier exhibited a regular morphology, with a particle size of 232.16 ± 19.82 nm, as indicated by dynamic light scattering (DLS), and demonstrated low toxicity to both HUVEC and 4T1 cells. With an encapsulation efficiency of 41.82 ± 5.67%, the carrier exhibited a slow release pattern in vitro after STING loading. Targeting results indicated that iRGD modification enhanced the system's ability to target 4T1 cells. The iRGD-STING-PFP@liposomes group demonstrated significant tumor growth inhibition in the subcutaneous breast cancer mouse model with effective activation of the immune system, resulting in the highest populations of matured dendritic cells (71.2 ± 5.4%), increased presentation of tumor-related antigens, promoted CD8+ T cell infiltration at the tumor site, and enhanced NK cell activity. Conclusions: The iRGD-STING-PFP@liposomes targeted drug delivery system effectively targets breast cancer cells, providing a new strategy for breast cancer immunotherapy. These findings indicate that iRGD-STING-PFP@liposomes could successfully deliver STING agonists to tumor tissue, trigger the innate immune response, and may serve as a potential platform for targeted immunotherapy.
Collapse
Affiliation(s)
- Cong Hu
- Department of Radiation Oncology, the First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China;
- Department of Ultrasound, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; (Y.J.); (Y.C.); (Z.W.); (Q.Z.)
| | - Yuancheng Jiang
- Department of Ultrasound, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; (Y.J.); (Y.C.); (Z.W.); (Q.Z.)
| | - Yixin Chen
- Department of Ultrasound, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; (Y.J.); (Y.C.); (Z.W.); (Q.Z.)
| | - Ying Wang
- Teaching Office, Zhongnan Hospital of Wuhan University, Wuhan 430071, China;
| | - Ziling Wu
- Department of Ultrasound, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; (Y.J.); (Y.C.); (Z.W.); (Q.Z.)
| | - Qi Zhang
- Department of Ultrasound, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; (Y.J.); (Y.C.); (Z.W.); (Q.Z.)
| | - Meng Wu
- Department of Ultrasound, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; (Y.J.); (Y.C.); (Z.W.); (Q.Z.)
| |
Collapse
|
2
|
Yu J, Li X, Li J, Sun N, Cheng P, Huang J, Li S, Kuai R. Single-Dose Physically Cross-Linked Hyaluronic Acid and Lipid Hybrid Nanoparticles Containing Cyclic Guanosine Monophosphate-Adenosine Monophosphate Eliminate Established Tumors. ACS NANO 2024; 18:29942-29955. [PMID: 39418110 DOI: 10.1021/acsnano.4c10673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Activating the STING pathway in the cytosol of tumor-infiltrating antigen-presenting cells (APCs) represents a promising strategy to elicit potent antitumor immune responses for cancer therapy. However, STING agonists are mostly small hydrophilic molecules that suffer from rapid clearance and poor cytosolic delivery following systemic administration. While various nanoparticles have been developed to promote cytosolic delivery, they often exhibit premature drug release during circulation. Alternatively, stable nanoparticles with sustained release during circulation have poor cytosolic delivery. In this study, we have developed physically cross-linked hyaluronic acid (HA) and lipid hybrid nanoparticles containing cyclic guanosine monophosphate-adenosine monophosphate (cGAMP), denoted as HLHC, to address these challenges. The HLH delivery system has sustained drug release due to multiple lipid layers physically cross-linked by HA. HLHC efficiently delivers cGAMP to the cytosol of APCs, inducing more IFNβ than cGAMP and liposomal cGAMP. HLH also improves the drug circulation time and biodistribution to the tumor compared with the liposomal formulation and free drug. Strikingly, a single dose of HLHC, but not liposomal cGAMP or free cGAMP, elicits potent antitumor immunity and regresses established MC38 tumors. A single dose of HLHC even regresses established B16F10 tumors upon combination with αPD-L1. Moreover, cured animals were protected from rechallenge with the same tumor cells. HLHC represents an efficient strategy to address delivery challenges associated with STING agonists and may have broad applications for the delivery of drugs acting in the cytosol.
Collapse
Affiliation(s)
- Jinchao Yu
- School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China
- Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
| | - Xinyan Li
- School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China
| | - Junyao Li
- School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China
| | - Nan Sun
- School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China
| | - Peng Cheng
- Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
- School of Life Sciences, Tsinghua University, Beijing 100084, China
- Frontier Research Center for Biological Structure & State Key Laboratory of Membrane Biology, Beijing 100084, China
| | - Jiayi Huang
- School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China
| | - Sai Li
- Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
- School of Life Sciences, Tsinghua University, Beijing 100084, China
- Frontier Research Center for Biological Structure & State Key Laboratory of Membrane Biology, Beijing 100084, China
| | - Rui Kuai
- School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China
- Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
| |
Collapse
|
3
|
Zheng S, Guo Y, Han Q, Peng X, Sheng R, Liu S, Li Z. STING agonists and PI3Kγ inhibitor co-loaded ferric ion-punicalagin networks for comprehensive cancer therapy. Int J Biol Macromol 2024; 282:136776. [PMID: 39454928 DOI: 10.1016/j.ijbiomac.2024.136776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 10/10/2024] [Accepted: 10/19/2024] [Indexed: 10/28/2024]
Abstract
Nanoparticles-based drug delivery system has been a promising approach for the treatment of colorectal cancer (CRC), which can be combined with chemotherapy, targeted therapy and immunotherapy to improve the treatment of CRC. 2'3' cyclic guanosine monophosphate-adenosine monophosphate (cGAMP) is an agonist of the STING signaling pathway activating antitumor immunity. IPI-549 is a small-molecule inhibitor for phosphatidylinositol 3-kinase γ (PI3Kγ), which can induce M1 macrophages polarization to provide pro-inflammatory microenvironment to suppress tumors. Here, we developed a ferric ion-punicalagin network (Fe-PU), which can be not only used as an inducer of ferroptosis, but also serve as a carrier to load cGAMP and IPI-549 to obtain nanohybrid (Fe-PU/CD-IPI). In order to improve the delivery effect and targeted ability to CRC, a cyclic arginine-glycine-aspartic acid peptide linked-bovine serum albumin were utilized to modify Fe-PU/CD-IPI to prepare nanohybrid Fe-PU/CD-IPI@cBSA. The therapeutic effect of various nanohybrids were validated in the mice with spontaneous tumor in the colorectal area and tumor-bearing mice, which lead to the increase of ferroptosis, the activation of STING signaling pathway, and the repolarization of macrophages. Collectively, the cGAMP and IPI-549 co-loaded nanohybrids effectively reshaped the tumor immune microenvironment, and exhibited prominent treatment effect of anti-colorectal cancer in vitro, patient-derived organoids, and in vivo.
Collapse
Affiliation(s)
- Shaoqin Zheng
- College of Life and Health Sciences, Northeastern University, Shenyang 110819, China
| | - Yitong Guo
- College of Life and Health Sciences, Northeastern University, Shenyang 110819, China
| | - Qing Han
- College of Life and Health Sciences, Northeastern University, Shenyang 110819, China
| | - Xueqiang Peng
- Department of General Surgery, the Fourth Affiliated Hospital, China Medical University, Shenyang 110032, China
| | - Ren Sheng
- College of Life and Health Sciences, Northeastern University, Shenyang 110819, China.
| | - Siyu Liu
- College of Life and Health Sciences, Northeastern University, Shenyang 110819, China.
| | - Zhuang Li
- Department of Anorectal Surgery, The First Hospital of China Medical University, Shenyang 110001, China.
| |
Collapse
|
4
|
Chada NC, Wilson JT. Jump-starting chimeric antigen receptor-T cells to go the extra mile with nanotechnology. Curr Opin Biotechnol 2024; 89:103179. [PMID: 39168033 DOI: 10.1016/j.copbio.2024.103179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 11/16/2023] [Accepted: 07/22/2024] [Indexed: 08/23/2024]
Abstract
Despite success in treating hematologic malignancies, chimeric antigen receptor-T cell (CAR-T) therapy still faces multiple challenges that have halted progress, especially against solid tumors. Recent advances in nanoscale engineeirng provide several avenues for overcoming these challenges, including more efficienct programming of CAR-Ts ex vivo, promoting immune responsiveness in the tumor microenvironment (TME) in vivo, and boosting CAR-T function in situ. Here, we summarize recent innovations that leverage nanotechnology to directly address the major obstacles that impede CAR-T therapy from reaching its full potential across various cancer types. We conclude with a commentary on the state of the field and how nanotechnology can shape the future of CAR-T and adoptive cell therapy in immuno-oncology.
Collapse
Affiliation(s)
- Neil C Chada
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA; Medical Scientist Training Program, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - John T Wilson
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA; Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN 37235, USA; Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Vanderbilt Institute of Chemical Biology, 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
|
5
|
Wang R, Kumar P, Reda M, Wallstrum AG, Crumrine NA, Ngamcherdtrakul W, Yantasee W. Nanotechnology Applications in Breast Cancer Immunotherapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2308639. [PMID: 38126905 PMCID: PMC11493329 DOI: 10.1002/smll.202308639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 11/21/2023] [Indexed: 12/23/2023]
Abstract
Next-generation cancer treatments are expected not only to target cancer cells but also to simultaneously train immune cells to combat cancer while modulating the immune-suppressive environment of tumors and hosts to ensure a robust and lasting response. Achieving this requires carriers that can codeliver multiple therapeutics to the right cancer and/or immune cells while ensuring patient safety. Nanotechnology holds great potential for addressing these challenges. This article highlights the recent advances in nanoimmunotherapeutic development, with a focus on breast cancer. While immune checkpoint inhibitors (ICIs) have achieved remarkable success and lead to cures in some cancers, their response rate in breast cancer is low. The poor response rate in solid tumors is often associated with the low infiltration of anti-cancer T cells and an immunosuppressive tumor microenvironment (TME). To enhance anti-cancer T-cell responses, nanoparticles are employed to deliver ICIs, bispecific antibodies, cytokines, and agents that induce immunogenic cancer cell death (ICD). Additionally, nanoparticles are used to manipulate various components of the TME, such as immunosuppressive myeloid cells, macrophages, dendritic cells, and fibroblasts to improve T-cell activities. Finally, this article discusses the outlook, challenges, and future directions of nanoimmunotherapeutics.
Collapse
Affiliation(s)
- Ruijie Wang
- Department of Biomedical Engineering, Oregon Health & Science University, 3303 S Bond Ave, Portland, OR 97239, USA
| | - Pramod Kumar
- Department of Biomedical Engineering, Oregon Health & Science University, 3303 S Bond Ave, Portland, OR 97239, USA
| | - Moataz Reda
- PDX Pharmaceuticals, 3303 S Bond Ave, CH13B, Portland, OR 97239, USA
| | | | - Noah A. Crumrine
- PDX Pharmaceuticals, 3303 S Bond Ave, CH13B, Portland, OR 97239, USA
| | | | - Wassana Yantasee
- Department of Biomedical Engineering, Oregon Health & Science University, 3303 S Bond Ave, Portland, OR 97239, USA
- PDX Pharmaceuticals, 3303 S Bond Ave, CH13B, Portland, OR 97239, USA
| |
Collapse
|
6
|
Sheehy T, Kwiatkowski AJ, Arora K, Kimmel BR, Schulman JA, Gibson-Corley KN, Wilson JT. STING-Activating Polymer-Drug Conjugates for Cancer Immunotherapy. ACS CENTRAL SCIENCE 2024; 10:1765-1781. [PMID: 39345818 PMCID: PMC11428287 DOI: 10.1021/acscentsci.4c00579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 08/09/2024] [Accepted: 08/12/2024] [Indexed: 10/01/2024]
Abstract
The stimulator of interferon genes (STING) pathway links innate and adaptive antitumor immunity and therefore plays an important role in cancer immune surveillance. This has prompted widespread development of STING agonists for cancer immunotherapy, but pharmacological barriers continue to limit the clinical impact of STING agonists and motivate the development of drug delivery systems to improve their efficacy and/or safety. We developed SAPCon, a STING-activating polymer-drug conjugate platform based on strain-promoted azide-alkyne cycloaddition of a novel dimeric amidobenzimidazole (diABZI) STING prodrug to hydrophilic poly(dimethylacrylamide-co-azido-ethylmethacrylate) polymer chains through a cathepsin B-responsive linker to increase circulation time and enable passive tumor accumulation. We found that intravenously administered SAPCon accumulated at tumor sites, where it was endocytosed by tumor-associated myeloid cells, resulting in increased STING activation in the tumor tissue. Consequently, SAPCon promoted an immunogenic tumor microenvironment characterized by increased frequency of activated macrophages and dendritic cells and improved infiltration of CD8+ T cells, resulting in inhibition of tumor growth, prolonged survival, and enhanced response to anti-PD-1 immune checkpoint blockade in orthotopic breast cancer models. Collectively, these studies position SAPCon as a modular and programmable platform for improving the efficacy of systemically administered STING agonists for cancer immunotherapy.
Collapse
Affiliation(s)
- Taylor
L. Sheehy
- Department
of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Alexander J. Kwiatkowski
- Department
of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Karan Arora
- Department
of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Blaise R. Kimmel
- Department
of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Jacob A. Schulman
- Department
of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Katherine N. Gibson-Corley
- Department
of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
| | - John T. Wilson
- Department
of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37232, United States
- Department
of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, Tennessee 37232, United States
- Department
of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
- Vanderbilt
Ingram Cancer Center, Vanderbilt University
Medical Center, Nashville, Tennessee 37232, United States
- Vanderbilt
Institute of Chemical Biology, Vanderbilt
University, Nashville, Tennessee 37232, United States
- Vanderbilt
Institute for Infection, Immunology and Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
- Vanderbilt
Center for Immunobiology, Vanderbilt University
Medical Center, Nashville, Tennessee 37232, United States
| |
Collapse
|
7
|
Sefat KMSR, Kumar M, Kehl S, Kulkarni R, Leekha A, Paniagua MM, Ackart DF, Jones N, Spencer C, Podell BK, Ouellet H, Varadarajan N. An intranasal nanoparticle vaccine elicits protective immunity against Mycobacterium tuberculosis. Vaccine 2024; 42:125909. [PMID: 38704256 DOI: 10.1016/j.vaccine.2024.04.055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 04/15/2024] [Accepted: 04/18/2024] [Indexed: 05/06/2024]
Abstract
Mucosal vaccines have the potential to elicit protective immune responses at the point of entry of respiratory pathogens, thus preventing even the initial seed infection. Unlike licensed injectable vaccines, mucosal vaccines comprising protein subunits are only in development. One of the primary challenges associated with mucosal vaccines has been identifying and characterizing safe yet effective mucosal adjuvants that can effectively prime multi-factorial mucosal immunity. In this study, we tested NanoSTING, a liposomal formulation of the endogenous activator of the stimulator of interferon genes (STING) pathway, cyclic guanosine adenosine monophosphate (cGAMP), as a mucosal adjuvant. We formulated a vaccine based on the H1 antigen (fusion protein of Ag85b and ESAT-6) adjuvanted with NanoSTING. Intranasal immunization of NanoSTING-H1 elicited a strong T-cell response in the lung of vaccinated animals characterized by (a) CXCR3+ KLRG1- lung resident T cells that are known to be essential for controlling bacterial infection, (b) IFNγ-secreting CD4+ T cells which is necessary for intracellular bactericidal activity, and (c) IL17-secreting CD4+ T cells that can confer protective immunity against multiple clinically relevant strains of Mtb. Upon challenge with aerosolized Mycobacterium tuberculosis Erdman strain, intranasal NanoSTING-H1 provides protection comparable to subcutaneous administration of the live attenuated Mycobacterium bovis vaccine strain Bacille-Calmette-Guérin (BCG). Our results indicate that NanoSTING adjuvanted protein vaccines can elicit a multi-factorial immune response that protects from infection by M. tuberculosis.
