1
|
Ji K, Wang L, Liu W, Li G, Lian X, Fan J, Song C, Jian Y. IDO1-mediated M2 macrophage polarization alleviates the progression of ankylosing spondylitis. Autoimmunity 2025; 58:2441134. [PMID: 39692554 DOI: 10.1080/08916934.2024.2441134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 10/31/2024] [Accepted: 12/08/2024] [Indexed: 12/19/2024]
Abstract
Indoleamine 2,3-dioxygenase 1 (IDO1) plays an anti-inflammatory role in autoimmune disease. However, its specific function in ankylosing spondylitis (AS) remain unclear. This study aimed to investigate the potential role of IDO1 in AS. Immunofluorescence, RT-qPCR, and western blot assays were employed to measure gene expression, while ELISA was used to quantify the release of M1 macrophage and M2 macrophage markers. CCK-8, EdU, flow cytometry, ALP staining, and Alizarin red staining (ARS) assays were conducted for functional analysis. JASPAR predicted the binding sites between PPARγ and the promoter, which were further validated by luciferase and ChIP assays. Our findings revealed that the expression of IDO1 was markedly elevated in AS patients. IDO1 overexpression promoted the proliferation of THP-1 cells and M2 macrophage polarization. Conversely, IDO1 knockdown facilitated the osteogenic differentiation of BMSCs. Furthermore, IDO1-mediated upregulation of PPARγ modulated RUNX2 transcription. PPARγ overexpression counteracted the effects of IDO1 knockdown, thereby inhibiting the osteogenic differentiation of BMSCs. In conclusion, the IDO1/PPARγ/RUNX2 signaling pathway may protect against AS by promoting M2 macrophage polarization and inhibiting osteogenic differentiation.
Collapse
Affiliation(s)
- Kangqi Ji
- Department of Spine Surgery, Central Hospital of Xuchang City, Xuchang, China
| | - Lingfei Wang
- Department of Spine Surgery, Central Hospital of Xuchang City, Xuchang, China
| | - Weijie Liu
- Department of Spine Surgery, Central Hospital of Xuchang City, Xuchang, China
| | - Genfeng Li
- Department of Spine Surgery, Central Hospital of Xuchang City, Xuchang, China
| | - Xiaoyu Lian
- Department of Spine Surgery, Central Hospital of Xuchang City, Xuchang, China
| | - Jun Fan
- Department of Spine Surgery, Central Hospital of Xuchang City, Xuchang, China
| | - Chen Song
- Hematology Laboratory, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Yanpeng Jian
- Department of Spine Surgery, Central Hospital of Xuchang City, Xuchang, China
| |
Collapse
|
2
|
Hiraga T. Immune microenvironment of cancer bone metastasis. Bone 2025; 191:117328. [PMID: 39549899 DOI: 10.1016/j.bone.2024.117328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 11/06/2024] [Accepted: 11/08/2024] [Indexed: 11/18/2024]
Abstract
Bone is a common and frequent site of metastasis in cancer patients, leading to a significant reduction in quality of life and increased mortality. Bone marrow, the primary site of hematopoiesis, also serves as both a primary and secondary lymphoid organ. It harbors and supports a diverse array of immune cells, thereby creating a distinct immune microenvironment. These immune cells engage in a range of activities, including anti-tumor, pro-tumor, or a combination of both, which influence the development and progression of bone metastases. Rapid advances in cancer immunotherapy have underscored its potential to eradicate bone metastases. However, clinical outcomes have not yet met expectations. To improve the efficacy of immunotherapy, it is crucial to gain a comprehensive and in-depth understanding of the immune microenvironment within bone metastases. This review provides an overview of the current understanding of the role of different immune cells, their anti-tumor and pro-tumor activities, and their overall contribution to bone metastasis.
Collapse
Affiliation(s)
- Toru Hiraga
- Department of Histology and Cell Biology, Matsumoto Dental University, Shiojiri, Nagano, Japan.
| |
Collapse
|
3
|
Zhang MG, Gallo RA, Tan CH, Camacho M, Fasih-Ahmad S, Moeyersoms AHM, Sayegh Y, Dubovy SR, Pelaez D, Rong AJ. Single-Cell RNA Profiling of Ocular Adnexal Sebaceous Carcinoma Reveals a Complex Tumor Microenvironment and Identifies New Biomarkers. Am J Ophthalmol 2025; 270:8-18. [PMID: 39393421 PMCID: PMC11735305 DOI: 10.1016/j.ajo.2024.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 09/23/2024] [Accepted: 10/03/2024] [Indexed: 10/13/2024]
Abstract
PURPOSE Ocular adnexal sebaceous carcinoma (OaSC) is an aggressive malignancy that often necessitates orbital exenteration. Its tumor composition and transcriptional profile remain largely unknown, which poses a significant barrier to medical advances. Here, we report the first in-depth transcriptomic analysis of OaSC at the single-cell resolution and discern mechanisms underlying cancer progression for the discovery of potential globe-sparing immunotherapies, targeted therapies, and biomarkers to guide clinical management. DESIGN Laboratory investigation with a retrospective observational case series. METHODS Single-cell RNA sequencing was performed on six patient specimens: three primary tumors, two tumors with pagetoid spread, and a normal tarsus sample. Cellular components were identified via gene signatures. Molecular pathways underlying tumorigenesis and pagetoid spread were discerned via gene ontology analysis of the differentially expressed genes between specimens. CALML5 immunohistochemistry was performed on an archival cohort of OaSC, squamous cell carcinoma, ocular surface squamous neoplasia (OSSN), and basal cell carcinoma cases. RESULTS Analysis of 29,219 cells from OaSC specimens revealed tumor, immune, and stromal cells. Tumor-infiltrating immune cells include a diversity of cell types, including exhausted T-cell populations. In primary OaSC tumors, mitotic nuclear division and oxidative phosphorylation pathways are upregulated, while lipid biosynthesis and metabolism pathways are downregulated. Epithelial tissue migration pathways are upregulated in tumor cells undergoing pagetoid spread. Single-cell RNA sequencing analyses also revealed that CALML5 is upregulated in OaSC tumor cells. Diffuse nuclear and cytoplasmic CALML5 staining was present in 28 of 28 (100%) OaSC cases. Diffuse nuclear and membranous CALML5 staining was present in 5 of 25 (20%) squamous cell carcinoma and OSSN cases, while diffuse nuclear staining was present in 1 of 12 (8%) basal cell carcinoma cases. CONCLUSIONS This study reveals a complex OaSC tumor microenvironment and confirms that the CALML5 immunohistochemical stain is a sensitive diagnostic marker.
Collapse
Affiliation(s)
- Michelle G Zhang
- From the Dr. Nasser Ibrahim Al-Rashid Orbital Vision Research Center (M.G.Z., R.A.G., A.H.M., D.P., and A.J.R.), Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, USA; Sylvester Comprehensive Cancer Center (M.G.Z., R.A.G., D.P., and A.J.R.), University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Ryan A Gallo
- From the Dr. Nasser Ibrahim Al-Rashid Orbital Vision Research Center (M.G.Z., R.A.G., A.H.M., D.P., and A.J.R.), Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, USA; Sylvester Comprehensive Cancer Center (M.G.Z., R.A.G., D.P., and A.J.R.), University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Charissa H Tan
- Department of Ophthalmology (C.H.T., M.C., S.F.A., Y.S., and S.R.D.), Florida Lions Ocular Pathology Laboratory, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Matthew Camacho
- Department of Ophthalmology (C.H.T., M.C., S.F.A., Y.S., and S.R.D.), Florida Lions Ocular Pathology Laboratory, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Sohaib Fasih-Ahmad
- Department of Ophthalmology (C.H.T., M.C., S.F.A., Y.S., and S.R.D.), Florida Lions Ocular Pathology Laboratory, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Acadia H M Moeyersoms
- From the Dr. Nasser Ibrahim Al-Rashid Orbital Vision Research Center (M.G.Z., R.A.G., A.H.M., D.P., and A.J.R.), Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, USA; Sylvester Comprehensive Cancer Center (M.G.Z., R.A.G., D.P., and A.J.R.), University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Yoseph Sayegh
- Department of Ophthalmology (C.H.T., M.C., S.F.A., Y.S., and S.R.D.), Florida Lions Ocular Pathology Laboratory, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Sander R Dubovy
- Department of Ophthalmology (C.H.T., M.C., S.F.A., Y.S., and S.R.D.), Florida Lions Ocular Pathology Laboratory, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Daniel Pelaez
- From the Dr. Nasser Ibrahim Al-Rashid Orbital Vision Research Center (M.G.Z., R.A.G., A.H.M., D.P., and A.J.R.), Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, USA; Sylvester Comprehensive Cancer Center (M.G.Z., R.A.G., D.P., and A.J.R.), University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Andrew J Rong
- From the Dr. Nasser Ibrahim Al-Rashid Orbital Vision Research Center (M.G.Z., R.A.G., A.H.M., D.P., and A.J.R.), Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, USA; Sylvester Comprehensive Cancer Center (M.G.Z., R.A.G., D.P., and A.J.R.), University of Miami Miller School of Medicine, Miami, Florida, USA; Division of Oculofacial Plastic, Reconstructive, and Orbital Surgery (A.J.R.), Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, USA.
| |
Collapse
|
4
|
Jeong M, Yoon J, Kim K, Wang J, Koo Y, Sailor MJ, Joo J, Park JH. Programmable Porous Silicon Microparticles for Temporally Staged Drug Delivery in Combination Cancer Immunotherapy. ACS APPLIED MATERIALS & INTERFACES 2025. [PMID: 39847761 DOI: 10.1021/acsami.4c19425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/25/2025]
Abstract
Combination therapies using checkpoint inhibitors with immunostimulatory agonists have attracted great attention due to their synergistic therapeutic effects for cancer treatment. However, such combination immunotherapies require specific timing of doses to show sufficient antitumor efficacy. Sequential treatment usually requires multiple administrations of the individual drugs at specific time points, thus increasing the complexity of the drug regimen and compromising patient compliance. Here, we introduce an injectable porous silicon microparticle (pSiMP) for combination cancer immunotherapy where its multilayered nanopore structure was electrochemically programmed to achieve release of three distinct immunomodulatory drugs in the right sequence at the desired time. We find the optimal sequential treatment timeline of stimulator of interferon genes (STING) agonist, anti-OX40 antibody (aOX40), and anti-PD-1 antibody (aPD-1) for immunosuppressive tumors. We show that a single intratumoral injection of a cocktail of release-programmed pSiMPs coloaded with each antibody and a STING agonist significantly suppresses the tumor growth compared to conventional treatment involving sequential bolus injections, or an injection of pSiMPs configured to release all drugs at the same time, with no delay. With the timely release of immunomodulatory drugs, the programmable pSiMPs offer an effective treatment strategy for combination immunotherapy.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Jinmyoung Joo
- Center for Genomic Integrity, Institute for Basic Science, Ulsan 44919, Republic of Korea
| | | |
Collapse
|
5
|
Bian X, Zhou L, Luo Z, Liu G, Hang Z, Li H, Li F, Wen Y. Emerging Delivery Systems for Enabling Precision Nucleic Acid Therapeutics. ACS NANO 2025. [PMID: 39834294 DOI: 10.1021/acsnano.4c11858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Nucleic acid therapeutics represent a highly promising treatment approach in modern medicine, treating diseases at the genetic level. However, these therapeutics face numerous challenges in practical applications, particularly regarding their stability, effectiveness, cellular uptake efficiency, and limitations in delivering them specifically to target tissues. To overcome these obstacles, researchers have developed various innovative delivery systems, including viral vectors, lipid nanoparticles, polymer nanoparticles, inorganic nanoparticles, protein carriers, exosomes, antibody oligonucleotide conjugates, and DNA nanostructure-based delivery systems. These systems enhance the therapeutic efficacy of nucleic acid drugs by improving their stability, targeting specificity, and half-life in vivo. In this review, we systematically discuss different types of nucleic acid drugs, analyze the major barriers encountered in their delivery, and summarize the current research progress in emerging delivery systems. We also highlight the latest advancements in the application of these systems for treating genetic diseases, infectious diseases, cancer, brain diseases, and wound healing. This review aims to provide a comprehensive overview of nucleic acid drug delivery systems' current status and future directions by integrating the latest advancements in nanotechnology, biomaterials science, and gene editing technologies, emphasizing their transformative potential in precision medicine.
Collapse
Affiliation(s)
- Xiaochun Bian
- Beijing Key Laboratory for Bioengineering and Sensing Technology, Daxing Research Institute, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Liping Zhou
- Beijing Key Laboratory for Bioengineering and Sensing Technology, Daxing Research Institute, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Zhiwei Luo
- Beijing Key Laboratory for Bioengineering and Sensing Technology, Daxing Research Institute, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Guotao Liu
- Beijing Key Laboratory for Bioengineering and Sensing Technology, Daxing Research Institute, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Zhongci Hang
- Beijing Key Laboratory for Bioengineering and Sensing Technology, Daxing Research Institute, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Haohao Li
- Beijing Key Laboratory for Bioengineering and Sensing Technology, Daxing Research Institute, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Fengyong Li
- Plastic Surgery Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Yongqiang Wen
- Beijing Key Laboratory for Bioengineering and Sensing Technology, Daxing Research Institute, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China
| |
Collapse
|
6
|
Nie J, Yang N, Sun S, Wang L, Pei Z, Wu J, Yu Q, Han Z, Chen Y, Cheng L. Antimony Component Schottky Nanoheterojunctions as Ultrasound-Heightened Pyroptosis Initiators for Sonocatalytic Immunotherapy. Angew Chem Int Ed Engl 2025; 64:e202416426. [PMID: 39305135 DOI: 10.1002/anie.202416426] [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: 08/27/2024] [Indexed: 11/08/2024]
Abstract
Pyroptosis, an inflammatory modality of programmed cell death associated with the immune response, can be initiated by bioactive ions and reactive oxygen species (ROS). However, bioactive ion-induced pyroptosis lacks specificity, and further exploration of other ions that can induce pyroptosis in cancer cells is needed. Sonocatalytic therapy (SCT) holds promise due to its exceptional penetration depth; however, the rapid recombination of electron-hole (e--h+) pairs and the complex tumor microenvironment (TME) impede its broader application. Herein, we discovered that antimony (Sb)-based nanomaterials induced pyroptosis in cancer cells. Therefore, Schottky heterojunctions containing Sb component (Sb2Se3@Pt) were effectively designed and constructed via in situ growth of platinum (Pt) nanoparticles (NPs) on Sb2Se3 semiconductor with narrow band gaps, which were utilized as US-heightened pyroptosis initiators to induce highly effective pyroptosis in cancer cells to boost SCT-immunotherapy. Under US irradiation, excited electrons were transferred from Sb2Se3 nanorods (NRs) to the co-catalyst Pt via Schottky junctions, and band bending effectively prevented electron backflow and achieved efficient ROS generation. Moreover, the pores oxidized and depleted the overexpressed GSH in the TME, potentially amplifying ROS generation. The biological effects of the Sb2Se3@Pt nanoheterojunction itself combined with the sonocatalytic amplification of oxidative stress significantly induced Caspase-1/GSDMD-dependent pyroptosis in cancer cells. Therefore, SCT treatment with Sb2Se3@Pt not only effectively restrained tumor proliferation but also induced potent immune memory responses and suppressed tumor recurrence. Furthermore, the integration of this innovative strategy with immune checkpoint blockade (ICB) treatment elicited a systemic immune response, effectively augmenting therapeutic effects and impeding the growth of abscopal tumors. Overall, this study provides further opportunities to explore pyroptosis-mediated SCT-immunotherapy.
Collapse
Affiliation(s)
- Jihu Nie
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, China
| | - Nailin Yang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, China
- Macao Institute of Materials Science and Engineering, Macau University of Science and Technology, Taipa, 999078, Macau SAR, China
| | - Shumin Sun
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, China
| | - Li Wang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, China
| | - Zifan Pei
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, China
| | - Jie Wu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, China
| | - Qiao Yu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, China
| | - Zhihui Han
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, China
| | - Youdong Chen
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, China
| | - Liang Cheng
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, China
- Macao Institute of Materials Science and Engineering, Macau University of Science and Technology, Taipa, 999078, Macau SAR, China
| |
Collapse
|
7
|
Yu Q, Sun S, Yang N, Pei Z, Chen Y, Nie J, Lei H, Wang L, Gong F, Cheng L. Self-Cascaded Pyroptosis-STING Initiators for Catalytic Metalloimmunotherapy. J Am Chem Soc 2025. [PMID: 39818788 DOI: 10.1021/jacs.4c12552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2025]
Abstract
Gasdermin (GSDM)-mediated pyroptosis involves the induction of mitochondrial damage and the subsequent release of mitochondrial DNA (mtDNA), which is anticipated to activate the cGAS-STING pathway, thereby augmenting the antitumor immune response. However, challenges lie in effectively triggering pyroptosis in cancer cells and subsequently enhancing the cGAS-STING activation with specificity. Herein, we developed intelligent self-cascaded pyroptosis-STING initiators of cobalt fluoride (CoF2) nanocatalysts for catalytic metalloimmunotherapy. CoF2 nanocatalysts with a semiconductor structure and enzyme-like activity generated a substantial amount of reactive oxygen species (ROS) under stimulation by endogenous H2O2 and exogenous ultrasound. Importantly, we discovered that Co-based nanomaterials themselves induce pyroptosis in cancer cells. Therefore, CoF2 nanocatalysts initially acted as pyroptosis inducers, triggering caspase-1/GSDMD-dependent pyroptosis in cancer cells via Co2+ and ROS, leading to mtDNA release. Subsequently, CoF2 nanocatalysts were further utilized as intelligent STING agonists that were specifically capable of detecting mtDNA and augmenting the activation of the cGAS-STING pathway. These cascade events triggered a robust immune response, effectively modulating the immunosuppressive tumor microenvironment into an immune-supportive state, thereby providing favorable support for antitumor therapy. This innovative strategy not only significantly impeded the growth of the primary tumor but also elicited an immune response to further augment the efficacy of immune checkpoint inhibitors in preventing distant tumor progression. Overall, this study proposed a self-cascade strategy for activating and amplifying the cGAS-STING pathway with specificity mediated by pyroptosis, representing a valuable avenue for future cancer catalytic metalloimmunotherapy.
Collapse
Affiliation(s)
- Qiao Yu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou 215123, China
- State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang Provincial Key Laboratory for Advanced Drug Delivery Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Shumin Sun
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou 215123, China
| | - Nailin Yang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou 215123, China
- Macao Institute of Materials Science and Engineering, Macau University of Science and Technology, Taipa, Macau SAR 999078, China
| | - Zifan Pei
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou 215123, China
| | - Youdong Chen
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou 215123, China
| | - Jihu Nie
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou 215123, China
| | - Huali Lei
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou 215123, China
| | - Li Wang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou 215123, China
| | - Fei Gong
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou 215123, China
| | - Liang Cheng
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou 215123, China
- Macao Institute of Materials Science and Engineering, Macau University of Science and Technology, Taipa, Macau SAR 999078, China
| |
Collapse
|
8
|
Deng Z, Li L, Meng Z, Zeng G, Cao R, Liu R. Pan-cancer analysis shows that TNFSF4 is a potential prognostic and immunotherapeutic biomarker for multiple cancer types including liver cancer. BMC Cancer 2025; 25:100. [PMID: 39825223 PMCID: PMC11740393 DOI: 10.1186/s12885-025-13479-4] [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: 04/12/2024] [Accepted: 01/08/2025] [Indexed: 01/20/2025] Open
Abstract
BACKGROUND As a member of the tumor necrosis factor (TNF) superfamily, tumor necrosis factor superfamily member 4 (TNFSF4) is expressed on antigen-presenting cells and activated T cells by binding to its receptor TNFRSF4. However, tumorigenicity of TNFSF4 has not been studied in pan-cancer. Therefore, comprehensive bioinformatics analysis of pan-cancer was performed to determine the mechanisms through which TNFSF4 regulates tumorigenesis. METHODS RNA-seq data for 33 cancers were analyzed from UCSC XENA database. Online websites and databases were used to investigate TNFSF4's biological function, epigenetic modifications, genetic alterations, and tumor immunity. Furthermore, cell phenotype experiment and tumor xenotransplantation experiment were performed to determine the biological functions of TNFSF4. RESULTS The pan-cancer analysis showed that TNFSF4 was upregulated in several tumors. Significant relationships between TNFSF4 expression and single-cells data were also observed in numerous cancer types. TNFSF4 expression correlated with the expression of immune checkpoint genes and could influence various drug sensitivity. Vitro and vivo experiments showed that TNFSF4 could promote the development and progression of Hepatocellular Carcinoma (HCC). CONCLUSIONS TNFSF4 was upregulated in multiple cancer types and promoted the development and progression of cancers through several mechanisms including regulation of the tumor-infiltration of immune cells. Our study shows that TNFSF4 is a promising prognostic and immunotherapeutic biomarker in some malignant tumors.
