1
|
Tan M, Gao Z, Wang X, Wang X, Lin C, Huang Y, Chen W, Zhang Y, Hou Z. MnO 2@CeO x-GAMP radiosensitizer with oxygen vacancies depended mimicking enzyme-like activities for radiosensitization-mediated STING pathway activation. Biomaterials 2025; 314:122797. [PMID: 39255531 DOI: 10.1016/j.biomaterials.2024.122797] [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: 05/30/2024] [Revised: 08/31/2024] [Accepted: 08/31/2024] [Indexed: 09/12/2024]
Abstract
Activation of the stimulator of interferon genes (STING) pathway by radiotherapy (RT) has a significant effect on eliciting antitumor immune responses. The generation of hydroxyl radical (·OH) storm and the sensitization of STING-relative catalytic reactions could improve radiosensitization-mediated STING activation. Herein, multi-functional radiosensitizer with oxygen vacancies depended mimicking enzyme-like activities was fabricated to produce more dsDNA which benefits intracellular 2', 3'-cyclic GMP-AMP (cGAMP) generation, together with introducing exogenous cGAMP to activate immune response. MnO2@CeOx nanozymes present enhanced superoxide dismutase (SOD)-like and peroxidase (POD)-like activities due to induced oxygen vacancies accelerate the redox cycles from Ce4+ to Ce3+ via intermetallic charge transfer. CeOx shells not only serve as radiosensitizer, but also provide the conjugation site for AMP/GMP to form MnO2@CeOx-GAMP (MCG). Upon X-ray irradiation, MCG with SOD-like activity facilitates the conversion of superoxide anions generated by Ce-sensitization into H2O2 within tumor microenvironment (TME). The downstream POD-like activity catalyzes the elevated H2O2 into a profusion of ·OH for producing more damage DNA fragments. TME-responsive decomposed MCG could supply exogenous cGAMP, meanwhile the releasing Mn2+ improve the sensitivity of cyclic GMP-AMP synthase to dsDNA for producing more cGAMP, resulting in the promotion of STING pathway activation.
Collapse
Affiliation(s)
- Meiling Tan
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, PR China; Guangzhou Institute of Cancer Research, The Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, 510095, PR China
| | - Zhimin Gao
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, PR China
| | - Xinyi Wang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, PR China
| | - Xiaozhao Wang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, PR China
| | - Chen Lin
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, PR China
| | - Yongxin Huang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, PR China
| | - Wei Chen
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, PR China; The Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, 511518, PR China
| | - Yaru Zhang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, PR China; The Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, 511518, PR China
| | - Zhiyao Hou
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, PR China; Guangzhou Institute of Cancer Research, The Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, 510095, PR China; The Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, 511518, PR China.
| |
Collapse
|
2
|
Zhang J, Lu L, Zhang W, Miao Y, Du H, Xia H, Tao Z, Du Z, Tang Y, Fang Q. Gadolinium ion-loaded mesoporous organosilica nanoplatform for enhanced radiotherapy in breast tumor treatment. Colloids Surf B Biointerfaces 2025; 246:114374. [PMID: 39541910 DOI: 10.1016/j.colsurfb.2024.114374] [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: 08/19/2024] [Revised: 10/22/2024] [Accepted: 11/10/2024] [Indexed: 11/17/2024]
Abstract
Triple-negative breast cancer (TNBC) is a highly aggressive subtype with limited therapeutic options, often exhibiting resistance to standard radiotherapy (RT) and chemotherapy. Recent advancements in nanomedicine provide an opportunity to enhance treatment efficacy through innovative drug delivery systems and radiosensitizers. In this study, we present a novel nanotheranostic platform, MOs-G@DOX, engineered to enhance the therapeutic efficacy of RT in the treatment of TNBC. This platform consists of gadolinium-containing mesoporous organosilica nanoparticles (MOs-G) that serve a dual function as a drug carrier and a radiosensitizer. The MOs-G were synthesized via a surfactant-mediated sol-gel process, followed by gadolinium incorporation through nanoprecipitation. The antitumor drug doxorubicin (DOX) was subsequently loaded into the mesoporous structure, forming the MOs-G@DOX nanoplatform. Comprehensive in vitro and in vivo studies demonstrated that MOs-G@DOX exhibits excellent biocompatibility and significantly enhances the radiosensitivity of TNBC cells, leading to superior tumor growth inhibition compared to conventional treatments. The stability of MOs-G, with minimal gadolinium ion leakage, further underscores its potential as a safe and effective nanomedicine. Additionally, the combination of MOs-G@DOX with RT showed a marked increase in reactive oxygen species (ROS) generation and tumor cell apoptosis, which were confirmed through histological analyses. These findings suggest that MOs-G@DOX is a promising candidate for advancing cancer therapy, particularly in the context of RT for TNBC.
Collapse
Affiliation(s)
- Junjie Zhang
- School of Fundamental Sciences, Bengbu Medical University, Bengbu, Anhui Province 233030, China.
| | - Li Lu
- School of Fundamental Sciences, Bengbu Medical University, Bengbu, Anhui Province 233030, China
| | - Wenqing Zhang
- School of Fundamental Sciences, Bengbu Medical University, Bengbu, Anhui Province 233030, China
| | - Yuchen Miao
- School of Fundamental Sciences, Bengbu Medical University, Bengbu, Anhui Province 233030, China
| | - Hengda Du
- School of Fundamental Sciences, Bengbu Medical University, Bengbu, Anhui Province 233030, China
| | - Hui Xia
- Department of Microbiology and Parasitology, Bengbu Medical University, Bengbu, Anhui Province 233030, China; Anhui Key Laboratory of Infection and Immunity, Bengbu Medical University, Bengbu, Anhui Province 233030, China
| | - Zhiyong Tao
- Department of Microbiology and Parasitology, Bengbu Medical University, Bengbu, Anhui Province 233030, China; Anhui Key Laboratory of Infection and Immunity, Bengbu Medical University, Bengbu, Anhui Province 233030, China
| | - Zhaofeng Du
- School of Fundamental Sciences, Bengbu Medical University, Bengbu, Anhui Province 233030, China
| | - Yulong Tang
- School of Fundamental Sciences, Bengbu Medical University, Bengbu, Anhui Province 233030, China
| | - Qiang Fang
- School of Fundamental Sciences, Bengbu Medical University, Bengbu, Anhui Province 233030, China; Department of Microbiology and Parasitology, Bengbu Medical University, Bengbu, Anhui Province 233030, China; Anhui Key Laboratory of Infection and Immunity, Bengbu Medical University, Bengbu, Anhui Province 233030, China.
| |
Collapse
|
3
|
Yao X, Huo W, Wang Y, Xia D, Chen Y, Tang Y, Tang H, Yang W, Liu Y, Xue J, Yuan Q, Gao X, Cao K. Environmental Low-Dose Radiation Activates Th1 Immunity through the Mitochondria-STING Pathway. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2024. [PMID: 39689952 DOI: 10.1021/acs.est.4c08009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2024]
Abstract
The presence of low-dose radiation (LDR) in the environment has become more prevalent. However, the effect of LDR exposure on the immune system remains elusive. Here, we interestingly found that LDR specifically elevated the percentage of CD4+IFNγ+ Th1 splenocytes, both in vitro and in vivo, without affecting the percentage of CD8+IFNγ+ Tc1 cells and regulatory T cells. A similar phenomenon was found in T cells from peripheral blood. Mechanistically, we found that LDR can induce mitochondrial damage, which stimulated the STING signaling pathway, leading to the enhanced expression of T-bet, the master transcriptional factor of Th1-cell differentiation. The specific STING signal inhibitor can abrogate the effect of LDR on Th1 differentiation, confirming the central role of the STING pathway. To further validate the immunoregulatory role of LDR, we exposed mice with whole body LDR and evaluated if LDR could protect mice against triple-negative breast cancer through enhanced antitumor immunity. As expected, LDR significantly delayed tumor development and promoted cell death. Meanwhile, LDR resulted in increased tumor-infiltrating Th1 cells, while the proportion of Tc1 and Treg cells remained unchanged. Furthermore, the infiltration of antitumor macrophages was also increased. In summary, we revealed that environmental LDR could specifically regulate Th1 T-cell activities, providing critical information for the potential application of LDR in both clinical and nonclinical settings.