Collapse
Affiliation(s)
- K M Samiur Rahman Sefat
- Department of Chemical and Biomolecular Engineering, University of Houston, Houston, TX 77054, USA
| | - Monish Kumar
- Department of Chemical and Biomolecular Engineering, University of Houston, Houston, TX 77054, USA
| | - Stephanie Kehl
- Department of Biological Sciences, University of Texas, El Paso, TX 79968, USA
| | - Rohan Kulkarni
- Department of Chemical and Biomolecular Engineering, University of Houston, Houston, TX 77054, USA
| | - Ankita Leekha
- Department of Chemical and Biomolecular Engineering, University of Houston, Houston, TX 77054, USA
| | - Melisa-Martinez Paniagua
- Department of Chemical and Biomolecular Engineering, University of Houston, Houston, TX 77054, USA
| | - David F Ackart
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO 80523, USA
| | - Nicole Jones
- Department of Biological Sciences, University of Texas, El Paso, TX 79968, USA
| | - Charles Spencer
- Department of Biological Sciences, University of Texas, El Paso, TX 79968, USA
| | - Brendan K Podell
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO 80523, USA
| | - Hugues Ouellet
- Department of Biological Sciences, University of Texas, El Paso, TX 79968, USA
| | - Navin Varadarajan
- Department of Chemical and Biomolecular Engineering, University of Houston, Houston, TX 77054, USA.
| |
Collapse
|
8
|
Li Y, Yi J, Ma R, Wang Y, Lou X, Dong Y, Cao Y, Li X, Wang M, Dang X, Li R, Lei N, Song H, Qin Z, Yang W. A polymeric nanoplatform enhances the cGAS-STING pathway in macrophages to potentiate phagocytosis for cancer immunotherapy. J Control Release 2024; 373:447-462. [PMID: 39038546 DOI: 10.1016/j.jconrel.2024.07.039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 07/13/2024] [Accepted: 07/16/2024] [Indexed: 07/24/2024]
Abstract
Immunosuppressive tumor-associated macrophages (TAMs) account for a high proportion of the tumor tissue and significantly impede immunoefficacy. Furthermore, the signal regulatory protein α (SIRPα) expressed in TAMs adversely correlates with macrophage activation and phagocytosis, resulting in immunosurveillance escape. To address these difficulties, a mannose-modified, pH-responsive nanoplatform with resiquimod (R848) and 2', 3'-cyclic GMP-AMP (cGAMP) co-encapsulation (named M-PNP@R@C) is designed to polarize TAMs and lower SIRPα expression. The co-delivery of R848 and cGAMP synergistically facilitates the polarization of TAMs from the anti-inflammatory M2 phenotype into the pro-inflammatory M1 phenotype, thereby enhancing antitumor immunotherapy. Remarkably, activation of the cGAMP-mediated stimulator of interferon genes (STING) in TAMs significantly downregulates the expression of SIRPα, which synergizes with the cluster of differentiation 47 (CD47) antibody for the dual blockade of the CD47-SIRPα axis. Further analysis of single-cell RNA sequencing indicates that STING activation downregulates SIRPα by regulating intracellular fatty acid oxidation metabolism. In vivo studies indicate that M-PNP@R@C significantly inhibits tumor growth with a potent antitumor immune response in melanoma graft tumor models. After synergy with anti-CD47, the double blockade strategies of the SIRPα/CD47 axis result in a notable inhibition of lung metastasis. A prolonged survival rate is observed after combination treatment with CD47 and programmed death ligand-1 antibodies for the triple immune checkpoint blockade. In summary, our study provides original insights into the potential role of the STING pathway in macrophage-based immunotherapy, thus offering a potential combinatorial strategy for cancer therapy.
Collapse
Affiliation(s)
- Yongjuan Li
- Medical Research Center, The First Affiliated Hospital of Zhengzhou University, The center of Infection and Immunity, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China; School of basic medical sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Jinmeng Yi
- Medical Research Center, The First Affiliated Hospital of Zhengzhou University, The center of Infection and Immunity, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China; School of basic medical sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Rong Ma
- School of Pharmaceutical Sciences, Henan Key Laboratory of Nanomedicine for Targeting Diagnosis and Treatment, Zhengzhou University, Zhengzhou 450001, China
| | - Yayun Wang
- School of Pharmaceutical Sciences, Henan Key Laboratory of Nanomedicine for Targeting Diagnosis and Treatment, Zhengzhou University, Zhengzhou 450001, China
| | - Xiaohan Lou
- Medical Research Center, The First Affiliated Hospital of Zhengzhou University, The center of Infection and Immunity, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Ya Dong
- Medical Research Center, The First Affiliated Hospital of Zhengzhou University, The center of Infection and Immunity, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Yongjian Cao
- School of Pharmaceutical Sciences, Henan Key Laboratory of Nanomedicine for Targeting Diagnosis and Treatment, Zhengzhou University, Zhengzhou 450001, China
| | - Xinyan Li
- Medical Research Center, The First Affiliated Hospital of Zhengzhou University, The center of Infection and Immunity, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Ming Wang
- Medical Research Center, The First Affiliated Hospital of Zhengzhou University, The center of Infection and Immunity, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Xiaowei Dang
- Medical Research Center, The First Affiliated Hospital of Zhengzhou University, The center of Infection and Immunity, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Rui Li
- Medical Research Center, The First Affiliated Hospital of Zhengzhou University, The center of Infection and Immunity, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Ningjing Lei
- School of basic medical sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Haiwei Song
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), Singapore, 138673, Singapore.
| | - Zhihai Qin
- Medical Research Center, The First Affiliated Hospital of Zhengzhou University, The center of Infection and Immunity, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China.
| | - Weijing Yang
- Medical Research Center, The First Affiliated Hospital of Zhengzhou University, The center of Infection and Immunity, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China; School of Pharmaceutical Sciences, Henan Key Laboratory of Nanomedicine for Targeting Diagnosis and Treatment, Zhengzhou University, Zhengzhou 450001, China.
| |
Collapse
|
9
|
Kametani Y, Ito R, Manabe Y, Kulski JK, Seki T, Ishimoto H, Shiina T. PBMC-engrafted humanized mice models for evaluating immune-related and anticancer drug delivery systems. Front Mol Biosci 2024; 11:1447315. [PMID: 39228913 PMCID: PMC11368775 DOI: 10.3389/fmolb.2024.1447315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 08/07/2024] [Indexed: 09/05/2024] Open
Abstract
Immune-related drug delivery systems (DDSs) in humanized mouse models are at the forefront of cancer research and serve as bridges between preclinical studies and clinical applications. These systems offer unique platforms for exploring new therapies and understanding their interactions with human cells and the immune system. Here, we focus on a DDS and a peripheral blood mononuclear cell (PBMC)-engrafted humanized mouse model that we recently developed, and consider some of the key components, challenges, and applications to advance these systems towards better cancer treatment on the basis of a better understanding of the immune response. Our DDS is unique and has a dual function, an anticancer effect and a capacity to fine-tune the immune reaction. The PBL-NOG-hIL-4-Tg mouse system is superior to other available humanized mouse systems for the development of such multifunctional DDSs because it supports the rapid reconstruction of an individual donor's immunity and avoids the onset of graft-versus-host disease.
Collapse
Affiliation(s)
- Yoshie Kametani
- Department of Molecular Life Science, Division of Basic Medical Science, Tokai University School of Medicine, Isehara, Japan
- Institute of Advanced Biosciences, Tokai University, Hiratsuka, Japan
| | - Ryoji Ito
- Central Institute for Experimental Medicine and Life Science (CIEM), Kawasaki, Japan
| | - Yoshiyuki Manabe
- Department of Chemistry, Graduate School of Science, Osaka University, Osaka, Japan
| | - Jerzy K. Kulski
- Department of Molecular Life Science, Division of Basic Medical Science, Tokai University School of Medicine, Isehara, Japan
- Faculty of Health and Medical Sciences, School of Biomedical Science, The University of Western Australia, Crawley, WA, Australia
| | - Toshiro Seki
- Department of Internal Medicine, Division of Nephrology, Endocrinology, and Metabolism, Tokai University School of Medicine, Isehara, Japan
| | - Hitoshi Ishimoto
- Department of Obstetrics and Gynecology, Tokai University School of Medicine, Isehara, Japan
| | - Takashi Shiina
- Department of Molecular Life Science, Division of Basic Medical Science, Tokai University School of Medicine, Isehara, Japan
- Institute of Advanced Biosciences, Tokai University, Hiratsuka, Japan
| |
Collapse
|
10
|
Endo R, Ueda T, Nagaoki T, Shima N, Sato Y, Harashima H, Nakamura T. Impact of in vivo fate of STING agonist-loaded lipid nanoparticles on antitumor immunity. J Control Release 2024; 372:609-618. [PMID: 38942082 DOI: 10.1016/j.jconrel.2024.06.064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 06/24/2024] [Accepted: 06/26/2024] [Indexed: 06/30/2024]
Abstract
Therapeutically manipulating the stimulator of interferon genes (STING) pathway has promising potential for enhancing antitumor immunity. Agonists of this pathway (STING agonists) are being evaluated in clinical trials. Loading the STING agonists into lipid nanoparticles (LNPs) increases their safety and efficacy. We previously developed STING agonists loaded LNPs consisting of the ionizable lipid YSK12-C4 (YSK12-LNPs), which showed significant antitumor effects. However, it is largely unclear how the in vivo fate of STING agonists loaded LNPs affects the antitumor immune responses. In this study, we compared the YSK12-LNPs with LNPs composed of DLin-MC3-DMA (MC3-LNPs) showing different in vivo fates. Biodistribution and flow cytometry analyses of mouse tissues revealed that the MC3-LNPs delivered higher amounts of STING agonists to the liver than the YSK12-LNPs. Additionally, significantly more liver leukocytes internalized the MC3-LNPs than the YSK12-LNPs. In contrast, the YSK12-LNPs delivered higher amounts of STING agonists to the liver leukocytes than the MC3-LNPs, leading to the effective induction of innate immunity and inflammation in the tumors. However, the antitumor effects in the B16-F10 lung metastasis and CT26 tumor models were comparable. Interestingly, flow cytometry analyses suggested that the YSK12-LNPs were more likely to activate natural killer cells and M1 macrophages, while the MC3-LNPs were more likely to activate CD8+ T cells. Our data suggest that different antitumor immune response mechanisms may operate depending on the characteristics and distribution of the LNPs.
Collapse
Affiliation(s)
- Rikito Endo
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812, Japan
| | - Tomoki Ueda
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812, Japan
| | - Takumi Nagaoki
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812, Japan
| | - Natsumi Shima
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812, Japan
| | - Yusuke Sato
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812, Japan
| | - Hideyoshi Harashima
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812, Japan.
| | - Takashi Nakamura
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812, Japan.
| |
Collapse
|
11
|
Zahid AA, Chakraborty A, Shamiya Y, Wilson RB, Borradaile N, Paul A. Cell Membrane-Derived Nanoparticles as Biomimetic Nanotherapeutics to Alleviate Fatty Liver Disease. ACS APPLIED MATERIALS & INTERFACES 2024; 16:39117-39128. [PMID: 39022877 DOI: 10.1021/acsami.4c08240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
The prevalence of metabolic dysfunction associated-steatotic liver disease (MASLD) (formerly known as nonalcoholic fatty liver disease; NAFLD) is estimated at around 32% of the world's population, resulting in a major healthcare concern in recent times. Current pharmaceutical methods lack efficacy for the treatment of the disease because of suboptimal pharmacokinetic parameters including poor bioavailability, short half-life, and premature clearance. Designing an efficient drug delivery system that provides a protective environment is critical for addressing these challenges. Such a system should aim to enhance the cellular uptake of drugs, improve their bioavailability, and reduce the chances of rapid clearance. Here, we developed nanoengineered natural cell membrane-derived nanoparticles (CMNs) incorporated with a model drug, rosuvastatin, in the bilayer assembly of CMNs to reduce the accumulation of lipids in hepatocytes, a hallmark of MASLD. We used a cell extrusion technique to develop self-assembled CMNs with precise size control compared to the cell shearing method. Interestingly, the prepared CMNs were found to be nonphagocytic, representing around 1.13% of phosphatidylserine receptors on healthy cells, which allows the possibility of their use as stealth nanoparticles for drug delivery. Furthermore, CMNs exhibit higher drug-loading efficiency, excellent cytocompatibility, and enhanced cellular internalization capabilities. Moreover, we show that the delivery of rosuvastatin-loaded CMNs in the in vitro MASLD model efficiently reduced hepatocyte lipid accumulation, including total cholesterol (26.8 ± 3.1%) and triglycerides (11.8 ± 0.8%), compared to the negative control. Taken together, the nanoengineered biomimetic CMNs enhance the drug's bioactivity in hepatic cells, establishing a foundation for further investigation of this drug delivery system in treating MASLD.
Collapse
Affiliation(s)
- Alap Ali Zahid
- Department of Chemical and Biochemical Engineering, The University of Western Ontario, London, Ontario N6A 5B9, Canada
| | - Aishik Chakraborty
- Department of Chemical and Biochemical Engineering, The University of Western Ontario, London, Ontario N6A 5B9, Canada
- Collaborative Specialization in Musculoskeletal Health Research and Bone and Joint Institute, The University of Western Ontario, London, Ontario N6A 5B9, Canada
| | - Yasmeen Shamiya
- Department of Chemistry, The University of Western Ontario, London, Ontario N6A 5B9, Canada
| | - Rachel B Wilson
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, Ontario N6A 3K7, Canada
| | - Nica Borradaile
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, Ontario N6A 3K7, Canada
| | - Arghya Paul
- Department of Chemical and Biochemical Engineering, The University of Western Ontario, London, Ontario N6A 5B9, Canada
- School of Biomedical Engineering, The University of Western Ontario, London, Ontario N6A 5B9, Canada
- Collaborative Specialization in Musculoskeletal Health Research and Bone and Joint Institute, The University of Western Ontario, London, Ontario N6A 5B9, Canada
- Department of Chemistry, The University of Western Ontario, London, Ontario N6A 5B9, Canada
| |
Collapse
|
12
|
Duan X, Zhao Y, Hu H, Wang X, Yan J, Li S, Zhang Y, Jiao J, Zhang G. Amino Acid Metabolism-Regulated Nanomedicine for Enhanced Tumor Immunotherapy through Synergistic Regulation of Immune Microenvironment. Biomater Res 2024; 28:0048. [PMID: 38966855 PMCID: PMC11223770 DOI: 10.34133/bmr.0048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 05/13/2024] [Indexed: 07/06/2024] Open
Abstract
The reprogramming of tumor metabolism presents a substantial challenge for effective immunotherapy, playing a crucial role in developing an immunosuppressive microenvironment. In particular, the degradation of the amino acid L-tryptophan (Trp) to kynurenine (Kyn) by indoleamine-pyrrole 2,3-dioxygenase 1 (IDO1) is one of the most clinically validated pathways for immune suppression. Thus, regulating the Trp/Kyn metabolism by IDO1 inhibition represents a promising strategy for enhancing immunotherapy. Herein, metabolism-regulated nanoparticles are prepared through metal coordination-driven assembly of an IDO1 inhibitor (NLG919) and a stimulator of interferon genes (STING) agonist (MSA-2) for enhanced immunotherapy. After intravenous administration, the assembled nanoparticles could efficiently accumulate in tumors, enhancing the bioavailability of NLG919 and down-regulating the metabolism of Trp to Kyn to remodel the immunosuppressive tumor microenvironment. Meanwhile, the released MSA-2 evoked potent STING pathway activation in tumors, triggering an effective immune response. The antitumor immunity induced by nanoparticles significantly inhibited the development of primary and metastatic tumors, as well as B16 melanoma. Overall, this study provided a novel paradigm for enhancing tumor immunotherapy through synergistic amino acid metabolism and STING pathway activation.
Collapse
Affiliation(s)
- Xiuying Duan
- Medical Science and Technology Innovation Center,
Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
- School of Life Sciences,
Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Yilei Zhao
- Medical Science and Technology Innovation Center,
Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Houyang Hu
- Medical Science and Technology Innovation Center,
Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Xuechun Wang
- Medical Science and Technology Innovation Center,
Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Jie Yan
- Medical Science and Technology Innovation Center,
Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Songyan Li
- Medical Science and Technology Innovation Center,
Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Yueying Zhang
- School of Clinical and Basic Medical Sciences,
Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Jianwei Jiao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology,
Chinese Academy of Sciences, Beijing 100101, China
| | - Guiqiang Zhang
- Medical Science and Technology Innovation Center,
Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| |
Collapse
|
13
|
Tian X, Ai J, Tian X, Wei X. cGAS-STING pathway agonists are promising vaccine adjuvants. Med Res Rev 2024; 44:1768-1799. [PMID: 38323921 DOI: 10.1002/med.22016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 12/10/2023] [Accepted: 01/09/2024] [Indexed: 02/08/2024]
Abstract
Adjuvants are of critical value in vaccine development as they act on enhancing immunogenicity of antigen and inducing long-lasting immunity. However, there are only a few adjuvants that have been approved for clinical use, which highlights the need for exploring and developing new adjuvants to meet the growing demand for vaccination. Recently, emerging evidence demonstrates that the cGAS-STING pathway orchestrates innate and adaptive immunity by generating type I interferon responses. Many cGAS-STING pathway agonists have been developed and tested in preclinical research for the treatment of cancer or infectious diseases with promising results. As adjuvants, cGAS-STING agonists have demonstrated their potential to activate robust defense immunity in various diseases, including COVID-19 infection. This review summarized the current developments in the field of cGAS-STING agonists with a special focus on the latest applications of cGAS-STING agonists as adjuvants in vaccination. Potential challenges were also discussed in the hope of sparking future research interests to further the development of cGAS-STING as vaccine adjuvants.