Collapse
Affiliation(s)
- Zhaoda Deng
- Medical School of Chinese PLA, Beijing, China
- Faculty of Hepato-Pancreato-Biliary Surgery, The First Medical Center, Chinese PLA General Hospital, Institute of Hepatobiliary Surgery of Chinese PLA, Beijing, China
- Key Laboratory of Digital Hepatobiliary Surgery, PLA, Beijing, China
| | - Lincheng Li
- Department of Surgery, Second Mobile Corps Hospital of Chinese People's Armed Police Force, Wuxi, China
| | - Zihe Meng
- Inner Mongolia Medical University, Hohhot, China
| | - Guineng Zeng
- Faculty of Hepato-Pancreato-Biliary Surgery, The First Medical Center, Chinese PLA General Hospital, Institute of Hepatobiliary Surgery of Chinese PLA, Beijing, China
- Key Laboratory of Digital Hepatobiliary Surgery, PLA, Beijing, China
- Nankai University School of Medicine, Nankai University, Tianjin, 300300, China
| | - Rui Cao
- Inner Mongolia Medical University, Hohhot, China
| | - Rong Liu
- Medical School of Chinese PLA, Beijing, China.
- Faculty of Hepato-Pancreato-Biliary Surgery, The First Medical Center, Chinese PLA General Hospital, Institute of Hepatobiliary Surgery of Chinese PLA, Beijing, China.
- Key Laboratory of Digital Hepatobiliary Surgery, PLA, Beijing, China.
| |
Collapse
|
9
|
Wang F, Zhao Q, Liu W, Zhang D, Dai X, Zhou W, Zeng X, Zhang Y, Cheng L, Shen G, Gu Y. A humanized anti-b7h3×4-1BB bispecific antibody exerts potent antitumour effects through the activation of innate and adaptive immunity. Biochem Biophys Res Commun 2025; 749:151347. [PMID: 39847994 DOI: 10.1016/j.bbrc.2025.151347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 01/10/2025] [Accepted: 01/15/2025] [Indexed: 01/25/2025]
Abstract
Agonistic monoclonal antibodies targeting 4-1BB have shown much preclinical promise, but their clinical development has been limited by obvious toxicity or unremarkable efficacy. Here, we generated two humanized anti-B7H3 × 4-1BB bsAbs (HK056-001/002) by fusing an anti-4-1BB scFv to the C-terminus of an anti-B7H3 with an intact Fc fragment from human IgG1 or IgG4. The two bsAbs were able to stimulate the 4-1BB signaling pathway, which was strictly dependent on B7H3 expression. In particular, HK056-001 retained Fc function and induced an ADCC effect in tumor cells, whereas HK056-002 did not. Strikingly, HK056-001 showed superior antitumour activity to HK056-002 both in vitro and in vivo. HK056-001 enhanced antitumour immunity and induced lasting antigen-specific immune memory to prevent tumor regrowth upon rechallenge, even at a dose as low as 2 mg/kg. Furthermore, HK056-001 did not induce nonspecific production of proinflammatory cytokines and had no apparent ability to induce ADA production. In addition, HK056-001 has no significant liver toxicity in human 4-1BB-KI BALB/c mice bearing CT26-B7H3 tumors. The optimal anti-B7H3 × 4-1BB bsAb HK056-001 exhibited synergistic antitumour effects by inducing an ADCC effect (innate immunity) and activating the 4-1BB signaling pathway (adaptive immunity) upon cross-bridging with B7H3 with no obvious toxicity, which could potentially provide a better therapeutic window compared to what is seen with 4-1BB agonists.
Collapse
Affiliation(s)
- Fengrong Wang
- Department of Pharmacology, Life Science and Biopharmaceutical Institution, Shenyang Pharmaceutical University, Shenyang, 110016, Liaoning Province, China; Hefei HankeMab Biotechnology Co., Ltd., Anhui Province Key Laboratory of Gene Engineering Pharmaceutical, Biomedicine Technology Innovation Center of Hefei, Anhui Anke Biotechnology (Group) Co., Ltd., Hefei, 230088, Anhui, China
| | - Qun Zhao
- Hefei HankeMab Biotechnology Co., Ltd., Anhui Province Key Laboratory of Gene Engineering Pharmaceutical, Biomedicine Technology Innovation Center of Hefei, Anhui Anke Biotechnology (Group) Co., Ltd., Hefei, 230088, Anhui, China
| | - Wenting Liu
- Hefei HankeMab Biotechnology Co., Ltd., Anhui Province Key Laboratory of Gene Engineering Pharmaceutical, Biomedicine Technology Innovation Center of Hefei, Anhui Anke Biotechnology (Group) Co., Ltd., Hefei, 230088, Anhui, China
| | - Dayan Zhang
- Hefei HankeMab Biotechnology Co., Ltd., Anhui Province Key Laboratory of Gene Engineering Pharmaceutical, Biomedicine Technology Innovation Center of Hefei, Anhui Anke Biotechnology (Group) Co., Ltd., Hefei, 230088, Anhui, China
| | - Xuejing Dai
- Hefei HankeMab Biotechnology Co., Ltd., Anhui Province Key Laboratory of Gene Engineering Pharmaceutical, Biomedicine Technology Innovation Center of Hefei, Anhui Anke Biotechnology (Group) Co., Ltd., Hefei, 230088, Anhui, China
| | - Weiming Zhou
- Hefei HankeMab Biotechnology Co., Ltd., Anhui Province Key Laboratory of Gene Engineering Pharmaceutical, Biomedicine Technology Innovation Center of Hefei, Anhui Anke Biotechnology (Group) Co., Ltd., Hefei, 230088, Anhui, China
| | - Xiaoli Zeng
- Hefei HankeMab Biotechnology Co., Ltd., Anhui Province Key Laboratory of Gene Engineering Pharmaceutical, Biomedicine Technology Innovation Center of Hefei, Anhui Anke Biotechnology (Group) Co., Ltd., Hefei, 230088, Anhui, China
| | - Yan Zhang
- Hefei HankeMab Biotechnology Co., Ltd., Anhui Province Key Laboratory of Gene Engineering Pharmaceutical, Biomedicine Technology Innovation Center of Hefei, Anhui Anke Biotechnology (Group) Co., Ltd., Hefei, 230088, Anhui, China
| | - Liansheng Cheng
- Hefei HankeMab Biotechnology Co., Ltd., Anhui Province Key Laboratory of Gene Engineering Pharmaceutical, Biomedicine Technology Innovation Center of Hefei, Anhui Anke Biotechnology (Group) Co., Ltd., Hefei, 230088, Anhui, China.
| | - Guodong Shen
- Anhui Province Key Laboratory of Geriatric Immunotherapy and Nutrition Therapy, Gerontology Institute of Anhui Province, Hefei, 230001, Anhui, China; Department of Geriatrics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China.
| | - Yanting Gu
- Department of Pharmacology, Life Science and Biopharmaceutical Institution, Shenyang Pharmaceutical University, Shenyang, 110016, Liaoning Province, China.
| |
Collapse
|
10
|
Cheng J, Xiao Y, Peng T, Zhang Z, Qin Y, Wang Y, Shi J, Yan J, Zhao Z, Zheng L, He Z, Wang J, Zhang Z, Li C, Zhu H, Jiang P. ETV7 limits the antiviral and antitumor efficacy of CD8 + T cells by diverting their fate toward exhaustion. NATURE CANCER 2025:10.1038/s43018-024-00892-0. [PMID: 39805956 DOI: 10.1038/s43018-024-00892-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 12/06/2024] [Indexed: 01/16/2025]
Abstract
Terminal exhaustion is a critical barrier to antitumor immunity. By integrating and analyzing single-cell RNA-sequencing and single-cell assay for transposase-accessible chromatin with sequencing data, we found that ETS variant 7 (ETV7) is indispensable for determining CD8+ T cell fate in tumors. ETV7 introduction drives T cell differentiation from memory to terminal exhaustion, limiting antiviral and antitumor efficacy in male mice. Mechanistically, ETV7 acts as a central transcriptional node by binding to specific memory genes and exhaustion genes and functionally skewing these transcriptional programs toward exhaustion. Clinically, ETV7 expression is negatively correlated with progression and responsiveness to immune checkpoint blockade in various human cancers. ETV7 depletion strongly enhances the antitumor efficacy of CD8+ T cells and engineered chimeric antigen receptor T cells in solid tumors. Thus, these findings demonstrate a decisive role for ETV7 in driving CD8+ T cell terminal exhaustion and reveal that ETV7 may be a promising target and biomarker for improving the efficacy of cancer immunotherapy.
Collapse
Affiliation(s)
- Jie Cheng
- State Key Laboratory of Molecular Oncology, School of Life Sciences, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China
- Department of Pathology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, China
| | - Yifeng Xiao
- State Key Laboratory of Molecular Oncology, School of Life Sciences, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China
| | - Ting Peng
- State Key Laboratory of Molecular Oncology, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Molecular Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Zijian Zhang
- Institute of Biology and Medicine, College of Life and Health Sciences, Wuhan University of Science and Technology, Wuhan, China
| | - You Qin
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuqian Wang
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
| | - Jiangzhou Shi
- Institute of Biology and Medicine, College of Life and Health Sciences, Wuhan University of Science and Technology, Wuhan, China
| | - Jinxin Yan
- State Key Laboratory of Molecular Oncology, School of Life Sciences, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China
| | - Zihao Zhao
- School of Life Sciences, Center for Statistical Science, Peking University, Beijing, China
| | - Liangtao Zheng
- School of Life Sciences, Center for Statistical Science, Peking University, Beijing, China
| | - Zhijun He
- State Key Laboratory of Molecular Oncology, School of Life Sciences, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China
| | - Jianwei Wang
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
| | - Zemin Zhang
- School of Life Sciences, Center for Statistical Science, Peking University, Beijing, China.
| | - Cheng Li
- School of Life Sciences, Center for Statistical Science, Peking University, Beijing, China.
| | - Haichuan Zhu
- Institute of Biology and Medicine, College of Life and Health Sciences, Wuhan University of Science and Technology, Wuhan, China.
| | - Peng Jiang
- State Key Laboratory of Molecular Oncology, School of Life Sciences, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China.
| |
Collapse
|
11
|
Mao J, Zhou L, Wu Y, Wang K, Ye X, Wang T, Yang J, Tong J, Miao Q, Jiang S, Xiao Y, Zhang K. Discovery of 1,2,4-benzotriazine derivatives as new hematopoietic progenitor kinase 1 (HPK1) inhibitors. Bioorg Chem 2025; 156:108158. [PMID: 39826501 DOI: 10.1016/j.bioorg.2025.108158] [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: 10/25/2024] [Revised: 12/26/2024] [Accepted: 01/08/2025] [Indexed: 01/22/2025]
Abstract
Hematopoietic progenitor kinase 1 (HPK1), which negatively regulates immune signaling, has emerged as an attractive small-molecule drug target for tumor immunotherapy. Herein, we report the discovery of the 1,2,4-benzotriazine derivatives as new potent HPK1 inhibitors. Notably, compound A29 exhibited improved HPK1 inhibitory activity relative to compound 1 in the ADP-Glo kinase assay (IC50 = 2.70 and 13.6 nM, respectively). The pronounced inhibitory activity of A29 against downstream p-SLP76 in Jurkat T cells (IC50 = 8.1 nM) as well as the ability to induce the production of interleukin 2 (IL-2) in human peripheral blood mononuclear cells (PBMCs) confirmed its cellular target engagement and immune stimulatory effect. Consistently, this lead compound significantly enhanced T-cell killing ability against murine colon cancer cells CT26 or MC38 in a co-culture system. Furthermore, A29 was efficacious in a CT26 xenograft mouse model alone, and significantly enhanced the antitumor efficacy of an anti-PD-1 antibody. This work provides a promising lead for the development of effective HPK1 inhibitors for tumor immunotherapy.
Collapse
Affiliation(s)
- Jie Mao
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Lixin Zhou
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China; Department of Hematology, Tongji Hospital, Frontier Science Center for Stem Cell Research, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Yuxing Wu
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Kaizhen Wang
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Xiuquan Ye
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Tianyu Wang
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Jiamei Yang
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Jun Tong
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Qi Miao
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Sheng Jiang
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China.
| | - Yibei Xiao
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China.
| | - Kuojun Zhang
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
12
|
Tomita K, Yamashita M, Ikegami K, Shimizu Y, Amino N, Nakao S. Combination with oxaliplatin improves abscopal effect of oncolytic virotherapy through reorganization of intratumoral macrophages. Mol Ther 2025; 33:401-414. [PMID: 39663702 DOI: 10.1016/j.ymthe.2024.12.007] [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: 05/14/2024] [Revised: 10/18/2024] [Accepted: 12/06/2024] [Indexed: 12/13/2024] Open
Abstract
Intratumoral administration is a widely used method for oncolytic virotherapy, as it enables immediate access of virus particles to the target tumor and potentially lead to the suppression of untreated distant tumors via in situ vaccination. However, because the injection volume and concentration of the virus solution are physically limited, the dose level cannot be increased. Additionally, efficacy in distant tumors needs improvement to prolong patient survival. Here, we demonstrate the benefit of oxaliplatin, with detailed mechanisms revealed through transcriptome analysis, which may provide a solution for the crucial deficiencies of oncolytic virotherapy. In virus-injected tumors, oxaliplatin improved virus retention through suppression of type I interferons. In distant virus-naive tumors, oxaliplatin induced alterations in the intratumoral macrophage characteristics, leading to the chemotaxis and recruitment of activated T cells and subsequently inducing an inflammatory state in the non-injected tumors. Our findings can be a trigger to change the therapeutic paradigm of oncolytic virotherapy for patients with systemic metastases.
Collapse
Affiliation(s)
- Kyoko Tomita
- Immuno-Oncology, Astellas Pharma Inc., Tsukuba, Japan
| | | | | | | | - Nobuaki Amino
- Immuno-Oncology, Astellas Pharma Inc., Tsukuba, Japan
| | | |
Collapse
|
13
|
Markova E, Wolowczyk C, Mohamed A, Sofias AM, Martin-Armas M, Sundset R, Berndtsson J, Hak S, Škalko-Basnet N. Liposomal Nω-hydroxy-l-norarginine, a proof-of-concept: Arginase inhibitors can be incorporated in liposomes while retaining their therapeutic activity ex vivo. Eur J Pharm Sci 2025; 204:106959. [PMID: 39521192 DOI: 10.1016/j.ejps.2024.106959] [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: 06/07/2024] [Revised: 10/02/2024] [Accepted: 11/07/2024] [Indexed: 11/16/2024]
Abstract
Cancer immunotherapy has evolved significantly over the last decade, with therapeutics targeting the adaptive immune system showing exciting effects in clinics. Yet, the modulation of the innate immune system, particularly the tumor-associated innate immune cells which are an integral part of immune responses in cancer, remains less understood. The arginase 1 (Arg1) pathway is a pivotal metabolic pathway that tumor-associated innate immune cells exploit to create an immunosuppressive tumor microenvironment, leading to the evasion of immune surveillance. The inhibition of Arg1 presents a therapeutic opportunity to reverse this immunosuppression, and Nω‑hydroxy-l-norarginine (nor-NOHA) has emerged as a potent arginase inhibitor with promising in vivo efficacy. However, the rapid systemic clearance of nor-NOHA poses a significant challenge for its therapeutic application. This study pioneers the encapsulation of nor-NOHA in liposomes, aiming to enhance its bioavailability and prolong its inhibitory activity against Arg1. Historically, the extensive interaction between innate immune cells and nanoparticles has been one of the biggest drawbacks in nanomedicine. Here we seek to utilize this effect and deliver liposomal nor-NOHA to the arginase 1 expressing innate immune cells. We systematically investigated the effect of lipid composition, acyl chain length, manufacturing and loading methodology on the encapsulation efficiency (EE%) and release profile of nor-NOHA. Our results indicate that while the manufacturing method and lipid acyl chain length do not significantly impact EE%, they crucially influence the release kinetics of nor-NOHA, with longer acyl chains demonstrating a more sustained release of nor-NOHA from liposomes enabling continuous inhibition of Arg1. Our findings suggest that liposomal nor-NOHA retains its functional inhibitory activity and could offer improved pharmacokinetic properties, making it a compelling base for iterations for further innovative cancer immunotherapeutic strategies in preclinical and clinical evaluations.
Collapse
Affiliation(s)
- Elena Markova
- Nuclear Medicine and Radiation Biology Research Group, Department of Clinical Medicine, Faculty of Health Science, UiT The Arctic University of Norway, Tromsø, Norway; PET Imaging Center Tromsø, University Hospital of North Norway (UNN), Tromsø, Norway.
| | - Camilla Wolowczyk
- Department of Biomedical Laboratory Science, Faculty of Natural Sciences & Centre of Molecular Inflammation Research (CEMIR), Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | - Aly Mohamed
- Drug Transport and Delivery Research Group, Department of Pharmacy, Faculty of Health Science, UiT The Arctic University of Norway, Tromsø, Norway
| | - Alexandros Marios Sofias
- Institute for Experimental Molecular Imaging (ExMI), RWTH Aachen University Hospital, Aachen, Germany
| | - Montserrat Martin-Armas
- Nuclear Medicine and Radiation Biology Research Group, Department of Clinical Medicine, Faculty of Health Science, UiT The Arctic University of Norway, Tromsø, Norway; PET Imaging Center Tromsø, University Hospital of North Norway (UNN), Tromsø, Norway
| | - Rune Sundset
- Nuclear Medicine and Radiation Biology Research Group, Department of Clinical Medicine, Faculty of Health Science, UiT The Arctic University of Norway, Tromsø, Norway; PET Imaging Center Tromsø, University Hospital of North Norway (UNN), Tromsø, Norway
| | - Jens Berndtsson
- Centre for Cellular Imaging, Core Facility, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Sjoerd Hak
- Department of Biotechnology and Nanomedicine, SINTEF Industry & Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway
| | - Nataša Škalko-Basnet
- Drug Transport and Delivery Research Group, Department of Pharmacy, Faculty of Health Science, UiT The Arctic University of Norway, Tromsø, Norway.
| |
Collapse
|
14
|
Wang J, Hu X, Wang Y, A R, Li X, Sun Y, Guan Z, Li X, Wu Y, Wang J, Zhao F, Liu Y, Wang H, Yu H, Wang T, Zhu M, Li X, Zhang D, Chen W, Han Z, Sun X. Development and characterisation of [ 18F]TTDP, a novel T cell immunoglobulin and ITIM domain tracer, in humanised mice and non-human primates. Eur J Nucl Med Mol Imaging 2025; 52:416-426. [PMID: 39297961 DOI: 10.1007/s00259-024-06911-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 08/28/2024] [Indexed: 09/21/2024]
Abstract
PURPOSE The T cell immunoglobulin and ITIM domain (TIGIT) blockade immunotherapy response is directly associated with individual differences of TIGIT expression on tumour-infiltrating lymphocytes (TILs) in tumour immune microenvironment (TIME) of non-small cell lung cancer (NSCLC). Here, we developed a TIGIT-targeted PET tracer to evaluate its feasibility in predicting immunotherapy efficacy, aiming to manage NSCLC patients accurately. METHODS We synthesised a 18F-labeled TIGIT-targeted D-peptide, [18F]TTDP, and investigated the specificity of [18F]TTDP both to murine TIGIT and human TIGIT by a series of in vitro and in vivo assays. [18F]TTDP PET imaging was performed in humanised immune system (HIS) mice models bearing NSCLC patient-derived xenografts (PDXs) to evaluate the predictive value of FDA-approved combination immunotherapy of atezolizumab plus tiragolumab. Lastly, rhesus macaque was applied for [18F] TTDP PET to explore the tracer's in vivo distribution and translational potential in non-human primates. RESULTS [18F]TTDP showed high specificity for both murine TIGIT and human TIGIT in vitro and in vivo. The HIS NSCLC PDX platform was successfully established for [18F]TTDP PET imaging, and tumour uptake of [18F]TTDP was significantly correlated with the TIGIT expression of TILs in the TIME. [18F]TTDP PET imaging, in predicting treatment response to the combination immunotherapy in NSCLC HIS-PDX models, showed a sensitivity of 83.33% and a specificity of 100%. In addition, [18F]TTDP PET also showed cross-species consistency of the tracer biodistribution between non-human primate and murine animals, and no adverse events were observed. CONCLUSION The combined implementation of the [18F]TTDP and HIS-PDX model creates a state-of-the-art preclinical platform that will impact the identification and validation of TIGIT-targeted PET image-guided diagnosis, treatment response prediction, beneficial patient screening, novel immunotherapies, and ultimately the outcome of NSCLC patients. We first provided in vivo biodistribution of [18F]TTDP PET imaging in rhesus macaque, indicating its excellent translational potential in the clinic.