Collapse
Affiliation(s)
- Xiuxiu Yao
- College of Materials Science and Engineering, Beijing University of Technology, Beijing 100124, China
| | - Wendi Huo
- College of Materials Science and Engineering, Beijing University of Technology, Beijing 100124, China
| | - Yuchen Wang
- Department of Chemistry, College of Chemistry and Life Science, Beijing University of Technology, Beijing 100124, China
| | - Dongfang Xia
- College of Materials Science and Engineering, Beijing University of Technology, Beijing 100124, China
| | - Yan Chen
- College of Materials Science and Engineering, Beijing University of Technology, Beijing 100124, China
| | - Yuhua Tang
- College of Materials Science and Engineering, Beijing University of Technology, Beijing 100124, China
| | - Huayong Tang
- Department of Chemistry, College of Chemistry and Life Science, Beijing University of Technology, Beijing 100124, China
| | - Wenjiang Yang
- Beijing Engineering Research Center of Radiographic Techniques and Equipment, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing 100049, China
| | - Yu Liu
- Beijing Engineering Research Center of Radiographic Techniques and Equipment, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing 100049, China
| | - Jingquan Xue
- Beijing Engineering Research Center of Radiographic Techniques and Equipment, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing 100049, China
| | - Qing Yuan
- Department of Chemistry, College of Chemistry and Life Science, Beijing University of Technology, Beijing 100124, China
| | - Xueyun Gao
- Department of Chemistry, College of Chemistry and Life Science, Beijing University of Technology, Beijing 100124, China
| | - Kai Cao
- Department of Chemistry, College of Chemistry and Life Science, Beijing University of Technology, Beijing 100124, China
| |
Collapse
|
4
|
Zhan S, Cao Z, Li J, Chen F, Lai X, Yang W, Teng Y, Li Z, Zhang W, Xie J. Iron Oxide Nanoparticles Induce Macrophage Secretion of ATP and HMGB1 to Enhance Irradiation-Led Immunogenic Cell Death. Bioconjug Chem 2024. [PMID: 39680043 DOI: 10.1021/acs.bioconjchem.4c00488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
ATP (adenosine triphosphate) and HMGB1 (high mobility group box 1 protein) are key players in treatments that induce immunogenic cell death (ICD). However, conventional therapies, including radiotherapy, are often insufficient to induce ICD. In this study, we explore a strategy using nanoparticle-loaded macrophages as a source of ATP and HMGB1 to complement radiation-induced intrinsic and adaptive immune responses. To this end, we tested three inorganic particles, namely, iron oxide nanoparticles (ION), aluminum oxide nanoparticles (AON), and zinc oxide nanoparticles (ZON), in vitro with bone marrow-derived dendritic cells (BMDCs) and then in vivo in syngeneic tumor models. Our results showed that ION was the most effective of the three nanoparticles in promoting the secretion of ATP and HMGB1 from macrophages without negatively affecting macrophage survival. Secretions from ION-loaded macrophages can activate BMDCs. Intratumoral injection of ION-loaded macrophages significantly enhanced tumor infiltration and activation of dendritic cells and cytotoxic T cells. Moreover, exogenous ION macrophages can enhance the efficacy of radiotherapy. In addition, direct injection of ION can also enhance the efficacy of radiotherapy, which is attributed to ION uptake by and stimulation of endogenous macrophages. Instead of directly targeting cancer cells, our strategy targets macrophages and uses them as a secretory source of ATP and HMGB1 to enhance radiation-induced ICD. Our research introduces a new nanoparticle-based immunomodulatory approach that may have applications in radiotherapy and beyond.
Collapse
Affiliation(s)
- Shuyue Zhan
- Department of Chemistry, University of Georgia, Athens, Georgia 30602, United States
| | - Zhengwei Cao
- Department of Chemistry, University of Georgia, Athens, Georgia 30602, United States
| | - Jianwen Li
- Department of Chemistry, University of Georgia, Athens, Georgia 30602, United States
| | - Fanghui Chen
- Department of Hematology and Medical Oncology & Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia 30322, United States
| | - Xinning Lai
- Department of Chemistry, University of Georgia, Athens, Georgia 30602, United States
| | - Wei Yang
- Department of Chemistry, University of Georgia, Athens, Georgia 30602, United States
| | - Yong Teng
- Department of Hematology and Medical Oncology & Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia 30322, United States
| | - Zibo Li
- Department of Radiology, Biomedical Research Imaging Center, and Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Weizhong Zhang
- Department of Chemistry, University of Georgia, Athens, Georgia 30602, United States
| | - Jin Xie
- Department of Chemistry, University of Georgia, Athens, Georgia 30602, United States
| |
Collapse
|
5
|
Mao Q, Wu Z, Lai Y, Wang L, Zhao Q, Xu X, Lu X, Qiu W, Zhang Z, Wu J, Wang G, Zhou R, Wu J, Sun H, Huang N, Huang X, Jiang L, Fang Y, Kong Y, Liang L, Bin J, Liao Y, Shi M, Liao W, Zeng D. Dynamic single-cell sequencing unveils the tumor microenvironment evolution of gastric cancer abdominal wall metastases during radiotherapy. Cancer Sci 2024; 115:3859-3874. [PMID: 39327670 PMCID: PMC11611773 DOI: 10.1111/cas.16308] [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: 03/29/2024] [Revised: 07/12/2024] [Accepted: 07/29/2024] [Indexed: 09/28/2024] Open
Abstract
Although the combination of immunotherapy and radiotherapy (RT) for the treatment of malignant tumors has shown rapid development, the insight of how RT remodels the tumor microenvironment to prime antitumor immunity involves a complex interplay of cell types and signaling pathways, much of which remains to be elucidated. Four tumor samples were collected from the same abdominal wall metastasis site of the patient with gastric cancer at baseline and during fractionated RT for single-cell RNA and T-cell receptor sequencing. The Seurat analysis pipeline and immune receptor analysis were used to characterize the gastric cancer metastasis ecosystem and investigated its dynamic changes of cell proportion, cell functional profiles and cell-to-cell communication during RT. Immunohistochemical and immunofluorescent staining and bulk RNA sequencing were applied to validate the key results. We found tumor cells upregulated immune checkpoint genes in response to RT. The infiltration and clonal expansion of T lymphocytes declined within tumors undergoing irradiation. Moreover, RT led to the accumulation of proinflammatory macrophages and natural killer T cells with enhanced cytotoxic gene expression signature. In addition, subclusters of dendritic cells and endothelial cells showed decrease in the expression of antigen present features in post-RT samples. More ECM component secreted by myofibroblasts during RT. These findings indicate that RT induced the dynamics of the immune response that should be taken into consideration when designing and clinically implementing innovative multimodal cancer treatment regimens of different RT and immunotherapy approaches.
Collapse
Affiliation(s)
- Qianqian Mao
- Department of Oncology, Nanfang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
- Cancer Center, The Sixth Affiliated Hospital, School of MedicineSouth China University of TechnologyFoshanChina
- Foshan Key Laboratory of Translational Medicine in Oncology, The Sixth Affiliated Hospital, School of MedicineSouth China University of TechnologyFoshanChina
| | - Zhenzhen Wu
- Department of Oncology, Nanfang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| | - Yonghong Lai
- Department of Oncology, Nanfang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| | - Ling Wang
- Department of Oncology, Nanfang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| | - Qiongzhi Zhao
- Department of Oncology, Nanfang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| | - Xi Xu
- Department of Oncology, Nanfang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| | - Xiansheng Lu
- Department of Pathology, Nanfang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| | - Wenjun Qiu
- Department of Oncology, Nanfang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| | - Zhihua Zhang
- Department of Oncology, Nanfang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| | - Jiani Wu
- Department of Oncology, Nanfang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| | - Gaofeng Wang
- Department of Plastic and Aesthetic SurgeryNanfang Hospital of Southern Medical UniversityGuangzhouGuangdongChina
- Department of DermatologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Rui Zhou
- Department of Oncology, Nanfang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| | - Jianhua Wu
- Department of Oncology, Nanfang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| | - Huiying Sun
- Department of Oncology, Nanfang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| | - Na Huang
- Department of Oncology, Nanfang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| | - Xiatong Huang
- Department of Oncology, Nanfang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| | - Luyang Jiang
- Department of Oncology, Nanfang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| | - Yiran Fang
- Department of Oncology, Nanfang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| | - Yuyun Kong
- Department of Oncology, Nanfang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| | - Li Liang
- Department of Pathology, Nanfang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| | - Jianping Bin
- Department of Cardiology, Nanfang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| | - Yulin Liao
- Department of Cardiology, Nanfang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| | - Min Shi
- Department of Oncology, Nanfang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| | - Wangjun Liao
- Department of Oncology, Nanfang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
- Cancer Center, The Sixth Affiliated Hospital, School of MedicineSouth China University of TechnologyFoshanChina
- Foshan Key Laboratory of Translational Medicine in Oncology, The Sixth Affiliated Hospital, School of MedicineSouth China University of TechnologyFoshanChina
| | - Dongqiang Zeng
- Department of Oncology, Nanfang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
- Cancer Center, The Sixth Affiliated Hospital, School of MedicineSouth China University of TechnologyFoshanChina
- Foshan Key Laboratory of Translational Medicine in Oncology, The Sixth Affiliated Hospital, School of MedicineSouth China University of TechnologyFoshanChina
| |
Collapse
|
6
|
Thapa R, Gupta S, Gupta G, Bhat AA, Smriti, Singla M, Ali H, Singh SK, Dua K, Kashyap MK. Epithelial-mesenchymal transition to mitigate age-related progression in lung cancer. Ageing Res Rev 2024; 102:102576. [PMID: 39515620 DOI: 10.1016/j.arr.2024.102576] [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: 09/05/2024] [Revised: 10/27/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024]
Abstract
Epithelial-Mesenchymal Transition (EMT) is a fundamental biological process involved in embryonic development, wound healing, and cancer progression. In lung cancer, EMT is a key regulator of invasion and metastasis, significantly contributing to the fatal progression of the disease. Age-related factors such as cellular senescence, chronic inflammation, and epigenetic alterations exacerbate EMT, accelerating lung cancer development in the elderly. This review describes the complex mechanism among EMT and age-related pathways, highlighting key regulators such as TGF-β, WNT/β-catenin, NOTCH, and Hedgehog signalling. We also discuss the mechanisms by which oxidative stress, mediated through pathways involving NRF2 and ROS, telomere attrition, regulated by telomerase activity and shelterin complex, and immune system dysregulation, driven by alterations in cytokine profiles and immune cell senescence, upregulate or downregulate EMT induction. Additionally, we highlighted pathways of transcription such as SNAIL, TWIST, ZEB, SIRT1, TP53, NF-κB, and miRNAs regulating these processes. Understanding these mechanisms, we highlight potential therapeutic interventions targeting these critical molecules and pathways.