Collapse
Affiliation(s)
- Xinyu Tian
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Centre for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, P.R. China
| | - Jiayuan Ai
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Centre for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, P.R. China
| | - Xiaohe Tian
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Centre for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, P.R. China
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Centre for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, P.R. China
| |
Collapse
|
14
|
Yu H, Chen Q, Zheng M, Wang R, Wang H, Cheng L, Hu Y, Dai M, Du C, Luo W, Tan M, Cao Y, Guo Y, Ran H. Combination of MHI148 Targeted Photodynamic Therapy and STING Activation Inhibits Tumor Metastasis and Recurrence. ACS APPLIED MATERIALS & INTERFACES 2024; 16:29672-29685. [PMID: 38813586 DOI: 10.1021/acsami.4c02528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/31/2024]
Abstract
Metastasis and recurrence are notable contributors to mortality associated with breast cancer. Although immunotherapy has shown promise in mitigating these risks after conventional treatments, its effectiveness remains constrained by significant challenges, such as impaired antigen presentation by dendritic cells (DCs) and inadequate T cell infiltration into tumor tissues. To address these limitations, we developed a multifunctional nanoparticle platform, termed GM@P, which consisted of a hydrophobic shell encapsulating the photosensitizer MHI148 and a hydrophilic core containing the STING agonist 2'3'-cGAMP. This design elicited robust type I interferon responses to activate antitumor immunity. The GM@P nanoparticles loaded with MHI148 specifically targeted breast cancer cells. Upon exposure to 808 nm laser irradiation, the MHI148-loaded nanoparticles produced toxic reactive oxygen species (ROS) to eradicate tumor cells through photodynamic therapy (PDT). Notably, PDT stimulated immunogenic cell death (ICD) to foster the potency of antitumor immune responses. Furthermore, the superior photoacoustic imaging (PAI) capabilities of MHI148 enabled the simultaneous visualization of diagnostic and therapeutic procedures. Collectively, our findings uncovered that the combination of PDT and STING activation facilitated a more conducive immune microenvironment, characterized by enhanced DC maturation, infiltration of CD8+ T cells, and proinflammatory cytokine release. This strategy stimulated local immune responses to augment systemic antitumor effects, offering a promising approach to suppress tumor growth, inhibit metastasis, and prevent recurrence.
Collapse
Affiliation(s)
- Huilin Yu
- Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, People's Republic of China
- Chongqing Key Laboratory of Ultrasound Molecular Imaging & State Key Laboratory of Ultrasound in Medicine and Engineering, Chongqing Medical University, Chongqing 400010, People's Republic of China
| | - Qiaoqi Chen
- Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, People's Republic of China
- Chongqing Key Laboratory of Ultrasound Molecular Imaging & State Key Laboratory of Ultrasound in Medicine and Engineering, Chongqing Medical University, Chongqing 400010, People's Republic of China
| | - Min Zheng
- Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, People's Republic of China
- Chongqing Key Laboratory of Ultrasound Molecular Imaging & State Key Laboratory of Ultrasound in Medicine and Engineering, Chongqing Medical University, Chongqing 400010, People's Republic of China
| | - Ruoyao Wang
- Chongqing Key Laboratory of Ultrasound Molecular Imaging & State Key Laboratory of Ultrasound in Medicine and Engineering, Chongqing Medical University, Chongqing 400010, People's Republic of China
- Department of Breast and Thyroid Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, People's Republic of China
| | - Haiyang Wang
- Chongqing Key Laboratory of Ultrasound Molecular Imaging & State Key Laboratory of Ultrasound in Medicine and Engineering, Chongqing Medical University, Chongqing 400010, People's Republic of China
- Department of Abdominal Wall, Hernia and Vascular Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, People's Republic of China
| | - Long Cheng
- Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, People's Republic of China
- Chongqing Key Laboratory of Ultrasound Molecular Imaging & State Key Laboratory of Ultrasound in Medicine and Engineering, Chongqing Medical University, Chongqing 400010, People's Republic of China
| | - Yaqin Hu
- Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, People's Republic of China
- Chongqing Key Laboratory of Ultrasound Molecular Imaging & State Key Laboratory of Ultrasound in Medicine and Engineering, Chongqing Medical University, Chongqing 400010, People's Republic of China
| | - Mingyuan Dai
- Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, People's Republic of China
- Chongqing Key Laboratory of Ultrasound Molecular Imaging & State Key Laboratory of Ultrasound in Medicine and Engineering, Chongqing Medical University, Chongqing 400010, People's Republic of China
| | - Chier Du
- Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, People's Republic of China
- Chongqing Key Laboratory of Ultrasound Molecular Imaging & State Key Laboratory of Ultrasound in Medicine and Engineering, Chongqing Medical University, Chongqing 400010, People's Republic of China
| | - Wenpei Luo
- Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, People's Republic of China
- Chongqing Key Laboratory of Ultrasound Molecular Imaging & State Key Laboratory of Ultrasound in Medicine and Engineering, Chongqing Medical University, Chongqing 400010, People's Republic of China
| | - Mixiao Tan
- Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, People's Republic of China
- Chongqing Key Laboratory of Ultrasound Molecular Imaging & State Key Laboratory of Ultrasound in Medicine and Engineering, Chongqing Medical University, Chongqing 400010, People's Republic of China
| | - Yang Cao
- Chongqing Key Laboratory of Ultrasound Molecular Imaging & State Key Laboratory of Ultrasound in Medicine and Engineering, Chongqing Medical University, Chongqing 400010, People's Republic of China
| | - Yuan Guo
- Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, People's Republic of China
- Chongqing Key Laboratory of Ultrasound Molecular Imaging & State Key Laboratory of Ultrasound in Medicine and Engineering, Chongqing Medical University, Chongqing 400010, People's Republic of China
| | - Haitao Ran
- Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, People's Republic of China
- Chongqing Key Laboratory of Ultrasound Molecular Imaging & State Key Laboratory of Ultrasound in Medicine and Engineering, Chongqing Medical University, Chongqing 400010, People's Republic of China
| |
Collapse
|
15
|
Wang J, Meng F, Yeo Y. Delivery of STING agonists for cancer immunotherapy. Curr Opin Biotechnol 2024; 87:103105. [PMID: 38461748 PMCID: PMC11162310 DOI: 10.1016/j.copbio.2024.103105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 02/15/2024] [Accepted: 02/18/2024] [Indexed: 03/12/2024]
Abstract
Agonists of the cyclic guanosine monophosphate-adenosine monophosphate synthase (cGAS)-Stimulator of Interferon Genes (STING) pathway, a critical mediator of innate immune response to foreign invaders with DNA, have gained significant interest in cancer immunotherapy. STING agonists are envisioned as a way of complementing the antitumor activity of the patient's immune system and immune checkpoint blockade therapy. However, their clinical development has been challenging due to the poor pharmacokinetic and physicochemical properties. This review discusses drug delivery efforts to circumvent the challenges, their accomplishment, and unmet needs based on the last five years of literature.
Collapse
Affiliation(s)
- Jianping Wang
- Department of Industrial and Molecular Pharmaceutics, Purdue University, 575 Stadium Mall Drive, West Lafayette, IN 47907, USA
| | - Fanfei Meng
- Department of Industrial and Molecular Pharmaceutics, Purdue University, 575 Stadium Mall Drive, West Lafayette, IN 47907, USA; Department of Biomedical and Nutritional Sciences, University of Massachusetts Lowell, 3 Solomont Way, Lowell, MA 01854, USA
| | - Yoon Yeo
- Department of Industrial and Molecular Pharmaceutics, Purdue University, 575 Stadium Mall Drive, West Lafayette, IN 47907, USA; Purdue University Institute for Cancer Research, 201 South University Street, West Lafayette, IN 47907, USA; Weldon School of Biomedical Engineering, Purdue University, 206 S Martin Jischke Drive, West Lafayette, IN 47907, USA.
| |
Collapse
|
16
|
Chellen T, Bausart M, Maus P, Vanvarenberg K, Limaye N, Préat V, Malfanti A. In situ administration of STING-activating hyaluronic acid conjugate primes anti-glioblastoma immune response. Mater Today Bio 2024; 26:101057. [PMID: 38660475 PMCID: PMC11040137 DOI: 10.1016/j.mtbio.2024.101057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 04/04/2024] [Accepted: 04/10/2024] [Indexed: 04/26/2024] Open
Abstract
Glioblastoma (GBM) is an aggressive brain tumor, with a highly immunosuppressive tumor immune microenvironment (TIME). In this work, we investigated the use of the STimulator of INterferon Genes (STING) pathway as an effective means to remodel the GBM TIME through the recruitment of both innate and adaptive immune cell populations. Using hyaluronic acid (HA), we developed a novel polymer-drug conjugate of a non-nucleotide STING agonist (MSA2), called HA-MSA2 for the in situ treatment of GBM. In JAWSII cells, HA-MSA2 exerted a greater increase of STING signaling and upregulation of STING-related downstream cyto-/chemokines in immune cells than the free drug. HA-MSA2 also elicited cancer cell-intrinsic immunostimulatory gene expression and promoted immunogenic cell death of GBM cells. In the SB28 GBM model, local delivery of HA-MSA2 induced a delay in tumor growth and a significant extension of survival. The analysis of the TIME showed a profound shift in the GBM immune landscape after HA-MSA2 treatment, with higher infiltration by innate and adaptive immune cells including dendritic, natural killer (NK) and CD8 T cell populations. The therapeutic potential of this novel polymer conjugate warrants further investigation, particularly with other chemo-immunotherapeutics or cancer vaccines as a promising combinatorial therapeutic approach.
Collapse
Affiliation(s)
- Teenesha Chellen
- UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue Mounier 73 B1.73.12, 1200, Brussels, Belgium
| | - Mathilde Bausart
- UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue Mounier 73 B1.73.12, 1200, Brussels, Belgium
| | - Pierre Maus
- UCLouvain, de Duve Institute, Genetics of Autoimmune Diseases and Cancer, Brussels, Belgium
| | - Kevin Vanvarenberg
- UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue Mounier 73 B1.73.12, 1200, Brussels, Belgium
| | - Nisha Limaye
- UCLouvain, de Duve Institute, Genetics of Autoimmune Diseases and Cancer, Brussels, Belgium
| | - Véronique Préat
- UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue Mounier 73 B1.73.12, 1200, Brussels, Belgium
| | - Alessio Malfanti
- UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue Mounier 73 B1.73.12, 1200, Brussels, Belgium
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via F. Marzolo 5, 35131 Padova, Italy
| |
Collapse
|
17
|
Wilson J, Kimmel B, Arora K, Chada N, Bharti V, Kwiatkowski A, Finklestein J, Hanna A, Arner E, Sheehy T, Pastora L, Yang J, Pagendarm H, Stone P, Taylor B, Hubert L, Gibson-Corley K, May J, McLean J, Rathmell J, Richmond A, Rathmell W, Balko J, Fingleton B, Hargrove-Wiley E. Programable Albumin-Hitchhiking Nanobodies Enhance the Delivery of STING Agonists to Potentiate Cancer Immunotherapy. RESEARCH SQUARE 2024:rs.3.rs-3243545. [PMID: 38766114 PMCID: PMC11100900 DOI: 10.21203/rs.3.rs-3243545/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Stimulator of interferon genes (STING) is a promising target for potentiating antitumor immunity, but multiple pharmacological barriers limit the clinical utility, efficacy, and/or safety of STING agonists. Here we describe a modular platform for systemic administration of STING agonists based on nanobodies engineered for in situ hitchhiking of agonist cargo on serum albumin. Using site-selective bioconjugation chemistries to produce molecularly defined products, we found that covalent conjugation of a STING agonist to anti-albumin nanobodies improved pharmacokinetics and increased cargo accumulation in tumor tissue, stimulating innate immune programs that increased the infiltration of activated natural killer cells and T cells, which potently inhibited tumor growth in multiple mouse tumor models. We also demonstrated the programmability of the platform through the recombinant integration of a second nanobody domain that targeted programmed cell death ligand-1 (PD-L1), which further increased cargo delivery to tumor sites while also blocking immunosuppressive PD-1/PD-L1 interactions. This bivalent nanobody carrier for covalently conjugated STING agonists stimulated robust antigen-specific T cell responses and long-lasting immunological memory, conferred enhanced therapeutic efficacy, and was effective as a neoadjuvant treatment for improving responses to adoptive T cell transfer therapy. Albumin-hitchhiking nanobodies thus offer an enabling, multimodal, and programmable platform for systemic delivery of STING agonists with potential to augment responses to multiple immunotherapeutic modalities.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Ann Hanna
- Vanderbilt University Medical Center
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Wang-Bishop L, Wehbe M, Pastora LE, Yang J, Kimmel BR, Garland KM, Becker KW, Carson CS, Roth EW, Gibson-Corley KN, Ulkoski D, Krishnamurthy V, Fedorova O, Richmond A, Pyle AM, Wilson JT. Nanoparticle Retinoic Acid-Inducible Gene I Agonist for Cancer Immunotherapy. ACS NANO 2024; 18:11631-11643. [PMID: 38652829 PMCID: PMC11080455 DOI: 10.1021/acsnano.3c06225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 04/03/2024] [Accepted: 04/10/2024] [Indexed: 04/25/2024]
Abstract
Pharmacological activation of the retinoic acid-inducible gene I (RIG-I) pathway holds promise for increasing tumor immunogenicity and improving the response to immune checkpoint inhibitors (ICIs). However, the potency and clinical efficacy of 5'-triphosphate RNA (3pRNA) agonists of RIG-I are hindered by multiple pharmacological barriers, including poor pharmacokinetics, nuclease degradation, and inefficient delivery to the cytosol where RIG-I is localized. Here, we address these challenges through the design and evaluation of ionizable lipid nanoparticles (LNPs) for the delivery of 3p-modified stem-loop RNAs (SLRs). Packaging of SLRs into LNPs (SLR-LNPs) yielded surface charge-neutral nanoparticles with a size of ∼100 nm that activated RIG-I signaling in vitro and in vivo. SLR-LNPs were safely administered to mice via both intratumoral and intravenous routes, resulting in RIG-I activation in the tumor microenvironment (TME) and the inhibition of tumor growth in mouse models of poorly immunogenic melanoma and breast cancer. Significantly, we found that systemic administration of SLR-LNPs reprogrammed the breast TME to enhance the infiltration of CD8+ and CD4+ T cells with antitumor function, resulting in enhanced response to αPD-1 ICI in an orthotopic EO771 model of triple-negative breast cancer. Therapeutic efficacy was further demonstrated in a metastatic B16.F10 melanoma model, with systemically administered SLR-LNPs significantly reducing lung metastatic burden compared to combined αPD-1 + αCTLA-4 ICI. Collectively, these studies have established SLR-LNPs as a translationally promising immunotherapeutic nanomedicine for potent and selective activation of RIG-I with the potential to enhance response to ICIs and other immunotherapeutic modalities.