Collapse
Affiliation(s)
- Jing Wang
- Department of Nuclear Medicine, the Fourth Affiliated Hospital of Harbin Medical University, Xiangan N Street, Songbei District, Harbin, 150028, Heilongjiang, China
- NHC Key Laboratory of Molecular Probe and Targeted Diagnosis and Therapy, Molecular Imaging Research Center (MIRC) of Harbin Medical University, Harbin, Heilongjiang, China
| | - Xinxin Hu
- Department of Nuclear Medicine, the Fourth Affiliated Hospital of Harbin Medical University, Xiangan N Street, Songbei District, Harbin, 150028, Heilongjiang, China
- NHC Key Laboratory of Molecular Probe and Targeted Diagnosis and Therapy, Molecular Imaging Research Center (MIRC) of Harbin Medical University, Harbin, Heilongjiang, China
| | - Yueqi Wang
- Department of Nuclear Medicine & Laboratory of Clinical Nuclear Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Rong A
- Department of Nuclear Medicine, the Fourth Affiliated Hospital of Harbin Medical University, Xiangan N Street, Songbei District, Harbin, 150028, Heilongjiang, China
- NHC Key Laboratory of Molecular Probe and Targeted Diagnosis and Therapy, Molecular Imaging Research Center (MIRC) of Harbin Medical University, Harbin, Heilongjiang, China
| | - Xiaoqian Li
- Department of Nuclear Medicine, the Fourth Affiliated Hospital of Harbin Medical University, Xiangan N Street, Songbei District, Harbin, 150028, Heilongjiang, China
- NHC Key Laboratory of Molecular Probe and Targeted Diagnosis and Therapy, Molecular Imaging Research Center (MIRC) of Harbin Medical University, Harbin, Heilongjiang, China
| | - Ying Sun
- Department of Nuclear Medicine, the Fourth Affiliated Hospital of Harbin Medical University, Xiangan N Street, Songbei District, Harbin, 150028, Heilongjiang, China
- NHC Key Laboratory of Molecular Probe and Targeted Diagnosis and Therapy, Molecular Imaging Research Center (MIRC) of Harbin Medical University, Harbin, Heilongjiang, China
| | - Zhengqi Guan
- Department of Nuclear Medicine, the Fourth Affiliated Hospital of Harbin Medical University, Xiangan N Street, Songbei District, Harbin, 150028, Heilongjiang, China
- NHC Key Laboratory of Molecular Probe and Targeted Diagnosis and Therapy, Molecular Imaging Research Center (MIRC) of Harbin Medical University, Harbin, Heilongjiang, China
| | - Xiaona Li
- Department of Nuclear Medicine, the Fourth Affiliated Hospital of Harbin Medical University, Xiangan N Street, Songbei District, Harbin, 150028, Heilongjiang, China
- NHC Key Laboratory of Molecular Probe and Targeted Diagnosis and Therapy, Molecular Imaging Research Center (MIRC) of Harbin Medical University, Harbin, Heilongjiang, China
| | - Yongyi Wu
- Department of Nuclear Medicine, the Fourth Affiliated Hospital of Harbin Medical University, Xiangan N Street, Songbei District, Harbin, 150028, Heilongjiang, China
- NHC Key Laboratory of Molecular Probe and Targeted Diagnosis and Therapy, Molecular Imaging Research Center (MIRC) of Harbin Medical University, Harbin, Heilongjiang, China
| | - Jiannan Wang
- Department of Nuclear Medicine, the Fourth Affiliated Hospital of Harbin Medical University, Xiangan N Street, Songbei District, Harbin, 150028, Heilongjiang, China
- NHC Key Laboratory of Molecular Probe and Targeted Diagnosis and Therapy, Molecular Imaging Research Center (MIRC) of Harbin Medical University, Harbin, Heilongjiang, China
| | - Fangyu Zhao
- Department of Nuclear Medicine, the Fourth Affiliated Hospital of Harbin Medical University, Xiangan N Street, Songbei District, Harbin, 150028, Heilongjiang, China
- NHC Key Laboratory of Molecular Probe and Targeted Diagnosis and Therapy, Molecular Imaging Research Center (MIRC) of Harbin Medical University, Harbin, Heilongjiang, China
| | - Yang Liu
- Department of Nuclear Medicine, the Fourth Affiliated Hospital of Harbin Medical University, Xiangan N Street, Songbei District, Harbin, 150028, Heilongjiang, China
- NHC Key Laboratory of Molecular Probe and Targeted Diagnosis and Therapy, Molecular Imaging Research Center (MIRC) of Harbin Medical University, Harbin, Heilongjiang, China
| | - Hongbin Wang
- Department of Nuclear Medicine, the Fourth Affiliated Hospital of Harbin Medical University, Xiangan N Street, Songbei District, Harbin, 150028, Heilongjiang, China
- NHC Key Laboratory of Molecular Probe and Targeted Diagnosis and Therapy, Molecular Imaging Research Center (MIRC) of Harbin Medical University, Harbin, Heilongjiang, China
| | - Hong Yu
- Department of Nuclear Medicine, the Fourth Affiliated Hospital of Harbin Medical University, Xiangan N Street, Songbei District, Harbin, 150028, Heilongjiang, China
- NHC Key Laboratory of Molecular Probe and Targeted Diagnosis and Therapy, Molecular Imaging Research Center (MIRC) of Harbin Medical University, Harbin, Heilongjiang, China
| | - Tianyi Wang
- Department of Nuclear Medicine, the Fourth Affiliated Hospital of Harbin Medical University, Xiangan N Street, Songbei District, Harbin, 150028, Heilongjiang, China
- NHC Key Laboratory of Molecular Probe and Targeted Diagnosis and Therapy, Molecular Imaging Research Center (MIRC) of Harbin Medical University, Harbin, Heilongjiang, China
| | - Mengyuan Zhu
- Department of Nuclear Medicine, the Fourth Affiliated Hospital of Harbin Medical University, Xiangan N Street, Songbei District, Harbin, 150028, Heilongjiang, China
- NHC Key Laboratory of Molecular Probe and Targeted Diagnosis and Therapy, Molecular Imaging Research Center (MIRC) of Harbin Medical University, Harbin, Heilongjiang, China
| | - Xinyu Li
- Department of Nuclear Medicine, the Fourth Affiliated Hospital of Harbin Medical University, Xiangan N Street, Songbei District, Harbin, 150028, Heilongjiang, China
- NHC Key Laboratory of Molecular Probe and Targeted Diagnosis and Therapy, Molecular Imaging Research Center (MIRC) of Harbin Medical University, Harbin, Heilongjiang, China
| | - Duoyi Zhang
- Department of Nuclear Medicine, the Fourth Affiliated Hospital of Harbin Medical University, Xiangan N Street, Songbei District, Harbin, 150028, Heilongjiang, China
- NHC Key Laboratory of Molecular Probe and Targeted Diagnosis and Therapy, Molecular Imaging Research Center (MIRC) of Harbin Medical University, Harbin, Heilongjiang, China
| | - Wei Chen
- Department of Nuclear Medicine & Laboratory of Clinical Nuclear Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhaoguo Han
- Department of Nuclear Medicine, the Fourth Affiliated Hospital of Harbin Medical University, Xiangan N Street, Songbei District, Harbin, 150028, Heilongjiang, China.
- NHC Key Laboratory of Molecular Probe and Targeted Diagnosis and Therapy, Molecular Imaging Research Center (MIRC) of Harbin Medical University, Harbin, Heilongjiang, China.
| | - Xilin Sun
- Department of Nuclear Medicine, the Fourth Affiliated Hospital of Harbin Medical University, Xiangan N Street, Songbei District, Harbin, 150028, Heilongjiang, China.
- NHC Key Laboratory of Molecular Probe and Targeted Diagnosis and Therapy, Molecular Imaging Research Center (MIRC) of Harbin Medical University, Harbin, Heilongjiang, China.
| |
Collapse
|
15
|
Ren XH, Guo T, Xu MF, Huang Y, Liao XR, Qi LJ, Cheng SX. A Multiple Targeting Genome Editing System for Remodulation of Circulating Malignant Cells to Eliminate Cancer Immunosuppression and Restore Immune Responses. Adv Healthc Mater 2025; 14:e2401223. [PMID: 39440615 DOI: 10.1002/adhm.202401223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 09/29/2024] [Indexed: 10/25/2024]
Abstract
Cancer immunotherapy, which aims to eliminate cancer immunosuppression and reactivate anticancer immunity, holds great promise in oncology treatments. However, it is challenging to accurately study the efficacy of immunotherapy based on human-derived cells through animal experiments due to xenogeneic immune rejection. Herein, a personalized and precise strategy to evaluate the effectiveness of immunotherapy using the blood samples of cancer patients is presented. Through the utilization of multiple cancer-targeting delivery system decorated with the epidermal growth factor receptor (EGFR)-specific aptamer CL4 and the AXL-specific aptamer GL21.T to achieve superior efficiency in delivering the genome editing plasmid for MUC1 knockout, effective modulation on the behavior of circulating malignant cells (CMCs) is realized. After genome editing, both mucin 1 (MUC1) and programmed death-ligand 1 (PD-L1) are significantly downregulated in CMCs. The elimination of immunosuppression results in markedly enhanced secretion of pro-inflammatory anticancer cytokines encompassing interleukins 2, 12, and 15 and interferon-γ by immune cells. The study not only provides a strategy to overcome immunosuppression but also yields critical insights for personalized immunotherapy approaches.
Collapse
Affiliation(s)
- Xiao-He Ren
- School of Life Sciences, Anhui Medical University, Hefei, Anhui, 230011, China
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, Hubei, 430072, China
| | - Tao Guo
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital of Anhui Medical University, Anhui Public Health Clinical Center, Hefei, Anhui, 230011, China
| | - Ma-Fei Xu
- School of Life Sciences, Anhui Medical University, Hefei, Anhui, 230011, China
| | - Yun Huang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Anhui Public Health Clinical Center, Hefei, Anhui, 230011, China
| | - Xin-Ru Liao
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, Hubei, 430072, China
| | - Li-Jin Qi
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, Hubei, 430072, China
| | - Si-Xue Cheng
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, Hubei, 430072, China
| |
Collapse
|
16
|
Sirera R, Beltrán-Visiedo M, Galluzzi L. Targeting immune evasion in hepatocellular carcinoma-initiating cells. Trends Immunol 2025; 46:4-6. [PMID: 39721855 DOI: 10.1016/j.it.2024.12.002] [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: 11/21/2024] [Accepted: 12/06/2024] [Indexed: 12/28/2024]
Abstract
Tumor-initiating cells (TICs) are particularly efficient at evading detection and elimination by the human immune system. Recent data from Yang and collaborators demonstrate that - at least in preclinical hepatocellular carcinoma models - the immunological privilege of CD49f+ TICs can be limited by targeting CD155, resulting in restored sensitivity to immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Rafael Sirera
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, Philadelphia, PA, USA; Department of Biotechnology, Universitat Politècnica de València, Valencia, Spain; Unidad Mixta TRIAL, Centro Investigación Príncipe Felipe-Fundación Investigación, Hospital General Universitario de Valencia, Valencia, Spain; Centro de Investigación Biomédica en Red Cáncer, CIBERONC, Madrid, Spain
| | - Manuel Beltrán-Visiedo
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Lorenzo Galluzzi
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, Philadelphia, PA, USA.
| |
Collapse
|
17
|
Xu JG, Chen S, He Y, Zhu X, Wang Y, Ye Z, Zhou JC, Wu X, Zhang L, Ren X, Jia H, Yu H, Wei X, Feng Y, Chen X, Cui X, Pan X, Wang S, Xia S, Shang H, Pu Y, Xu W, Li H, Chen Q, Chen Z, Wang M, Yan X, Shi H, Li M, Xia Y, Bellelli R, Dong S, He J, Huang J, Cai CL, Zhu X, Zhan Y, Wan L. An antibody cocktail targeting two different CD73 epitopes enhances enzyme inhibition and tumor control. Nat Commun 2024; 15:10872. [PMID: 39738003 DOI: 10.1038/s41467-024-55207-9] [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: 10/26/2023] [Accepted: 12/05/2024] [Indexed: 01/01/2025] Open
Abstract
CD73, an ectoenzyme responsible for adenosine production, is often elevated in immuno-suppressive tumor environments. Inhibition of CD73 activity holds great promise as a therapeutic strategy for CD73-expressing cancers. In this study, we have developed a therapeutic anti-human CD73 antibody cocktail, HB0045. HB0045 is a 1:1 mixture of two humanized monoclonal IgG1 antibodies (mAbs), HB0038 and HB0039. The cocktail not only harnesses the advantages of its parental mAbs in enzyme inhibition but also shows a significantly greater capability of promoting T cell proliferation in vitro. Structural analyses show that HB0045 effectively locks the CD73 dimer in a "partially open" non-active conformation through a double lock mechanism. In various animal models of syngeneic and xenograft tumors, HB0045 inhibits tumor growth more potently than the single mAbs. Collectively, our findings provide functional and structural insights into the mechanism of a CD73-targeting antibody cocktail.
Collapse
MESH Headings
- 5'-Nucleotidase/immunology
- 5'-Nucleotidase/antagonists & inhibitors
- Animals
- Humans
- Mice
- GPI-Linked Proteins/immunology
- GPI-Linked Proteins/antagonists & inhibitors
- GPI-Linked Proteins/metabolism
- Epitopes/immunology
- Cell Line, Tumor
- Xenograft Model Antitumor Assays
- Cell Proliferation/drug effects
- Antibodies, Monoclonal, Humanized/immunology
- Antibodies, Monoclonal, Humanized/pharmacology
- Antibodies, Monoclonal, Humanized/chemistry
- Neoplasms/immunology
- Neoplasms/drug therapy
- Female
- T-Lymphocytes/immunology
- T-Lymphocytes/drug effects
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal/pharmacology
- Antibodies, Monoclonal/chemistry
- Mice, Inbred BALB C
- Combined Antibody Therapeutics
Collapse
Affiliation(s)
- Jin-Gen Xu
- Key Laboratory of Immune Response and Immunotherapy, Guangzhou Institutes of Biomedicine and Health (GIBH), Chinese Academy of Scienes, Guangzhou, China
- Drug Discovery, Shanghai Huaota Biopharmaceutical Co. Ltd., Shanghai, China
| | - Shi Chen
- Drug Discovery, Shanghai Huaota Biopharmaceutical Co. Ltd., Shanghai, China
| | - Yang He
- Institute of Molecular and Cell Biology, Singapore, Singapore
| | - Xi Zhu
- Drug Discovery, Shanghai Huaota Biopharmaceutical Co. Ltd., Shanghai, China
| | - Yanting Wang
- Drug Discovery, Shanghai Huaota Biopharmaceutical Co. Ltd., Shanghai, China
| | - Zhifeng Ye
- Key Laboratory of Immune Response and Immunotherapy, Guangzhou Institutes of Biomedicine and Health (GIBH), Chinese Academy of Scienes, Guangzhou, China
| | - Jin Chuan Zhou
- Department of Biochemistry, University of Oxford, Oxford, UK
| | - Xuanhui Wu
- Key Laboratory of Immune Response and Immunotherapy, Guangzhou Institutes of Biomedicine and Health (GIBH), Chinese Academy of Scienes, Guangzhou, China
| | - Lei Zhang
- Drug Discovery, Shanghai Huaota Biopharmaceutical Co. Ltd., Shanghai, China
| | - Xiaochen Ren
- Drug Discovery, Shanghai Huaota Biopharmaceutical Co. Ltd., Shanghai, China
| | - Huifeng Jia
- Drug Discovery, Shanghai Huaota Biopharmaceutical Co. Ltd., Shanghai, China
| | - Haijia Yu
- Drug Discovery, Shanghai Huaota Biopharmaceutical Co. Ltd., Shanghai, China
| | - Xiaoyue Wei
- Drug Discovery, Shanghai Huaota Biopharmaceutical Co. Ltd., Shanghai, China
| | - Yujie Feng
- Drug Discovery, Shanghai Huaota Biopharmaceutical Co. Ltd., Shanghai, China
| | - Xiaofang Chen
- Drug Discovery, Shanghai Huaota Biopharmaceutical Co. Ltd., Shanghai, China
| | - Xiaopei Cui
- Drug Discovery, Shanghai Huaota Biopharmaceutical Co. Ltd., Shanghai, China
| | - Xianfei Pan
- Drug Discovery, Shanghai Huaota Biopharmaceutical Co. Ltd., Shanghai, China
| | - Shaojie Wang
- Drug Discovery, Shanghai Huaota Biopharmaceutical Co. Ltd., Shanghai, China
| | - Simin Xia
- Drug Discovery, Shanghai Huaota Biopharmaceutical Co. Ltd., Shanghai, China
| | - Hongjie Shang
- Drug Discovery, Shanghai Huaota Biopharmaceutical Co. Ltd., Shanghai, China
| | - Yueqing Pu
- Drug Discovery, Shanghai Huaota Biopharmaceutical Co. Ltd., Shanghai, China
| | - Wei Xu
- Drug Discovery, Shanghai Huaota Biopharmaceutical Co. Ltd., Shanghai, China
| | - Haidong Li
- College of Biology and Pharmacy, Yulin Normal University, Yulin, China
| | - Qian Chen
- Drug Discovery, Shanghai Huaota Biopharmaceutical Co. Ltd., Shanghai, China
| | - Zeyu Chen
- Biortus Biosciences Co. Ltd, Jiangyin, China
| | - Manfu Wang
- Biortus Biosciences Co. Ltd, Jiangyin, China
| | | | - Hui Shi
- Biortus Biosciences Co. Ltd, Jiangyin, China
| | - Mingwei Li
- Drug Discovery, Shanghai Huaota Biopharmaceutical Co. Ltd., Shanghai, China
| | - Yisui Xia
- Medical School, Guangdong Key Laboratory for Genome Stability & Disease Prevention, Shenzhen University, Shenzhen, China
| | - Roberto Bellelli
- Centre for Cancer Cell and Molecular Biology, Barts Cancer Institute, Charterhouse Square, Barbican, London, UK
| | - Shunli Dong
- Huzhou Key Laboratory of Molecular Medicine, Huzhou Central Hospital, Huzhou, China
- Huzhou Key Laboratory of Precision Diagnosis and Treatment in Respiratory Diseases, Huzhou, China
| | - Jun He
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Jun Huang
- The MOE Key Laboratory of Biosystems Homeostasis & Protection, Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang, China
| | - Chen-Leng Cai
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Xiangyang Zhu
- Drug Discovery, Shanghai Huaota Biopharmaceutical Co. Ltd., Shanghai, China.
| | - Yifan Zhan
- Drug Discovery, Shanghai Huaota Biopharmaceutical Co. Ltd., Shanghai, China.
| | - Li Wan
- Key Laboratory of Immune Response and Immunotherapy, Guangzhou Institutes of Biomedicine and Health (GIBH), Chinese Academy of Scienes, Guangzhou, China.
- Institute of Drug Discovery, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.
| |
Collapse
|
18
|
Xu H, Wang Y, Liu G, Zhu Z, Shahbazi MA, Reis RL, Kundu SC, Shi X, Zu M, Xiao B. Nano-Armed Limosilactobacillus reuteri for Enhanced Photo-Immunotherapy and Microbiota Tryptophan Metabolism against Colorectal Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024:e2410011. [PMID: 39739630 DOI: 10.1002/advs.202410011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 12/10/2024] [Indexed: 01/02/2025]
Abstract
Despite being a groundbreaking approach to treating colorectal cancer (CRC), the efficacy of immunotherapy is significantly compromised by the immunosuppressive tumor microenvironment and dysbiotic intestinal microbiota. Here, leveraging the superior carrying capacity and innate immunity-stimulating property of living bacteria, a nanomedicine-engineered bacterium, LR-S-CD/CpG@LNP, with optical responsiveness, immune-stimulating activity, and the ability to regulate microbiota metabolome is developed. Immunoadjuvant (CpG) and carbon dot (CD) co-loaded plant lipid nanoparticles (CD/CpG@LNPs) are constructed and conjugated to the surface of Limosilactobacillus reuteri (LR) via reactive oxygen species (ROS)-responsive linkers. The inherent photothermal and photodynamic properties of oral CD/CpG@LNPs induce in situ cytotoxic ROS generation and immunogenic cell death of colorectal tumor cells. The generated neoantigens and the released CpG function as a potent in situ vaccine that stimulates the maturation of immature dendritic cells. The mature dendritic cells and metabolites secreted by LR subsequently facilitated the tumor infiltration of cytotoxic T lymphocytes to eradicate colorectal tumors. The further in vivo results demonstrate that the photo-immunotherapy and intestinal microbial metabolite regulation of LR-S-CD/CpG@LNPs collectively suppressed the growth of orthotopic colorectal tumors and their liver metastases, presenting a promising avenue for synergistic treatment of CRC via the oral route.