Collapse
Affiliation(s)
- Riya Thapa
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Saurabh Gupta
- Chameli Devi Institute of Pharmacy, Department of Pharmacology, Indore, Madhya Pradesh, India
| | - Gaurav Gupta
- Centre for Research Impact & Outcome-Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India.
| | - Asif Ahmad Bhat
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Smriti
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Madhav Singla
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Haider Ali
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144411, India; Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, Australia
| | - Kamal Dua
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, Australia; Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, NSW 2007, Australia
| | - Manoj Kumar Kashyap
- Molecular Oncology Laboratory, Amity Stem Cell Institute, Amity Medical School, Amity University Haryana, Panchgaon (Manesar), Gurugram, Haryana, India.
| |
Collapse
|
7
|
Wang Y, Wang J, Ma X, Li H, Sun X, Kang M, Zhang H, Xue Y, Fang Y. CD312 Promotes Paediatric Acute Lymphoblastic Leukaemia Through GNA15-Mediated Non-Classical GPCR Signalling Pathway. J Cell Mol Med 2024; 28:e70283. [PMID: 39656442 PMCID: PMC11629795 DOI: 10.1111/jcmm.70283] [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: 08/29/2024] [Revised: 11/06/2024] [Accepted: 11/11/2024] [Indexed: 12/12/2024] Open
Abstract
The bone marrow-infiltrated immune microenvironment plays a crucial role in blood system diseases, such as leukaemia. In this study, we aimed to investigate the critical role of the immune microenvironment in the onset and progression of childhood acute lymphoblastic leukaemia (ALL). Through high-throughput detection and screening of the GPCR database in the childhood ALL immune microenvironment, we identified CD312 as a candidate target. CD312 is associated with the distribution of Treg and CTL cells within the bone marrow immune microenvironment of ALL children. After CD312 knockdown, the proportion of the Treg subgroup in immune cells was significantly reduced, whereas the proportion of CTL subgroup cells was increased. CD312 exhibited good affinity with GNA15 in the transmembrane intracellular segment, and it could interact with GNA15. The BrdU staining assay revealed that the proliferation of leukaemia cells was enhanced in the CD312-overexpressed CD3+ T cells group via the phosphorylation of ERK, JNK and p38, whereas it was decreased by GNA15 knockdown in the co-culture system. In conclusion, our study suggests that CD312 fosters a suppressive immune microenvironment in the onset and progression of paediatric ALL through a GNA15-mediated non-classical GPCR signalling pathway.
Collapse
Affiliation(s)
- Yaping Wang
- Department of Hematology and Oncology, Children's Hospital of Nanjing Medical UniversityNanjing Medical UniversityNanjingChina
| | - Jiali Wang
- Department of Hematology and Oncology, Children's Hospital of Nanjing Medical UniversityNanjing Medical UniversityNanjingChina
| | - Xiaopeng Ma
- Department of Hematology and Oncology, Children's Hospital of Nanjing Medical UniversityNanjing Medical UniversityNanjingChina
| | - Huimin Li
- Department of Hematology and Oncology, Children's Hospital of Nanjing Medical UniversityNanjing Medical UniversityNanjingChina
| | - Xiaoyan Sun
- Department of Hematology and Oncology, Children's Hospital of Nanjing Medical UniversityNanjing Medical UniversityNanjingChina
| | - Meiyun Kang
- Department of Hematology and Oncology, Children's Hospital of Nanjing Medical UniversityNanjing Medical UniversityNanjingChina
| | - Heng Zhang
- Department of Hematology and Oncology, Children's Hospital of Nanjing Medical UniversityNanjing Medical UniversityNanjingChina
| | - Yao Xue
- Department of Hematology and Oncology, Children's Hospital of Nanjing Medical UniversityNanjing Medical UniversityNanjingChina
| | - Yongjun Fang
- Department of Hematology and Oncology, Children's Hospital of Nanjing Medical UniversityNanjing Medical UniversityNanjingChina
| |
Collapse
|
8
|
Kim Y, Jeon SH, Kim S, Kang MH, Han MG, Lee SY, Kim IA. In vitro-irradiated cancer vaccine enhances anti-tumor efficacy of radiotherapy and PD-L1 blockade in a syngeneic murine breast cancer model. Radiother Oncol 2024; 200:110480. [PMID: 39159681 DOI: 10.1016/j.radonc.2024.110480] [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: 03/18/2024] [Revised: 07/18/2024] [Accepted: 08/15/2024] [Indexed: 08/21/2024]
Abstract
BACKGROUND AND PURPOSE Local radiotherapy (RT) exerts immunostimulatory effects by inducing immunogenic cell death. However, it remains unknown whether in vitro-irradiated tumor cells can elicit anti-tumor responses and enhance the efficacy of local RT and immune checkpoint inhibitors when injected in vivo. METHODS AND MATERIALS We tested the "in vitro-irradiated cancer vaccine (ICV)", wherein tumor cells killed by varying doses of irradiation and their supernatants are intravenously injected. We examined the efficacy of combining local RT (24 Gy in three fractions), PD-L1 blockade, and the ICV in a murine breast cancer model. The immune cell profiles were analyzed via flow cytometry and immunohistochemistry. The cytokine levels were measured by multiplex immunoassays. RESULTS The ICV significantly increased the effector memory phenotype and interferon-γ production capacity in splenic CD8+ T cells. The in vitro-irradiated products contained immune response-related molecules. When combined with local RT and PD-L1 blockade, the ICV significantly delayed the growth of irradiated and non-irradiated tumors. The triple combination therapy increased the proportions of CD8+ T cells and effector memory CD8+ T cells while decreasing the proportion of CTLA-4+ exhausted CD8+ T cells within tumor microenvironment. Additionally, plasma level of interferon-γ and proliferation of effector T cells in the spleen and tumor-draining lymph nodes were significantly increased by the triple combination therapy. CONCLUSIONS The ICV enhanced the therapeutic efficacy of local RT and PD-L1 blockade by augmenting anti-tumor immune responses. Our findings suggest a therapeutic potential of in vitro-irradiation products of tumor cells.
Collapse
Affiliation(s)
- Yoomin Kim
- Department of Tumor Biology and Cancer Research Institute, Graduate School of Medicine, Seoul National University, Seoul, Republic of Korea; Medical Science Research Institute, Seoul National University Bundang Hospital, Seongnam, Republic of Korea; Department of Radiation Oncology, Seoul National University Bundang Hospital, Seongnam, Republic of Korea.
| | - Seung Hyuck Jeon
- Department of Radiation Oncology, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Seongmin Kim
- Department of Tumor Biology and Cancer Research Institute, Graduate School of Medicine, Seoul National University, Seoul, Republic of Korea; Medical Science Research Institute, Seoul National University Bundang Hospital, Seongnam, Republic of Korea; Department of Radiation Oncology, Seoul National University Bundang Hospital, Seongnam, Republic of Korea; Integrated Major in Innovative Medical Science, Seoul National University, Seoul, Republic of Korea
| | - Mi Hyun Kang
- Medical Science Research Institute, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Min Guk Han
- Department of Tumor Biology and Cancer Research Institute, Graduate School of Medicine, Seoul National University, Seoul, Republic of Korea; Medical Science Research Institute, Seoul National University Bundang Hospital, Seongnam, Republic of Korea; Department of Radiation Oncology, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Se Yup Lee
- Korea Nuclear Engineering Co., Ltd, Seoul, Republic of Korea
| | - In Ah Kim
- Department of Tumor Biology and Cancer Research Institute, Graduate School of Medicine, Seoul National University, Seoul, Republic of Korea; Medical Science Research Institute, Seoul National University Bundang Hospital, Seongnam, Republic of Korea; Department of Radiation Oncology, Seoul National University Bundang Hospital, Seongnam, Republic of Korea; Integrated Major in Innovative Medical Science, Seoul National University, Seoul, Republic of Korea; Department of Radiation Oncology, Seoul National University, Seoul, Republic of Korea.
| |
Collapse
|
9
|
Hu Y, Miao Y, Zhang Y, Wang X, Liu X, Zhang W, Deng D. Co-Assembled Binary Polyphenol Natural Products for the Prevention and Treatment of Radiation-Induced Skin Injury. ACS NANO 2024; 18:27557-27569. [PMID: 39329362 DOI: 10.1021/acsnano.4c08508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/28/2024]
Abstract
Radiation therapy, a fundamental treatment for tumors, is often accompanied by radiation-induced skin injury (RISI). Excessive production of reactive oxygen species (ROS) and subsequent inflammation are two key factors in RISI development that will cause skin injury and affect radiotherapy. Herein, the co-assembled binary polyphenol natural products inspired the development of a dual-functional cascade microneedle system for prevention and treatment of RISI. Specifically, epigallocatechin gallate (EGCG) and curcumin (CUR) were co-assembled into nanoparticles (CEPG) by intermolecular interactions and then incorporated with catalase (CAT) to achieve a cascade system in the microneedles (this microneedle system was conducive to penetrate into the epidermal keratinocytes where RISI had the greatest impact). When using microneedles, the tip dissolved rapidly and delivered CEPG and CAT into the dermis, where CEPG NPs were able to respond to ROS and decompose into EGCG and CUR. More importantly, EGCG and CAT formed a cascade that converts superoxide anions into water step-by-step, which can reduce cell damage caused by free radicals in the early stages of radiation for prevention; meanwhile, CUR inhibited inflammatory pathways, achieving the treatment of skin inflammation in the post-radiotherapy period. These explorations broaden the strategy for the application of natural products in RISI.
Collapse
Affiliation(s)
- Yanwei Hu
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 211198, China
| | - Yuhang Miao
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 211198, China
| | - Yan Zhang
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 211198, China
| | - Xin Wang
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 211198, China
| | - Xin Liu
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 211198, China
| | - Wei Zhang
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 211198, China
| | - Dawei Deng
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 211198, China
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 211198, China
| |
Collapse
|
10
|
Huang Z, Huang S, Song S, Ding Y, Zhou H, Zhang S, Weng L, Zhang Y, Hu Y, Yuan A, Dai Y, Luo Z, Wang L. Two-dimensional coordination risedronate-manganese nanobelts as adjuvant for cancer radiotherapy and immunotherapy. Nat Commun 2024; 15:8692. [PMID: 39375342 PMCID: PMC11458765 DOI: 10.1038/s41467-024-53084-w] [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: 03/27/2024] [Accepted: 10/01/2024] [Indexed: 10/09/2024] Open
Abstract
The irradiated tumor itself represents an opportunity to establish endogenous in situ vaccines. However, such in situ cancer vaccination (ISCV) triggered by radiation therapy (RT) alone is very weak and hardly elicits systemic anticancer immunity. In this study, we develop two-dimensional risedronate-manganese nanobelts (RMn-NBs) as an adjuvant for RT to address this issue. RMn-NBs exhibit good T2 magnetic resonance imaging performance and enhanced Fenton-like catalytic activity, which induces immunogenic cell death. RMn-NBs can inhibit the HIF-1α/VEGF axis to empower RT and synchronously activate the cGAS/STING pathway for promoting the secretion of type I interferon, thereby boosting RT-triggered ISCV and immune checkpoint blockade therapy against primary and metastatic tumors. RMn-NBs as a nano-adjuvant for RT show good biocompatibility and therapeutic efficacy, presenting a promising prospect for cancer radiotherapy and immunotherapy.