Collapse
Affiliation(s)
- Lihong Wang-Bishop
- Department
of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, Tennessee 37212, United States
| | - Mohamed Wehbe
- Department
of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, Tennessee 37212, United States
| | - Lucinda E. Pastora
- Department
of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, Tennessee 37212, United States
| | - Jinming Yang
- Department
of Pharmacology, Vanderbilt University Medical
Center, Nashville, Tennessee 37232, United States
- Department
of Veterans Affairs, Tennessee Valley Healthcare
System, Nashville, Tennessee 37212, United States
| | - Blaise R. Kimmel
- Department
of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, Tennessee 37212, United States
| | - Kyle M. Garland
- Department
of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, Tennessee 37212, United States
| | - Kyle W. Becker
- Department
of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, Tennessee 37212, United States
| | - Carcia S. Carson
- Department
of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37212, United States
| | - Eric W. Roth
- Northwestern
University Atomic and Nanoscale Characterization Experimental (NUANCE)
Center, Northwestern University, Evanston, Illinois 60208, United States
| | - Katherine N. Gibson-Corley
- Department
of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
- Department
of Medicine, Vanderbilt University Medical
Center, Nashville, Tennessee 37232, United States
| | - David Ulkoski
- Advanced
Drug Delivery, Pharmaceutical Sciences, R&D, AstraZeneca, Boston, Massachusetts 02451, United States
| | - Venkata Krishnamurthy
- Advanced
Drug Delivery, Pharmaceutical Sciences, R&D, AstraZeneca, Boston, Massachusetts 02451, United States
| | - Olga Fedorova
- Department
of Molecular, Cellular and Developmental Biology, Yale University, New Haven, Connecticut 06520, United States
- Howard
Hughes Medical Institute, Chevy Chase, Maryland 20815, United States
| | - Ann Richmond
- Department
of Pharmacology, Vanderbilt University Medical
Center, Nashville, Tennessee 37232, United States
- Department
of Veterans Affairs, Tennessee Valley Healthcare
System, Nashville, Tennessee 37212, United States
| | - Anna Marie Pyle
- Department
of Molecular, Cellular and Developmental Biology, Yale University, New Haven, Connecticut 06520, United States
- Howard
Hughes Medical Institute, Chevy Chase, Maryland 20815, United States
- Department
of Chemistry, Yale University, New Haven, Connecticut 06520, United States
| | - John T. Wilson
- Department
of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, Tennessee 37212, United States
- Department
of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37212, United States
- Department
of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
- Vanderbilt
Institute of Chemical Biology, Vanderbilt
University, Nashville, Tennessee 37212, United States
- Vanderbilt
Institute of Nanoscale Science and Engineering, Vanderbilt University, Nashville, Tennessee 37212, United States
- Vanderbilt
Institute for Infection, Immunology, and Inflammation, Vanderbilt University, Nashville, Tennessee 37212, United States
- Vanderbilt
Center for Immunobiology, Vanderbilt University
Medical Center, Nashville, Tennessee 37232, United States
- Vanderbilt
Ingram Cancer Center, Nashville, Tennessee 37232, United States
| |
Collapse
|
19
|
Li J, Yu H, Kang Y, Niu K, Wang M, Jiang Y, Jiang N, Ding Z, Gan Z, Yu Q. STING Membrane Prevents Post-Surgery Tissue Adhesion and Tumor Recurrence of Colorectal Cancer. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2309655. [PMID: 38517062 DOI: 10.1002/adma.202309655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 02/21/2024] [Indexed: 03/23/2024]
Abstract
Surgery is the standard treatment regimen for resectable colorectal cancer (CRC). However, it is very hard to completely remove all cancer cells in clinical practice, leading to the high recurrence rates of the disease. Moreover, the post-surgery tissue adhesion greatly prevents the possibility of reoperation, significantly limiting the long-term surviving of CRC patients. To overcome CRC recurrence and avoid the post-surgery tissue adhesion, this work develops a novel stimulator of interferon genes "STING" membrane based on the coaxial electrospinning technology and hyaluronic acid modification. A reactive oxygen species responsive prodrug of gambogic acid (GB) and a potent STING agonist (CDN) are coloaded in the core-shell structure of the membrane, which endows the loaded drug with sustained and sequential release patterns. The localized delivery of GB and CDN can selectively induce efficient immunogenic cell death of cancer cells and then evoke the systemic anticancer immunity by activating the Cyclic GMP-AMP (cGAMP) synthase/STING pathway. As-designed "STING" membrane not only safely prevents tumor recurrence through the synergistic chemoimmunotherapy but also efficiently avoids the post-surgery tissue adhesion, facilitating the clinical intervention of CRC.
Collapse
Affiliation(s)
- Jianlin Li
- The State Key Laboratory of Organic-inorganic Composites, Beijing Laboratory of Biomedical Materials, Key Laboratory of Biomedical Materials of Natural Macromolecules (Ministry of Education), College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Haiwang Yu
- The State Key Laboratory of Organic-inorganic Composites, Beijing Laboratory of Biomedical Materials, Key Laboratory of Biomedical Materials of Natural Macromolecules (Ministry of Education), College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Yan Kang
- The State Key Laboratory of Organic-inorganic Composites, Beijing Laboratory of Biomedical Materials, Key Laboratory of Biomedical Materials of Natural Macromolecules (Ministry of Education), College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Kun Niu
- The State Key Laboratory of Organic-inorganic Composites, Beijing Laboratory of Biomedical Materials, Key Laboratory of Biomedical Materials of Natural Macromolecules (Ministry of Education), College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Ming Wang
- The State Key Laboratory of Organic-inorganic Composites, Beijing Laboratory of Biomedical Materials, Key Laboratory of Biomedical Materials of Natural Macromolecules (Ministry of Education), College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Yitong Jiang
- The State Key Laboratory of Organic-inorganic Composites, Beijing Laboratory of Biomedical Materials, Key Laboratory of Biomedical Materials of Natural Macromolecules (Ministry of Education), College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Ni Jiang
- The State Key Laboratory of Organic-inorganic Composites, Beijing Laboratory of Biomedical Materials, Key Laboratory of Biomedical Materials of Natural Macromolecules (Ministry of Education), College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Zhenshan Ding
- Department of Urology, China-Japan Friendship Hospitals, Beijing, 100029, China
| | - Zhihua Gan
- The State Key Laboratory of Organic-inorganic Composites, Beijing Laboratory of Biomedical Materials, Key Laboratory of Biomedical Materials of Natural Macromolecules (Ministry of Education), College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Qingsong Yu
- The State Key Laboratory of Organic-inorganic Composites, Beijing Laboratory of Biomedical Materials, Key Laboratory of Biomedical Materials of Natural Macromolecules (Ministry of Education), College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| |
Collapse
|
20
|
Sheehy TL, Kwiatkowski AJ, Arora K, Kimmel BR, Schulman JA, Gibson-Corley K, Wilson JT. STING-Activating Polymer-Drug Conjugates for Cancer Immunotherapy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.23.585817. [PMID: 38585879 PMCID: PMC10996458 DOI: 10.1101/2024.03.23.585817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
The stimulator of interferon genes (STING) pathway links innate and adaptive antitumor immunity and therefore plays an important role in cancer immune surveillance. This has prompted widespread development of STING agonists for cancer immunotherapy, but pharmacological barriers continue to limit the clinical impact of STING agonists and motivate the development of drug delivery systems to improve their efficacy and/or safety. To address this challenge, we developed SAPCon, a STING-activating polymer-drug conjugate platform based on strain-promoted azide-alkyne cycloaddition of dimeric-amidobenzimidazole (diABZI) STING agonists to hydrophilic polymer chains through an enzyme-responsive chemical linker. To synthesize a first-generation SAPCon, we designed a diABZI prodrug modified with a DBCO reactive handle a cathepsin B-cleavable spacer for intracellular drug release and conjugated this to pendant azide groups on a 100 kDa poly(dimethyla acrylamide-co-azide methacrylate) copolymer backbone to increase circulation time and enable passive tumor accumulation. We found that intravenously administered SAPCon accumulated at tumor sites where they it was endocytosed by tumor-associated myeloid cells, resulting in increased STING activation in tumor tissue compared to a free diABZI STING agonist. Consequently, SAPCon promoted an immunogenic tumor microenvironment, characterized by increased frequency of activated macrophages and dendritic cells and improved infiltration of CD8+ T cells, resulting in inhibition of tumor growth, prolonged survival, and increased response to anti-PD-1 immune checkpoint blockade in orthotopic models of breast cancer. Collectively, these studies position SAPCon as a modular and programmable platform for improving the efficacy of systemically administered STING agonists for cancer immunotherapy.
Collapse
|
21
|
Nguyen DC, Song K, Jokonya S, Yazdani O, Sellers DL, Wang Y, Zakaria ABM, Pun SH, Stayton PS. Mannosylated STING Agonist Drugamers for Dendritic Cell-Mediated Cancer Immunotherapy. ACS CENTRAL SCIENCE 2024; 10:666-675. [PMID: 38559305 PMCID: PMC10979423 DOI: 10.1021/acscentsci.3c01310] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 01/22/2024] [Accepted: 02/06/2024] [Indexed: 04/04/2024]
Abstract
The Stimulator of Interferon Genes (STING) pathway is a promising target for cancer immunotherapy. Despite recent advances, therapies targeting the STING pathway are often limited by routes of administration, suboptimal STING activation, or off-target toxicity. Here, we report a dendritic cell (DC)-targeted polymeric prodrug platform (polySTING) that is designed to optimize intracellular delivery of a diamidobenzimidazole (diABZI) small-molecule STING agonist while minimizing off-target toxicity after parenteral administration. PolySTING incorporates mannose targeting ligands as a comonomer, which facilitates its uptake in CD206+/mannose receptor+ professional antigen-presenting cells (APCs) in the tumor microenvironment (TME). The STING agonist is conjugated through a cathepsin B-cleavable valine-alanine (VA) linker for selective intracellular drug release after receptor-mediated endocytosis. When administered intravenously in tumor-bearing mice, polySTING selectively targeted CD206+/mannose receptor+ APCs in the TME, resulting in increased cross-presenting CD8+ DCs, infiltrating CD8+ T cells in the TME as well as maturation across multiple DC subtypes in the tumor-draining lymph node (TDLN). Systemic administration of polySTING slowed tumor growth in a B16-F10 murine melanoma model as well as a 4T1 murine breast cancer model with an acceptable safety profile. Thus, we demonstrate that polySTING delivers STING agonists to professional APCs after systemic administration, generating efficacious DC-driven antitumor immunity with minimal side effects. This new polymeric prodrug platform may offer new opportunities for combining efficient targeted STING agonist delivery with other selective tumor therapeutic strategies.
Collapse
Affiliation(s)
- Dinh Chuong Nguyen
- Molecular
Engineering & Sciences Institute, University
of Washington, Seattle, Washington 98195, United States
| | - Kefan Song
- Department
of Bioengineering, University of Washington, Seattle, Washington 98195, United States
| | - Simbarashe Jokonya
- Department
of Bioengineering, University of Washington, Seattle, Washington 98195, United States
| | - Omeed Yazdani
- Department
of Bioengineering, University of Washington, Seattle, Washington 98195, United States
| | - Drew L. Sellers
- Department
of Bioengineering, University of Washington, Seattle, Washington 98195, United States
| | - Yonghui Wang
- Department
of Bioengineering, University of Washington, Seattle, Washington 98195, United States
| | - ABM Zakaria
- Department
of Bioengineering, University of Washington, Seattle, Washington 98195, United States
| | - Suzie H. Pun
- Molecular
Engineering & Sciences Institute, University
of Washington, Seattle, Washington 98195, United States
- Department
of Bioengineering, University of Washington, Seattle, Washington 98195, United States
| | - Patrick S. Stayton
- Molecular
Engineering & Sciences Institute, University
of Washington, Seattle, Washington 98195, United States
- Department
of Bioengineering, University of Washington, Seattle, Washington 98195, United States
| |
Collapse
|
22
|
Kane G, Lusi C, Brassil M, Atukorale P. Engineering approaches for innate immune-mediated tumor microenvironment remodeling. IMMUNO-ONCOLOGY TECHNOLOGY 2024; 21:100406. [PMID: 38213392 PMCID: PMC10777078 DOI: 10.1016/j.iotech.2023.100406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/13/2024]
Abstract
Cancer immunotherapy offers transformative promise particularly for the treatment of lethal cancers, since a correctly trained immune system can comprehensively orchestrate tumor clearance with no need for continued therapeutic intervention. Historically, the majority of immunotherapies have been T cell-focused and have included immune checkpoint inhibitors, chimeric antigen receptor T cells, and T-cell vaccines. Unfortunately T-cell-focused therapies have failed to achieve optimal efficacy in most solid tumors largely because of a highly immunosuppressed 'cold' or immune-excluded tumor microenvironment (TME). Recently, a rapidly growing treatment paradigm has emerged that focuses on activation of tumor-resident innate antigen-presenting cells, such as dendritic cells and macrophages, which can drive a proinflammatory immune response to remodel the TME from 'cold' or immune-excluded to 'hot'. Early strategies for TME remodeling centered on free cytokines and agonists, but these approaches have faced significant hurdles in both delivery and efficacy. Systemic toxicity from off-target inflammation is a paramount concern in these therapies. To address this critical gap, engineering approaches have provided the opportunity to add 'built-in' capabilities to cytokines, agonists, and other therapeutic agents to mediate improved delivery and efficacy. Such capabilities have included protective encapsulation to shield them from degradation, targeting to direct them with high specificity to tumors, and co-delivery strategies to harness synergistic proinflammatory pathways. Here, we review innate immune-mediated TME remodeling engineering approaches that focus on cytokines, innate immune agonists, immunogenic viruses, and cell-based methods, highlighting emerging preclinical approaches and strategies that are either being tested in clinical trials or already Food and Drug Administration approved.
Collapse
Affiliation(s)
- G.I. Kane
- Department of Biomedical Engineering, University of Massachusetts Amherst, Amherst
- University of Massachusetts Cancer Center, Worcester
| | - C.F. Lusi
- Department of Biomedical Engineering, University of Massachusetts Amherst, Amherst
- University of Massachusetts Cancer Center, Worcester
| | - M.L. Brassil
- Department of Biomedical Engineering, University of Massachusetts Amherst, Amherst
- University of Massachusetts Cancer Center, Worcester
| | - P.U. Atukorale
- Department of Biomedical Engineering, University of Massachusetts Amherst, Amherst
- University of Massachusetts Cancer Center, Worcester
- Division of Innate Immunity, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, USA
| |
Collapse
|
23
|
Liu X, Cheng Y, Mu Y, Zhang Z, Tian D, Liu Y, Hu X, Wen T. Diverse drug delivery systems for the enhancement of cancer immunotherapy: an overview. Front Immunol 2024; 15:1328145. [PMID: 38298192 PMCID: PMC10828056 DOI: 10.3389/fimmu.2024.1328145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 01/03/2024] [Indexed: 02/02/2024] Open
Abstract
Despite the clear benefits demonstrated by immunotherapy, there is still an inevitable off-target effect resulting in serious adverse immune reactions. In recent years, the research and development of Drug Delivery System (DDS) has received increased prominence. In decades of development, DDS has demonstrated the ability to deliver drugs in a precisely targeted manner to mitigate side effects and has the advantages of flexible control of drug release, improved pharmacokinetics, and drug distribution. Therefore, we consider that combining cancer immunotherapy with DDS can enhance the anti-tumor ability. In this paper, we provide an overview of the latest drug delivery strategies in cancer immunotherapy and briefly introduce the characteristics of DDS based on nano-carriers (liposomes, polymer nano-micelles, mesoporous silica, extracellular vesicles, etc.) and coupling technology (ADCs, PDCs and targeted protein degradation). Our aim is to show readers a variety of drug delivery platforms under different immune mechanisms, and analyze their advantages and limitations, to provide more superior and accurate targeting strategies for cancer immunotherapy.
Collapse
Affiliation(s)
- Xu Liu
- Department of Respiratory and Infectious Disease of Geriatrics, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yang Cheng
- Department of Respiratory and Infectious Disease of Geriatrics, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yao Mu
- Department of Respiratory and Infectious Disease of Geriatrics, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | | | - Dan Tian
- Department of Thoracic Surgery, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Yunpeng Liu
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, Liaoning, China
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, Liaoning, China
- Liaoning Province Clinical Research Center for Cancer, The First Hospital of China Medical University, Shenyang, Liaoning, China
- Clinical Cancer Treatment and Research Center of Shenyang, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xuejun Hu
- Department of Respiratory and Infectious Disease of Geriatrics, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Ti Wen
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, Liaoning, China
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, Liaoning, China
- Liaoning Province Clinical Research Center for Cancer, The First Hospital of China Medical University, Shenyang, Liaoning, China
- Clinical Cancer Treatment and Research Center of Shenyang, The First Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
24
|
Chen X, Xu Z, Li T, Thakur A, Wen Y, Zhang K, Liu Y, Liang Q, Liu W, Qin JJ, Yan Y. Nanomaterial-encapsulated STING agonists for immune modulation in cancer therapy. Biomark Res 2024; 12:2. [PMID: 38185685 PMCID: PMC10773049 DOI: 10.1186/s40364-023-00551-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 12/19/2023] [Indexed: 01/09/2024] Open
Abstract
The cGAS-STING signaling pathway has emerged as a critical mediator of innate immune responses, playing a crucial role in improving antitumor immunity through immune effector responses. Targeting the cGAS-STING pathway holds promise for overcoming immunosuppressive tumor microenvironments (TME) and promoting effective tumor elimination. However, systemic administration of current STING agonists faces challenges related to low bioavailability and potential adverse effects, thus limiting their clinical applicability. Recently, nanotechnology-based strategies have been developed to modulate TMEs for robust immunotherapeutic responses. The encapsulation and delivery of STING agonists within nanoparticles (STING-NPs) present an attractive avenue for antitumor immunotherapy. This review explores a range of nanoparticles designed to encapsulate STING agonists, highlighting their benefits, including favorable biocompatibility, improved tumor penetration, and efficient intracellular delivery of STING agonists. The review also summarizes the immunomodulatory impacts of STING-NPs on the TME, including enhanced secretion of pro-inflammatory cytokines and chemokines, dendritic cell activation, cytotoxic T cell priming, macrophage re-education, and vasculature normalization. Furthermore, the review offers insights into co-delivered nanoplatforms involving STING agonists alongside antitumor agents such as chemotherapeutic compounds, immune checkpoint inhibitors, antigen peptides, and other immune adjuvants. These platforms demonstrate remarkable versatility in inducing immunogenic responses within the TME, ultimately amplifying the potential for antitumor immunotherapy.