Collapse
Affiliation(s)
- Haiting Xu
- State Key Laboratory of Resource Insects, College of Sericulture, Textile, and Biomass Sciences, Southwest University, Chongqing, 400715, China
| | - Yajun Wang
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology, Chengdu, 610054, China
| | - Ga Liu
- State Key Laboratory of Resource Insects, College of Sericulture, Textile, and Biomass Sciences, Southwest University, Chongqing, 400715, China
| | - Zhenhua Zhu
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Mohammad-Ali Shahbazi
- Department of Biomedical Engineering, University Medical Center Groningen, University of Groningen, Antonius Deusinglaan 1, Groningen, 9713 AV, Netherlands
- W.J. Kolff Institute for Biomedical Engineering and Materials Science, University of Groningen, Antonius Deusinglaan 1, Groningen, 9713 AV, Netherlands
| | - Rui L Reis
- 3Bs Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Barco, Guimarães, 4805-017, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga, Guimarães, 4800-058, Portugal
| | - Subhas C Kundu
- 3Bs Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Barco, Guimarães, 4805-017, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga, Guimarães, 4800-058, Portugal
| | - Xiaoxiao Shi
- State Key Laboratory of Resource Insects, College of Sericulture, Textile, and Biomass Sciences, Southwest University, Chongqing, 400715, China
| | - Menghang Zu
- State Key Laboratory of Resource Insects, College of Sericulture, Textile, and Biomass Sciences, Southwest University, Chongqing, 400715, China
| | - Bo Xiao
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology, Chengdu, 610054, China
| |
Collapse
|
19
|
Chen Y, Tian H, Zhang X, Nice EC, Huang C, Zhang H, Zheng S. Copper-coordination driven brain-targeting nanoassembly for efficient glioblastoma multiforme immunotherapy by cuproptosis-mediated tumor immune microenvironment reprogramming. J Nanobiotechnology 2024; 22:801. [PMID: 39731102 DOI: 10.1186/s12951-024-03059-2] [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: 09/02/2024] [Accepted: 12/02/2024] [Indexed: 12/29/2024] Open
Abstract
Limited drug accumulation and an immunosuppressive microenvironment are the major bottlenecks in the treatment of glioblastoma multiforme (GBM). Herein, we report a copper-coordination driven brain-targeting nanoassembly (TCe6@Cu/TP5 NPs) for site-specific delivery of therapeutic agents and efficient immunotherapy by activating the cGAS-STING pathway and downregulating the expression of PD-L1. To achieve this, the mitochondria-targeting triphenylphosphorus (TPP) was linked to photosensitizer Chlorin e6 (Ce6) to form TPP-Ce6 (TCe6), which was then self-assembled with copper ions and thymopentin (TP5) to obtain TCe6@Cu/TP5 NPs. This nanoassembly effectively accumulated in tumor sites through the copper transport mechanism. Meanwhile, TCe6@Cu/TP5 could induce mitochondrial impairment by photodynamic therapy (PDT) mediated reactive oxygen species (ROS) accumulation and Cu2+ triggered cuproptosis, resulting in evoking the AMP-activated protein kinase (AMPK) pathway to degrade PD-L1, and activating the cGAS-STING pathway to enhance anti-tumor immunity. Moreover, TP5 significantly promoted the proliferation and differentiation of dendritic cells (DCs) and T lymphocytes to further amplify the cancer immunity cycle. Collectively, our TCe6@Cu/TP5 NPs effectively facilitate drug accumulation and activate systemic antitumor immunity in vitro and in vivo, providing an innovative solution across the BBB that potentiates GBM immunotherapy.
Collapse
Affiliation(s)
- Yang Chen
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Hailong Tian
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu, 610041, China
| | - Xiaodian Zhang
- Key Laboratory of Emergency and Trauma of Ministry of Education, Engineering Research Center for Hainan Biological Sample Resources of Major Diseases, the Hainan Branch of National Clinical Research Center for Cancer, the First Affiliated Hospital, Hainan Medical University, Haikou, 570102, China
| | - Edouard C Nice
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, 3800, Australia
| | - Canhua Huang
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu, 610041, China
| | - Haiyuan Zhang
- School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, 434023, China.
| | - Shaojiang Zheng
- Key Laboratory of Emergency and Trauma of Ministry of Education, Engineering Research Center for Hainan Biological Sample Resources of Major Diseases, the Hainan Branch of National Clinical Research Center for Cancer, the First Affiliated Hospital, Hainan Medical University, Haikou, 570102, China.
| |
Collapse
|
20
|
Wei J, Li W, Zhang P, Guo F, Liu M. Current trends in sensitizing immune checkpoint inhibitors for cancer treatment. Mol Cancer 2024; 23:279. [PMID: 39725966 DOI: 10.1186/s12943-024-02179-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 11/20/2024] [Indexed: 12/28/2024] Open
Abstract
Immune checkpoint inhibitors (ICIs) have dramatically transformed the treatment landscape for various malignancies, achieving notable clinical outcomes across a wide range of indications. Despite these advances, resistance to immune checkpoint blockade (ICB) remains a critical clinical challenge, characterized by variable response rates and non-durable benefits. However, growing research into the complex intrinsic and extrinsic characteristics of tumors has advanced our understanding of the mechanisms behind ICI resistance, potentially improving treatment outcomes. Additionally, robust predictive biomarkers are crucial for optimizing patient selection and maximizing the efficacy of ICBs. Recent studies have emphasized that multiple rational combination strategies can overcome immune checkpoint resistance and enhance susceptibility to ICIs. These findings not only deepen our understanding of tumor biology but also reveal the unique mechanisms of action of sensitizing agents, extending clinical benefits in cancer immunotherapy. In this review, we will explore the underlying biology of ICIs, discuss the significance of the tumor immune microenvironment (TIME) and clinical predictive biomarkers, analyze the current mechanisms of resistance, and outline alternative combination strategies to enhance the effectiveness of ICIs, including personalized strategies for sensitizing tumors to ICIs.
Collapse
Grants
- ZYJC21043 the 1.3.5 Project for Disciplines of Excellence, West China Hospital, Sichuan University
- ZYJC21043 the 1.3.5 Project for Disciplines of Excellence, West China Hospital, Sichuan University
- ZYJC21043 the 1.3.5 Project for Disciplines of Excellence, West China Hospital, Sichuan University
- ZYJC21043 the 1.3.5 Project for Disciplines of Excellence, West China Hospital, Sichuan University
- ZYJC21043 the 1.3.5 Project for Disciplines of Excellence, West China Hospital, Sichuan University
- 2023YFS0111 Social Development Science and Technology Project of Sichuan Province on Science and Technology
- 2023YFS0111 Social Development Science and Technology Project of Sichuan Province on Science and Technology
- 2023YFS0111 Social Development Science and Technology Project of Sichuan Province on Science and Technology
- 2023YFS0111 Social Development Science and Technology Project of Sichuan Province on Science and Technology
- 2023YFS0111 Social Development Science and Technology Project of Sichuan Province on Science and Technology
Collapse
Affiliation(s)
- Jing Wei
- Department of Medical Oncology, Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province, 610041, China
| | - Wenke Li
- Department of Medical Oncology, Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province, 610041, China
| | - Pengfei Zhang
- Department of Medical Oncology, Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province, 610041, China
| | - Fukun Guo
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH, 45229, USA
| | - Ming Liu
- Department of Medical Oncology, Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province, 610041, China.
| |
Collapse
|
21
|
Wang X, Jing Z, Huang X, Liu X, Zhang Y, Wang Z, Ma P. PD-L1 antibody conjugated dihydrotanshinone I-loaded polymeric nanoparticle for targeted cancer immunotherapy combining PD-L1 blockade with immunogenic cell death. Int J Pharm 2024; 667:125004. [PMID: 39608587 DOI: 10.1016/j.ijpharm.2024.125004] [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: 09/03/2024] [Revised: 11/07/2024] [Accepted: 11/24/2024] [Indexed: 11/30/2024]
Abstract
PURPOSE Combination immune checkpoint inhibitors (ICI) with chemotherapeutic agents has proven to be highly promising in cancer therapy. However, low response rate, immune-related adverse events, and lack of effectively targeted co-delivery strategy are still major hurdles to overcome for this combination therapeutic regimen. Herein, programmed death-L1 (PD-L1) antibody modified and dihydrotanshinone I (DHT) loaded nanoparticle was prepared for tumor targeting drug delivery, thus achieving immune checkpoint blockade (ICB) and immunogenic cell death (ICD) synergistic anti-tumor effects. METHODS The DHT-loaded nanoparticle (DHT NP) was prepared by the emulsion solvent diffusion method. Atezolizumab (ATEZO) was thiolated with 2-iminothiolane and conjugated to the surface of DHT NP to prepare the ATEZO DHT NP. The drug encapsulation efficiency, drug loading, particle size and drug release were determined. The in vitro cellular uptake, cell proliferation inhibition and apoptosis were evaluated on the HGC-27 tumor cell. The in vivo tumor targeting, anti-tumor efficiency and immune regulation were assessed on tumor bearing mice. RESULTS The optimized ATEZO DHT NP was a spherical nanoparticle of about 250 nm with a continuous drug release profile. It was selectively taken up by the tumor cells through PD-L1 receptor-mediated endocytosis, which resulted in enhanced cytotoxicity and cell apoptosis. In vivo imaging further demonstrated its superior tumor tissue targeting ability. When tumor bearing mice were treated with the ATEZO DHT NP, its synergistic anti-tumor effect was much stronger than that of a single drug. Moreover, the tumor targeting delivery of DHT caused tumor necrosis and initiated ICD with release of tumor-associated antigens, which efficiently up-regulated the population of CD4+ and CD8+ T cells. Notably, there were no obvious system toxicity or tissue damage occur during the whole treatment period. CONCLUSION The ATEZO DHT NP could specifically target to tumor and enhance treatment efficiency through combination of PD-L1 blockade with ICD effect.
Collapse
Affiliation(s)
- Xue Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Ziqi Jing
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaobin Huang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaoya Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Yujie Zhang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China.
| | - Zhijun Wang
- Department of Geriatric Medicine &National Clinical Research Center of Geriatric Disease, The 2nd Medical Center of Chinese PLA General Hospital, Beijing, China; Department of Interventional Radiology, The 1st & 5th Medical Center of Chinese PLA General Hospital, Beijing, China.
| | - Pengkai Ma
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China.
| |
Collapse
|
22
|
Ciernikova S, Sevcikova A, Novisedlakova M, Mego M. Insights into the Relationship Between the Gut Microbiome and Immune Checkpoint Inhibitors in Solid Tumors. Cancers (Basel) 2024; 16:4271. [PMID: 39766170 PMCID: PMC11674129 DOI: 10.3390/cancers16244271] [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: 11/25/2024] [Revised: 12/16/2024] [Accepted: 12/20/2024] [Indexed: 01/11/2025] Open
Abstract
Immunotherapy with immune checkpoint inhibitors represents a revolutionary approach to the treatment of solid tumors, including malignant melanoma, lung cancer, and gastrointestinal malignancies. Anti-CTLA-4 and anti-PD-1/PDL-1 therapies provide prolonged survival for cancer patients, but their efficacy and safety are highly variable. This review focuses on the crucial role of the gut microbiome in modulating the efficacy and toxicity of immune checkpoint blockade. Studies suggest that the composition of the gut microbiome may influence the response to immunotherapy, with specific bacterial strains able to promote an anti-tumor immune response. On the other hand, dysbiosis may increase the risk of adverse effects, such as immune-mediated colitis. Interventions aimed at modulating the microbiome, including the use of probiotics, prebiotics, fecal microbial transplantation, or dietary modifications, represent promising strategies to increase treatment efficacy and reduce toxicity. The combination of immunotherapy with the microbiome-based strategy opens up new possibilities for personalized treatment. In addition, factors such as physical activity and nutritional supplementation may indirectly influence the gut ecosystem and consequently improve treatment outcomes in refractory patients, leading to enhanced patient responses and prolonged survival.
Collapse
Affiliation(s)
- Sona Ciernikova
- Department of Genetics, Cancer Research Institute, Biomedical Research Center of the Slovak Academy of Sciences, Dubravska Cesta 9, 845 05 Bratislava, Slovakia;
| | - Aneta Sevcikova
- Department of Genetics, Cancer Research Institute, Biomedical Research Center of the Slovak Academy of Sciences, Dubravska Cesta 9, 845 05 Bratislava, Slovakia;
| | - Maria Novisedlakova
- Department of Oncology, Hospital Bory, Ivana Bukovčana 6118, 841 08 Bratislava, Slovakia;
| | - Michal Mego
- 2nd Department of Oncology, Faculty of Medicine, Comenius University, Bratislava and National Cancer Institute, Klenova 1, 833 10 Bratislava, Slovakia;
| |
Collapse
|
23
|
Shao C, Zhang Y, Li H, Chen J, Huang T, Li J, Wen S, Wang S, Fan S, Zhao Y. Radiotherapy-resistant prostate cancer cells escape immune checkpoint blockade through the senescence-related ataxia telangiectasia and Rad3-related protein. Cancer Commun (Lond) 2024. [PMID: 39698847 DOI: 10.1002/cac2.12636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Revised: 10/31/2024] [Accepted: 11/27/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND The majority of patients with prostate cancer (PCa) exhibit intrinsic resistance to immune checkpoint blockade (ICB) following radiotherapy (RT). This resistance is generally attributed to the limited antigen presentation of heterogeneous cells within tumors. Here, we aimed to isolate and characterize these diverse subgroups of tumor post-RT to understand the molecular mechanisms of their resistance to ICB. METHODS Single-cell RNA-sequencing (scRNA-seq) was used to profile senescent cancer cell clusters induced by RT in LNCaP cells. The expression and phosphorylation levels of ataxia telangiectasia and Rad3-related protein (ATR) were assessed by immunohistochemistry in clinical samples from patients with or without RT. Co-immunoprecipitation, mutagenesis, and Western blotting were used to measure the interactions between proteins. Xenograft experiments were performed to assess the tumor immune response in the mice. RESULTS We identified a subset of PCa cells that exhibited resistance to RT, characterized by a reduced antigen presentation capability, which enhanced their ability to evade immune detection and resist cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) blockade. scRNA-seq revealed that the senescent state was a transient phase of PCa cells post-RT, particularly in CTLA-4 blockade treatment-resistant cells. This state was marked by increased cytosolic ATR level. Cytosolic ATR phosphorylated CD86 in its cytosolic domain and enhanced the interaction between CD86 and its E3 ligase MARCH1 through electrostatic attraction. Depletion or inhibition of Atr increased the sensitivity to immune attack and improved responses to anti-Ctla-4 antibody treatment in a mouse model. CONCLUSIONS Our findings indicate that the activation of cytosolic ATR, which is associated with cellular senescence, impedes the effectiveness of combined RT and ICB treatments. This discovery may provide valuable insights for improving the efficacy of combined RT and ICB therapies in PCa.
Collapse
Affiliation(s)
- Chenyi Shao
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, P. R. China
| | - Yingyi Zhang
- School of Disaster and Emergency Medicine, Tianjin University, Tianjin, P. R. China
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Hang Li
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, P. R. China
| | - Jiajia Chen
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, P. R. China
| | - Ting Huang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, P. R. China
| | - Jiaze Li
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, P. R. China
| | - Simeng Wen
- Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin Medical University, Tianjin, P. R. China
| | - Sen Wang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, P. R. China
| | - Saijun Fan
- Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, P. R. China
| | - Yu Zhao
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, P. R. China
| |
Collapse
|
24
|
Schad F, Thronicke A, Hofheinz RD, Klein R, Grabowski P, Oei SL, Wüstefeld H, Grah C. Immune Checkpoint Blockade Combined with AbnobaViscum ® Therapy Is Linked to Improved Survival in Advanced or Metastatic Non-Small-Cell Lung Cancer Patients: A Registry Study in Accordance with the ESMO Guidance for Reporting Real-World Evidence. Pharmaceuticals (Basel) 2024; 17:1713. [PMID: 39770555 PMCID: PMC11677823 DOI: 10.3390/ph17121713] [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: 11/18/2024] [Revised: 12/16/2024] [Accepted: 12/17/2024] [Indexed: 01/11/2025] Open
Abstract
Background: Recent advancements in cancer treatment have shown the potential of immune checkpoint blockade (ICB) plus Viscum album L. therapy in improving survival rates for patients with advanced or metastatic non-small-cell lung cancer (NSCLC). The objective of this study was to investigate factors associated with improved survival in NSCLC patients treated with a combination of ICB and abnobaViscum®. Methods: Patients with advanced or metastatic NSCLC from the accredited Network Oncology registry were included in this real-world data study adhering to ESMO-GROW criteria with ethics approval. Survival outcomes were compared between patients receiving ICB therapy alone versus those receiving combinational ICB plus abnobaViscum® therapy using Kaplan-Meier and multivariable Cox proportional hazard analysis. Results: Among 300 patients (median age 68 years; male/female ratio 1.19), 222 received ICB alone (CTRL group) and 78 received combinational therapy (COMB group). Overall survival was significantly prolonged in the COMB group by 7 months compared to CTRL (13.8 months vs. 6.8 months, p = 0.005) with a survival rate of 16.5% in the COMB group vs. 8.0% in the CTRL group. In programmed death-ligand 1 positive (≥1%) patients treated with first-line ICB, the addition of abnobaViscum® reduced the adjusted hazard of death by 75% (aHR: 0.25; 95%CI: 0.11-0.60, p = 0.02). Conclusions: The addition of abnobaViscum® to ICB is significantly associated with improved survival in patients with advanced or metastatic NSCLC patients, irrespective of age, stage, Eastern cooperative oncology group status, surgery, or radiation. Potential mechanisms include immune modulation, reduced primary ICB resistance, and tumor microenvironment modifications. The findings warrant further validation in randomized controlled trials or registry-based randomized controlled trials. Trial registration: The study was registered (DRKS00013335).
Collapse
Affiliation(s)
- Friedemann Schad
- Network Oncology Registry, Research Institute Havelhöhe, Kladower Damm 221, 14089 Berlin, Germany;
- Interdisciplinary Oncological Centre, Hospital Havelhöhe, Kladower Damm 221, 14089 Berlin, Germany;
| | - Anja Thronicke
- Network Oncology Registry, Research Institute Havelhöhe, Kladower Damm 221, 14089 Berlin, Germany;
| | - Ralf-Dieter Hofheinz
- Mannheim Cancer Center, Mannheim University Hospital, Theodor-Kutzer Ufer 1-3, 68167 Mannheim, Germany;
| | - Reinhild Klein
- Department of Internal Medicine II, University Hospital Tübingen, Otfried-Müller Straße 10, 72076 Tübingen, Germany;
| | - Patricia Grabowski
- Interdisciplinary Oncological Centre, Hospital Havelhöhe, Kladower Damm 221, 14089 Berlin, Germany;
- Charité Fatigue Centrum, Charité—Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Shiao Li Oei
- Network Oncology Registry, Research Institute Havelhöhe, Kladower Damm 221, 14089 Berlin, Germany;
| | - Hannah Wüstefeld
- Lung Cancer Center, Hospital Havelhöhe, Kladower Damm 221, 14089 Berlin, Germany; (H.W.); (C.G.)
| | - Christian Grah
- Lung Cancer Center, Hospital Havelhöhe, Kladower Damm 221, 14089 Berlin, Germany; (H.W.); (C.G.)
| |
Collapse
|
25
|
Guo X, Cui T, Sun L, Fu Y, Cheng C, Wu C, Zhu Y, Liang S, Liu Y, Zhou S, Li X, Ji C, Ma K, Zhang N, Chu Q, Xing C, Deng S, Wang J, Liu Y, Liu L. A STT3A-dependent PD-L1 glycosylation modification mediated by GMPS drives tumor immune evasion in hepatocellular carcinoma. Cell Death Differ 2024:10.1038/s41418-024-01432-0. [PMID: 39690246 DOI: 10.1038/s41418-024-01432-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 12/01/2024] [Accepted: 12/05/2024] [Indexed: 12/19/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is a malignant tumor characterized by rapid progression. To explore the regulatory mechanism of rapid tumor growth and metastasis, we conducted proteomic and scRNA-Seq analyses on advanced HCC tissues and identified a significant molecule, guanine monophosphate synthase (GMPS), closely associated with the immune evasion in HCC. We analyzed the immune microenvironment characteristics remodeled by GMPS using scRNA-Seq and found GMPS induced tumor immune evasion in HCC by impairing the tumor-killing function of CD8 + T cells. Further investigation revealed that GMPS increased PD-L1 expression by regulating its ubiquitination and glycosylation modification. Mechanistically, GMPS enhanced the bond between PD-L1 and the catalytic subunit STT3A of oligosaccharyltransferase (OST) by acting as an additional module connecting the Sec61 channel complex and STT3A, which aided in the translocation and modification of nascent peptides. Increased PD-L1 impaired the tumor-killing function of CD8 + T cells, leading to the immune evasion. Importantly, targeting GMPS with angustmycin A, an inhibitor of GMPS activity, significantly suppressed PD-L1 expression and tumor growth in HCC, which also increased the sensitivity to anti-CTLA-4 immunotherapy. These findings suggested the potential of targeting GMPS as a promising therapeutic approach for HCC.