Collapse
Affiliation(s)
- Zhusheng Huang
- State Key Laboratory for Organic Electronics and Information Displays (SKLOEID), School of Chemistry and Life Sciences, Nanjing University of Posts and Telecommunications, Nanjing, China
- Jiangsu Key Laboratory of Smart Biomaterials and Theranostic Technology, College of Optical Engineering & Flexible Electronics (Future Technology), Nanjing University of Posts and Telecommunications, Nanjing, China
- Cancer Centre and Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China
- MoE Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macau SAR, China
| | - Shiqian Huang
- State Key Laboratory for Organic Electronics and Information Displays (SKLOEID), School of Chemistry and Life Sciences, Nanjing University of Posts and Telecommunications, Nanjing, China
| | - Simin Song
- State Key Laboratory for Organic Electronics and Information Displays (SKLOEID), School of Chemistry and Life Sciences, Nanjing University of Posts and Telecommunications, Nanjing, China
| | - Yankui Ding
- State Key Laboratory for Organic Electronics and Information Displays (SKLOEID), School of Chemistry and Life Sciences, Nanjing University of Posts and Telecommunications, Nanjing, China
| | - Hao Zhou
- State Key Laboratory for Organic Electronics and Information Displays (SKLOEID), School of Chemistry and Life Sciences, Nanjing University of Posts and Telecommunications, Nanjing, China
| | - Shaoyin Zhang
- State Key Laboratory for Organic Electronics and Information Displays (SKLOEID), School of Chemistry and Life Sciences, Nanjing University of Posts and Telecommunications, Nanjing, China
| | - Lixing Weng
- State Key Laboratory for Organic Electronics and Information Displays (SKLOEID), School of Chemistry and Life Sciences, Nanjing University of Posts and Telecommunications, Nanjing, China
- Jiangsu Key Laboratory of Smart Biomaterials and Theranostic Technology, College of Optical Engineering & Flexible Electronics (Future Technology), Nanjing University of Posts and Telecommunications, Nanjing, China
| | - Ying Zhang
- State Key Laboratory for Organic Electronics and Information Displays (SKLOEID), School of Chemistry and Life Sciences, Nanjing University of Posts and Telecommunications, Nanjing, China
- Jiangsu Key Laboratory of Smart Biomaterials and Theranostic Technology, College of Optical Engineering & Flexible Electronics (Future Technology), Nanjing University of Posts and Telecommunications, Nanjing, China
| | - Yiqiao Hu
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University, Nanjing, China
| | - Ahu Yuan
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University, Nanjing, China
| | - Yunlu Dai
- Cancer Centre and Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China.
- MoE Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macau SAR, China.
| | - Zhimin Luo
- State Key Laboratory for Organic Electronics and Information Displays (SKLOEID), School of Chemistry and Life Sciences, Nanjing University of Posts and Telecommunications, Nanjing, China.
- Jiangsu Key Laboratory of Smart Biomaterials and Theranostic Technology, College of Optical Engineering & Flexible Electronics (Future Technology), Nanjing University of Posts and Telecommunications, Nanjing, China.
| | - Lianhui Wang
- State Key Laboratory for Organic Electronics and Information Displays (SKLOEID), School of Chemistry and Life Sciences, Nanjing University of Posts and Telecommunications, Nanjing, China.
- Jiangsu Key Laboratory of Smart Biomaterials and Theranostic Technology, College of Optical Engineering & Flexible Electronics (Future Technology), Nanjing University of Posts and Telecommunications, Nanjing, China.
| |
Collapse
|
11
|
Khan AQ, Al-Tamimi M, Anver R, Agha MV, Anamangadan G, Raza SS, Ahmad F, Ahmad A, Alam M, Buddenkotte J, Steinhoff M, Uddin S. Targeting of S-phase kinase associated protein 2 stabilized tumor suppressors leading to apoptotic cell death in squamous skin cancer cells. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167286. [PMID: 38866114 DOI: 10.1016/j.bbadis.2024.167286] [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: 11/21/2023] [Revised: 05/29/2024] [Accepted: 06/04/2024] [Indexed: 06/14/2024]
Abstract
S-phase kinase-associated protein 2 (Skp2) is an F-box protein overexpressed in human cancers and linked with poor prognosis. It triggers cancer pathogenesis, including stemness and drug resistance. In this study, we have explored the potential role of Skp2 targeting in restoring the expression of tumor suppressors in human cutaneous squamous cell carcinoma (cSCC) cells. Our results showed that genetic and pharmacological Skp2 targeting markedly suppressed cSCC cell proliferation, colony growth, spheroid formation, and enhanced sensitization to chemotherapeutic drugs. Further, western blot results demonstrated restoration of tumor suppressor (KLF4) and CDKI (p21) and suppression of vimentin and survivin in Skp2-knocked-down cSCC cells. Importantly, we also explored that Skp2 targeting potentiates apoptosis of cSCC cells through MAPK signaling. Moreover, co-targeting of Skp2 and PI3K/AKT resulted in increased cancer cell death. Interestingly, curcumin, a well-known naturally derived anticancer agent, also inhibits Skp2 expression with concomitant CDKI upregulation. In line, curcumin suppressed cSCC cell growth through ROS-mediated apoptosis, while the use of N-acetyl cysteine (NAC) reversed curcumin-induced cell death. Curcumin treatment also sensitized cSCC cells to conventional anticancer drugs, such as cisplatin and doxorubicin. Altogether, these data suggest that Skp2 targeting restores the functioning of tumor suppressors, inhibits the expression of genes associated with cell proliferation and stemness, and sensitizes cancer cells to anticancer drugs. Thus, genetic, and pharmacological ablation of Skp2 can be an important strategy for attenuating cancer pathogenesis and associated complications in skin squamous cell carcinoma.
Collapse
Affiliation(s)
- Abdul Q Khan
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar.
| | - Maha Al-Tamimi
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Rasheeda Anver
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Maha Victor Agha
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Gazala Anamangadan
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Syed Shadab Raza
- Department of Stem Cell Biology and Regenerative Medicine, Era University, Lucknow 226003, India
| | - Fareed Ahmad
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar; Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar; Department of Dermatology and Venereology, Rumailah Hospital, Hamad Medical Corporation, Doha 3050, Qatar
| | - Aamir Ahmad
- Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar; Department of Dermatology and Venereology, Rumailah Hospital, Hamad Medical Corporation, Doha 3050, Qatar
| | - Majid Alam
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar; Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar; Department of Dermatology and Venereology, Rumailah Hospital, Hamad Medical Corporation, Doha 3050, Qatar
| | - Joerg Buddenkotte
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar; Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar; Department of Dermatology and Venereology, Rumailah Hospital, Hamad Medical Corporation, Doha 3050, Qatar
| | - Martin Steinhoff
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar; Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar; Department of Dermatology and Venereology, Rumailah Hospital, Hamad Medical Corporation, Doha 3050, Qatar; Department of Medicine, Weill Cornell Medicine Qatar, Qatar Foundation-Education City, Doha 24144, Qatar; Department of Medicine, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA; College of Medicine, Qatar University, Doha 2713, Qatar.
| | - Shahab Uddin
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar; Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar; Department of Dermatology and Venereology, Rumailah Hospital, Hamad Medical Corporation, Doha 3050, Qatar; Laboratory Animal Research Center, Qatar University, Doha 2713, Qatar.
| |
Collapse
|
12
|
He J, Wang C, Fang X, Li J, Shen X, Zhang J, Peng C, Li H, Li S, Karp JM, Kuai R. Tuning the fluidity and protein corona of ultrasound-responsive liposomal nanovaccines to program T cell immunity in mice. Nat Commun 2024; 15:8121. [PMID: 39284814 PMCID: PMC11405680 DOI: 10.1038/s41467-024-52104-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 08/27/2024] [Indexed: 09/22/2024] Open
Abstract
Inducing high levels of antigen-specific CD8α+ T cells in the tumor is beneficial for cancer immunotherapy, but achieving this in a safe and effective manner remains challenging. Here, we have developed a designer liposomal nanovaccine containing a sonosensitizer (LNVS) to efficiently program T cell immunity in mice. Following intravenous injection, LNVS accumulates in the spleen in a protein corona and fluidity-dependent manner, leading to greater frequencies of antigen-specific CD8α+ T cells than soluble vaccines (the mixture of antigens and adjuvants). Meanwhile, some LNVS passively accumulates in the tumor, where it responds to ultrasound (US) to increase the levels of chemokines and adhesion molecules that are beneficial for recruiting CD8α+ T cells to the tumor. LNVS + US induces higher levels of intratumoral antitumor T cells than traditional sonodynamic therapy, regresses established mouse MC38 tumors and orthotopic cervical cancer, and protects cured mice from relapse. Our platform sheds light on the importance of tuning the fluidity and protein corona of naovaccines to program T cell immunity in mice and may inspire new strategies for cancer immunotherapy.