Collapse
Affiliation(s)
- Xi Chen
- Department of Pharmacy, Xiangya Hospital, Central South University, 410008, Changsha, Hunan, China
| | - Zhijie Xu
- Department of Pathology, Xiangya Hospital, Central South University, 410008, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, 410008, Changsha, Hunan, China
| | - Tongfei Li
- Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, 442000, Shiyan, Hubei, China
| | - Abhimanyu Thakur
- Pritzker School of Molecular Engineering, Ben May Department for Cancer Research, University of Chicago, 60637, Chicago, IL, USA
| | - Yu Wen
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Furong Laboratory, Central South University, 410008, Changsha, Hunan, China
| | - Kui Zhang
- Pritzker School of Molecular Engineering, Ben May Department for Cancer Research, University of Chicago, 60637, Chicago, IL, USA
| | - Yuanhong Liu
- Department of Pharmacy, Xiangya Hospital, Central South University, 410008, Changsha, Hunan, China
| | - Qiuju Liang
- Department of Pharmacy, Xiangya Hospital, Central South University, 410008, Changsha, Hunan, China
| | - Wangrui Liu
- Department of Interventional Oncology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 200127, Shanghai, China.
| | - Jiang-Jiang Qin
- Hangzhou Institute of Medicine (HIM), Zhejiang Cancer Hospital, Chinese Academy of Sciences, 310022, Hangzhou, Zhejiang, China.
| | - Yuanliang Yan
- Department of Pharmacy, Xiangya Hospital, Central South University, 410008, Changsha, Hunan, China.
| |
Collapse
|
25
|
Zhang J, Yu S, Peng Q, Wang P, Fang L. Emerging mechanisms and implications of cGAS-STING signaling in cancer immunotherapy strategies. Cancer Biol Med 2024; 21:j.issn.2095-3941.2023.0440. [PMID: 38172538 PMCID: PMC10875285 DOI: 10.20892/j.issn.2095-3941.2023.0440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 12/14/2023] [Indexed: 01/05/2024] Open
Abstract
The intricate interplay between the human immune system and cancer development underscores the central role of immunotherapy in cancer treatment. Within this landscape, the innate immune system, a critical sentinel protecting against tumor incursion, is a key player. The cyclic GMP-AMP synthase (cGAS) and stimulator of interferon genes (STING) pathway has been found to be a linchpin of innate immunity: activation of this signaling pathway orchestrates the production of type I interferon (IFN-α/β), thus fostering the maturation, differentiation, and mobilization of immune effectors in the tumor microenvironment. Furthermore, STING activation facilitates the release and presentation of tumor antigens, and therefore is an attractive target for cancer immunotherapy. Current strategies to activate the STING pathway, including use of pharmacological agonists, have made substantial advancements, particularly when combined with immune checkpoint inhibitors. These approaches have shown promise in preclinical and clinical settings, by enhancing patient survival rates. This review describes the evolving understanding of the cGAS-STING pathway's involvement in tumor biology and therapy. Moreover, this review explores classical and non-classical STING agonists, providing insights into their mechanisms of action and potential for optimizing immunotherapy strategies. Despite challenges and complexities, the cGAS-STING pathway, a promising avenue for enhancing cancer treatment efficacy, has the potential to revolutionize patient outcomes.
Collapse
Affiliation(s)
- Jiawen Zhang
- Tongji University Cancer Center, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai 200072, China
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201620, China
| | - Sihui Yu
- Department of Obstetrics and Gynecology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Qiao Peng
- Tongji University Cancer Center, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai 200072, China
| | - Ping Wang
- Tongji University Cancer Center, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai 200072, China
| | - Lan Fang
- Tongji University Cancer Center, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai 200072, China
| |
Collapse
|
26
|
Xuan C, Hu R. Chemical Biology Perspectives on STING Agonists as Tumor Immunotherapy. ChemMedChem 2023; 18:e202300405. [PMID: 37794702 DOI: 10.1002/cmdc.202300405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 10/02/2023] [Accepted: 10/03/2023] [Indexed: 10/06/2023]
Abstract
Stimulator of interferon genes (STING) is a crucial adaptor protein in the innate immune response. STING activation triggers cytokine secretion, including type I interferon and initiates T cell-mediated adaptive immunity. The activated immune system converts "cold tumors" into "hot tumors" that are highly responsive to T cells by recruiting them to the tumor microenvironment, ultimately leading to potent and long-lasting antitumor effects. Unlike most immune checkpoint inhibitors, STING agonists represent a groundbreaking class of innate immune agonists that hold great potential for effectively targeting various cancer populations and are poised to become a blockbuster in tumor immunotherapy. This review will focus on the correlation between the STING signaling pathway and tumor immunity, as well as explore the impact of STING activation on other biological processes. Ultimately, we will summarize the development and optimization of STING agonists from a medicinal chemistry perspective, evaluate their potential in cancer therapy, and identify possible challenges for future advancement.
Collapse
Affiliation(s)
- Chenyuan Xuan
- Department of Pharmacology, China Pharmaceutical University, No 24, TongJiaXiang, Gulou District, Nanjing, 210009, P. R. China
| | - Rong Hu
- Department of Pharmacology, China Pharmaceutical University, No 24, TongJiaXiang, Gulou District, Nanjing, 210009, P. R. China
| |
Collapse
|
27
|
Huang C, Shao N, Huang Y, Chen J, Wang D, Hu G, Zhang H, Luo L, Xiao Z. Overcoming challenges in the delivery of STING agonists for cancer immunotherapy: A comprehensive review of strategies and future perspectives. Mater Today Bio 2023; 23:100839. [PMID: 38024837 PMCID: PMC10630661 DOI: 10.1016/j.mtbio.2023.100839] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 10/12/2023] [Accepted: 10/19/2023] [Indexed: 12/01/2023] Open
Abstract
STING (Stimulator of Interferon Genes) agonists have emerged as promising agents in the field of cancer immunotherapy, owing to their excellent capacity to activate the innate immune response and combat tumor-induced immunosuppression. This review provides a comprehensive exploration of the strategies employed to develop effective formulations for STING agonists, with particular emphasis on versatile nano-delivery systems. The recent advancements in delivery systems based on lipids, natural/synthetic polymers, and proteins for STING agonists are summarized. The preparation methodologies of nanoprecipitation, self-assembly, and hydrogel, along with their advantages and disadvantages, are also discussed. Furthermore, the challenges and opportunities in developing next-generation STING agonist delivery systems are elaborated. This review aims to serve as a reference for researchers in designing novel and effective STING agonist delivery systems for cancer immunotherapy.
Collapse
Affiliation(s)
- Cuiqing Huang
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, The First Affiliated Hospital of Jinan University, Guangzhou, 510632, China
- Department of Ultrasound, Guangdong Women and Children Hospital, Guangzhou, 511400, China
| | - Ni Shao
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, The First Affiliated Hospital of Jinan University, Guangzhou, 510632, China
| | - Yanyu Huang
- Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, CA, 95817, USA
| | - Jifeng Chen
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, The First Affiliated Hospital of Jinan University, Guangzhou, 510632, China
| | - Duo Wang
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, The First Affiliated Hospital of Jinan University, Guangzhou, 510632, China
| | - Genwen Hu
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, The First Affiliated Hospital of Jinan University, Guangzhou, 510632, China
- Department of Radiology, Shenzhen People's Hospital, The Second Clinical Medical College of Jinan University, Shenzhen, 518020, China
| | - Hong Zhang
- Department of Interventional Vascular Surgery, The Sixth Affiliated Hospital of Jinan University, Dongguan, 523560, China
| | - Liangping Luo
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, The First Affiliated Hospital of Jinan University, Guangzhou, 510632, China
| | - Zeyu Xiao
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, The First Affiliated Hospital of Jinan University, Guangzhou, 510632, China
| |
Collapse
|
28
|
Dosta P, Cryer AM, Dion MZ, Shiraishi T, Langston SP, Lok D, Wang J, Harrison S, Hatten T, Ganno ML, Appleman VA, Taboada GM, Puigmal N, Ferber S, Kalash S, Prado M, Rodríguez AL, Kamoun WS, Abu-Yousif AO, Artzi N. Investigation of the enhanced antitumour potency of STING agonist after conjugation to polymer nanoparticles. NATURE NANOTECHNOLOGY 2023; 18:1351-1363. [PMID: 37443252 DOI: 10.1038/s41565-023-01447-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 05/31/2023] [Indexed: 07/15/2023]
Abstract
Intravenously administered cyclic dinucleotides and other STING agonists are hampered by low cellular uptake and poor circulatory half-life. Here we report the covalent conjugation of cyclic dinucleotides to poly(β-amino ester) nanoparticles through a cathepsin-sensitive linker. This is shown to increase stability and loading, thereby expanding the therapeutic window in multiple syngeneic tumour models, enabling the study of how the long-term fate of the nanoparticles affects the immune response. In a melanoma mouse model, primary tumour clearance depends on the STING signalling by host cells-rather than cancer cells-and immune memory depends on the spleen. The cancer cells act as a depot for the nanoparticles, releasing them over time to activate nearby immune cells to control tumour growth. Collectively, this work highlights the importance of nanoparticle structure and nano-biointeractions in controlling immunotherapy efficacy.
Collapse
Affiliation(s)
- Pere Dosta
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Medicine, Division of Engineering in Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA.
| | - Alexander M Cryer
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Medicine, Division of Engineering in Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Michelle Z Dion
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Medicine, Division of Engineering in Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
- Harvard-MIT Division of Health Sciences & Technology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | | | | | - David Lok
- Takeda Development Center Americas, Inc. (TDCA), Lexington, MA, USA
| | - Jianing Wang
- Takeda Development Center Americas, Inc. (TDCA), Lexington, MA, USA
| | - Sean Harrison
- Takeda Development Center Americas, Inc. (TDCA), Lexington, MA, USA
| | - Tiquella Hatten
- Takeda Development Center Americas, Inc. (TDCA), Lexington, MA, USA
| | - Michelle L Ganno
- Takeda Development Center Americas, Inc. (TDCA), Lexington, MA, USA
| | - Vicky A Appleman
- Takeda Development Center Americas, Inc. (TDCA), Lexington, MA, USA
| | | | - Núria Puigmal
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Medicine, Division of Engineering in Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Shiran Ferber
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Medicine, Division of Engineering in Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Santhosh Kalash
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Medicine, Division of Engineering in Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Michaela Prado
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Medicine, Division of Engineering in Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Alma L Rodríguez
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Medicine, Division of Engineering in Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Walid S Kamoun
- Takeda Development Center Americas, Inc. (TDCA), Lexington, MA, USA
| | | | - Natalie Artzi
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Medicine, Division of Engineering in Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA.
| |
Collapse
|
29
|
Pagendarm HM, Stone PT, Kimmel BR, Baljon JJ, Aziz MH, Pastora LE, Hubert L, Roth EW, Almunif S, Scott EA, Wilson JT. Engineering endosomolytic nanocarriers of diverse morphologies using confined impingement jet mixing. NANOSCALE 2023; 15:16016-16029. [PMID: 37753868 PMCID: PMC10568979 DOI: 10.1039/d3nr02874g] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 09/18/2023] [Indexed: 09/28/2023]
Abstract
The clinical translation of many biomolecular therapeutics has been hindered by undesirable pharmacokinetic (PK) properties, inadequate membrane permeability, poor endosomal escape and cytosolic delivery, and/or susceptibility to degradation. Overcoming these challenges merits the development of nanoscale drug carriers (nanocarriers) to improve the delivery of therapeutic cargo. Herein, we implement a flash nanoprecipitation (FNP) approach to produce nanocarriers of diverse vesicular morphologies by using various molecular weight PEG-bl-DEAEMA-co-BMA (PEG-DB) polymers. We demonstrated that FNP can produce uniform (PDI < 0.1) particles after 5 impingements, and that by varying the copolymer hydrophilic mass fraction, FNP enables access to a diverse variety of nanoarchitectures including micelles, unilamellar vesicles (polymersomes), and multi-compartment vesicles (MCVs). We synthesized a library of 2 kDa PEG block copolymers, with DEAEMA-co-BMA second block molecular weights of 3, 6, 12, 15, 20, and 30 kDa. All formulations were both pH responsive, endosomolytic, and capable of loading and cytosolically delivering small negatively charged molecules - albeit to different degrees. Using a B16.F10 melanoma model, we showcased the therapeutic potential of a lead FNP formulated PEG-DB nanocarrier, encapsulating the cyclic dinucleotide (CDN) cGAMP to activate the stimulator of interferon genes (STING) pathway in a therapeutically relevant context. Collectively, these data demonstrate that an FNP process can be used to formulate pH-responsive nanocarriers of diverse morphologies using a PEG-DB polymer system. As FNP is an industrially scalable process, these data address the critical translational challenge of producing PEG-DB nanoparticles at scale. Furthermore, the diverse morphologies produced may specialize in the delivery of distinct biomolecular cargos for other therapeutic applications, implicating the therapeutic potential of this platform in an array of disease applications.
Collapse
Affiliation(s)
- Hayden M Pagendarm
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA.
| | - Payton T Stone
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Blaise R Kimmel
- 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.
| | - Mina H Aziz
- Department of Biochemistry, Vanderbilt University, Nashville, TN 37235, USA
- Department of Neuroscience, Vanderbilt University, Nashville, TN 37235, USA
| | - Lucinda E Pastora
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Lauren Hubert
- Department of Chemical Engineering, The University of Rhode Island, Kingston, RI 02881, USA
| | - Eric W Roth
- NUANCE BioCryo, Northwestern University, Evanston, IL 60208, USA
| | - Sultan Almunif
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Evan A Scott
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
- Interdisciplinary Biological Sciences, Northwestern University, Evanston, IL 60208, USA
- Chemistry of Life Processes Institute, Northwestern University, Evanston, IL 60208, USA
- Simpson Querrey Institute, Northwestern University, Chicago, IL 60611, USA
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL 60611, USA
| | - John T Wilson
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA.
- Department of Chemical and Biomolecular 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 37235, USA
- Vanderbilt Institute of Nanoscale Science and Engineering, Vanderbilt University, Nashville, TN 37235, USA
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| |
Collapse
|
30
|
Go EJ, Yang H, Park W, Lee SJ, Han JH, Kong SJ, Lee WS, Han DK, Chon HJ, Kim C. Systemic Delivery of a STING Agonist-Loaded Positively Charged Liposome Selectively Targets Tumor Immune Microenvironment and Suppresses Tumor Angiogenesis. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2300544. [PMID: 37381624 DOI: 10.1002/smll.202300544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 06/13/2023] [Indexed: 06/30/2023]
Abstract
Although stimulator of interferon genes (STING) agonists has shown great promise in preclinical studies, the clinical development of STING agonist therapy is challenged by its limited systemic delivery. Here, positively charged fusogenic liposomes loaded with a STING agonist (PoSTING) are designed for systemic delivery and to preferentially target the tumor microenvironment. When PoSTING is administered intravenously, it selectively targets not only tumor cells but also immune and tumor endothelial cells (ECs). In particular, delivery of STING agonists to tumor ECs normalizes abnormal tumor vasculatures, induces intratumoral STING activation, and elicits robust anti-tumor T cell immunity within the tumor microenvironment. Therefore, PoSTING can be used as a systemic delivery platform to overcome the limitations of using STING agonists in clinical trials.
Collapse
Affiliation(s)
- Eun-Jin Go
- Medical Oncology, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, Gyeonggi, 13496, Republic of Korea
- Laboratory of Translational Immuno-Oncology, CHA University School of Medicine, Seongnam, Gyeonggi, 13496, Republic of Korea
| | - Hannah Yang
- Medical Oncology, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, Gyeonggi, 13496, Republic of Korea
- Laboratory of Translational Immuno-Oncology, CHA University School of Medicine, Seongnam, Gyeonggi, 13496, Republic of Korea
| | - Wooram Park
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Seoburo 2066, Suwon, Gyeonggi, 16419, Republic of Korea
| | - Seung Joon Lee
- Medical Oncology, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, Gyeonggi, 13496, Republic of Korea
- Laboratory of Translational Immuno-Oncology, CHA University School of Medicine, Seongnam, Gyeonggi, 13496, Republic of Korea
| | - Jun-Hyeok Han
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Seoburo 2066, Suwon, Gyeonggi, 16419, Republic of Korea
| | - So Jung Kong
- Medical Oncology, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, Gyeonggi, 13496, Republic of Korea
- Laboratory of Translational Immuno-Oncology, CHA University School of Medicine, Seongnam, Gyeonggi, 13496, Republic of Korea
| | - Won Suk Lee
- Medical Oncology, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, Gyeonggi, 13496, Republic of Korea
- Laboratory of Translational Immuno-Oncology, CHA University School of Medicine, Seongnam, Gyeonggi, 13496, Republic of Korea
| | - Dong Keun Han
- Department of Biomedical Science, CHA University, Seongnam, Gyeonggi, 13496, Republic of Korea
| | - Hong Jae Chon
- Medical Oncology, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, Gyeonggi, 13496, Republic of Korea
- Laboratory of Translational Immuno-Oncology, CHA University School of Medicine, Seongnam, Gyeonggi, 13496, Republic of Korea
| | - Chan Kim
- Medical Oncology, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, Gyeonggi, 13496, Republic of Korea
- Laboratory of Translational Immuno-Oncology, CHA University School of Medicine, Seongnam, Gyeonggi, 13496, Republic of Korea
| |
Collapse
|
31
|
Zhang J, Wang S, Zhang D, He X, Wang X, Han H, Qin Y. Nanoparticle-based drug delivery systems to enhance cancer immunotherapy in solid tumors. Front Immunol 2023; 14:1230893. [PMID: 37600822 PMCID: PMC10435760 DOI: 10.3389/fimmu.2023.1230893] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 07/19/2023] [Indexed: 08/22/2023] Open
Abstract
Immunotherapy has developed rapidly in solid tumors, especially in the areas of blocking inhibitory immune checkpoints and adoptive T-cell transfer for immune regulation. Many patients benefit from immunotherapy. However, the response rate of immunotherapy in the overall population are relatively low, which depends on the characteristics of the tumor and individualized patient differences. Moreover, the occurrence of drug resistance and adverse reactions largely limit the development of immunotherapy. Recently, the emergence of nanodrug delivery systems (NDDS) seems to improve the efficacy of immunotherapy by encapsulating drug carriers in nanoparticles to precisely reach the tumor site with high stability and biocompatibility, prolonging the drug cycle of action and greatly reducing the occurrence of toxic side effects. In this paper, we mainly review the advantages of NDDS and the mechanisms that enhance conventional immunotherapy in solid tumors, and summarize the recent advances in NDDS-based therapeutic strategies, which will provide valuable ideas for the development of novel tumor immunotherapy regimen.