Collapse
Affiliation(s)
- Xinyu Guo
- Department of Hepatic Surgery, Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang, China
| | - Tianming Cui
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China
| | - Linmao Sun
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China
| | - Yumin Fu
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China
| | - Cheng Cheng
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China
| | - Chenghui Wu
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China
| | - Yitong Zhu
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China
| | - Shuhang Liang
- Department of Gastrointestinal Surgery, Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China
| | - Yufeng Liu
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China
| | - Shuo Zhou
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China
| | - Xianying Li
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China
| | - Changyong Ji
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China
| | - Kun Ma
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China
| | - Ning Zhang
- Department of Hepatic Surgery, Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang, China
| | - Qi Chu
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China
| | - Changjian Xing
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China
| | - Shumin Deng
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China
| | - Jiabei Wang
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China.
| | - Yao Liu
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China.
| | - Lianxin Liu
- Department of Hepatic Surgery, Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang, China.
| |
Collapse
|
26
|
Cui C, Zhang H, Yang C, Yin M, Teng X, Yang M, Kong D, Zhang J, Peng W, Chu Z, Wang J, Sun Y, Kang L, Lyu B, Gao Q, Wu M, Wang Y, Li Y. Inhibition of JNK Signaling Overcomes Cancer-Associated Fibroblast-Mediated Immunosuppression and Enhances the Efficacy of Immunotherapy in Bladder Cancer. Cancer Res 2024; 84:4199-4213. [PMID: 39292817 DOI: 10.1158/0008-5472.can-24-0940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 07/15/2024] [Accepted: 09/11/2024] [Indexed: 09/20/2024]
Abstract
Currently, only 20% to 40% of patients with cancer benefit from immune checkpoint inhibitors. Understanding the mechanisms underlying the immunosuppressive tumor microenvironment (TME) and characterizing dynamic changes in the immunologic landscape during treatment are critical for improving responsiveness to immunotherapy. In this study, we identified JNK signaling in cancer-associated fibroblasts (CAF) as a regulator of the immunosuppressive TME. Single-cell RNA sequencing of bladder cancer samples treated with a JNK inhibitor revealed enhanced cytotoxicity and effector functions of CD8+ T cells. In untreated tumors, CAFs interacted frequently with CD8+ T cells and mediated their exhaustion. JNK inhibition abrogated the immunosuppression function of CAFs by downregulating the expression of thymic stromal lymphopoietin (TSLP), thereby restoring CD8+ T-cell cytotoxicity. In addition, blockade of CAF-derived TSLP in combination with anti-PD-1 treatment promoted tumor elimination by CD8+ T cells in vivo. Collectively, these results indicate that JNK signaling plays an important immunosuppressive role in the TME by promoting expression of TSLP in CAFs and suggest that inhibiting JNK signaling could be a promising immunotherapeutic strategy for cancer treatment. Significance: JNK signaling promotes the secretion of TSLP by bladder cancer-associated fibroblasts to impede CD8+ T-cell activity, which can be circumvented by combination treatment targeting JNK signaling and PD-1.
Collapse
Affiliation(s)
- Chengying Cui
- Department of Genetics, School of Life Science, Anhui Medical University, Hefei, China
| | - Haojie Zhang
- Department of Urology, Huadong Hospital, Fudan University, Shanghai, China
| | - Congcong Yang
- Department of Genetics, School of Life Science, Anhui Medical University, Hefei, China
| | - Mingwei Yin
- Department of Genetics, School of Life Science, Anhui Medical University, Hefei, China
- Department of Pathophysiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Xinkun Teng
- Department of Genetics, School of Life Science, Anhui Medical University, Hefei, China
| | - Miaomiao Yang
- The First Affiliated Hospital of Anhui Medical University, Pathology Center, Hefei, China
- Anhui Public Health Clinical Center, Pathology Center, Hefei, China
| | - Dejie Kong
- Department of Genetics, School of Life Science, Anhui Medical University, Hefei, China
| | - Jinzhi Zhang
- Department of Genetics, School of Life Science, Anhui Medical University, Hefei, China
| | - Weidong Peng
- Department of Epidemiology and Health Statistics, School of Public Health, Anhui Medical University, Hefei, China
| | - Zhimin Chu
- Department of Genetics, School of Life Science, Anhui Medical University, Hefei, China
| | - Jingjing Wang
- Department of Genetics, School of Life Science, Anhui Medical University, Hefei, China
| | - Yating Sun
- Department of Genetics, School of Life Science, Anhui Medical University, Hefei, China
| | - Liping Kang
- Department of Genetics, School of Life Science, Anhui Medical University, Hefei, China
| | - Bin Lyu
- Department of Genetics, School of Life Science, Anhui Medical University, Hefei, China
| | - Qian Gao
- Department of Genetics, School of Life Science, Anhui Medical University, Hefei, China
| | - Mingqing Wu
- Department of Genetics, School of Life Science, Anhui Medical University, Hefei, China
| | - Yongqiang Wang
- Department of Urology, South China Hospital, Medical School, Shenzhen University, Shenzhen, China
| | - Yang Li
- Department of Genetics, School of Life Science, Anhui Medical University, Hefei, China
- Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Anhui Medical University, Hefei, China
| |
Collapse
|
27
|
Zhang R, Yang Y, Li X, Jiao C, Lou M, Mi W, Mao-Ying QL, Chu Y, Wang Y. Exploring shared targets in cancer immunotherapy and cancer-induced bone pain: Insights from preclinical studies. Cancer Lett 2024; 611:217399. [PMID: 39689823 DOI: 10.1016/j.canlet.2024.217399] [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: 09/05/2024] [Revised: 11/13/2024] [Accepted: 12/14/2024] [Indexed: 12/19/2024]
Abstract
Cancer casts a profound shadow on global health, with pain emerging as one of the dominant and severe complications, particularly in advanced stages. The effective management of cancer-induced pain remains an unmet need. Emerging preclinical evidence suggests that targets related to tumor immunotherapy may also modulate cancer-related pain pathways, thus offering a promising therapeutic direction. This review, focusing on more than ten molecular targets that link cancer immunotherapy and cancer-induced bone pain, underscores their potential to tackle both aspects in the context of comprehensive cancer care. Emphasizing factors such as types of cancer, drug administration methods, and sex differences in the analgesic efficacy of immunotherapeutic agents provides neuroscientific insights into personalized pain management for patients with cancer.
Collapse
Affiliation(s)
- Ruofan Zhang
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Yachen Yang
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Xiang Li
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200443, China
| | - Chunmeng Jiao
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Mengping Lou
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Wenli Mi
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Qi-Liang Mao-Ying
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Yuxia Chu
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Yanqing Wang
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China; State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, Fudan University, Shanghai 200032, China.
| |
Collapse
|
28
|
Yu K, Tang Y, Wang C, Liu W, Hu M, Hu A, Kuang Y, Zacksenhaus E, Yu XZ, Xiao X, Ben-David Y. The Astragalus Membranaceus Herb Attenuates Leukemia by Inhibiting the FLI1 Oncogene and Enhancing Anti-Tumor Immunity. Int J Mol Sci 2024; 25:13426. [PMID: 39769192 PMCID: PMC11676164 DOI: 10.3390/ijms252413426] [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: 11/12/2024] [Revised: 12/06/2024] [Accepted: 12/10/2024] [Indexed: 01/03/2025] Open
Abstract
Astragalus membranaceus (AM) herb is a component of traditional Chinese medicine used to treat various cancers. Herein, we demonstrate a strong anti-leukemic effect of AM injected (Ai) into the mouse model of erythroleukemia induced by Friend virus. Chemical analysis combined with mass spectrometry of AM/Ai identified the compounds Betulinic acid, Kaempferol, Hederagenin, and formononetin, all major mediators of leukemia inhibition in culture and in vivo. Docking analysis demonstrated binding of these four compounds to FLI1, resulting in downregulation of its targets, induction of apoptosis, differentiation, and suppression of cell proliferation. Chemical composition analysis identified other compounds previously known having anti-tumor activity independent of the FLI1 blockade. Among these, Astragaloside-A (As-A) has marginal effect on cells in culture, but strongly inhibits leukemogenesis in vivo, likely through improvement of anti-tumor immunity. Indeed, both IDO1 and TDO2 were identified as targets of As-A, leading to suppression of tryptophane-mediated Kyn production and leukemia suppression. Moreover, As-A interacts with histamine decarboxylase (HDC), leading to suppression of anti-inflammatory genes TNF, IL1B/IL1A, TNFAIP3, and CXCR2, but not IL6. These results implicate HDC as a novel immune checkpoint mediator, induced in the tumor microenvironment to promote leukemia. Functional analysis of AM components may allow development of combination therapy with optimal anti-leukemia effect.
Collapse
Affiliation(s)
- Kunlin Yu
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 561113, China; (K.Y.); (Y.T.); (C.W.); (W.L.); (M.H.); (A.H.); (Y.K.)
- Natural Products Research Center of Guizhou Province, Guiyang 550014, China
| | - Yao Tang
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 561113, China; (K.Y.); (Y.T.); (C.W.); (W.L.); (M.H.); (A.H.); (Y.K.)
- Natural Products Research Center of Guizhou Province, Guiyang 550014, China
| | - Chunlin Wang
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 561113, China; (K.Y.); (Y.T.); (C.W.); (W.L.); (M.H.); (A.H.); (Y.K.)
- Natural Products Research Center of Guizhou Province, Guiyang 550014, China
| | - Wuling Liu
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 561113, China; (K.Y.); (Y.T.); (C.W.); (W.L.); (M.H.); (A.H.); (Y.K.)
- Natural Products Research Center of Guizhou Province, Guiyang 550014, China
| | - Maoting Hu
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 561113, China; (K.Y.); (Y.T.); (C.W.); (W.L.); (M.H.); (A.H.); (Y.K.)
- Natural Products Research Center of Guizhou Province, Guiyang 550014, China
| | - Anling Hu
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 561113, China; (K.Y.); (Y.T.); (C.W.); (W.L.); (M.H.); (A.H.); (Y.K.)
- Natural Products Research Center of Guizhou Province, Guiyang 550014, China
| | - Yi Kuang
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 561113, China; (K.Y.); (Y.T.); (C.W.); (W.L.); (M.H.); (A.H.); (Y.K.)
- Natural Products Research Center of Guizhou Province, Guiyang 550014, China
| | - Eldad Zacksenhaus
- Division of Advanced Diagnostics, Toronto General Research Institute, University Health Network, Toronto, ON M5G 1L7, Canada;
| | - Xue-Zhong Yu
- Department of Microbiology & Immunology, Medical College of Wisconsin, Milwaukee, WI 53226, USA;
| | - Xiao Xiao
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 561113, China; (K.Y.); (Y.T.); (C.W.); (W.L.); (M.H.); (A.H.); (Y.K.)
- Natural Products Research Center of Guizhou Province, Guiyang 550014, China
| | - Yaacov Ben-David
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 561113, China; (K.Y.); (Y.T.); (C.W.); (W.L.); (M.H.); (A.H.); (Y.K.)
- Natural Products Research Center of Guizhou Province, Guiyang 550014, China
| |
Collapse
|
29
|
Akdogan O, Turkmen S, Uyar GC, Yucel KB, Tufekci B, Gurler F, Yazici O, Ozdemir N, Ozet A, Karakaya C, Sutcuoglu O. Impact of Serum GDF-15 and IL-6 on Immunotherapy Response in Cancer: A Prospective Study. Cancers (Basel) 2024; 16:4146. [PMID: 39766045 PMCID: PMC11674841 DOI: 10.3390/cancers16244146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 12/07/2024] [Accepted: 12/10/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Immunotherapy has transformed cancer treatment; however, predicting treatment response remains challenging. Serum biomarkers can help identify patients who are most likely to benefit from immunotherapy. OBJECTIVE This study evaluates the relationship between serum growth differentiation factor 15 (GDF-15) and interleukin-6 (IL-6) levels and treatment outcomes in cancer patients undergoing second-line immunotherapy. METHODS We conducted a prospective observational study involving 85 patients with non-small-cell lung cancer (NSCLC), renal cell carcinoma (RCC), or malignant melanoma treated with nivolumab. The baseline serum levels of GDF-15 and IL-6 were measured by using ELISA kits. The primary endpoints were progression-free survival (PFS) and overall survival (OS), with cachexia as a secondary outcome. RESULTS Elevated GDF-15 levels were significantly associated with shorter PFS (HR: 0.55, 95% CI: 0.32-0.96, p = 0.032) and OS (HR: 0.47, 95% CI: 0.25-0.90, p = 0.020). Higher IL-6 levels correlated with shorter PFS, though statistical significance was not achieved. Additionally, high GDF-15 levels were linked to increased cachexia incidence (p = 0.037). CONCLUSION Our findings indicate that GDF-15 could serve as a prognostic biomarker for immunotherapy response and may also be a target for cachexia management. Further studies should explore its potential to guide clinical decision making in oncology.
Collapse
Affiliation(s)
- Orhun Akdogan
- Department of Medical Oncology, Gazi University, 06560 Ankara, Türkiye; (F.G.); (O.Y.); (N.O.); (A.O.)
| | - Sena Turkmen
- Department of Medical Biochemistry, Gazi University, 06560 Ankara, Türkiye; (S.T.); (C.K.)
| | - Galip Can Uyar
- Department of Medical Oncology, Ankara Etlik City Hospital, 06170 Ankara, Türkiye; (G.C.U.); (K.B.Y.); (O.S.)
| | - Kadriye Bir Yucel
- Department of Medical Oncology, Ankara Etlik City Hospital, 06170 Ankara, Türkiye; (G.C.U.); (K.B.Y.); (O.S.)
| | - Busra Tufekci
- Department of Internal Medicine, Gazi University, 06560 Ankara, Türkiye;
| | - Fatih Gurler
- Department of Medical Oncology, Gazi University, 06560 Ankara, Türkiye; (F.G.); (O.Y.); (N.O.); (A.O.)
| | - Ozan Yazici
- Department of Medical Oncology, Gazi University, 06560 Ankara, Türkiye; (F.G.); (O.Y.); (N.O.); (A.O.)
| | - Nuriye Ozdemir
- Department of Medical Oncology, Gazi University, 06560 Ankara, Türkiye; (F.G.); (O.Y.); (N.O.); (A.O.)
| | - Ahmet Ozet
- Department of Medical Oncology, Gazi University, 06560 Ankara, Türkiye; (F.G.); (O.Y.); (N.O.); (A.O.)
| | - Cengiz Karakaya
- Department of Medical Biochemistry, Gazi University, 06560 Ankara, Türkiye; (S.T.); (C.K.)
| | - Osman Sutcuoglu
- Department of Medical Oncology, Ankara Etlik City Hospital, 06170 Ankara, Türkiye; (G.C.U.); (K.B.Y.); (O.S.)
| |
Collapse
|
30
|
Triantafyllou E, Gudd CLC, Possamai LA. Immune-mediated liver injury from checkpoint inhibitors: mechanisms, clinical characteristics and management. Nat Rev Gastroenterol Hepatol 2024:10.1038/s41575-024-01019-7. [PMID: 39663461 DOI: 10.1038/s41575-024-01019-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/05/2024] [Indexed: 12/13/2024]
Abstract
Immunotherapy has changed the treatment landscape for patients with cancer in the past decade. Immune checkpoint inhibitor (ICI)-based therapies have proven effective in a range of malignancies, including liver and gastrointestinal cancers, but they can cause diverse off-target organ toxicities. With the increasingly wider application of these drugs, immune-mediated liver injury from ICIs has become a commonly encountered challenge in clinical hepatology and gastroenterology. In this Review, we discuss the evidence from human and animal studies on the immunological mechanisms of immune-mediated liver injury from ICIs and summarize its clinical features and practical considerations for its management.
Collapse
Affiliation(s)
- Evangelos Triantafyllou
- Section of Hepatology and Gastroenterology, Division of Digestive Diseases, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom.
| | - Cathrin L C Gudd
- Section of Hepatology and Gastroenterology, Division of Digestive Diseases, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Lucia A Possamai
- Section of Hepatology and Gastroenterology, Division of Digestive Diseases, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom.
- Liver and Antiviral Unit, St Mary's Hospital, Imperial College Healthcare NHS Trust, London, United Kingdom.
| |
Collapse
|
31
|
Wang M, Liu Y, Li Y, Lu T, Wang M, Cheng Z, Chen L, Wen T, Pan M, Hu G. Tumor Microenvironment-Responsive Nanoparticles Enhance IDO1 Blockade Immunotherapy by Remodeling Metabolic Immunosuppression. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024:e2405845. [PMID: 39661740 DOI: 10.1002/advs.202405845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 10/25/2024] [Indexed: 12/13/2024]
Abstract
The clinical efficacy of immune checkpoint blockade (ICB) therapy is significantly compromised in the metabolically disordered tumor microenvironment (TME), posing a formidable challenge that cannot be ignored in current antitumor strategies. In this study, TME-responsive nanoparticles (HMP1G NPs) loaded with 1-methyltryptophan (1-MT; an indoleamine 2,3-dioxygenase 1 [IDO1] inhibitor,) and S-nitrosoglutathione (GSNO; a nitric oxide donor) is developed to enhance the therapeutic efficacy of 1-MT-mediated ICB. The HMP1G NPs responded to H+ and glutathione in the TME, releasing Mn2+, GSNO, and 1-MT. The released Mn2+ catalyzed the production of abundant reactive oxygen species and nitric oxide from hydrogen peroxide and GSNO, and the generated nitric oxide, synergistically with 1-MT, inhibited the accumulation of kynurenine mediated by IDO1 in the tumor. Mechanistically, HMP1G NPs downregulated tumor cell-derived IDO1 via the aryl hydrocarbon receptor/signal transducer and activator of transcription 3/interleukin signaling axis to improve kynurenine/tryptophan metabolism and immunosuppression. In a murine breast cancer model, treatment with HMP1G NPs elicited effective antitumor immunity and enhanced survival outcomes. This study highlights a novel nano-platform that simultaneously improves metabolism and enhances ICB efficacy to achieve a new and efficient antitumor strategy.
Collapse
Affiliation(s)
- Mengna Wang
- Department of Otorhinolaryngology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, P. R. China
- The First Clinical College, Chongqing Medical University, Chongqing, 400016, P. R. China
| | - Yuhong Liu
- Department of Otorhinolaryngology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, P. R. China
- The First Clinical College, Chongqing Medical University, Chongqing, 400016, P. R. China
| | - Yanshi Li
- Department of Otorhinolaryngology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, P. R. China
| | - Tao Lu
- Department of Otorhinolaryngology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, P. R. China
| | - Min Wang
- Department of Otorhinolaryngology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, P. R. China
| | - Zhaobo Cheng
- Department of Otorhinolaryngology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, P. R. China
- The First Clinical College, Chongqing Medical University, Chongqing, 400016, P. R. China
| | - Lin Chen
- Department of Otorhinolaryngology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, P. R. China
| | - Tongling Wen
- Department of Otorhinolaryngology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, P. R. China
- The First Clinical College, Chongqing Medical University, Chongqing, 400016, P. R. China
| | - Min Pan
- Department of Otorhinolaryngology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, P. R. China
| | - Guohua Hu
- Department of Otorhinolaryngology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, P. R. China
| |
Collapse
|
32
|
Monteagudo M, Calsina B, Salazar-Hidalgo ME, Martínez-Montes ÁM, Piñeiro-Yáñez E, Caleiras E, Martín MC, Rodríguez-Perales S, Letón R, Gil E, Buffet A, Burnichon N, Fernández-Sanromán Á, Díaz-Talavera A, Mellid S, Arroba E, Reglero C, Martínez-Puente N, Roncador G, Del Olmo MI, Corrales PJP, Oliveira CL, Álvarez-Escolá C, Gutiérrez MC, López-Fernández A, García NP, Regojo RM, Díaz LR, Laorden NR, Guadarrama OS, Bechmann N, Beuschlein F, Canu L, Eisenhofer G, Fassnacht M, Nölting S, Quinkler M, Rapizzi E, Remde H, Timmers HJ, Favier J, Gimenez-Roqueplo AP, Rodriguez-Antona C, Currás-Freixes M, Al-Shahrour F, Cascón A, Leandro-García LJ, Montero-Conde C, Robledo M. MAML3-fusions modulate vascular and immune tumour microenvironment and confer high metastatic risk in pheochromocytoma and paraganglioma. Best Pract Res Clin Endocrinol Metab 2024; 38:101931. [PMID: 39218714 DOI: 10.1016/j.beem.2024.101931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Pheochromocytomas and paragangliomas are rare neuroendocrine tumours. Around 20-25 % of patients develop metastases, for which there is an urgent need of prognostic markers and therapeutic stratification strategies. The presence of a MAML3-fusion is associated with increased metastatic risk, but neither the processes underlying disease progression, nor targetable vulnerabilities have been addressed. We have compiled a cohort of 850 patients, which has shown a 3.65 % fusion prevalence and represents the largest MAML3-positive series reported to date. While MAML3-fusions mainly cause single pheochromocytomas, we also observed somatic post-zygotic events, resulting in multiple tumours in the same patient. MAML3-tumours show increased expression of neuroendocrine-to-mesenchymal transition markers, MYC-targets, and angiogenesis-related genes, leading to a distinct tumour microenvironment with unique vascular and immune profiles. Importantly, our findings have identified MAML3-tumours specific vulnerabilities beyond Wnt-pathway dysregulation, such as a rich vascular network, and overexpression of PD-L1 and CD40, suggesting potential therapeutic targets.