Collapse
Affiliation(s)
- Jia He
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
- Tsinghua-Peking Center for Life Sciences, Beijing, China
| | - Chaoyu Wang
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
- Tsinghua-Peking Center for Life Sciences, Beijing, China
| | - Xiao Fang
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
- Tsinghua-Peking Center for Life Sciences, Beijing, China
| | - Junyao Li
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
- Tsinghua-Peking Center for Life Sciences, Beijing, China
| | - Xueying Shen
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
- Tsinghua-Peking Center for Life Sciences, Beijing, China
| | - Junxia Zhang
- Tsinghua-Peking Center for Life Sciences, Beijing, China
- School of Life Sciences, Tsinghua University, Beijing, China
- Frontier Research Center for Biological Structure & State Key Laboratory of Membrane Biology, Beijing, China
| | - Cheng Peng
- Tsinghua-Peking Center for Life Sciences, Beijing, China
- School of Life Sciences, Tsinghua University, Beijing, China
- Frontier Research Center for Biological Structure & State Key Laboratory of Membrane Biology, Beijing, China
| | - Hongjian Li
- School of Medicine, Tsinghua University, Beijing, China
| | - Sai Li
- Tsinghua-Peking Center for Life Sciences, Beijing, China
- School of Life Sciences, Tsinghua University, Beijing, China
- Frontier Research Center for Biological Structure & State Key Laboratory of Membrane Biology, Beijing, China
| | - Jeffrey M Karp
- Department of Anesthesiology, Perioperative, and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Harvard-MIT Program in Health Sciences and Technology, MIT, Cambridge, MA, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Rui Kuai
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, China.
- Tsinghua-Peking Center for Life Sciences, Beijing, China.
| |
Collapse
|
13
|
Li L, Liu X, Han C, Tian L, Wang Y, Han B. Ferroptosis in radiation-induced brain injury: roles and clinical implications. Biomed Eng Online 2024; 23:93. [PMID: 39261942 PMCID: PMC11389269 DOI: 10.1186/s12938-024-01288-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 08/31/2024] [Indexed: 09/13/2024] Open
Abstract
Radiation-induced brain injury (RBI) presents a significant challenge for patients undergoing radiation therapy for head, neck, and intracranial tumors. This review aims to elucidate the role of ferroptosis in RBI and its therapeutic implications. Specifically, we explore how ferroptosis can enhance the sensitivity of tumor cells to radiation while also examining strategies to mitigate radiation-induced damage to normal brain tissues. By investigating the mechanisms through which radiation increases cellular reactive oxygen species (ROS) and initiates ferroptosis, we aim to develop targeted therapeutic strategies that maximize treatment efficacy and minimize neurotoxicity. The review highlights key regulatory factors in the ferroptosis pathway, including glutathione peroxidase 4 (GPX4), cystine/glutamate antiporter system Xc- (System Xc-), nuclear factor erythroid 2-related factor 2 (NRF2), Acyl-CoA synthetase long-chain family member 4 (ACSL4), and others, and their interactions in the context of RBI. Furthermore, we discuss the clinical implications of modulating ferroptosis in radiation therapy, emphasizing the potential for selective induction of ferroptosis in tumor cells and inhibition in healthy cells. The development of advanced diagnostic tools and therapeutic strategies targeting ferroptosis offers a promising avenue for enhancing the safety and efficacy of radiation therapy, underscoring the need for further research in this burgeoning field.
Collapse
Affiliation(s)
- Lifang Li
- Department of Radiotherapy, Tianjin Medical University Baodi Hospital, Tianjin, 301800, China
| | - Xia Liu
- Department of Radiotherapy, Tianjin Medical University Baodi Hospital, Tianjin, 301800, China
| | - Chunfeng Han
- Department of Pharmacy, Tianjin Medical University Baodi Hospital, Tianjin, 301800, China
| | - Licheng Tian
- Department of Radiotherapy, Tianjin Medical University Baodi Hospital, Tianjin, 301800, China
| | - Yongzhi Wang
- Department of Radiotherapy, Tianjin Medical University Baodi Hospital, Tianjin, 301800, China
| | - Baolin Han
- Department of Radiotherapy, Tianjin Medical University Baodi Hospital, Tianjin, 301800, China.
| |
Collapse
|
14
|
Yang R, Zhang S, Wang L, Chen Y, Chen X, Xia J, Ren X, Cheng B, Chen X. Radiation-induced exosomes promote oral squamous cell carcinoma progression via enhancing SLC1A5-glutamine metabolism. J Oral Pathol Med 2024; 53:458-467. [PMID: 38802300 DOI: 10.1111/jop.13561] [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: 09/02/2023] [Revised: 04/16/2024] [Accepted: 05/11/2024] [Indexed: 05/29/2024]
Abstract
BACKGROUND Radiotherapy (RT) can drive cancer cells to enter a state of cellular senescence in which cells can secrete senescence-associated secretory phenotype (SASP) and produce small extracellular vesicles (sEVs) to interact with cells in the tumor microenvironment (TME). Tumor-derived sEVs that are taken up by recipient cells contribute to cancer cell metabolic plasticity, resistance to anticancer therapy, and adaptation to the TME. However, how radiation-induced sEVs support oral squamous cell carcinoma (OSCC) progression remains unclear. METHODS Beta-galactosidase staining and SASP mRNA expression analysis were used to evaluate the senescence-associated activity of OSCC cells after irradiation. Nanoparticle tracking analysis was performed to identify radiation-induced sEVs. Liquid chromatography-tandem mass spectrometry (LC-MS) was used to explore changes in the levels of proteins in radiation-induced sEVs. Cell Counting Kit-8 and colony formation assays were performed to investigate the function of radiation-induced SASP and sEVs in vitro. A xenograft tumor model was established to investigate the functions of radiation-induced sEVs and V-9302 in vivo as well as the underlying mechanisms. Bioinformatics analysis was performed to determine the relationship between glutamine metabolism and OSCC recurrence. RESULTS We determined that the radiation-induced SASP triggered OSCC cell proliferation. Additionally, radiation-induced sEVs exacerbated OSCC cell malignancy. LC-MS/MS and bioinformatics analyses revealed that SLC1A5, which is a cellular receptor that participates in glutamine uptake, was significantly enriched in radiation-induced sEVs. In vitro and in vivo, inhibiting SLC1A5 could block the oncogenic effects of radiation-induced sEVs in OSCC. CONCLUSION Radiation-induced sEVs might promote the proliferation of unirradiated cancer cells by enhancing glutamine metabolism; this might be a novel molecular mechanism underlying radiation resistance in OSCC patients.
Collapse
Affiliation(s)
- Rongchun Yang
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
- Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Siyuan Zhang
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
- Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Lixuan Wang
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
- Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Yingyao Chen
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
- Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Xiaobing Chen
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
- Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Juan Xia
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
- Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Xianyue Ren
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
- Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Bin Cheng
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
- Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Xijuan Chen
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
- Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
15
|
Amit U, Uslu U, Verginadis II, Kim MM, Motlagh SAO, Diffenderfer ES, Assenmacher CA, Bicher S, Atoche SJ, Ben-Josef E, Young RM, June CH, Koumenis C. Proton radiation boosts the efficacy of mesothelin-targeting chimeric antigen receptor T cell therapy in pancreatic cancer. Proc Natl Acad Sci U S A 2024; 121:e2403002121. [PMID: 39047033 PMCID: PMC11294999 DOI: 10.1073/pnas.2403002121] [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: 02/12/2024] [Accepted: 06/24/2024] [Indexed: 07/27/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) represents a challenge in oncology, with limited treatment options for advanced-stage patients. Chimeric antigen receptor T cell (CAR T) therapy targeting mesothelin (MSLN) shows promise, but challenges such as the hostile immunosuppressive tumor microenvironment (TME) hinder its efficacy. This study explores the synergistic potential of combining proton radiation therapy (RT) with MSLN-targeting CAR T therapy in a syngeneic PDAC model. Proton RT significantly increased MSLN expression in tumor cells and caused a significant increase in CAR T cell infiltration into tumors. The combination therapy reshaped the immunosuppressive TME, promoting antitumorigenic M1 polarized macrophages and reducing myeloid-derived suppressor cells (MDSC). In a flank PDAC model, the combination therapy demonstrated superior attenuation of tumor growth and improved survival compared to individual treatments alone. In an orthotopic PDAC model treated with image-guided proton RT, tumor growth was significantly reduced in the combination group compared to the RT treatment alone. Further, the combination therapy induced an abscopal effect in a dual-flank tumor model, with increased serum interferon-γ levels and enhanced proliferation of extratumoral CAR T cells. In conclusion, combining proton RT with MSLN-targeting CAR T therapy proves effective in modulating the TME, enhancing CAR T cell trafficking, and exerting systemic antitumor effects. Thus, this combinatorial approach could present a promising strategy for improving outcomes in unresectable PDAC.
Collapse
Affiliation(s)
- Uri Amit
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
- Department of Radiation Oncology, Tel Aviv Medical Center, Tel Aviv64239, Israel
| | - Ugur Uslu
- Department of Pathology and Laboratory Medicine, Center for Cellular Immunotherapies, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA19104
- Parker Institute for Cancer Immunotherapy at University of Pennsylvania, Philadelphia, PA19104
| | - Ioannis I. Verginadis
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
| | - Michele M. Kim
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
| | - Seyyedeh Azar Oliaei Motlagh
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
| | - Eric S. Diffenderfer
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
| | - Charles-Antoine Assenmacher
- Department of Pathobiology, School of Veterinary Medicine, Comparative Pathology Core, University of Pennsylvania, Philadelphia, PA19104
| | - Sandra Bicher
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
| | - Sebastian J. Atoche
- Department of Pathology and Laboratory Medicine, Center for Cellular Immunotherapies, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA19104
- Parker Institute for Cancer Immunotherapy at University of Pennsylvania, Philadelphia, PA19104
| | - Edgar Ben-Josef
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
| | - Regina M. Young
- Department of Pathology and Laboratory Medicine, Center for Cellular Immunotherapies, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA19104
- Parker Institute for Cancer Immunotherapy at University of Pennsylvania, Philadelphia, PA19104
| | - Carl H. June
- Department of Pathology and Laboratory Medicine, Center for Cellular Immunotherapies, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA19104
- Parker Institute for Cancer Immunotherapy at University of Pennsylvania, Philadelphia, PA19104
| | - Constantinos Koumenis
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
| |
Collapse
|
16
|
Jassi C, kuo WW, Kuo CH, Chang CM, Chen MC, Shih TC, Li CC, Huang CY. Mediation of radiation-induced bystander effect and epigenetic modification: The role of exosomes in cancer radioresistance. Heliyon 2024; 10:e34460. [PMID: 39114003 PMCID: PMC11304029 DOI: 10.1016/j.heliyon.2024.e34460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 03/20/2024] [Accepted: 07/09/2024] [Indexed: 08/10/2024] Open
Abstract
Exosomes are nano-sized extracellular vesicles produced by almost all mammalian cells. They play an important role in cell-to-cell communication by transferring biologically active molecules from the cell of origin to the recipient cells. Ionizing radiation influences exosome production and molecular cargo loading. In cancer management, ionizing radiation is a form of treatment that exerts its cancer cytotoxicity by induction of DNA damage and other alterations to the targeted tissue cells. However, normal bystander non-targeted cells may exhibit the effects of ionizing radiation, a phenomenon called radiation-induced bystander effect (RIBE). The mutual communication between the two groups of cells (targeted and non-targeted) via radiation-influenced exosomes enables the exchange of radiosensitive molecules. This facilitates indirect radiation exposure, leading, among other effects, to epigenetic remodeling and subsequent adaptation to radiation. This review discusses the role exosomes play in epigenetically induced radiotherapy resistance through the mediation of RIBE.