Collapse
Affiliation(s)
- Jiaxin Zhang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Siyuan Wang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Daidi Zhang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xin He
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xue Wang
- Academy of Medical Science, School of Basic Medical Science, Zhengzhou University, Zhengzhou, China
| | - Huiqiong Han
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yanru Qin
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
32
|
Jin X, Wang W, Zhao X, Jiang W, Shao Q, Chen Z, Huang C. The battle between the innate immune cGAS-STING signaling pathway and human herpesvirus infection. Front Immunol 2023; 14:1235590. [PMID: 37600809 PMCID: PMC10433641 DOI: 10.3389/fimmu.2023.1235590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 07/21/2023] [Indexed: 08/22/2023] Open
Abstract
The incidence of human herpesvirus (HHVs) is gradually increasing and has affected a wide range of population. HHVs can result in serious consequences such as tumors, neonatal malformations, sexually transmitted diseases, as well as pose an immense threat to the human health. The cGAS-STING pathway is one of the innate immune pattern-recognition receptors discovered recently. This article discusses the role of the cGAS-STING pathway in human diseases, especially in human herpesvirus infections, as well as highlights how these viruses act on this pathway to evade the host immunity. Moreover, the author provides a comprehensive overview of modulators of the cGAS-STING pathway. By focusing on the small molecule compounds based on the cGAS-STING pathway, novel targets and concepts have been proposed for the development of antiviral drugs and vaccines, while also providing a reference for the investigation of disease models related to the cGAS-STING pathway. HHV is a double-stranded DNA virus that can trigger the activation of intracellular DNA sensor cGAS, after which the host cells initiate a cascade of reactions that culminate in the secretion of type I interferon to restrict the viral replication. Meanwhile, the viral protein can interact with various molecules in the cGAS-STING pathway. Viruses can evade immune surveillance and maintain their replication by inhibiting the enzyme activity of cGAS and reducing the phosphorylation levels of STING, TBK1 and IRF3 and suppressing the interferon gene activation. Activators and inhibitors of the cGAS-STING pathway have yielded numerous promising research findings in vitro and in vivo pertaining to cGAS/STING-related disease models. However, there remains a dearth of small molecule modulators that have been successfully translated into clinical applications, which serves as a hurdle to be overcome in the future.
Collapse
Affiliation(s)
- Ximing Jin
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wenjia Wang
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xinwei Zhao
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wenhua Jiang
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qingqing Shao
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhuo Chen
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Cong Huang
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
33
|
Hao Y, Ji Z, Zhou H, Wu D, Gu Z, Wang D, ten Dijke P. Lipid-based nanoparticles as drug delivery systems for cancer immunotherapy. MedComm (Beijing) 2023; 4:e339. [PMID: 37560754 PMCID: PMC10407046 DOI: 10.1002/mco2.339] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Revised: 06/26/2023] [Accepted: 07/04/2023] [Indexed: 08/11/2023] Open
Abstract
Immune checkpoint inhibitors (ICIs) have shown remarkable success in cancer treatment. However, in cancer patients without sufficient antitumor immunity, numerous data indicate that blocking the negative signals elicited by immune checkpoints is ineffective. Drugs that stimulate immune activation-related pathways are emerging as another route for improving immunotherapy. In addition, the development of nanotechnology presents a promising platform for tissue and cell type-specific delivery and improved uptake of immunomodulatory agents, ultimately leading to enhanced cancer immunotherapy and reduced side effects. In this review, we summarize and discuss the latest developments in nanoparticles (NPs) for cancer immuno-oncology therapy with a focus on lipid-based NPs (lipid-NPs), including the characteristics and advantages of various types. Using the agonists targeting stimulation of the interferon genes (STING) transmembrane protein as an exemplar, we review the potential of various lipid-NPs to augment STING agonist therapy. Furthermore, we present recent findings and underlying mechanisms on how STING pathway activation fosters antitumor immunity and regulates the tumor microenvironment and provide a summary of the distinct STING agonists in preclinical studies and clinical trials. Ultimately, we conduct a critical assessment of the obstacles and future directions in the utilization of lipid-NPs to enhance cancer immunotherapy.
Collapse
Affiliation(s)
- Yang Hao
- Department of Laboratory AnimalsCollege of Animal SciencesJilin UniversityChangchunChina
- Department of Basic MedicineChangzhi Medical CollegeChangzhiChina
- Department of Cell and Chemical Biology and Oncode InstituteLeiden University Medical CenterLeidenThe Netherlands
| | - Zhonghao Ji
- Department of Laboratory AnimalsCollege of Animal SciencesJilin UniversityChangchunChina
- Department of Basic MedicineChangzhi Medical CollegeChangzhiChina
| | - Hengzong Zhou
- Department of Laboratory AnimalsCollege of Animal SciencesJilin UniversityChangchunChina
| | - Dongrun Wu
- Departure of Philosophy, Faculty of HumanitiesLeiden UniversityLeidenThe Netherlands
| | - Zili Gu
- Department of RadiologyLeiden University Medical CenterLeidenThe Netherlands
| | - Dongxu Wang
- Department of Laboratory AnimalsCollege of Animal SciencesJilin UniversityChangchunChina
| | - Peter ten Dijke
- Department of Cell and Chemical Biology and Oncode InstituteLeiden University Medical CenterLeidenThe Netherlands
| |
Collapse
|
34
|
Chen X, Meng F, Xu Y, Li T, Chen X, Wang H. Chemically programmed STING-activating nano-liposomal vesicles improve anticancer immunity. Nat Commun 2023; 14:4584. [PMID: 37524727 PMCID: PMC10390568 DOI: 10.1038/s41467-023-40312-y] [Citation(s) in RCA: 34] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 07/20/2023] [Indexed: 08/02/2023] Open
Abstract
The often immune-suppressive tumor microenvironment (TME) may hinder immune evasion and response to checkpoint blockade therapies. Pharmacological activation of the STING pathway does create an immunologically hot TME, however, systemic delivery might lead to undesired off-target inflammatory responses. Here, we generate a small panel of esterase-activatable pro-drugs based on the structure of the non-nucleotide STING agonist MSA-2 that are subsequently stably incorporated into a liposomal vesicle for intravenous administration. The pharmacokinetic properties and immune stimulatory capacity of pro-drugs delivered via liposomes (SAProsomes) are enhanced compared to the free drug form. By performing efficacy screening among the SAProsomes incorporating different pro-drugs in syngeneic mouse tumor models, we find that superior therapeutic performance relies on improved delivery to the desired tumor and lymphoid compartments. The best candidate, SAProsome-3, highly stimulates secretion of inflammatory cytokines and creates a tumoricidal immune landscape. Notably, upon application to breast cancer or melanoma mouse models, SAProsome-3 elicits durable remission of established tumors and postsurgical tumor-free survival while decreasing metastatic burden without significant systemic toxicity. In summary, our work establishes the proof of principle for a better targeted and more efficient and safe STING agonist therapy.
Collapse
Affiliation(s)
- Xiaona Chen
- The First Affiliated Hospital, NHC Key Laboratory of Combined Multi-Organ Transplantation, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Zhejiang University School of Medicine, 310003, Hangzhou, Zhejiang Province, P. R. China
| | - Fanchao Meng
- The First Affiliated Hospital, NHC Key Laboratory of Combined Multi-Organ Transplantation, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Zhejiang University School of Medicine, 310003, Hangzhou, Zhejiang Province, P. R. China
| | - Yiting Xu
- The First Affiliated Hospital, NHC Key Laboratory of Combined Multi-Organ Transplantation, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Zhejiang University School of Medicine, 310003, Hangzhou, Zhejiang Province, P. R. China
| | - Tongyu Li
- The First Affiliated Hospital, NHC Key Laboratory of Combined Multi-Organ Transplantation, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Zhejiang University School of Medicine, 310003, Hangzhou, Zhejiang Province, P. R. China
| | - Xiaolong Chen
- The First Affiliated Hospital, NHC Key Laboratory of Combined Multi-Organ Transplantation, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Zhejiang University School of Medicine, 310003, Hangzhou, Zhejiang Province, P. R. China
| | - Hangxiang Wang
- The First Affiliated Hospital, NHC Key Laboratory of Combined Multi-Organ Transplantation, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Zhejiang University School of Medicine, 310003, Hangzhou, Zhejiang Province, P. R. China.
- Jinan Microecological Biomedicine Shandong Laboratory, 250117, Jinan, Shandong Province, P. R. China.
| |
Collapse
|
35
|
Ying-Rui M, Bu-Fan B, Deng L, Rong S, Qian-Mei Z. Targeting the stimulator of interferon genes (STING) in breast cancer. Front Pharmacol 2023; 14:1199152. [PMID: 37448962 PMCID: PMC10338072 DOI: 10.3389/fphar.2023.1199152] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 06/21/2023] [Indexed: 07/18/2023] Open
Abstract
Breast cancer has a high occurrence rate globally and its treatment has demonstrated clinical efficacy with the use of systemic chemotherapy and immune checkpoint blockade. Insufficient cytotoxic T lymphocyte infiltration and the accumulation of immunosuppressive cells within tumours are the primary factors responsible for the inadequate clinical effectiveness of breast cancer treatment. The stimulator of interferon genes (STING) represents a pivotal protein in the innate immune response. Upon activation, STING triggers the activation and enhancement of innate and adaptive immune functions, resulting in therapeutic benefits for malignant tumours. The STING signalling pathway in breast cancer is influenced by various factors such as deoxyribonucleic acid damage response, tumour immune microenvironment, and mitochondrial function. The use of STING agonists is gaining momentum in breast cancer research. This review provides a comprehensive overview of the cyclic guanosine monophosphate-adenosine monophosphate synthase-STING pathway, its agonists, and the latest findings related to their application in breast cancer.
Collapse
Affiliation(s)
- Ma Ying-Rui
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Bai Bu-Fan
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Liu Deng
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Shi Rong
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhou Qian-Mei
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, China
| |
Collapse
|
36
|
Zahid AA, Chakraborty A, Luo W, Coyle A, Paul A. Tailoring the Inherent Properties of Biobased Nanoparticles for Nanomedicine. ACS Biomater Sci Eng 2023. [PMID: 37378614 DOI: 10.1021/acsbiomaterials.3c00364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/29/2023]
Abstract
Biobased nanoparticles are at the leading edge of the rapidly developing field of nanomedicine and biotherapeutics. Their unique size, shape, and biophysical properties make them attractive tools for biomedical research, including vaccination, targeted drug delivery, and immune therapy. These nanoparticles are engineered to present native cell receptors and proteins on their surfaces, providing a biomimicking camouflage for therapeutic cargo to evade rapid degradation, immune rejection, inflammation, and clearance. Despite showing promising clinical relevance, commercial implementation of these biobased nanoparticles is yet to be fully realized. In this perspective, we discuss advanced biobased nanoparticle designs used in medical applications, such as cell membrane nanoparticles, exosomes, and synthetic lipid-derived nanoparticles, and highlight their benefits and potential challenges. Moreover, we critically assess the future of preparing such particles using artificial intelligence and machine learning. These advanced computational tools will be able to predict the functional composition and behavior of the proteins and cell receptors present on the nanoparticle surfaces. With more advancement in designing new biobased nanoparticles, this field of research could play a key role in dictating the future rational design of drug transporters, thereby ultimately improving overall therapeutic outcomes.
Collapse
Affiliation(s)
- Alap Ali Zahid
- Department of Chemical and Biochemical Engineering, The University of Western Ontario, London, Ontario N6A 5B9, Canada
| | - Aishik Chakraborty
- Department of Chemical and Biochemical Engineering, The University of Western Ontario, London, Ontario N6A 5B9, Canada
| | - Wei Luo
- Department of Chemical and Biochemical Engineering, The University of Western Ontario, London, Ontario N6A 5B9, Canada
| | - Ali Coyle
- School of Biomedical Engineering, The University of Western Ontario, London, Ontario N6A 5B9, Canada
| | - Arghya Paul
- Department of Chemical and Biochemical Engineering, The University of Western Ontario, London, Ontario N6A 5B9, Canada
- School of Biomedical Engineering, The University of Western Ontario, London, Ontario N6A 5B9, Canada
- Department of Chemistry, The Centre for Advanced Materials and Biomaterials Research, The University of Western Ontario, London, Ontario N6A 5B9, Canada
| |
Collapse
|
37
|
Seok JK, Kim M, Kang HC, Cho YY, Lee HS, Lee JY. Beyond DNA sensing: expanding the role of cGAS/STING in immunity and diseases. Arch Pharm Res 2023:10.1007/s12272-023-01452-3. [PMID: 37354378 DOI: 10.1007/s12272-023-01452-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Accepted: 06/13/2023] [Indexed: 06/26/2023]
Abstract
Cyclic guanosine monophosphate-adenosine monophosphate (cGAMP) synthase (cGAS) is a DNA sensor that elicits a robust type I interferon response by recognizing ubiquitous danger-associated molecules. The cGAS/stimulator of interferon genes (cGAS/STING) is activated by endogenous DNA, including DNA released from mitochondria and extranuclear chromatin, as well as exogenous DNA derived from pathogenic microorganisms. cGAS/STING is positioned as a key axis of autoimmunity, the inflammatory response, and cancer progression, suggesting that the cGAS/STING signaling pathway represents an efficient therapeutic target. Based on the accumulated evidence, we present insights into the prevention and treatment of cGAS/STING-related chronic immune and inflammatory diseases. This review presents the current state of clinical and nonclinical development of modulators targeting cGAS/STING, providing useful information on the design of therapeutic strategies.