Collapse
Affiliation(s)
- María Monteagudo
- Hereditary Endocrine Cancer Group; Human Cancer Genetics Program Spanish National Cancer Research Centre (CNIO), Madrid, Spain; PhD Program in Neuroscience, Universidad Autonoma de Madrid-Cajal Institute, Madrid, Spain
| | - Bruna Calsina
- Familial Cancer Clinical Unit, Human Cancer Genetics Program Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Milton E Salazar-Hidalgo
- Hereditary Endocrine Cancer Group; Human Cancer Genetics Program Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Ángel M Martínez-Montes
- Hereditary Endocrine Cancer Group; Human Cancer Genetics Program Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Elena Piñeiro-Yáñez
- Bioinformatics Unit, Structural Biology Program, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Eduardo Caleiras
- Histopathology Core Unit Biotechnology Program Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Maria Carmen Martín
- Molecular Citogenetic Unit Human Cancer Genetics Program Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Sandra Rodríguez-Perales
- Molecular Citogenetic Unit Human Cancer Genetics Program Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Rocío Letón
- Hereditary Endocrine Cancer Group; Human Cancer Genetics Program Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Eduardo Gil
- Familial Cancer Clinical Unit, Human Cancer Genetics Program Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Alexandre Buffet
- Département de médecine génomique des tumeurs et des cancers, AP-HP, Hôpital Européen Georges Pompidou, Paris, France; Université Paris Cité, Inserm, PARCC, Paris, France
| | - Nelly Burnichon
- Département de médecine génomique des tumeurs et des cancers, AP-HP, Hôpital Européen Georges Pompidou, Paris, France; Université Paris Cité, Inserm, PARCC, Paris, France
| | - Ángel Fernández-Sanromán
- Hereditary Endocrine Cancer Group; Human Cancer Genetics Program Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Alberto Díaz-Talavera
- Hereditary Endocrine Cancer Group; Human Cancer Genetics Program Spanish National Cancer Research Centre (CNIO), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain
| | - Sara Mellid
- Hereditary Endocrine Cancer Group; Human Cancer Genetics Program Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Ester Arroba
- Hereditary Endocrine Cancer Group; Human Cancer Genetics Program Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Clara Reglero
- Hereditary Endocrine Cancer Group; Human Cancer Genetics Program Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Natalia Martínez-Puente
- Hereditary Endocrine Cancer Group; Human Cancer Genetics Program Spanish National Cancer Research Centre (CNIO), Madrid, Spain; PhD Program in Neuroscience, Universidad Autonoma de Madrid-Cajal Institute, Madrid, Spain
| | - Giovanna Roncador
- Monoclonal Antibodies Core Unit Biotechnology Program Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Maria Isabel Del Olmo
- Department of Endocrinology and Nutrition, University Hospital La Fe, Valencia, Spain
| | | | - Cristina Lamas Oliveira
- Department of Endocrinology and Nutrition Albacete University Hospital, SESCAM, Albacete, Spain
| | | | | | | | | | | | - Luis Robles Díaz
- Department of Oncology, 12 de Octubre University Hospital, Madrid, Spain
| | | | | | - Nicole Bechmann
- Institute for Clinical Chemistry and Laboratory Medicine Faculty of Medicine and University Hospital Carl Gustav Carus Technische Universität Dresden, Dresden Germany, Germany
| | - Felix Beuschlein
- Medizinische Klinik und Poliklinik IV Klinikum der Universität München, Munich, Germany; Klinik für Endokrinologie Diabetologie und Klinische Ernährung UniversitätsSpital Zürich, Zürich, Switzerland; LOOP Zurich - Medical Research Center, Zurich, Switzerland
| | - Letizia Canu
- Department of Experimental and Clinical Medicine University of Florence, Florence, Italy
| | - Graeme Eisenhofer
- Department of Medicine III University Hospital Carl Gustav Carus Technische Universität Dresden, Dresden, Germany
| | - Martin Fassnacht
- Department of Internal Medicine I Division of Endocrinology and Diabetes University Hospital Würzburg University of Würzburg, Würzburg, Germany; Comprehensive Cancer Center Mainfranken University of Würzburg, Würzburg, Germany
| | - Svenja Nölting
- Klinik für Endokrinologie Diabetologie und Klinische Ernährung UniversitätsSpital Zürich, Zürich, Switzerland
| | - Marcus Quinkler
- Endocrinology in Charlottenburg Stuttgarter Platz 1, Berlin, Germany
| | - Elena Rapizzi
- Department of Experimental and Clinical Medicine University of Florence, Florence, Italy
| | - Hanna Remde
- Comprehensive Cancer Center Mainfranken University of Würzburg, Würzburg, Germany
| | - Henri J Timmers
- Department of Internal Medicine, Radboud University Medical Centre, Nijmegen, the Netherlands
| | - Judith Favier
- Département de médecine génomique des tumeurs et des cancers, AP-HP, Hôpital Européen Georges Pompidou, Paris, France; Université Paris Cité, Inserm, PARCC, Paris, France
| | - Anne-Paule Gimenez-Roqueplo
- Département de médecine génomique des tumeurs et des cancers, AP-HP, Hôpital Européen Georges Pompidou, Paris, France; Université Paris Cité, Inserm, PARCC, Paris, France
| | - Cristina Rodriguez-Antona
- Hereditary Endocrine Cancer Group; Human Cancer Genetics Program Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Maria Currás-Freixes
- Familial Cancer Clinical Unit, Human Cancer Genetics Program Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Fatima Al-Shahrour
- Bioinformatics Unit, Structural Biology Program, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Alberto Cascón
- Hereditary Endocrine Cancer Group; Human Cancer Genetics Program Spanish National Cancer Research Centre (CNIO), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain
| | - Luis J Leandro-García
- Hereditary Endocrine Cancer Group; Human Cancer Genetics Program Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Cristina Montero-Conde
- Hereditary Endocrine Cancer Group; Human Cancer Genetics Program Spanish National Cancer Research Centre (CNIO), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain
| | - Mercedes Robledo
- Hereditary Endocrine Cancer Group; Human Cancer Genetics Program Spanish National Cancer Research Centre (CNIO), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain.
| |
Collapse
|
33
|
Zhang Z, Lu Y, Liu W, Huang Y. Nanomaterial-assisted delivery of CpG oligodeoxynucleotides for boosting cancer immunotherapy. J Control Release 2024; 376:184-199. [PMID: 39368710 DOI: 10.1016/j.jconrel.2024.09.044] [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: 05/09/2024] [Revised: 08/03/2024] [Accepted: 09/26/2024] [Indexed: 10/07/2024]
Abstract
Cancer immunotherapy aims to improve immunity to not only eliminate the primary tumor but also inhibit metastasis and recurrence. It is considered an extremely promising therapeutic approach that breaks free from the traditional paradigm of oncological treatment. As the medical community learns more about the immune system's mechanisms that "turn off the brake" and "step on the throttle", there is increasingly successful research on immunomodulators. However, there are still more restrictions than countermeasures with immunotherapy related to immunomodulators, such as low responsiveness and immune-related adverse events that cause multiple adverse reactions. Therefore, medical experts and materials scientists attempted to the efficacy of immunomodulatory treatments through various methods, especially nanomaterial-assisted strategies. CpG oligodeoxynucleotides (CpG) not only act as an adjuvant to promote immune responses, but also induce autophagy. In this review, the enhancement of immunotherapy using nanomaterial-based CpG formulations is systematically elaborated, with a focus on the delivery, protection, synergistic promotion of CpG efficacy by nanomaterials, and selection of the timing of treatment. In addition, we also discuss and prospect the existing problems and future directions of research on nanomaterials in auxiliary CpG therapy.
Collapse
Affiliation(s)
- Zhiyu Zhang
- Department of Pharmacology, Beijing Chest Hospital, Capital Medical University/Beijing Key Laboratory of Drug Resistance Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Yu Lu
- Department of Pharmacology, Beijing Chest Hospital, Capital Medical University/Beijing Key Laboratory of Drug Resistance Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China.
| | - Wenjing Liu
- Department of Pharmacology, Beijing Chest Hospital, Capital Medical University/Beijing Key Laboratory of Drug Resistance Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China.
| | - Yuanyu Huang
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, Key Laboratory of Molecular Medicine and Biotherapy, Beijing Institute of Technology, Beijing 100081, China
| |
Collapse
|
34
|
Lin J, Zhou Y, Li C, Li B, Hao H, Tian F, Li H, Liu Z, Wang G, Shen XC, Tang R, Wang X. Hydrogel activation of Mincle receptors for tumor cell processing: A novel approach in cancer immunotherapy. Biomaterials 2024; 311:122703. [PMID: 39002516 DOI: 10.1016/j.biomaterials.2024.122703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 07/02/2024] [Accepted: 07/08/2024] [Indexed: 07/15/2024]
Abstract
An obstacle in current tumor immunotherapies lies in the challenge of achieving sustained and tumor-targeting T cell immunity, impeded by the limited antigen processing and cross-presentation of tumor antigens. Here, we propose a hydrogel-based multicellular immune factory within the body that autonomously converts tumor cells into an antitumor vaccine. Within the body, the scaffold, formed by a calcium-containing chitosan hydrogel complex (ChitoCa) entraps tumor cells and attracts immune cells to establish a durable and multicellular microenvironment. Within this context, tumor cells are completely eliminated by antigen-presenting cells (APCs) and processed for cross-antigen presentation. The regulatory mechanism relies on the Mincle receptor, a cell-phagocytosis-inducing C-type lectin receptor specifically activated on ChitoCa-recruited APCs, which serves as a recognition synapse, facilitating a tenfold increase in tumor cell engulfment and subsequent elimination. The ChitoCa-induced tumor cell processing further promotes the cross-presentation of tumor antigens to prime protective CD8+ T cell responses. Therefore, the ChitoCa treatment establishes an immune niche within the tumor microenvironment, resulting in effective tumor regression either used alone or in combination with other immunotherapies. This hydrogel-induced immune factory establishes a functional organ-like multicellular colony for tumor-specific immunotherapy, paving the way for innovative strategies in cancer treatment.
Collapse
Affiliation(s)
- Jiake Lin
- Department of Chemistry, Zhejiang University, Hangzhou, Zhejiang, 310058, China; Liangzhu Laboratory, Hangzhou, Zhejiang, 311113, China
| | - Yuemin Zhou
- Qiushi Academy for Advanced Studies, Zhejiang University, Hangzhou, Zhejiang, 310058, China; Department of Chemistry, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Chen Li
- Qiushi Academy for Advanced Studies, Zhejiang University, Hangzhou, Zhejiang, 310058, China; Department of Chemistry, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Benke Li
- Qiushi Academy for Advanced Studies, Zhejiang University, Hangzhou, Zhejiang, 310058, China; Department of Chemistry, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Haibin Hao
- Qiushi Academy for Advanced Studies, Zhejiang University, Hangzhou, Zhejiang, 310058, China; Department of Chemistry, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Fengchao Tian
- Qiushi Academy for Advanced Studies, Zhejiang University, Hangzhou, Zhejiang, 310058, China; Department of Chemistry, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Huixin Li
- Qiushi Academy for Advanced Studies, Zhejiang University, Hangzhou, Zhejiang, 310058, China; Department of Chemistry, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Zhenyu Liu
- Department of Chemistry, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Guangchuan Wang
- State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Xing-Can Shen
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, 541004, China
| | - Ruikang Tang
- Department of Chemistry, Zhejiang University, Hangzhou, Zhejiang, 310058, China; Liangzhu Laboratory, Hangzhou, Zhejiang, 311113, China; Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310016, China.
| | - Xiaoyu Wang
- Qiushi Academy for Advanced Studies, Zhejiang University, Hangzhou, Zhejiang, 310058, China; Liangzhu Laboratory, Hangzhou, Zhejiang, 311113, China; Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310016, China.
| |
Collapse
|
35
|
Zhang Q, Wang X, Chen J, Wu J, Zhou M, Xia R, Wang W, Zheng X, Xie Z. Recent progress of porphyrin metal-organic frameworks for combined photodynamic therapy and hypoxia-activated chemotherapy. Chem Commun (Camb) 2024; 60:13641-13652. [PMID: 39497649 DOI: 10.1039/d4cc04512b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2024]
Abstract
Nanoscale metal-organic frameworks integrated with porphyrins (Por-nMOFs) have emerged as efficient nanoplatforms for photodynamic therapy (PDT), which relies on the conversion of molecular oxygen into cytotoxic singlet oxygen. However, the hypoxic microenvironment within tumors significantly limits the efficacy of PDT. To address this challenge, researchers have explored various strategies to either alter or exploit the hypoxic conditions in tumors. One such strategy involves leveraging the porous structure of Por-nMOFs to load hypoxia-activated prodrugs (HAPs) like tirapazamine (TPZ), thereby utilizing the tumor's intrinsic hypoxic environment to trigger a chemotherapeutic effect that synergizes with PDT. Advances in nanoscience have enabled the development of porphyrin-based nMOFs capable of simultaneously loading both porphyrin photosensitizers and TPZ, ensuring effective release within cancer cells under high-phosphate conditions. The subsequent activation of co-loaded TPZ, by the tumor's own hypoxic microenvironment, and that created during PDT, facilitates a combined PDT and chemotherapy approach. This method not only enhances the suppression of cancer cell proliferation but also improves control over tumor metastasis while mitigating the negative impact of hypoxia on singular Por-nMOFs in PDT. This review summarizes recent advances in Por-nMOFs research, focusing on the design strategies for enhancing water dispersibility, circulatory stability, and targeting specificity through post-synthetic modifications. Additionally, this review highlights the bioapplication of Por-nMOFs by integrating TPZ chemotherapy and other therapeutic modalities to combat hypoxic and metastatic malignancies. We anticipate that this review will inspire further research into Por-nMOFs and advance their application in biomedicine.
Collapse
Affiliation(s)
- Qiuyun Zhang
- School of Pharmacy, Nantong University, Nantong, Jiangsu Province 226001, China.
| | - Xiaohui Wang
- School of Public Health, Nantong University, Nantong, Jiangsu Province 226001, China.
| | - Jiayi Chen
- School of Pharmacy, Nantong University, Nantong, Jiangsu Province 226001, China.
| | - Junjie Wu
- School of Pharmacy, Nantong University, Nantong, Jiangsu Province 226001, China.
| | - Mengjiao Zhou
- School of Pharmacy, Nantong University, Nantong, Jiangsu Province 226001, China.
| | - Rui Xia
- School of Public Health, Nantong University, Nantong, Jiangsu Province 226001, China.
| | - Weiqi Wang
- School of Pharmacy, Nantong University, Nantong, Jiangsu Province 226001, China.
| | - Xiaohua Zheng
- School of Pharmacy, Nantong University, Nantong, Jiangsu Province 226001, China.
| | - Zhigang Xie
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun, Jilin 130022, P. R. China
| |
Collapse
|
36
|
Galassi C, Chan TA, Vitale I, Galluzzi L. The hallmarks of cancer immune evasion. Cancer Cell 2024; 42:1825-1863. [PMID: 39393356 DOI: 10.1016/j.ccell.2024.09.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 08/27/2024] [Accepted: 09/16/2024] [Indexed: 10/13/2024]
Abstract
According to the widely accepted "three Es" model, the host immune system eliminates malignant cell precursors and contains microscopic neoplasms in a dynamic equilibrium, preventing cancer outgrowth until neoplastic cells acquire genetic or epigenetic alterations that enable immune escape. This immunoevasive phenotype originates from various mechanisms that can be classified under a novel "three Cs" conceptual framework: (1) camouflage, which hides cancer cells from immune recognition, (2) coercion, which directly or indirectly interferes with immune effector cells, and (3) cytoprotection, which shields malignant cells from immune cytotoxicity. Blocking the ability of neoplastic cells to evade the host immune system is crucial for increasing the efficacy of modern immunotherapy and conventional therapeutic strategies that ultimately activate anticancer immunosurveillance. Here, we review key hallmarks of cancer immune evasion under the "three Cs" framework and discuss promising strategies targeting such immunoevasive mechanisms.
Collapse
Affiliation(s)
- Claudia Galassi
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY, USA
| | - Timothy A Chan
- Department of Radiation Oncology, Taussig Cancer Center, Cleveland Clinic, Cleveland, OH, USA; Center for Immunotherapy and Precision Immuno-Oncology, Cleveland Clinic, Cleveland, OH, USA; National Center for Regenerative Medicine, Cleveland, OH, USA; Case Comprehensive Cancer Center, Cleveland, OH, USA
| | - Ilio Vitale
- Italian Institute for Genomic Medicine, c/o IRCSS Candiolo, Torino, Italy; Candiolo Cancer Institute, FPO - IRCCS, Candiolo, Italy.
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY, USA; Sandra and Edward Meyer Cancer Center, New York, NY, USA; Caryl and Israel Englander Institute for Precision Medicine, New York, NY, USA; Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, Philadelphia, PA, USA.
| |
Collapse
|
37
|
Zhao L, Liu Y, Jin F, Hu K, Lv M, Zhou Y, Zhao W, Hu Y, Wu J, Yang Y, Wang W. Multifunctional nanoparticles potentiate in-situ tumor vaccines via reversing insufficient Photothermal therapy by disrupting tumor vasculature. J Control Release 2024; 376:842-860. [PMID: 39401677 DOI: 10.1016/j.jconrel.2024.10.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 09/25/2024] [Accepted: 10/11/2024] [Indexed: 11/08/2024]
Abstract
Photothermal therapy can trigger immunogenic cell death and release personalized in-situ tumor vaccine, activating immune responses to eliminate systemic tumors beyond the irradiated zone. However, the immune response of the in-situ tumor vaccines is often undermined by the residual tumor cells and their induced immunosuppressive tumor microenvironment (TME), which is attributed to insufficient photothermal effects stemming from the limited accumulation of photosensitizers. To overcome these limitations, we developed multi-functional nanoparticles (VI@Gd-NPs) that integrate a tumor vasculature-specific disrupting agent (Vadimezan, Phase III clinical drug), a photosensitizer (Indocyanine Green, ICG), and a magnetic resonance imaging contrast agent (Gadolinium, Gd) through chemical self-assembly. By selectively disrupting the tumor vasculature, these nanoparticles enhance the intratumoral delivery of photosensitizers (ICG and blood cells), and Gd. With the guidance of Gd-enhanced MRI, the improved delivery facilitates comprehensive photothermal ablation and regulates the TME, further initiating the in-situ tumor vaccine. Notably, this approach significantly enhances anti-tumor immune responses, improves survival rates, and reduces tumor recurrence and metastasis in various animal models. Moreover, depleting CD8+ T cells reverses these therapeutic benefits, highlighting the critical role of adaptive T cell immunity. Therefore, the VI@Gd-NPs treatment holds great potential for reigniting the in-situ tumor vaccine of photothermal therapy.