Collapse
Affiliation(s)
- Chikondi Jassi
- Department of Biological Sciences and Technology, China Medical University, Taichung, Taiwan
| | - Wei-Wen kuo
- Department of Biological Sciences and Technology, China Medical University, Taichung, Taiwan
| | - Chia-Hua Kuo
- Laboratory of Exercise Biochemistry, University of Taipei, Taipei, Taiwan
- Department of Kinesiology and Health Science, College of William and Mary, Williamsburg, VA, USA
| | - Chun-Ming Chang
- Department of General Surgery, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
- School of Medicine Tzu Chi University, 701, Section 3, Chung-Yang Road, Hualien 97004, Taiwan
| | - Ming-Cheng Chen
- Division of Colorectal Surgery, Department of Surgery, Taichung Veterans General Hospital, Taichung 40705, Taiwan
- Faculty of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Tzu-Ching Shih
- Department of Biomedical Imaging & Radiological Science College of Medicine, China Medical University, Taichung 404, Taiwan
| | - Chi-Cheng Li
- School of Medicine Tzu Chi University, 701, Section 3, Chung-Yang Road, Hualien 97004, Taiwan
- Department of Hematology and Oncology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
- Center of Stem Cell & Precision Medicine, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Chih-Yang Huang
- Cardiovascular and Mitochondria Related Diseases Research Center, Hualien Tzu Chi Hospital, Hualien 970, Taiwan
- Graduate Institute of Biomedicine, China Medical University, Taichung, Taiwan
- Department of Biotechnology, Asia University, Taichung 413, Taiwan
- Center of General Education, Buddhist Tzu Chi Medical Foundation, Tzu Chi University of Science and Technology, Hualien 970, Taiwan
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung 404, Taiwan
| |
Collapse
|
17
|
Luo X, Liu H, Wen J, Hu J, Li Y, Li G, Dai G, Li Y, Li J. Composite hydrogels with antioxidant and robust adhesive properties for the prevention of radiation-induced dermatitis. J Mater Chem B 2024; 12:6927-6939. [PMID: 38904166 DOI: 10.1039/d4tb00511b] [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: 06/22/2024]
Abstract
Radiotherapy is a pivotal means of cancer treatment, but it often leads to radiation dermatitis, a skin injury caused by radiation-induced excess reactive oxygen species (ROS). Scavenging free radicals in the course of radiation therapy will be an effective means to prevent radiation dermatitis. This study demonstrates a novel double network hydrogel doped with MoS2 nanosheets for the prevention of radiation-induced dermatitis. The resultant SPM hydrogel constructed from polyacrylamide (PAM) and sodium alginate (SA) nanofiber presented favorable mechanical and adhesion properties. It could conform well to the human body's irregular contours without secondary dressing fixation, making it suitable for skin protection applications. The in vitro and in vivo experiments showed that the antioxidant properties conferred by MoS2 nanosheets enable SPM to effectively mitigate excessive ROS and reduce oxidative stress, thereby preventing radiation dermatitis caused by oxidative damage. Biosafety assessments indicated good biocompatibility of the composite hydrogel, suggesting SPM's practicality and potential as an external dressing for skin radiation protection.
Collapse
Affiliation(s)
- Xue Luo
- Research Center for Nano-Biomaterials, Analytical and Testing Center, Sichuan University, Chengdu, 610065, P. R. China.
| | - Huan Liu
- Research Center for Nano-Biomaterials, Analytical and Testing Center, Sichuan University, Chengdu, 610065, P. R. China.
| | - Jing Wen
- Research Center for Nano-Biomaterials, Analytical and Testing Center, Sichuan University, Chengdu, 610065, P. R. China.
| | - Jiaxin Hu
- Research Center for Nano-Biomaterials, Analytical and Testing Center, Sichuan University, Chengdu, 610065, P. R. China.
| | - Yongzhi Li
- Research Center for Nano-Biomaterials, Analytical and Testing Center, Sichuan University, Chengdu, 610065, P. R. China.
| | - Guangjun Li
- Department of Radiotherapy Physics & Technology, West China Hospital, Sichuan University, Chengdu, 610041, P. R. China
| | - Guyu Dai
- Department of Radiotherapy Physics & Technology, West China Hospital, Sichuan University, Chengdu, 610041, P. R. China
| | - Yubao Li
- Research Center for Nano-Biomaterials, Analytical and Testing Center, Sichuan University, Chengdu, 610065, P. R. China.
| | - Jidong Li
- Research Center for Nano-Biomaterials, Analytical and Testing Center, Sichuan University, Chengdu, 610065, P. R. China.
| |
Collapse
|
18
|
Luo T, Jiang X, Fan Y, Yuan E, Li J, Tillman L, Lin W. STING agonist-conjugated metal-organic framework induces artificial leukocytoid structures and immune hotspots for systemic antitumor responses. Natl Sci Rev 2024; 11:nwae167. [PMID: 38887543 PMCID: PMC11182667 DOI: 10.1093/nsr/nwae167] [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: 01/11/2024] [Revised: 04/15/2024] [Accepted: 04/23/2024] [Indexed: 06/20/2024] Open
Abstract
Radiotherapy is widely used for cancer treatment, but its clinical utility is limited by radioresistance and its inability to target metastases. Nanoscale metal-organic frameworks (MOFs) have shown promise as high-Z nanoradiosensitizers to enhance radiotherapy and induce immunostimulatory regulation of the tumor microenvironment. We hypothesized that MOFs could deliver small-molecule therapeutics to synergize with radiotherapy for enhanced antitumor efficacy. Herein, we develop a robust nanoradiosensitizer, GA-MOF, by conjugating a STING agonist, 2',3'-cyclic guanosine monophosphate-adenosine monophosphate (GA), on MOFs for synergistic radiosensitization and STING activation. GA-MOF demonstrated strong anticancer efficacy by forming immune-cell-rich nodules (artificial leukocytoid structures) and transforming them into immunostimulatory hotspots with radiotherapy. Further combination with an immune checkpoint blockade suppressed distant tumors through systemic immune activation. Our work not only demonstrates the potent radiosensitization of GA-MOF, but also provides detailed mechanisms regarding MOF distribution, immune regulatory pathways and long-term immune effects.
Collapse
Affiliation(s)
- Taokun Luo
- Department of Chemistry, University of Chicago, Chicago 60637, USA
| | - Xiaomin Jiang
- Department of Chemistry, University of Chicago, Chicago 60637, USA
| | - Yingjie Fan
- Department of Chemistry, University of Chicago, Chicago 60637, USA
| | - Eric Yuan
- Department of Chemistry, University of Chicago, Chicago 60637, USA
| | - Jinhong Li
- Department of Chemistry, University of Chicago, Chicago 60637, USA
| | - Langston Tillman
- Department of Chemistry, University of Chicago, Chicago 60637, USA
| | - Wenbin Lin
- Department of Chemistry, University of Chicago, Chicago 60637, USA
- Department of Radiation and Cellular Oncology and the Ludwig Center for Metastasis Research, University of Chicago, Chicago 60637, USA
| |
Collapse
|
19
|
Meng Y, Huang J, Ding J, Zhou H, Li Y, Zhou W. Mn-phenolic networks as synergistic carrier for STING agonists in tumor immunotherapy. Mater Today Bio 2024; 26:101018. [PMID: 38516172 PMCID: PMC10952078 DOI: 10.1016/j.mtbio.2024.101018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 02/21/2024] [Accepted: 03/05/2024] [Indexed: 03/23/2024] Open
Abstract
The cGAS-STING pathway holds tremendous potential as a regulator of immune responses, offering a means to reshape the tumor microenvironment and enhance tumor immunotherapy. Despite the emergence of STING agonists, their clinical viability is hampered by stability and delivery challenges, as well as variations in STING expression within tumors. In this study, we present Mn-phenolic networks as a novel carrier for ADU-S100, a hydrophilic STING agonist, aimed at bolstering immunotherapy. These nanoparticles, termed TMA NMs, are synthesized through the coordination of tannic acid and manganese ions, with surface modification involving bovine serum albumin to enhance their colloidal stability. TMA NMs exhibit pH/GSH-responsive disintegration properties, enabling precise drug release. This effectively addresses drug stability issues and facilitates efficient intracellular drug delivery. Importantly, TMA NMs synergistically enhance the effects of ADU-S100 through the concurrent release of Mn2+, which serves as a sensitizer of the STING pathway, resulting in significant STING pathway activation. Upon systemic administration, these nanoparticles efficiently accumulate within tumors. The activation of STING pathways not only induces immunogenic cell death (ICD) in tumor cells but also orchestrates systemic remodeling of the immunosuppressive microenvironment. This includes the promotion of cytokine release, dendritic cell maturation, and T cell infiltration, leading to pronounced suppression of tumor growth. Combining with the excellent biocompatibility and biodegradability, this Mn-based nanocarrier represents a promising strategy for enhancing tumor immunotherapy through the cGAS-STING pathway.