Collapse
Affiliation(s)
- Jin Kyung Seok
- College of Pharmacy, The Catholic University of Korea, Bucheon, 14662, Republic of Korea
| | - Minhyuk Kim
- College of Pharmacy, The Catholic University of Korea, Bucheon, 14662, Republic of Korea
| | - Han Chang Kang
- College of Pharmacy, The Catholic University of Korea, Bucheon, 14662, Republic of Korea
| | - Yong-Yeon Cho
- College of Pharmacy, The Catholic University of Korea, Bucheon, 14662, Republic of Korea
| | - Hye Suk Lee
- College of Pharmacy, The Catholic University of Korea, Bucheon, 14662, Republic of Korea
| | - Joo Young Lee
- College of Pharmacy, The Catholic University of Korea, Bucheon, 14662, Republic of Korea.
| |
Collapse
|
38
|
Arango D, Cifuentes J, Puentes PR, Beltran T, Bittar A, Ocasión C, Muñoz-Camargo C, Bloch NI, Reyes LH, Cruz JC. Tailoring Magnetite-Nanoparticle-Based Nanocarriers for Gene Delivery: Exploiting CRISPRa Potential in Reducing Conditions. NANOMATERIALS (BASEL, SWITZERLAND) 2023; 13:nano13111782. [PMID: 37299685 DOI: 10.3390/nano13111782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 05/22/2023] [Accepted: 05/26/2023] [Indexed: 06/12/2023]
Abstract
Gene delivery has emerged as a promising alternative to conventional treatment approaches, allowing for the manipulation of gene expression through gene insertion, deletion, or alteration. However, the susceptibility of gene delivery components to degradation and challenges associated with cell penetration necessitate the use of delivery vehicles for effective functional gene delivery. Nanostructured vehicles, such as iron oxide nanoparticles (IONs) including magnetite nanoparticles (MNPs), have demonstrated significant potential for gene delivery applications due to their chemical versatility, biocompatibility, and strong magnetization. In this study, we developed an ION-based delivery vehicle capable of releasing linearized nucleic acids (tDNA) under reducing conditions in various cell cultures. As a proof of concept, we immobilized a CRISPR activation (CRISPRa) sequence to overexpress the pink1 gene on MNPs functionalized with polyethylene glycol (PEG), 3-[(2-aminoethyl)dithio]propionic acid (AEDP), and a translocating protein (OmpA). The nucleic sequence (tDNA) was modified to include a terminal thiol group and was conjugated to AEDP's terminal thiol via a disulfide exchange reaction. Leveraging the natural sensitivity of the disulfide bridge, the cargo was released under reducing conditions. Physicochemical characterizations, including thermogravimetric analysis (TGA) and Fourier-transform infrared (FTIR) spectroscopy, confirmed the correct synthesis and functionalization of the MNP-based delivery carriers. The developed nanocarriers exhibited remarkable biocompatibility, as demonstrated by the hemocompatibility, platelet aggregation, and cytocompatibility assays using primary human astrocytes, rodent astrocytes, and human fibroblast cells. Furthermore, the nanocarriers enabled efficient cargo penetration, uptake, and endosomal escape, with minimal nucleofection. A preliminary functionality test using RT-qPCR revealed that the vehicle facilitated the timely release of CRISPRa vectors, resulting in a remarkable 130-fold overexpression of pink1. We demonstrate the potential of the developed ION-based nanocarrier as a versatile and promising gene delivery vehicle with potential applications in gene therapy. The developed nanocarrier is capable of delivering any nucleic sequence (up to 8.2 kb) once it is thiolated using the methodology explained in this study. To our knowledge, this represents the first MNP-based nanocarrier capable of delivering nucleic sequences under specific reducing conditions while preserving functionality.
Collapse
Affiliation(s)
- David Arango
- Department of Biomedical Engineering, Universidad de Los Andes, Bogotá 111711, Colombia
| | - Javier Cifuentes
- Department of Biomedical Engineering, Universidad de Los Andes, Bogotá 111711, Colombia
| | - Paola Ruiz Puentes
- Department of Biomedical Engineering, Universidad de Los Andes, Bogotá 111711, Colombia
| | - Tatiana Beltran
- Department of Biomedical Engineering, Universidad de Los Andes, Bogotá 111711, Colombia
| | - Amaury Bittar
- Department of Biomedical Engineering, Universidad de Los Andes, Bogotá 111711, Colombia
| | - Camila Ocasión
- Department of Chemical and Food Engineering, Universidad de Los Andes, Bogotá 111711, Colombia
| | | | - Natasha I Bloch
- Department of Biomedical Engineering, Universidad de Los Andes, Bogotá 111711, Colombia
| | - Luis H Reyes
- Department of Chemical and Food Engineering, Universidad de Los Andes, Bogotá 111711, Colombia
| | - Juan C Cruz
- Department of Biomedical Engineering, Universidad de Los Andes, Bogotá 111711, Colombia
| |
Collapse
|
39
|
Brai A, Poggialini F, Pasqualini C, Trivisani CI, Vagaggini C, Dreassi E. Progress towards Adjuvant Development: Focus on Antiviral Therapy. Int J Mol Sci 2023; 24:9225. [PMID: 37298177 PMCID: PMC10253057 DOI: 10.3390/ijms24119225] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 05/12/2023] [Accepted: 05/22/2023] [Indexed: 06/12/2023] Open
Abstract
In recent decades, vaccines have been extraordinary resources to prevent pathogen diffusion and cancer. Even if they can be formed by a single antigen, the addition of one or more adjuvants represents the key to enhance the response of the immune signal to the antigen, thus accelerating and increasing the duration and the potency of the protective effect. Their use is of particular importance for vulnerable populations, such as the elderly or immunocompromised people. Despite their importance, only in the last forty years has the search for novel adjuvants increased, with the discovery of novel classes of immune potentiators and immunomodulators. Due to the complexity of the cascades involved in immune signal activation, their mechanism of action remains poorly understood, even if significant discovery has been recently made thanks to recombinant technology and metabolomics. This review focuses on the classes of adjuvants under research, recent mechanism of action studies, as well as nanodelivery systems and novel classes of adjuvants that can be chemically manipulated to create novel small molecule adjuvants.
Collapse
Affiliation(s)
- Annalaura Brai
- Department of Biotechnologies, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, I-53100 Siena, Italy; (A.B.); (F.P.); (C.P.); (C.V.)
| | - Federica Poggialini
- Department of Biotechnologies, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, I-53100 Siena, Italy; (A.B.); (F.P.); (C.P.); (C.V.)
| | - Claudia Pasqualini
- Department of Biotechnologies, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, I-53100 Siena, Italy; (A.B.); (F.P.); (C.P.); (C.V.)
| | - Claudia Immacolata Trivisani
- Department of Biotechnologies, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, I-53100 Siena, Italy; (A.B.); (F.P.); (C.P.); (C.V.)
- Department of Pharmaceutical Sciences, University of Vienna, 1090 Vienna, Austria
| | - Chiara Vagaggini
- Department of Biotechnologies, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, I-53100 Siena, Italy; (A.B.); (F.P.); (C.P.); (C.V.)
| | - Elena Dreassi
- Department of Biotechnologies, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, I-53100 Siena, Italy; (A.B.); (F.P.); (C.P.); (C.V.)
| |
Collapse
|
40
|
Wang-Bishop L, Kimmel BR, Ngwa VM, Madden MZ, Baljon JJ, Florian DC, Hanna A, Pastora LE, Sheehy TL, Kwiatkowski AJ, Wehbe M, Wen X, Becker KW, Garland KM, Schulman JA, Shae D, Edwards D, Wolf MM, Delapp R, Christov PP, Beckermann KE, Balko JM, Rathmell WK, Rathmell JC, Chen J, Wilson JT. STING-activating nanoparticles normalize the vascular-immune interface to potentiate cancer immunotherapy. Sci Immunol 2023; 8:eadd1153. [PMID: 37146128 PMCID: PMC10226150 DOI: 10.1126/sciimmunol.add1153] [Citation(s) in RCA: 25] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 04/13/2023] [Indexed: 05/07/2023]
Abstract
The tumor-associated vasculature imposes major structural and biochemical barriers to the infiltration of effector T cells and effective tumor control. Correlations between stimulator of interferon genes (STING) pathway activation and spontaneous T cell infiltration in human cancers led us to evaluate the effect of STING-activating nanoparticles (STANs), which are a polymersome-based platform for the delivery of a cyclic dinucleotide STING agonist, on the tumor vasculature and attendant effects on T cell infiltration and antitumor function. In multiple mouse tumor models, intravenous administration of STANs promoted vascular normalization, evidenced by improved vascular integrity, reduced tumor hypoxia, and increased endothelial cell expression of T cell adhesion molecules. STAN-mediated vascular reprogramming enhanced the infiltration, proliferation, and function of antitumor T cells and potentiated the response to immune checkpoint inhibitors and adoptive T cell therapy. We present STANs as a multimodal platform that activates and normalizes the tumor microenvironment to enhance T cell infiltration and function and augments responses to immunotherapy.
Collapse
Affiliation(s)
- Lihong Wang-Bishop
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN 37232, United States
| | - Blaise R. Kimmel
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN 37232, United States
| | - Verra M. Ngwa
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, United States
| | - Matthew Z. Madden
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, United States
| | - Jessalyn J. Baljon
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37232, United States
| | - David C. Florian
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN 37232, United States
| | - Ann Hanna
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, United States
| | - Lucinda E. Pastora
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN 37232, United States
| | - Taylor L. Sheehy
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37232, United States
| | - Alexander J. Kwiatkowski
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN 37232, United States
| | - Mohamed Wehbe
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN 37232, United States
| | - Xiaona Wen
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN 37232, United States
| | - Kyle W. Becker
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN 37232, United States
| | - Kyle M. Garland
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN 37232, United States
| | - Jacob A. Schulman
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37232, United States
| | - Daniel Shae
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN 37232, United States
| | - Deanna Edwards
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, United States
- Vanderbilt Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN 37232, United States
| | - Melissa M. Wolf
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, United States
| | - Rossane Delapp
- Department of Civil and Environmental Engineering, Vanderbilt University, Nashville, TN 37232, United States
| | - Plamen P. Christov
- Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN 37232, United States
| | - Kathryn E. Beckermann
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, United States
- Vanderbilt Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN 37232, United States
| | - Justin M. Balko
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, United States
- Vanderbilt Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN 37232, United States
- Vanderbilt Institute for Infection, Immunology and Inflammation, Vanderbilt University Medical Center, Nashville, TN 37232
- Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN 37232
| | - W. Kimryn Rathmell
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, United States
- Vanderbilt Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN 37232, United States
- Vanderbilt Institute for Infection, Immunology and Inflammation, Vanderbilt University Medical Center, Nashville, TN 37232
- Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Jeffrey C. Rathmell
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, United States
- Vanderbilt Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN 37232, United States
- Vanderbilt Institute for Infection, Immunology and Inflammation, Vanderbilt University Medical Center, Nashville, TN 37232
- Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Jin Chen
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, United States
- Vanderbilt Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN 37232, United States
- Vanderbilt Institute for Infection, Immunology and Inflammation, Vanderbilt University Medical Center, Nashville, TN 37232
| | - John T. Wilson
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN 37232, United States
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, United States
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37232, United States
- Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN 37232, United States
- Vanderbilt Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN 37232, United States
- Vanderbilt Institute for Infection, Immunology and Inflammation, Vanderbilt University Medical Center, Nashville, TN 37232
- Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN 37232
| |
Collapse
|
41
|
Sun X, Zhou X, Lei YL, Moon JJ. Unlocking the promise of systemic STING agonist for cancer immunotherapy. J Control Release 2023; 357:417-421. [PMID: 37001564 PMCID: PMC10476228 DOI: 10.1016/j.jconrel.2023.03.047] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 03/26/2023] [Indexed: 04/21/2023]
Abstract
Stimulator of interferon genes (STING) pathway is the key innate immune pathway involving in cancer immunity. Emerging new molecules and drug delivery systems have made systemic STING agonist immunotherapy possible and demonstrated efficient tumor eradication in preclinical studies. In this perspective, we will discuss the potential mechanisms of STING agonism as a multifaceted anti-cancer therapy and the pharmacological challenges associated with systemic delivery of STING agonists on the level of organs, tissues, cells, and intracellular compartments. We will present and discuss drug delivery strategies to address these challenges. New advances in the field can unlock the promise of systemic STING agonist as effective and safe cancer immunotherapy.
Collapse
Affiliation(s)
- Xiaoqi Sun
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA; Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA.
| | - Xingwu Zhou
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA; Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Yu Leo Lei
- Department of Periodontics and Oral Medicine, University of Michigan, Ann Arbor, MI 48109, USA; Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA
| | - James J Moon
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA; Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA; Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA; Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA; Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
42
|
Li Y, Li X, Yi J, Cao Y, Qin Z, Zhong Z, Yang W. Nanoparticle-Mediated STING Activation for Cancer Immunotherapy. Adv Healthc Mater 2023:e2300260. [PMID: 36905358 DOI: 10.1002/adhm.202300260] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 03/02/2023] [Indexed: 03/12/2023]
Abstract
As the first line of host defense against pathogenic infections, innate immunity plays a key role in antitumor immunotherapy. The cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) (cGAS-STING) pathway has attracted much attention because of the secretion of various proinflammatory cytokines and chemokines. Many STING agonists have been identified and applied into preclinical or clinical trials for cancer immunotherapy. However, the fast excretion, low bioavailability, nonspecificity, and adverse effects of the small molecule STING agonists limit their therapeutic efficacy and in vivo application. Nanodelivery systems with appropriate size, charge, and surface modification are capable of addressing these dilemmas. In this review, the mechanism of the cGAS-STING pathway is discussed and the STING agonists, focusing on nanoparticle-mediated STING therapy and combined therapy for cancers, are summarized. Finally, the future direction and challenges of nano-STING therapy are expounded, emphasizing the pivotal scientific problems and technical bottlenecks and hoping to provide general guidance for its clinical application.
Collapse
Affiliation(s)
- Yongjuan Li
- Medical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, 450001, China.,The Center of Infection and Immunity, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Xinyan Li
- Medical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, 450001, China.,The Center of Infection and Immunity, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Jinmeng Yi
- Medical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, 450001, China.,The Center of Infection and Immunity, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Yongjian Cao
- School of Pharmaceutical Sciences, Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Zhihai Qin
- Medical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, 450001, China.,The Center of Infection and Immunity, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Zhiyuan Zhong
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, State Key Laboratory of Radiation Medicine and Protection, and College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Weijing Yang
- School of Pharmaceutical Sciences, Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou University, Zhengzhou, Henan, 450001, China
| |
Collapse
|
43
|
Florian DC, Bennett NE, Odziomek M, Baljon JJ, Wehbe M, Merkel AR, Fischer MA, Savona MR, Rhoades JA, Guelcher SA, Wilson JT. Nanoparticle STING Agonist Reprograms the Bone Marrow to an Antitumor Phenotype and Protects Against Bone Destruction. CANCER RESEARCH COMMUNICATIONS 2023; 3:223-234. [PMID: 36968140 PMCID: PMC10035525 DOI: 10.1158/2767-9764.crc-22-0180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 11/23/2022] [Accepted: 01/11/2023] [Indexed: 01/15/2023]
Abstract
When breast cancer metastasizes to bone, treatment options are limited. Failure to treat bone metastases is thought to be due to therapy-resistant features of the bone marrow microenvironment. Using a murine model of bone metastatic mammary carcinoma, we demonstrate that systemic delivery of polymer nanoparticles loaded with cyclic dinucleotide (CDN) agonists of stimulator of interferon genes (STING) inhibited tumor growth and bone destruction after 7 days of treatment. Each dose of STING-activating nanoparticles trafficked to the bone marrow compartment and was retained within the tumor microenvironment for over 24 hours, enhancing antitumor immunity through proinflammatory cytokine production and early T-cell activation. While acquired resistance mechanisms, including increased levels of immunosuppressive cytokines and the infiltration of regulatory T cells, ultimately limited antitumor efficacy after 2 weeks of treatment, bone protective effects remained. Overall, these studies demonstrate that STING pathway activation, here enabled using a nanomedicine approach to enhance CDN delivery to bone metastatic sites, can reprogram the immune contexture of the bone marrow to an antitumor phenotype that inhibits bone colonization of metastatic breast cancer cells and protects from tumor-mediated bone destruction. Significance Bone metastases are difficult to treat due to the inaccessibility of the bone marrow compartment and the immunosuppressive microenvironment that protects resident stem cells. Packaging a STING agonist into a nanoparticle that enables systemic administration and drug accumulation at tumor sites overcomes both barriers to stymie metastatic breast cancer growth.
Collapse
Affiliation(s)
- David C. Florian
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee
- Center for Bone Biology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Natalie E. Bennett
- Center for Bone Biology, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, Tennessee
- Program in Cancer Biology, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Mateusz Odziomek
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee
- Center for Bone Biology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Jessalyn J. Baljon
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee
| | - Mohamed Wehbe
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Tennessee
| | - Alyssa R. Merkel
- Center for Bone Biology, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, Tennessee
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Tennessee
| | - Melissa A. Fischer
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Tennessee
| | - Michael R. Savona
- Program in Cancer Biology, Vanderbilt University School of Medicine, Nashville, Tennessee
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Tennessee
- Vanderbilt Ingram Cancer Center, Nashville, Tennessee
| | - Julie A. Rhoades
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee
- Center for Bone Biology, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, Tennessee
- Program in Cancer Biology, Vanderbilt University School of Medicine, Nashville, Tennessee
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Tennessee
| | - Scott A. Guelcher
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee
- Center for Bone Biology, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, Tennessee
| | - John T. Wilson
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee
- Program in Cancer Biology, Vanderbilt University School of Medicine, Nashville, Tennessee
- Vanderbilt Ingram Cancer Center, Nashville, Tennessee
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, Tennessee
| |
Collapse
|
44
|
Pimkova Polidarova M, Vanekova L, Brehova P, Dejmek M, Vavrina Z, Birkus G, Brazdova A. Synthetic Stimulator of Interferon Genes (STING) Agonists Induce a Cytokine-Mediated Anti-Hepatitis B Virus Response in Nonparenchymal Liver Cells. ACS Infect Dis 2023; 9:23-32. [PMID: 36472628 DOI: 10.1021/acsinfecdis.2c00424] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Chronic hepatitis B (CHB) remains a major public health problem worldwide, with limited treatment options, but inducing an antiviral response by innate immunity activation may provide a therapeutic alternative. We assessed the cytokine-mediated anti-hepatitis B virus (HBV) potential for stimulating the cyclic GMP-AMP synthase-stimulator of interferon genes (STING) pathway using STING agonists in primary human hepatocytes (PHH) and nonparenchymal liver cells (NPCs). The natural STING agonist, 2',3'-cyclic GMP-AMP, the synthetic analogue 3',3'-c-di(2'F,2'dAMP), and its bis(pivaloyloxymethyl) prodrug had strong indirect cytokine-mediated anti-HBV effects in PHH regardless of HBV genotype. Furthermore, STING agonists induced anti-HBV cytokine secretion in vitro, in both human and mouse NPCs, and triggered hepatic T cell activation. Cytokine secretion and lymphocyte activation were equally stimulated in NPCs isolated from control and HBV-persistent mice. Therefore, STING agonists modulate immune activation regardless of HBV persistence, paving the way toward a CHB therapy.