Collapse
Affiliation(s)
- Lili Zhao
- Vaccine Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing 211198, China
| | - Yiran Liu
- Vaccine Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing 211198, China
| | - Fangfei Jin
- Vaccine Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing 211198, China
| | - Kaiyuan Hu
- Vaccine Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing 211198, China
| | - Miao Lv
- Vaccine Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing 211198, China
| | - Yuehua Zhou
- Vaccine Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing 211198, China
| | - Weijun Zhao
- Vaccine Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing 211198, China
| | - Yiqiao Hu
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School of Nanjing University & School of Life Sciences, Nanjing University, Nanjing 210093, China
| | - Jinhui Wu
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School of Nanjing University & School of Life Sciences, Nanjing University, Nanjing 210093, China
| | - Yong Yang
- Vaccine Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing 211198, China
| | - Wenguang Wang
- Vaccine Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing 211198, China.
| |
Collapse
|
38
|
Jing C, Bai Z, Tong K, Yang X, Liu K, Wu H, Zhu J, Guo W, Zhang Z, Deng W. Efficacy and safety of camrelizumab, apatinib, and capecitabine combination therapy in advanced biliary tract cancer: a phase 2, nonrandomized, prospective study. Oncologist 2024; 29:e1565-e1574. [PMID: 39102756 PMCID: PMC11546768 DOI: 10.1093/oncolo/oyae154] [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: 03/30/2024] [Accepted: 05/22/2024] [Indexed: 08/07/2024] Open
Abstract
BACKGROUND Biliary tract cancer (BTC) is a highly malignant tumor, with limited therapy regimens and short response duration. In this study, we aim to assess the efficacy and safety of the combination of camrelizumab, apatinib, and capecitabine as the first- or second-line treatment in patients with advanced BTC. METHODS In this phase 2, nonrandomized, prospective study, eligible patients received camrelizumab (200 mg, d1, Q3W), apatinib (250 mg, qd, d1-d21, Q3W), and capecitabine (1000 mg/m², bid, d1-d14, Q3W) until trial discontinued. The primary endpoint was the objective response rate (ORR). The secondary endpoints were disease control rate, progression-free survival (PFS), overall survival (OS), and safety. RESULTS From July 2019 to April 2023, we enrolled a total of 28 patients, of whom 14 patients were in the first-line treatment setting and 14 patients were in the second-line setting. At the data cutoff (April 30, 2023), the median follow-up duration was 18.03 months. Eight of 28 patients reached objective response (ORR: 28.57%), with an ORR of 50% and 7.1% for first-line and second-line treatment patients (P = .033). The median PFS was 6.30 months and the median OS was 12.80 months. Grade 3 or 4 adverse events (AEs) occurred in 9 (32.14%) patients, including elevated transaminase, thrombocytopenia, etc. No serious treatment-related AEs or treatment-related deaths occurred. CONCLUSIONS In this trial, the combination of camrelizumab, apatinib, and capecitabine showed promising antitumor activity and manageable toxicity in patients with advanced BTC, especially in the first-line setting. CLINICAL TRIAL REGISTRATION NCT04720131.
Collapse
Affiliation(s)
- Chao Jing
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Zhigang Bai
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, People's Republic of China
- National Clinical Research Center for Digestive Diseases, Beijing, People's Republic of China
| | - Kuinan Tong
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Xiaobao Yang
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Kun Liu
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Hongwei Wu
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Jiegao Zhu
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Wei Guo
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, People's Republic of China
- National Clinical Research Center for Digestive Diseases, Beijing, People's Republic of China
| | - Zhongtao Zhang
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, People's Republic of China
- National Clinical Research Center for Digestive Diseases, Beijing, People's Republic of China
| | - Wei Deng
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, People's Republic of China
- National Clinical Research Center for Digestive Diseases, Beijing, People's Republic of China
| |
Collapse
|
39
|
Wang Z, Cui L, Lin Y, Huo B, Zhang H, Xie C, Zhang H, Liu Y, Jin H, Guo H, Li M, Wang X, Zhou P, Huang P, Liu J, Xia X. Cancer cell-intrinsic biosynthesis of itaconate promotes tumor immunogenicity. EMBO J 2024; 43:5530-5547. [PMID: 39349845 PMCID: PMC11574104 DOI: 10.1038/s44318-024-00217-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 07/16/2024] [Accepted: 08/07/2024] [Indexed: 11/20/2024] Open
Abstract
The Krebs cycle byproduct itaconate has recently emerged as an important metabolite regulating macrophage immune functions, but its role in tumor cells remains unknown. Here, we show that increased tumor-intrinsic cis-aconitate decarboxylase (ACOD1 or CAD, encoded by immune-responsive gene 1, Irg1) expression and itaconate production promote tumor immunogenicity and anti-tumor immune responses. Furthermore, we identify thimerosal, a vaccine preservative, as a specific inducer of IRG1 expression in tumor cells but not in macrophages, thereby enhancing tumor immunogenicity. Mechanistically, thimerosal induces itaconate production through a ROS-RIPK3-IRF1 signaling axis in tumor cells. Further, increased IRG1/itaconate upregulates antigen presentation-related gene expression via promoting TFEB nuclear translocation. Intratumoral injection of thimerosal induced itaconate production, activated the tumor immune microenvironment, and inhibited tumor growth in a T cell-dependent manner. Importantly, IRG1 deficiency markedly impaired tumor response to thimerosal treatment. Furthermore, itaconate induction by thimerosal potentiates the anti-tumor efficacy of adoptive T-cell therapy and anti-PD1 therapy in a mouse lymphoma model. Hence, our findings identify a new role for tumor intrinsic IRG1/itaconate in promoting tumor immunogenicity and provide a translational means to increase immunotherapy efficacy.
Collapse
Affiliation(s)
- Zining Wang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Lei Cui
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yanxun Lin
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Bitao Huo
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
- Metabolic Innovation Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Hongxia Zhang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Chunyuan Xie
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Huanling Zhang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yongxiang Liu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Huan Jin
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Hui Guo
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Mengyun Li
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Xiaojuan Wang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Penghui Zhou
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Peng Huang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
- Metabolic Innovation Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Jinyun Liu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
- Metabolic Innovation Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Hainan Academy of Medical Sciences, Hainan Medical University, Haikou, China
| | - Xiaojun Xia
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China.
- Hainan Academy of Medical Sciences, Hainan Medical University, Haikou, China.
| |
Collapse
|
40
|
Liu Z, Li S, Xiao Y, Liu X, Zhang B, Zeng Q, Ao Q, Zhang X. A Multi-Functional Nanoadjuvant Coupling Manganese with Toll-Like 9 Agonist Stimulates Potent Innate and Adaptive Anti-Tumor Immunity. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2402678. [PMID: 39258810 PMCID: PMC11538688 DOI: 10.1002/advs.202402678] [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: 03/14/2024] [Revised: 06/20/2024] [Indexed: 09/12/2024]
Abstract
The effectiveness of Toll-like 9 agonists (CpG) as an adjuvant for tumor immunotherapy is restricted due to their insufficient ability to activate anti-tumor immunity. To address that, the common nutrient metal ions are explored (Mn2+, Cu2+, Ca2+, Mg2+, Zn2+, Fe3+, and Al3+), identifying Mn2+ as a key enhancer of CpG to mediate immune activation by augmenting the STING-NF-κB pathway. Mn2+ and CpG are then self-assembled with epigallocatechin gallate (EGCG) into a nanoadjuvant MPN/CpG. Local delivery of MPN/CpG effectively inhibits tumor growth in a B16 melanoma-bearing mouse model, reshaping the tumor microenvironment (TME) by repolarizing M2-type tumor-associated macrophages (TAMs) to an M1-type and boosting intra-tumoral infiltration of CD8+/CD4+ T lymphocytes and DCs. Furthermore, compared to free CpG, MPN/CpG exhibits heightened accumulation in lymph nodes, enhancing CpG uptake and DC activation, consequently inducing significant antigen-specific cytotoxic CD8+ T cell immune response and humoral immunity. In a prophylactic tumor-bearing mouse model, MPN/CpG vaccination with OVA antigen significantly delays B16-OVA melanoma growth and extends mouse survival. These findings underscore the potential of MPN/CpG as a multifunctional adjuvant platform to drive powerful innate and adaptive immunity and regulate TME against tumors.
Collapse
Affiliation(s)
- Zhongjie Liu
- College of Biomedical EngineeringSichuan UniversityChengdu610064China
- NMPA Key Laboratory for Quality Research and Control of Tissue Regenerative Biomaterial & Institute of Regulatory Science for Medical Device & National Engineering Research Center for BiomaterialsSichuan UniversityChengduSichuan610064China
| | - Shu Li
- College of Biomedical EngineeringSichuan UniversityChengdu610064China
| | - Yang Xiao
- College of Biomedical EngineeringSichuan UniversityChengdu610064China
| | - Xiaoyang Liu
- Orthopedic Research Institution, Department of OrthopedicsWest China Hospital, Sichuan UniversityChengdu610041China
| | - Bin Zhang
- College of Biomedical EngineeringSichuan UniversityChengdu610064China
| | - Qin Zeng
- College of Biomedical EngineeringSichuan UniversityChengdu610064China
- NMPA Key Laboratory for Quality Research and Control of Tissue Regenerative Biomaterial & Institute of Regulatory Science for Medical Device & National Engineering Research Center for BiomaterialsSichuan UniversityChengduSichuan610064China
| | - Qiang Ao
- College of Biomedical EngineeringSichuan UniversityChengdu610064China
- NMPA Key Laboratory for Quality Research and Control of Tissue Regenerative Biomaterial & Institute of Regulatory Science for Medical Device & National Engineering Research Center for BiomaterialsSichuan UniversityChengduSichuan610064China
| | - Xingdong Zhang
- College of Biomedical EngineeringSichuan UniversityChengdu610064China
- NMPA Key Laboratory for Quality Research and Control of Tissue Regenerative Biomaterial & Institute of Regulatory Science for Medical Device & National Engineering Research Center for BiomaterialsSichuan UniversityChengduSichuan610064China
| |
Collapse
|
41
|
Laccourreye O, Garcia D, Holsinger FC, Weinstein GS. Ten-Year Outcome After Supracricoid Partial Laryngectomy in cT3M0 Laryngeal Squamous Cell Carcinoma-A STROBE Analysis. Laryngoscope 2024; 134:4557-4563. [PMID: 39152757 DOI: 10.1002/lary.31568] [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: 03/15/2024] [Revised: 05/10/2024] [Accepted: 05/28/2024] [Indexed: 08/19/2024]
Abstract
OBJECTIVES The aim of this study was to document 10-year outcomes after supracricoid partial laryngectomy (SCPL) in selected cT3M0 laryngeal squamous cell carcinoma (SCC) patients. METHODS This real-life retrospective observational study analyzed an inception cohort of 168 patients with isolated, untreated, selected cT3M0 laryngeal SCC, that were consecutively managed by SCPL during the period 1973-2013, and followed up until death or for a minimum of 10 years in 92% of cases at a single French academic and tertiary referral care center. Prior induction chemotherapy, arytenoid cartilage removal, level II-IV neck dissection, and postoperative radiation therapy were performed on 148, 77, 136, and 27 patients, respectively. The main objective was to determine 10-year actuarial local control and laryngeal preservation estimates. Secondary objectives included 10-year actuarial survival and cause-of-death analysis, and assessment of correlations between endpoints and clinical variables. The significance threshold was set at p < 0.005. RESULTS Ten-year actuarial local control, laryngeal preservation, and survival estimates were 90%, 85%, and 52%, respectively. Salvage treatment resulted in an overall 99% local control rate. Metachronous second primary cancer, intercurrent disease without evidence of SCC, SCPL-related death, and uncontrolled local recurrence accounted for 31%, 26%, 7%, and 2% of causes of death. On univariate analysis, overall local recurrence and laryngeal preservation rates varied significantly, from 5% to 54% and 90% to 46% when resection margins were R0 and R1, respectively. CONCLUSION The present study highlighted successful 10-year outcomes after SCPL, providing further evidence in favor of its integration into the conservative armamentarium for endolaryngeal cT3 SCC. LEVEL OF EVIDENCE 4 Laryngoscope, 134:4557-4563, 2024.
Collapse
Affiliation(s)
- Ollivier Laccourreye
- Service d'Otorhinolaryngologie et de Chirurgie Cervico-Faciale Hôpital Européen Georges Pompidou, Université Paris Cité, Paris, France
| | - Dominique Garcia
- Service d'Otorhinolaryngologie et de Chirurgie Cervico-Faciale Hôpital Européen Georges Pompidou, Université Paris Cité, Paris, France
| | - F Christopher Holsinger
- Division Head & Neck Surgery, Department of Otolaryngology, Stanford University, Palo Alto, California, U.S.A
| | - Gregory Steven Weinstein
- Penn Center for Head & Neck Cancer, Department of Otorhinolaryngology Head & Neck Surgery, Pennsylvania University, Philadelphia, Pennsylvania, U.S.A
| |
Collapse
|
42
|
Li X, Liu Y, Gui J, Gan L, Xue J. Cell Identity and Spatial Distribution of PD-1/PD-L1 Blockade Responders. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2400702. [PMID: 39248327 PMCID: PMC11538707 DOI: 10.1002/advs.202400702] [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: 01/19/2024] [Revised: 07/08/2024] [Indexed: 09/10/2024]
Abstract
The programmed death 1 (PD-1)/programmed death ligand 1 (PD-L1) axis inhibits T cell activity, impairing anti-tumor immunity. Blocking this axis with therapeutic antibodies is one of the most promising anti-tumor immunotherapies. It has long been recognized that PD-1/PD-L1 blockade reinvigorates exhausted T (TEX) cells already present in the tumor microenvironment (TME). However, recent advancements in high-throughput gene sequencing and bioinformatic tools have provided researchers with a more granular and dynamic insight into PD-1/PD-L1 blockade-responding cells, extending beyond the TME and TEX populations. This review provides an update on the cell identity, spatial distribution, and treatment-induced spatiotemporal dynamics of PD-1/PD-L1 blockade responders. It also provides a synopsis of preliminary reports of potential PD-1/PD-L1 blockade responders other than T cells to depict a panoramic picture. Important questions to answer in further studies and the translational and clinical potential of the evolving understandings are also discussed.
Collapse
Affiliation(s)
- Xintong Li
- Division of Thoracic Tumor Multimodality TreatmentState Key Laboratory of Biotherapy and Cancer CenterNational Clinical Research Center for GeriatricsWest China HospitalSichuan UniversityChengdu610041China
| | - Yuanxin Liu
- Division of Thoracic Tumor Multimodality TreatmentState Key Laboratory of Biotherapy and Cancer CenterNational Clinical Research Center for GeriatricsWest China HospitalSichuan UniversityChengdu610041China
| | - Jun Gui
- State Key Laboratory of Systems Medicine for CancerRenji‐Med X Clinical Stem Cell Research CenterRen Ji HospitalShanghai Jiao Tong University School of MedicineShanghai200127China
| | - Lu Gan
- Research Laboratory of Emergency MedicineDepartment of Emergency MedicineNational Clinical Research Center for GeriatricsWest China HospitalSichuan UniversityChengdu610041China
| | - Jianxin Xue
- Division of Thoracic Tumor Multimodality TreatmentState Key Laboratory of Biotherapy and Cancer CenterNational Clinical Research Center for GeriatricsLaboratory of Clinical Cell TherapyWest China HospitalSichuan UniversityChengdu610041China
| |
Collapse
|
43
|
Wang K, Zhu S, Zhang Y, Wang Y, Bian Z, Lu Y, Shao Q, Jin X, Xu X, Mo R. Targeting the GTPase RAN by liposome delivery for tackling cancer stemness-emanated therapeutic resistance. J Control Release 2024; 375:589-600. [PMID: 39245420 DOI: 10.1016/j.jconrel.2024.09.007] [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: 04/14/2024] [Revised: 09/02/2024] [Accepted: 09/03/2024] [Indexed: 09/10/2024]
Abstract
Cancer therapeutic resistance as a common hallmark of cancer is often responsible for treatment failure and poor patient survival. Cancer stem-like cells (CSCs) are one of the main contributors to therapeutic resistance, cancer relapse and metastasis. Through screening from our in-house library of natural products, we found polyphyllin II (PPII) as a potent anti-CSC compound for triple-negative breast cancer (TNBC). To enhance anti-CSC selectivity and improve druggability of PPII, we leverage the liposome-mediated delivery technique for increasing solubility of PPII, and more significantly, attaining broader therapeutic window. Liposomal PPII demonstrates its marked potency to inhibit tumor growth, post-surgical recurrence and metastasis compared to commercial liposomal chemotherapeutics in the mouse models of CSC-enriched TNBC tumor. We further identify PPII as an inhibitor of the Ras-related nuclear (RAN) protein whose upregulated expression is correlated with poor clinical outcomes. The direct binding of PPII to RAN reduces TNBC stemness, thereby suppressing tumor progression. Our work offers a significance from drug discovery to drug delivery benefiting from liposome technique for targeted treatment of high-stemness tumor.
Collapse
Affiliation(s)
- Kaili Wang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Jiangsu Key Laboratory of Drug Design and Optimization, Center of Advanced Pharmaceuticals and Biomaterials, School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China
| | - Sitong Zhu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Jiangsu Key Laboratory of Drug Design and Optimization, Center of Advanced Pharmaceuticals and Biomaterials, School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China
| | - Ying Zhang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Jiangsu Key Laboratory of Drug Design and Optimization, Center of Advanced Pharmaceuticals and Biomaterials, School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China
| | - Yuqian Wang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Jiangsu Key Laboratory of Drug Design and Optimization, Center of Advanced Pharmaceuticals and Biomaterials, School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China
| | - Zhenqian Bian
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Jiangsu Key Laboratory of Drug Design and Optimization, Center of Advanced Pharmaceuticals and Biomaterials, School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China
| | - Yougong Lu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Jiangsu Key Laboratory of Drug Design and Optimization, Center of Advanced Pharmaceuticals and Biomaterials, School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China
| | - Quanlin Shao
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Jiangsu Key Laboratory of Drug Design and Optimization, Center of Advanced Pharmaceuticals and Biomaterials, School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China
| | - Xiang Jin
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310032, China
| | - Xiaojun Xu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Jiangsu Key Laboratory of Drug Design and Optimization, Center of Advanced Pharmaceuticals and Biomaterials, School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China; Department of Pharmacy, the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Center for Innovative Traditional Chinese Medicine Target and New Drug Research, International Institutes of Medicine, Zhejiang University, Yiwu 322001, Zhejiang, China.
| | - Ran Mo
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Jiangsu Key Laboratory of Drug Design and Optimization, Center of Advanced Pharmaceuticals and Biomaterials, School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China.
| |
Collapse
|
44
|
Li J, Yi H, Fu Y, Zhuang J, Zhan Z, Guo L, Zheng J, Yu X, Zhang DY. Biodegradable iridium coordinated nanodrugs potentiate photodynamic therapy and immunotherapy of lung cancer. J Colloid Interface Sci 2024; 680:9-24. [PMID: 39488900 DOI: 10.1016/j.jcis.2024.10.156] [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: 09/05/2024] [Revised: 10/24/2024] [Accepted: 10/25/2024] [Indexed: 11/05/2024]
Abstract
Hypoxia, which is a common characteristic of most solid tumors, not only contributes to the immunosuppressive nature of the tumor microenvironment (TME) but also reduces the efficacy of many oxygen-depleting therapies, including photodynamic therapy (PDT). In this study, we developed acidity-responsive biodegradable iridium-coordinated (IPC) nanodrugs consisting of iridium ions, the photosensitizer chlorin e6 (Ce6), and polyvinylpyrrolidone to potentiate the effects of PDT and immunotherapy by modulating the TME. IPC nanodrugs that accumulate at high levels in tumors catalyze excess hydrogen peroxide to produce oxygen while depleting glutathione levels within cancer cells; thus, the released Ce6 is more efficient at producing reactive oxygen species (ROS) in response to laser irradiation. In addition, IPC nanodrugs alleviate tumor hypoxia, induce more immunogenic cell death by amplifying PDT responses, and synergistically inhibit tumor growth by initiating robust antitumor immunity and reversing the immunosuppressive nature of the TME. As a result, IPC nanodrugs exert pronounced combined therapeutic effects in vitro and in vivo, without obvious toxic effects due to acidity-responsive degradation. These iridium-coordinated nanodrugs have the potential to modulate the TME, amplify the effects of PDT, and substantially inhibit tumors, and they are expected to provide novel ideas for combination therapy of hypoxic cancer.