Collapse
Affiliation(s)
- Yingcai Meng
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan, China
- Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Jiaxin Huang
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan, China
| | - Jinsong Ding
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan, China
| | - Haiyan Zhou
- Department of Pathology, School of Basic Medicine, Central South University, China
- Department of Pathology, Xiangya Hospital, Central South University, China
| | - Yong Li
- Department of Pediatric Surgery, Hunan Children's Hospital, Changsha 410004, Hunan, China
| | - Wenhu Zhou
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan, China
| |
Collapse
|
20
|
Malla R, Srilatha M, Muppala V, Farran B, Chauhan VS, Nagaraju GP. Neoantigens and cancer-testis antigens as promising vaccine candidates for triple-negative breast cancer: Delivery strategies and clinical trials. J Control Release 2024; 370:707-720. [PMID: 38744346 DOI: 10.1016/j.jconrel.2024.05.020] [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: 11/30/2023] [Revised: 03/15/2024] [Accepted: 05/10/2024] [Indexed: 05/16/2024]
Abstract
Immunotherapy is gaining prominence as a promising strategy for treating triple-negative breast cancer (TNBC). Neoantigens (neoAgs) and cancer-testis antigens (CTAs) are tumor-specific targets originating from somatic mutations and epigenetic changes in cancer cells. These antigens hold great promise for personalized cancer vaccines, as supported by preclinical and early clinical evidence in TNBC. This review delves into the potential of neoAgs and CTAs as vaccine candidates, emphasizing diverse strategies and delivery approaches. It also highlights the current status of vaccination modalities undergoing clinical trials in TNBC therapy. A comprehensive understanding of neoAgs, CTAs, vaccination strategies, and innovative delivery methods is crucial for optimizing neoAg-based immunotherapies in clinical practice.
Collapse
Affiliation(s)
- RamaRao Malla
- Cancer Biology Lab, Department of Biochemistry and Bioinformatics, GITAM School of Science, GITAM (Deemed to be University), Visakhapatnam 530045, Andhra Pradesh, India
| | - Mundla Srilatha
- Department of Biotechnology, Sri Venkateswara University, Tirupati 517502, AP, India
| | - Veda Muppala
- Department of Neuroscience, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Batoul Farran
- Division of Hematology and Oncology, Department of Medicine, Henry Ford Health, Detroit, MI 48202, USA
| | - Virander Singh Chauhan
- Molecular Medicine Group, Molecular Medicines International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Ganji Purnachandra Nagaraju
- Department of Hematology and Oncology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35233, USA.
| |
Collapse
|
21
|
Qiu J, Cheng Z, Jiang Z, Gan L, Zhang Z, Xie Z. Immunomodulatory Precision: A Narrative Review Exploring the Critical Role of Immune Checkpoint Inhibitors in Cancer Treatment. Int J Mol Sci 2024; 25:5490. [PMID: 38791528 PMCID: PMC11122264 DOI: 10.3390/ijms25105490] [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: 03/24/2024] [Revised: 05/11/2024] [Accepted: 05/14/2024] [Indexed: 05/26/2024] Open
Abstract
An immune checkpoint is a signaling pathway that regulates the recognition of antigens by T-cell receptors (TCRs) during an immune response. These checkpoints play a pivotal role in suppressing excessive immune responses and maintaining immune homeostasis against viral or microbial infections. There are several FDA-approved immune checkpoint inhibitors (ICIs), including ipilimumab, pembrolizumab, and avelumab. These ICIs target cytotoxic T-lymphocyte-associated protein 4 (CTLA-4), programmed cell death protein 1 (PD-1), and programmed death ligand 1 (PD-L1). Furthermore, ongoing efforts are focused on developing new ICIs with emerging potential. In comparison to conventional treatments, ICIs offer the advantages of reduced side effects and durable responses. There is growing interest in the potential of combining different ICIs with chemotherapy, radiation therapy, or targeted therapies. This article comprehensively reviews the classification, mechanism of action, application, and combination strategies of ICIs in various cancers and discusses their current limitations. Our objective is to contribute to the future development of more effective anticancer drugs targeting immune checkpoints.
Collapse
Affiliation(s)
- Junyu Qiu
- College of Basic Medical, Nanchang University, Nanchang 330006, China; (J.Q.); (Z.C.); (Z.J.); (L.G.); (Z.Z.)
- Queen Mary School, Medical Department, Nanchang University, Nanchang 330031, China
| | - Zilin Cheng
- College of Basic Medical, Nanchang University, Nanchang 330006, China; (J.Q.); (Z.C.); (Z.J.); (L.G.); (Z.Z.)
- Queen Mary School, Medical Department, Nanchang University, Nanchang 330031, China
| | - Zheng Jiang
- College of Basic Medical, Nanchang University, Nanchang 330006, China; (J.Q.); (Z.C.); (Z.J.); (L.G.); (Z.Z.)
- Queen Mary School, Medical Department, Nanchang University, Nanchang 330031, China
| | - Luhan Gan
- College of Basic Medical, Nanchang University, Nanchang 330006, China; (J.Q.); (Z.C.); (Z.J.); (L.G.); (Z.Z.)
- Huan Kui School, Medical Department, Nanchang University, Nanchang 330031, China
| | - Zixuan Zhang
- College of Basic Medical, Nanchang University, Nanchang 330006, China; (J.Q.); (Z.C.); (Z.J.); (L.G.); (Z.Z.)
- Queen Mary School, Medical Department, Nanchang University, Nanchang 330031, China
| | - Zhenzhen Xie
- College of Basic Medical, Nanchang University, Nanchang 330006, China; (J.Q.); (Z.C.); (Z.J.); (L.G.); (Z.Z.)
| |
Collapse
|
22
|
Huang Z, Xiao Z, Yu L, Liu J, Yang Y, Ouyang W. Tumor-associated macrophages in non-small-cell lung cancer: From treatment resistance mechanisms to therapeutic targets. Crit Rev Oncol Hematol 2024; 196:104284. [PMID: 38311012 DOI: 10.1016/j.critrevonc.2024.104284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 01/20/2024] [Accepted: 01/31/2024] [Indexed: 02/06/2024] Open
Abstract
Non-small cell lung cancer (NSCLC) remains one of the leading causes of cancer-related deaths worldwide. Different treatment approaches are typically employed based on the stage of NSCLC. Common clinical treatment methods include surgical resection, drug therapy, and radiation therapy. However, with the introduction and utilization of immune checkpoint inhibitors, cancer treatment has entered a new era, completely revolutionizing the treatment landscape for various cancers and significantly improving overall patient survival. Concurrently, treatment resistance often poses a critical challenge, with many patients experiencing disease progression following an initial response due to treatment resistance. Increasing evidence suggests that the tumor microenvironment (TME) plays a pivotal role in treatment resistance. Tumor-associated macrophages (TAMs) within the TME can promote treatment resistance in NSCLC by secreting various cytokines activating signaling pathways, and interacting with other immune cells. Therefore, this article will focus on elucidating the key mechanisms of TAMs in treatment resistance and analyze how targeting TAMs can reduce the levels of treatment resistance in NSCLC, providing a comprehensive understanding of the principles and approaches to overcome treatment resistance in NSCLC.
Collapse
Affiliation(s)
- Zhenjun Huang
- Department of Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Ziqi Xiao
- The Second Clinical Medical College, Nanchang University, Nanchang 330006, China
| | - Liqing Yu
- The Second Clinical Medical College, Nanchang University, Nanchang 330006, China
| | - Jiayu Liu
- The Second Clinical Medical College, Nanchang University, Nanchang 330006, China
| | - Yihan Yang
- Jiangxi Institute of Respiratory Disease, The First Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi Province, China; Jiangxi Clinical Research Center for Respiratory Diseases, Nanchang 330006, Jiangxi Province, China.
| | - Wenhao Ouyang
- Department of Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China.
| |
Collapse
|
23
|
Khafaga AF, Gaballa MMS, Karam R, Shoulah SA, Shamma RN, Khalifa NE, Farrag NE, Noreldin AE. Synergistic therapeutic strategies and engineered nanoparticles for anti-vascular endothelial growth factor therapy in cancer. Life Sci 2024; 341:122499. [PMID: 38342375 DOI: 10.1016/j.lfs.2024.122499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 01/29/2024] [Accepted: 02/07/2024] [Indexed: 02/13/2024]
Abstract
Angiogenesis is one of the defining characteristics of cancer. Vascular endothelial growth factor (VEGF) is crucial for the development of angiogenesis. A growing interest in cancer therapy is being caused by the widespread use of antiangiogenic drugs in treating several types of human cancer. However, this therapeutic approach can worsen resistance, invasion, and overall survival. As we proceed, refining combination strategies and addressing the constraint of targeted treatments are paramount. Therefore, major challenges in using novel combinations of antiangiogenic agents with cytotoxic treatments are currently focused on illustrating the potential of synergistic therapeutic strategies, alongside advancements in nanomedicine and gene therapy, present opportunities for more precise interference with angiogenesis pathways and tumor environments. Nanoparticles have the potential to regulate several crucial activities and improve several drug limitations such as lack of selectivity, non-targeted cytotoxicity, insufficient drug delivery at tumor sites, and multi-drug resistance based on their unique features. The goal of this updated review is to illustrate the enormous potential of novel synergistic therapeutic strategies and the targeted nanoparticles as an alternate strategy for t treating a variety of tumors employing antiangiogenic therapy.