Collapse
Affiliation(s)
- Marketa Pimkova Polidarova
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo namesti 2, Prague 16000, Czech Republic
| | - Lenka Vanekova
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo namesti 2, Prague 16000, Czech Republic.,Faculty of Science, Charles University, Albertov 6, Prague 12800, Czech Republic
| | - Petra Brehova
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo namesti 2, Prague 16000, Czech Republic
| | - Milan Dejmek
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo namesti 2, Prague 16000, Czech Republic
| | - Zdenek Vavrina
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo namesti 2, Prague 16000, Czech Republic.,Faculty of Science, Charles University, Albertov 6, Prague 12800, Czech Republic
| | - Gabriel Birkus
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo namesti 2, Prague 16000, Czech Republic
| | - Andrea Brazdova
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo namesti 2, Prague 16000, Czech Republic
| |
Collapse
|
45
|
Li H, Luo Q, Zhang H, Ma X, Gu Z, Gong Q, Luo K. Nanomedicine embraces cancer radio-immunotherapy: mechanism, design, recent advances, and clinical translation. Chem Soc Rev 2023; 52:47-96. [PMID: 36427082 DOI: 10.1039/d2cs00437b] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Cancer radio-immunotherapy, integrating external/internal radiation therapy with immuno-oncology treatments, emerges in the current management of cancer. A growing number of pre-clinical studies and clinical trials have recently validated the synergistic antitumor effect of radio-immunotherapy, far beyond the "abscopal effect", but it suffers from a low response rate and toxicity issues. To this end, nanomedicines with an optimized design have been introduced to improve cancer radio-immunotherapy. Specifically, these nanomedicines are elegantly prepared by incorporating tumor antigens, immuno- or radio-regulators, or biomarker-specific imaging agents into the corresponding optimized nanoformulations. Moreover, they contribute to inducing various biological effects, such as generating in situ vaccination, promoting immunogenic cell death, overcoming radiation resistance, reversing immunosuppression, as well as pre-stratifying patients and assessing therapeutic response or therapy-induced toxicity. Overall, this review aims to provide a comprehensive landscape of nanomedicine-assisted radio-immunotherapy. The underlying working principles and the corresponding design strategies for these nanomedicines are elaborated by following the concept of "from bench to clinic". Their state-of-the-art applications, concerns over their clinical translation, along with perspectives are covered.
Collapse
Affiliation(s)
- Haonan Li
- Department of Radiology, Department of Biotherapy, Huaxi MR Research Center (HMRRC), Cancer Center, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu 610041, China.
| | - Qiang Luo
- Department of Radiology, Department of Biotherapy, Huaxi MR Research Center (HMRRC), Cancer Center, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu 610041, China.
| | - Hu Zhang
- Amgen Bioprocessing Centre, Keck Graduate Institute, Claremont, CA 91711, USA
| | - Xuelei Ma
- Department of Radiology, Department of Biotherapy, Huaxi MR Research Center (HMRRC), Cancer Center, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu 610041, China.
| | - Zhongwei Gu
- Department of Radiology, Department of Biotherapy, Huaxi MR Research Center (HMRRC), Cancer Center, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu 610041, China.
| | - Qiyong Gong
- Department of Radiology, Department of Biotherapy, Huaxi MR Research Center (HMRRC), Cancer Center, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu 610041, China. .,Functional and Molecular Imaging Key Laboratory of Sichuan Province and Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu 610041, China
| | - Kui Luo
- Department of Radiology, Department of Biotherapy, Huaxi MR Research Center (HMRRC), Cancer Center, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu 610041, China. .,Functional and Molecular Imaging Key Laboratory of Sichuan Province and Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu 610041, China
| |
Collapse
|
46
|
Hao M, Zhu L, Hou S, Chen S, Li X, Li K, Zhu N, Chen S, Xue L, Ju C, Zhang C. Sensitizing Tumors to Immune Checkpoint Blockage via STING Agonists Delivered by Tumor-Penetrating Neutrophil Cytopharmaceuticals. ACS NANO 2023; 17:1663-1680. [PMID: 36595464 DOI: 10.1021/acsnano.2c11764] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Immune checkpoint inhibitors (ICIs) have displayed potential efficacy in triple-negative breast cancer (TNBC) treatment, while only a minority of patients benefit from ICI therapy currently. Although activation of the innate immune stimulator of interferon genes (STING) pathway potentiates antitumor immunity and thus sensitizes tumors to ICIs, the efficient tumor penetration of STING agonists remains critically challenging. Herein, we prepare a tumor-penetrating neotype neutrophil cytopharmaceutical (NEs@STING-Mal-NP) with liposomal STING agonists conjugating on the surface of neutrophils, which is different from the typical neutrophil cytopharmaceutical that loads drugs inside the neutrophils. We show NEs@STING-Mal-NP that inherit the merits of neutrophils including proactive tumor vascular extravasation and tissue penetration significantly boost the tumor penetration of STING agonists. Moreover, the backpacked liposomal STING agonists can be released in response to hyaluronidase rich in the tumor environment, leading to enhanced uptake by tumor-infiltrating immune cells and tumor cells. Thus, NEs@STING-Mal-NP effectively activate the STING pathway and reinvigorate the tumor environment through converting macrophages and neutrophils to antitumor phenotypes, promoting the maturation of dendritic cells, and enhancing the infiltration and tumoricidal ability of T cells. Specifically, this cytopharmaceutical displays a significant inhibition on tumor growth and prolongs the survival of TNBC-bearing mice when combined with ICIs. We demonstrate that neutrophils serve as promising vehicles for delivering STING agonists throughout solid tumors and the developed neutrophil cytopharmaceuticals with backpacked STING agonists exhibit huge potential in boosting the immunotherapy of ICIs.
Collapse
Affiliation(s)
- Meixi Hao
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 210009, P.R. China
| | - Lulu Zhu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 210009, P.R. China
| | - Siyuan Hou
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 210009, P.R. China
| | - Sijia Chen
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 210009, P.R. China
| | - Xiuqi Li
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 210009, P.R. China
| | - Kaiming Li
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 210009, P.R. China
| | - Nianci Zhu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 210009, P.R. China
| | - Shanshan Chen
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 210009, P.R. China
| | - Lingjing Xue
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 210009, P.R. China
| | - Caoyun Ju
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 210009, P.R. China
| | - Can Zhang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 210009, P.R. China
| |
Collapse
|
47
|
Huang C, Zhou Y, Cheng J, Guo X, Shou D, Quan Y, Chen H, Chen H, Zhou Y. Pattern recognition receptors in the development of nonalcoholic fatty liver disease and progression to hepatocellular carcinoma: An emerging therapeutic strategy. Front Endocrinol (Lausanne) 2023; 14:1145392. [PMID: 37020586 PMCID: PMC10067914 DOI: 10.3389/fendo.2023.1145392] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 03/08/2023] [Indexed: 04/07/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is characterized by excessive lipid accumulation and has become the leading chronic liver disease worldwide. NAFLD is viewed as the hepatic manifestation of metabolic syndrome, ranging from simple steatosis and nonalcoholic steatohepatitis (NASH) to advanced fibrosis, eventually leading to cirrhosis and hepatocellular carcinoma (HCC). The pathogenesis of NAFLD progression is still not clear. Pattern recognition receptor (PRR)-mediated innate immune responses play a critical role in the initiation of NAFLD and the progression of NAFLD-related HCC. Toll-like receptors (TLRs) and the cyclic GMP-AMP (cGAMP) synthase (cGAS) are the two major PRRs in hepatocytes and resident innate immune cells in the liver. Increasing evidence indicates that the overactivation of TLRs and the cGAS signaling pathways may contribute to the development of liver disorders, including NAFLD progression. However, induction of PRRs is critical for the release of type I interferons (IFN-I) and the maturation of dendritic cells (DCs), which prime systemic antitumor immunity in HCC therapy. In this review, we will summarize the emerging evidence regarding the molecular mechanisms of TLRs and cGAS in the development of NAFLD and HCC. The dysfunction of PRR-mediated innate immune response is a critical determinant of NAFLD pathology; targeting and selectively inhibiting TLRs and cGAS signaling provides therapeutic potential for treating NALF-associated diseases in humans.
Collapse
Affiliation(s)
- Chen Huang
- Department of Gastroenterology and Hepatology, Guangzhou Key Laboratory of Digestive Diseases, Guangzhou Digestive Disease Center, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Youlian Zhou
- Department of Gastroenterology and Hepatology, Guangzhou Key Laboratory of Digestive Diseases, Guangzhou Digestive Disease Center, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Department of Gastroenterology and Hepatology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Jiemin Cheng
- Department of Gastroenterology and Hepatology, Guangzhou Key Laboratory of Digestive Diseases, Guangzhou Digestive Disease Center, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Department of Gastroenterology and Hepatology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Xue Guo
- Department of Gastroenterology and Hepatology, Guangzhou Key Laboratory of Digestive Diseases, Guangzhou Digestive Disease Center, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Department of Gastroenterology and Hepatology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Diwen Shou
- Department of Gastroenterology and Hepatology, Guangzhou Key Laboratory of Digestive Diseases, Guangzhou Digestive Disease Center, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Department of Gastroenterology and Hepatology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Ying Quan
- Department of Gastroenterology and Hepatology, Guangzhou Key Laboratory of Digestive Diseases, Guangzhou Digestive Disease Center, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Department of Gastroenterology and Hepatology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Hanqing Chen
- Department of Gastroenterology and Hepatology, Guangzhou Key Laboratory of Digestive Diseases, Guangzhou Digestive Disease Center, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Department of Gastroenterology and Hepatology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- *Correspondence: Yongjian Zhou, ; Huiting Chen, ; Hanqing Chen,
| | - Huiting Chen
- Department of Gastroenterology and Hepatology, Guangzhou Key Laboratory of Digestive Diseases, Guangzhou Digestive Disease Center, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- *Correspondence: Yongjian Zhou, ; Huiting Chen, ; Hanqing Chen,
| | - Yongjian Zhou
- Department of Gastroenterology and Hepatology, Guangzhou Key Laboratory of Digestive Diseases, Guangzhou Digestive Disease Center, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Department of Gastroenterology and Hepatology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- *Correspondence: Yongjian Zhou, ; Huiting Chen, ; Hanqing Chen,
| |
Collapse
|
48
|
Lee D, Huntoon K, Kang M, Lu Y, Gallup T, Jiang W, Kim BYS. Harnessing cGAS‐STING Pathway for Cancer Immunotherapy: From Bench to Clinic. ADVANCED THERAPEUTICS 2022. [DOI: 10.1002/adtp.202200040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- DaeYong Lee
- Department of Neurosurgery The University of Texas MD Anderson Cancer Center Houston TX 77030 USA
| | - Kristin Huntoon
- Department of Neurosurgery The University of Texas MD Anderson Cancer Center Houston TX 77030 USA
| | - Minjeong Kang
- Department of radiation oncology The University of Texas MD Anderson Cancer Center Houston TX 77030 USA
| | - Yifei Lu
- Department of Neurosurgery The University of Texas MD Anderson Cancer Center Houston TX 77030 USA
| | - Thomas Gallup
- Department of Neurosurgery The University of Texas MD Anderson Cancer Center Houston TX 77030 USA
| | - Wen Jiang
- Department of radiation oncology The University of Texas MD Anderson Cancer Center Houston TX 77030 USA
| | - Betty Y S Kim
- Department of Neurosurgery The University of Texas MD Anderson Cancer Center Houston TX 77030 USA
| |
Collapse
|
49
|
Yamada Y, Sato Y, Nakamura T, Harashima H. Innovative cancer nanomedicine based on immunology, gene editing, intracellular trafficking control. J Control Release 2022; 348:357-369. [PMID: 35623492 DOI: 10.1016/j.jconrel.2022.05.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 05/12/2022] [Accepted: 05/20/2022] [Indexed: 11/17/2022]
Abstract
The recent rapid progress in the area of drug delivery systems (DDS) has opened a new era in medicine with a strong linkage to understanding the molecular mechanisms associated with cancer survival. In this review, we summarize new cancer strategies that have recently been developed based on our DDS technology. Cancer immunotherapy will be improved based on the concept of the cancer immunity cycle, which focuses on dynamic interactions between various types of cancer and immune cells in our body. The new technology of genome editing will also be discussed with reference to how these new DDS technologies can be used to introduce therapeutic cargoes into our body. Lastly, a new organelle, mitochondria will be the focus of creating a new cancer treatment strategy by a MITO-Porter which can deliver macromolecules directly to mitochondria of cancer cells via a membrane fusion approach and the impact of controlled intracellular trafficking will be discussed.
Collapse
Affiliation(s)
- Yuma Yamada
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812, Japan; Japan Science and Technology Agency (JST) Fusion Oriented REsearch for disruptive Science and Technology (FOREST) Program, Japan
| | - Yusuke Sato
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812, Japan
| | - Takashi Nakamura
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812, Japan
| | - Hideyoshi Harashima
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812, Japan.
| |
Collapse
|
50
|
Dane EL, Belessiotis-Richards A, Backlund C, Wang J, Hidaka K, Milling LE, Bhagchandani S, Melo MB, Wu S, Li N, Donahue N, Ni K, Ma L, Okaniwa M, Stevens MM, Alexander-Katz A, Irvine DJ. STING agonist delivery by tumour-penetrating PEG-lipid nanodiscs primes robust anticancer immunity. NATURE MATERIALS 2022; 21:710-720. [PMID: 35606429 PMCID: PMC9156412 DOI: 10.1038/s41563-022-01251-z] [Citation(s) in RCA: 130] [Impact Index Per Article: 65.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Accepted: 04/07/2022] [Indexed: 05/13/2023]
Abstract
Activation of the innate immune STimulator of INterferon Genes (STING) pathway potentiates antitumour immunity, but systemic delivery of STING agonists to tumours is challenging. We conjugated STING-activating cyclic dinucleotides (CDNs) to PEGylated lipids (CDN-PEG-lipids; PEG, polyethylene glycol) via a cleavable linker and incorporated them into lipid nanodiscs (LNDs), which are discoid nanoparticles formed by self-assembly. Compared to state-of-the-art liposomes, intravenously administered LNDs carrying CDN-PEG-lipid (LND-CDNs) exhibited more efficient penetration of tumours, exposing the majority of tumour cells to STING agonist. A single dose of LND-CDNs induced rejection of established tumours, coincident with immune memory against tumour rechallenge. Although CDNs were not directly tumoricidal, LND-CDN uptake by cancer cells correlated with robust T-cell activation by promoting CDN and tumour antigen co-localization in dendritic cells. LNDs thus appear promising as a vehicle for robust delivery of compounds throughout solid tumours, which can be exploited for enhanced immunotherapy.
Collapse
Affiliation(s)
- Eric L Dane
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Alexis Belessiotis-Richards
- Department of Materials, Imperial College London, London, UK
- Department of Bioengineering, Imperial College London, London, UK
- Institute of Biomedical Engineering, Imperial College London, London, UK
| | - Coralie Backlund
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Jianing Wang
- Millennium Pharmaceuticals, Inc., Cambridge, MA, USA
| | - Kousuke Hidaka
- Immunology Unit, Takeda Pharmaceutical Company Limited, Fujisawa, Kanagawa, Japan
| | - Lauren E Milling
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Sachin Bhagchandani
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Mariane B Melo
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Shengwei Wu
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Na Li
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Nathan Donahue
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Kaiyuan Ni
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Leyuan Ma
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Masanori Okaniwa
- Oncology Drug Discovery Unit, Takeda Pharmaceuticals International Co., Cambridge, MA, USA
| | - Molly M Stevens
- Department of Materials, Imperial College London, London, UK
- Department of Bioengineering, Imperial College London, London, UK
- Institute of Biomedical Engineering, Imperial College London, London, UK
| | - Alfredo Alexander-Katz
- Department of Materials Science & Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Darrell J Irvine
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Materials Science & Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA.
- Howard Hughes Medical Institute, Chevy Chase, MD, USA.
| |
Collapse
|