Collapse
Affiliation(s)
- Jingyao Li
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, The Fifth Affiliated Hospital and School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Huixi Yi
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, The Fifth Affiliated Hospital and School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Yuanyuan Fu
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, The Fifth Affiliated Hospital and School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Jiani Zhuang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, The Fifth Affiliated Hospital and School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Zhixiong Zhan
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, The Fifth Affiliated Hospital and School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Liyou Guo
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, The Fifth Affiliated Hospital and School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Ji Zheng
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400037, China; State Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical University (Army Medical University), Chongqing 400042, China.
| | - Xiyong Yu
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, The Fifth Affiliated Hospital and School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China.
| | - Dong-Yang Zhang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, The Fifth Affiliated Hospital and School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China.
| |
Collapse
|
45
|
Sætersmoen M, Kotchetkov IS, Torralba-Raga L, Mansilla-Soto J, Sohlberg E, Krokeide SZ, Hammer Q, Sadelain M, Malmberg KJ. Targeting HLA-E-overexpressing cancers with a NKG2A/C switch receptor. MED 2024:100521. [PMID: 39423821 DOI: 10.1016/j.medj.2024.09.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 06/06/2024] [Accepted: 09/23/2024] [Indexed: 10/21/2024]
Abstract
BACKGROUND Human leukocyte antigen (HLA)-E is overexpressed by a large proportion of solid tumors, including malignant glioblastoma, and acts as a major checkpoint for NKG2A+ CD8+ T cells and natural killer (NK) cells in the tumor microenvironment and circulation. This axis operates alongside PD-L1 to inhibit effector responses by T and NK cells. METHODS We engineered a chimeric A/C switch receptor, combining the high HLA-E binding affinity of the NKG2A receptor ectodomain with the activating signaling of the NKG2C receptor endodomain. The cytotoxic function of A/C switch-transduced NK and T cells was evaluated against tumor cells with varying levels of HLA-E expression. In vivo efficacy was assessed using a xenograft model of glioblastoma. FINDINGS A/C switch-transduced NK and T cells exhibited superior and specific cytotoxicity against tumor cells with medium to high HLA-E expression. A/C switch-expressing human T cells demonstrated enhanced anti-tumor function in a glioblastoma xenograft model. The activity of the modified T cells was governed by an equilibrium between A/C switch levels and HLA-E expression, creating a therapeutic window to minimize on-target, off-tumor toxicities. Normal cells remained insensitive to A/C switch T cells, even after interferon (IFN)-γ pretreatment to induce HLA-E expression. CONCLUSIONS The A/C switch receptor effectively targets tumor cells expressing high levels of HLA-E, either alone or in combination with other engineered specificities, to overcome the suppressive NKG2A/HLA-E checkpoint. This approach offers a promising therapeutic strategy with a favorable safety profile for targeting HLA-E-overexpressing tumors. FUNDING This work was funded by The Research Council of Norway, the Norwegian Cancer Society, and the National Cancer Institute.
Collapse
Affiliation(s)
- Michelle Sætersmoen
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Ivan S Kotchetkov
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Center for Cell Engineering and Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Lamberto Torralba-Raga
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Jorge Mansilla-Soto
- Center for Cell Engineering and Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ebba Sohlberg
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Silje Zandstra Krokeide
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Quirin Hammer
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Michel Sadelain
- Center for Cell Engineering and Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Karl-Johan Malmberg
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway.
| |
Collapse
|
46
|
Zhou W, Qu M, Yue Y, Zhong Z, Nan K, Sun X, Wu Q, Zhang J, Chen W, Miao C. Acetylcysteine synergizes PD-1 blockers against colorectal cancer progression by promoting TCF1 +PD1 +CD8 + T cell differentiation. Cell Commun Signal 2024; 22:503. [PMID: 39420342 PMCID: PMC11484120 DOI: 10.1186/s12964-024-01848-8] [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: 05/22/2024] [Accepted: 09/23/2024] [Indexed: 10/19/2024] Open
Abstract
BACKGROUND Programmed cell death protein 1 (PD-1) blockade is essential in treating progressive colorectal cancer (CRC). However, some patients with CRC do not respond well to immunotherapy, possibly due to the exhaustion of CD8+ T cells in the tumor microenvironment. N-Acetylcysteine (NAC) can reduce CD8+ T cell exhaustion in vitro and induce their differentiation into long-lasting phenotypes, thus enhancing the anti-tumor effect of adoptive T cell transfer. However, whether NAC can be combined with PD-1 blockade in CRC treatment and how NAC regulates CD8+ T cell differentiation remain unclear. Hence, in this study, we aimed to investigate whether NAC has a synergistic effect with PD-1 blockers against CRC progression. METHODS We constructed a mouse CRC model to study the effect of NAC on tumors. The effect of NAC on CD8 + T cell differentiation and its potential mechanism were explored using cell flow assay and other studies in vitro and ex vivo. RESULTS We demonstrated that NAC synergized PD-1 antibodies to inhibit CRC progression in a mouse CRC model mediated by CD8+ T cells. We further found that NAC can induce TCF1+PD1+CD8+ T cell differentiation and reduce the formation of exhausted T cells in vitro and in vivo. Moreover, NAC enhanced the expression of Glut4 in CD8+ T cells, promoting the differentiation of TCF1+PD1+CD8+ T cells. CONCLUSIONS Our study provides a novel idea for immunotherapy for clinically progressive CRC and suggests that Glut4 may be a new immunometabolic molecular target for regulating CD8+ T cell differentiation.
Collapse
Affiliation(s)
- Wenchang Zhou
- Department of Anesthesiology, Zhongshan Hospital, Cancer Center, Zhongshan Hospital, Fudan University, Fudan University, 180# Feng-Lin Road, Shanghai, 200032, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Mengdi Qu
- Department of Anesthesiology, Zhongshan Hospital, Cancer Center, Zhongshan Hospital, Fudan University, Fudan University, 180# Feng-Lin Road, Shanghai, 200032, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Ying Yue
- Department of Anesthesiology, Zhongshan Hospital, Cancer Center, Zhongshan Hospital, Fudan University, Fudan University, 180# Feng-Lin Road, Shanghai, 200032, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Ziwen Zhong
- Department of Anesthesiology, Zhongshan Hospital, Cancer Center, Zhongshan Hospital, Fudan University, Fudan University, 180# Feng-Lin Road, Shanghai, 200032, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Ke Nan
- Department of Anesthesiology, Zhongshan Hospital, Cancer Center, Zhongshan Hospital, Fudan University, Fudan University, 180# Feng-Lin Road, Shanghai, 200032, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Xingfeng Sun
- Department of Anesthesiology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, 200438, China
| | - Qichao Wu
- Department of Anesthesiology, Zhongshan Hospital, Cancer Center, Zhongshan Hospital, Fudan University, Fudan University, 180# Feng-Lin Road, Shanghai, 200032, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Jie Zhang
- Department of Anesthesiology, Zhongshan Hospital, Cancer Center, Zhongshan Hospital, Fudan University, Fudan University, 180# Feng-Lin Road, Shanghai, 200032, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Wankun Chen
- Department of Anesthesiology, Zhongshan Hospital, Cancer Center, Zhongshan Hospital, Fudan University, Fudan University, 180# Feng-Lin Road, Shanghai, 200032, China.
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China.
| | - Changhong Miao
- Department of Anesthesiology, Zhongshan Hospital, Cancer Center, Zhongshan Hospital, Fudan University, Fudan University, 180# Feng-Lin Road, Shanghai, 200032, China.
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China.
| |
Collapse
|
47
|
Ding Q, Weng Y, Li Y, Lin W, Lin X, Lin T, Yang H, Xu W, Wang J, Ying H, Qiu S. Inhibition of PNCK inflames tumor microenvironment and sensitizes head and neck squamous cell carcinoma to immune checkpoint inhibitors. J Immunother Cancer 2024; 12:e009893. [PMID: 39395840 PMCID: PMC11474745 DOI: 10.1136/jitc-2024-009893] [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] [Accepted: 09/26/2024] [Indexed: 10/14/2024] Open
Abstract
BACKGROUND The landscape of the tumor microenvironment (TME) is intricately linked to the development of head and neck squamous cell carcinoma (HNSCC) and significantly influences immunotherapy efficacy. Recent research has underscored the pivotal role of PNCK in cancer progression, yet its relationship with immunotherapy remains elusive. METHODS We leveraged sequencing data from our cohort and public databases to evaluate PNCK expression, prognostic significance, and immune efficacy prediction. In vitro and in vivo experiments explored the role of PNCK in HNSCC progression. Animal models assessed the therapeutic effects and survival benefits of PNCK knockdown combined with immune checkpoint inhibitors (ICIs). Single-cell transcriptomics analyzed the impact of PNCK on the TME, and proteomic studies elucidated the mechanisms. RESULTS PNCK exerts multifaceted critical roles in the progression of HNSCC. Lower PNCK expression is associated with improved prognosis, enhanced immune cell infiltration, and increased responsiveness to ICIs. Conversely, PNCK promotes HNSCC cell migration, invasion, proliferation, colony formation, zebrafish angiogenesis, and tumor growth in mice. Moreover, targeting PNCK enhances sensitivity to ICIs and leads to significant alterations in the T-cell and B-cell ratios within the TME. These changes are attributed to the inhibition of nuclear transcription of PNCK-phosphorylated ZEB1, which restricts cytokine release and inflames the immune microenvironment to regulate the TME. CONCLUSIONS Inhibition of PNCK may be a potential strategy for treating HNSCC, as it may activate the immune response and improve the TME, thereby enhancing the efficacy of immunotherapy for HNSCC patients.
Collapse
Affiliation(s)
- Qin Ding
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital (Fujian Branch of Fudan University Shanghai Cancer Center), Fuzhou, Fujian, China
- Fujian Provincial Key Laboratory of Translational Cancer Medicine, Fuzhou, Fujian, China
| | - Youliang Weng
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital (Fujian Branch of Fudan University Shanghai Cancer Center), Fuzhou, Fujian, China
- Fujian Provincial Key Laboratory of Translational Cancer Medicine, Fuzhou, Fujian, China
| | - Ying Li
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital (Fujian Branch of Fudan University Shanghai Cancer Center), Fuzhou, Fujian, China
- Fujian Provincial Key Laboratory of Translational Cancer Medicine, Fuzhou, Fujian, China
| | - Wanzun Lin
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital (Fujian Branch of Fudan University Shanghai Cancer Center), Fuzhou, Fujian, China
- Fujian Provincial Key Laboratory of Translational Cancer Medicine, Fuzhou, Fujian, China
| | - Xiaosan Lin
- Department of Stomatology, Affiliated Fuzhou First Hospital of Fujian Medical University, Fuzhou, Fujian, China
| | - Tingting Lin
- Department of Medical and Radiation Oncology, Affiliated Sanming First Hospital, Fujian Medical University, Sanming, Fujian, China
| | - Hanxuan Yang
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital (Fujian Branch of Fudan University Shanghai Cancer Center), Fuzhou, Fujian, China
- Fujian Provincial Key Laboratory of Translational Cancer Medicine, Fuzhou, Fujian, China
| | - Wenqian Xu
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital (Fujian Branch of Fudan University Shanghai Cancer Center), Fuzhou, Fujian, China
- Fujian Provincial Key Laboratory of Translational Cancer Medicine, Fuzhou, Fujian, China
| | - Jianming Wang
- Innovation Center for Cancer Research, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital (Fujian Branch of Fudan University Shanghai Cancer Center), Fuzhou, Fujian, China
- Fujian Key Laboratory of Advanced Technology for Cancer Screening and Early Diagnosis, Fujian Cancer Hospital (Fujian Branch of Fudan University Shanghai Cancer Center), Fuzhou, Fujian, China
| | - Hongmei Ying
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
| | - Sufang Qiu
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital (Fujian Branch of Fudan University Shanghai Cancer Center), Fuzhou, Fujian, China
- Fujian Provincial Key Laboratory of Translational Cancer Medicine, Fuzhou, Fujian, China
| |
Collapse
|
48
|
He Q, Zhang Y, Li W, Chen S, Xiong J, Zhao R, Yuan K, Hu Q, Liu S, Gao G, Bedford MT, Tang DG, Xu B, Zou C, Zhang D. Inhibition of PRMT5 moderately suppresses prostate cancer growth in vivo but enhances its response to immunotherapy. Cancer Lett 2024; 602:217214. [PMID: 39218291 DOI: 10.1016/j.canlet.2024.217214] [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: 05/07/2024] [Revised: 08/11/2024] [Accepted: 08/28/2024] [Indexed: 09/04/2024]
Abstract
Protein arginine methylation is a common post-translational modification (PTM) catalyzed by nine protein arginine methyltransferases (PRMTs). As the major symmetric arginine methyltransferase that methylates both histone and non-histone substrates, PRMT5 plays key roles in a number of biological processes critical for development and tumorigenesis. PRMT5 overexpression has been reported in multiple cancer types including prostate cancer (PCa), but the exact biological and mechanistic understanding of PRMT5 in aggressive PCa remains ill-defined. Here, we show that PRMT5 is upregulated in PCa, correlates with worse patient survival, promotes corrupted RNA splicing, and functionally cooperates with an array of pro-tumorigenic pathways to enhance oncogenesis. PRMT5 inhibition via either genetic knockdown or pharmacological inhibition reduces stemness with paralleled differentiation and arrests cell cycle progression without causing appreciable apoptosis. Strikingly, the severity of antitumor effect of PRMT5 inhibition correlates with disease aggressiveness, with AR+ PCa being less affected. Molecular characterization pinpoints MYC, but not (or at least to a lesser degree) AR, as the main partner of PRMT5 to form a positive feedback loop to exacerbate malignancy in both AR+ and AR- PCa cells. Inspired by the surprising finding that PRMT5 negatively correlates with tumor immune infiltration and transcriptionally suppresses an immune-gene program, we further show that although PRMT5 inhibitor (PRMT5i) EPZ015666 or anti-PD-1 immunotherapy alone exhibits limited antitumor effects, combination of PRMT5i with anti-PD-1 displays superior efficacy in inhibiting castration-resistant PCa (CRPC) in vivo. Finally, to expand the potential use of PRMT5i through a synthetic lethality concept, we also perform a global CRISPR/Cas9 knockout screen to unravel that many clinical-grade drugs of known oncogenic pathways can be repurposed to target CRPC when used in combination with PRMT5i at low doses. Collectively, our findings establish a rationale to exploit PRMT5i in combination with immunotherapy or other targeted therapies to treat aggressive PCa.
Collapse
Affiliation(s)
- Qinju He
- Affiliated Hospital of Hunan University, School of Biomedical Sciences, Hunan University, Changsha, China; Hunan Key Laboratory of Animal Models and Molecular Medicine, Hunan University, Changsha, 410082, China
| | - Yuanzhen Zhang
- Affiliated Hospital of Hunan University, School of Biomedical Sciences, Hunan University, Changsha, China; Hunan Key Laboratory of Animal Models and Molecular Medicine, Hunan University, Changsha, 410082, China
| | - Wenchao Li
- Department of Urology, Affiliated Zhongda Hospital of Southeast University, Nanjing, 210009, China
| | - Saisai Chen
- Department of Urology, Affiliated Zhongda Hospital of Southeast University, Nanjing, 210009, China
| | - Jiangling Xiong
- Hunan Key Laboratory of Animal Models and Molecular Medicine, Hunan University, Changsha, 410082, China
| | - Ruizhe Zhao
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA; Current Address: Department of Urology, First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Kai Yuan
- Hunan Key Laboratory of Molecular Precision Medicine, Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, 410000, China
| | - Qiang Hu
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, 14263, New York, USA
| | - Song Liu
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, 14263, New York, USA
| | - Guozhen Gao
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Mark T Bedford
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Dean G Tang
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Bin Xu
- Department of Urology, Affiliated Zhongda Hospital of Southeast University, Nanjing, 210009, China.
| | - Cheng Zou
- Affiliated Hospital of Hunan University, School of Biomedical Sciences, Hunan University, Changsha, China; Hunan Key Laboratory of Animal Models and Molecular Medicine, Hunan University, Changsha, 410082, China; Shenzhen Research Institute, Hunan University, Shenzhen, 518000, China.
| | - Dingxiao Zhang
- Affiliated Hospital of Hunan University, School of Biomedical Sciences, Hunan University, Changsha, China; Hunan Key Laboratory of Animal Models and Molecular Medicine, Hunan University, Changsha, 410082, China; Shenzhen Research Institute, Hunan University, Shenzhen, 518000, China.
| |
Collapse
|
49
|
Cheng F, He L, Wang J, Lai L, Ma L, Qu K, Yang Z, Wang X, Zhao R, Weng L, Wang L. Synergistic immunotherapy with a calcium-based nanoinducer: evoking pyroptosis and remodeling tumor-associated macrophages for enhanced antitumor immune response. NANOSCALE 2024; 16:18570-18583. [PMID: 39291343 DOI: 10.1039/d4nr01497a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
The challenges posed by low immunogenicity and the immunosuppressive tumor microenvironment (TME) significantly hinder the efficacy of cancer immunotherapy. Pyroptosis, characterized as a pro-inflammatory cell death pathway, emerges as a promising approach to augment immunotherapy by promoting immunogenic cell death (ICD). The predominance of M2 phenotype tumor-associated macrophages (TAMs) in the TME underscores the critical need for TAM reprogramming to mitigate this immunosuppression. Herein, we introduce a calcium-based, intelligent-responsive nanoinducer (CaZCH NPs), designed to concurrently initiate pyroptosis and remodel TAMs, thereby amplifying antitumor immunotherapy effects. Modified with hyaluronic acid, CaZCH NPs can target tumor cells. Once internalized, CaZCH NPs respond to the acidic environment, releasing Ca2+, curcumin and H2O2 to induce mitochondrial Ca2+ overload and oxidation stress, leading to caspase-3/GSDME-mediated cell pyroptosis. Concurrently, O2 produced by CaZCH and pro-inflammatory cytokines from pyroptotic cells work together to shift TAM polarization towards the M1 phenotype, effectively countering TME's immunosuppressive effect. Notably, the synergistic effect of Ca2+-mediated pyroptosis and TAM remodeling demonstrates superior antitumor efficiency in colorectal cancer models. The induced ICD, coupled with M1-type TAMs, effectively enhances immunogenicity and mitigates immunosuppression, promoting dendritic cell maturation and activating CD8+ T cell-dependent systemic antitumor immunity. Our study presents a promising synergistic strategy for achieving highly efficient immunotherapy using a simple calcium-based nanoinducer.
Collapse
Affiliation(s)
- Fang Cheng
- State Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), Nanjing University of Posts and Telecommunications, Nanjing 210023, P. R. China.
| | - Lei He
- State Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), Nanjing University of Posts and Telecommunications, Nanjing 210023, P. R. China.
| | - Jiaqi Wang
- State Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), Nanjing University of Posts and Telecommunications, Nanjing 210023, P. R. China.
| | - Lunhui Lai
- State Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), Nanjing University of Posts and Telecommunications, Nanjing 210023, P. R. China.
| | - Li Ma
- State Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), Nanjing University of Posts and Telecommunications, Nanjing 210023, P. R. China.
| | - Kuiming Qu
- State Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), Nanjing University of Posts and Telecommunications, Nanjing 210023, P. R. China.
| | - Zicheng Yang
- State Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), Nanjing University of Posts and Telecommunications, Nanjing 210023, P. R. China.
| | - Xinyue Wang
- State Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), Nanjing University of Posts and Telecommunications, Nanjing 210023, P. R. China.
| | - Ruyu Zhao
- State Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), Nanjing University of Posts and Telecommunications, Nanjing 210023, P. R. China.
| | - Lixing Weng
- State Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), Nanjing University of Posts and Telecommunications, Nanjing 210023, P. R. China.
| | - Lianhui Wang
- State Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), Nanjing University of Posts and Telecommunications, Nanjing 210023, P. R. China.
| |
Collapse
|
50
|
Liu S, Guo H, Li D, Wang C. Immunologically effective biomaterials enhance immunotherapy of prostate cancer. J Mater Chem B 2024; 12:9821-9834. [PMID: 39239675 DOI: 10.1039/d3tb03044j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/07/2024]
Abstract
Prostate cancer (PCa) is one of the most common malignant neoplasms affecting the male population. The onset of the disease is insidious and often associated with severe consequences, such as bone metastases at the time of initial diagnosis. Once it advances to metastatic castration-resistant PCa (mCRPC), conventional treatment methods become ineffective. As research on the mechanism of tumor therapy advances, immunotherapy has been evolving rapidly. However, PCa is a solid tumor type that primarily faces the challenges of poor immunogenicity and inhibitory tumor microenvironment (TME). Fortunately, the extensive use of biomaterials has led to continuous advancement in PCa immunotherapy. These innovative materials aim to address intractable issues, such as immune escape and immune desert, to inhibit tumor progression and metastasis. This detailed review focuses on the regulation of different aspects of tumor immunity by immunologically effective biomaterials, including modulating adaptive immunity, innate immunity, and the immune microenvironment, to enhance the efficacy of PCa immunotherapy. In addition, this review provides a perspective on the future prospects of immunotherapeutic nanoplatforms based on biomaterials in the treatment of PCa.
Collapse
Affiliation(s)
- Siqi Liu
- Department of Urology, General Surgery Center, The First Hospital of Jilin University, 1 Xinmin Street, Changchun 130061, P. R. China
| | - Hui Guo
- Department of Urology, General Surgery Center, The First Hospital of Jilin University, 1 Xinmin Street, Changchun 130061, P. R. China
| | - Di Li
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, 1 Xinmin Street, Changchun 130061, P. R. China
| | - Chunxi Wang
- Department of Urology, General Surgery Center, The First Hospital of Jilin University, 1 Xinmin Street, Changchun 130061, P. R. China
| |
Collapse
|