Collapse
Affiliation(s)
- Asmaa F Khafaga
- Department of Pathology, Faculty of Veterinary Medicine, Alexandria University, Edfina 22758, Egypt.
| | - Mohamed M S Gaballa
- Department of Pathology, Faculty of Veterinary Medicine, Benha University, Toukh 13736, Egypt.
| | - Reham Karam
- Department of Virology, Faculty of Veterinary Medicine, Mansoura University, 35511, Egypt.
| | - Salma A Shoulah
- Department of Animal Medicine (Infectious Diseases), Faculty of Veterinary Medicine, Benha University, Toukh 13736, Egypt.
| | - Rehab N Shamma
- Department of Pharmaceutics & Industrial Pharmacy, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt.
| | - Norhan E Khalifa
- Department of Physiology, Faculty of Veterinary Medicine, Matrouh University, Matrouh 51511, Egypt.
| | - Nehal E Farrag
- Faculty of Veterinary Medicine, Alexandria University, Edfina 22758, Egypt.
| | - Ahmed E Noreldin
- Department of Histology and Cytology, Faculty of Veterinary Medicine, Damanhour University, Damanhour 22511, Egypt.
| |
Collapse
|
24
|
Wang CW, Biswas PK, Islam A, Chen MK, Chueh PJ. The Use of Immune Regulation in Treating Head and Neck Squamous Cell Carcinoma (HNSCC). Cells 2024; 13:413. [PMID: 38474377 DOI: 10.3390/cells13050413] [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: 12/19/2023] [Revised: 02/20/2024] [Accepted: 02/23/2024] [Indexed: 03/14/2024] Open
Abstract
Immunotherapy has emerged as a promising new treatment modality for head and neck cancer, offering the potential for targeted and effective cancer management. Squamous cell carcinomas pose significant challenges due to their aggressive nature and limited treatment options. Conventional therapies such as surgery, radiation, and chemotherapy often have limited success rates and can have significant side effects. Immunotherapy harnesses the power of the immune system to recognize and eliminate cancer cells, and thus represents a novel approach with the potential to improve patient outcomes. In the management of head and neck squamous cell carcinoma (HNSCC), important contributions are made by immunotherapies, including adaptive cell therapy (ACT) and immune checkpoint inhibitor therapy. In this review, we are focusing on the latter. Immune checkpoint inhibitors target proteins such as programmed cell death protein 1 (PD-1) and cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) to enhance the immune response against cancer cells. The CTLA-4 inhibitors, such as ipilimumab and tremelimumab, have been approved for early-stage clinical trials and have shown promising outcomes in terms of tumor regression and durable responses in patients with advanced HNSCC. Thus, immune checkpoint inhibitor therapy holds promise in overcoming the limitations of conventional therapies. However, further research is needed to optimize treatment regimens, identify predictive biomarkers, and overcome potential resistance mechanisms. With ongoing advancements in immunotherapy, the future holds great potential for transforming the landscape of oral tumor treatment and providing new hope for patients.
Collapse
Affiliation(s)
- Che-Wei Wang
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung 40227, Taiwan
- Department of Otorhinolaryngology-Head and Neck Surgery, Changhua Christian Hospital, Changhua 50006, Taiwan
| | - Pulak Kumar Biswas
- Institute of Molecular Medicine, National Cheng Kung University, Tainan 70101, Taiwan
| | - Atikul Islam
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung 40227, Taiwan
| | - Mu-Kuan Chen
- Department of Otorhinolaryngology-Head and Neck Surgery, Changhua Christian Hospital, Changhua 50006, Taiwan
| | - Pin Ju Chueh
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung 40227, Taiwan
| |
Collapse
|
25
|
Ma L, Deng L, Peng J, Yu J, Meng X. Chemotherapy-free radiotherapy combined with immune checkpoint inhibitors: a new regimen for locally advanced non-small cell lung cancer? Cancer Biol Med 2024; 20:j.issn.2095-3941.2023.0402. [PMID: 38318930 PMCID: PMC10845940 DOI: 10.20892/j.issn.2095-3941.2023.0402] [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: 11/01/2023] [Accepted: 12/22/2023] [Indexed: 02/07/2024] Open
Abstract
Maintenance immunotherapy after concurrent chemoradiotherapy remains the standard therapeutic approach in patients with unresectable locally advanced non-small cell lung cancer (LA-NSCLC). The efficacy of pembrolizumab without chemotherapy in stage IV NSCLC has incited interest in similar approaches for LA-NSCLC. Several recent investigations involving the synergistic potential of immunotherapy combined with radiotherapy (iRT) have generated encouraging results. This review discusses the existing studies and prospective directions of chemotherapy-free iRT strategies in unresectable LA-NSCLC. Although the initial findings of chemotherapy-free iRT strategies have shown promising efficacy, we must consider the methodologic limitations of current studies and the myriad of challenges that accompany the implementation of chemotherapy-free iRT. These challenges include determining the optimal dose and fractionation, precise target volume delineation, and identification of additional suitable patient cohorts. Furthermore, the feasibility of chemotherapy-free iRT as a novel treatment modality for select patients with LA-NSCLC is contingent upon validation through randomized phase III trials.
Collapse
Affiliation(s)
- Lin Ma
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan 430000, China
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, China
| | - Liufu Deng
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jianfeng Peng
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, China
| | - Jinming Yu
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan 430000, China
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, China
| | - Xiangjiao Meng
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, China
| |
Collapse
|
26
|
Wang L, Zhou H, Chen Q, Lin Z, Jiang C, Chen X, Chen M, Liu L, Shao L, Liu X, Pan J, Wu J, Song J, Wu J, Zhang D. STING Agonist-Loaded Nanoparticles Promotes Positive Regulation of Type I Interferon-Dependent Radioimmunotherapy in Rectal Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2307858. [PMID: 38063844 PMCID: PMC10870073 DOI: 10.1002/advs.202307858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 11/14/2023] [Indexed: 02/17/2024]
Abstract
Hypoxia-associated radioresistance in rectal cancer (RC) has severely hampered the response to radioimmunotherapy (iRT), necessitating innovative strategies to enhance RC radiosensitivity and improve iRT efficacy. Here, a catalytic radiosensitizer, DMPtNPS, and a STING agonist, cGAMP, are integrated to overcome RC radioresistance and enhance iRT. DMPtNPS promotes efficient X-ray energy transfer to generate reactive oxygen species, while alleviating hypoxia within tumors, thereby increasing radiosensitivity. Mechanistically, the transcriptomic and immunoassay analysis reveal that the combination of DMPtNPS and RT provokes bidirectional regulatory effects on the immune response, which may potentially reduce the antitumor efficacy. To mitigate this, cGAMP is loaded into DMPtNPS to reverse the negative impact of DMPtNPS and RT on the tumor immune microenvironment (TiME) through the type I interferon-dependent pathway, which promotes cancer immunotherapy. In a bilateral tumor model, the combination treatment of RT, DMPtNPS@cGAMP, and αPD-1 demonstrates a durable complete response at the primary site and enhanced abscopal effect at the distant site. This study highlights the critical role of incorporating catalytic radiosensitizers and STING agonists into the iRT approach for RC.
Collapse
Affiliation(s)
- Lei Wang
- Department of Radiation OncologyFujian Cancer HospitalFujian Medical UniversityFuzhou350025P. R. China
- Department of Oncologythe Second Affiliated Hospital of Nanchang UniversityNanchang360000P. R. China
| | - Han Zhou
- Department of Clinical OncologyThe University of Hong Kong‐Shenzhen HospitalShenzhenGuangdong518053P. R. China
| | - Qingjing Chen
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian ProvinceMengchao Hepatobiliary Hospital of Fujian Medical UniversityFuzhou350025P. R. China
- Department of Hepatopancreatobiliary SurgeryFirst Affiliated Hospital of Fujian Medical UniversityFuzhou350004P.R. China
| | - Zhiwen Lin
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian ProvinceMengchao Hepatobiliary Hospital of Fujian Medical UniversityFuzhou350025P. R. China
- Department of Hepatopancreatobiliary SurgeryFirst Affiliated Hospital of Fujian Medical UniversityFuzhou350004P.R. China
| | - Chenwei Jiang
- School of Biomedical EngineeringShanghai Jiao Tong UniversityShanghai200030P. R. China
| | - Xingte Chen
- Department of Radiation OncologyFujian Cancer HospitalFujian Medical UniversityFuzhou350025P. R. China
| | - Mingdong Chen
- Department of Radiation OncologyMengchao Hepatobiliary Hospital of Fujian Medical UniversityFuzhou350025P. R. China
| | - Libin Liu
- Department of Radiation OncologyFujian Cancer HospitalFujian Medical UniversityFuzhou350025P. R. China
| | - Lingdong Shao
- Department of Radiation OncologyFujian Cancer HospitalFujian Medical UniversityFuzhou350025P. R. China
| | - Xiaolong Liu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian ProvinceMengchao Hepatobiliary Hospital of Fujian Medical UniversityFuzhou350025P. R. China
- CAS Key Laboratory of Design and Assembly of Functional NanostructuresFujian Institute of Research on the Structure of MatterChinese Academy of SciencesFuzhou350002P. R. China
- Mengchao Med‐X CenterFuzhou UniversityFuzhou350116P. R. China
| | - Jianji Pan
- Department of Radiation OncologyFujian Cancer HospitalFujian Medical UniversityFuzhou350025P. R. China
| | - Jingcheng Wu
- Department of Health ScienceTechnology and EducationNational Health Commission of the People's Republic of ChinaBeijing100088China
| | - Jibin Song
- State Key Laboratory of Chemical Resource EngineeringCollege of ChemistryBeijing University of Chemical TechnologyBeijing10010P. R. China
| | - Junxin Wu
- Department of Radiation OncologyFujian Cancer HospitalFujian Medical UniversityFuzhou350025P. R. China
| | - Da Zhang
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian ProvinceMengchao Hepatobiliary Hospital of Fujian Medical UniversityFuzhou350025P. R. China
- Mengchao Med‐X CenterFuzhou UniversityFuzhou350116P. R. China
| |
Collapse
|
27
|
Zhang Q, Wang X, Zhao Y, Cheng Z, Fang D, Liu Y, Tian G, Li M, Luo Z. Nanointegrative In Situ Reprogramming of Tumor-Intrinsic Lipid Droplet Biogenesis for Low-Dose Radiation-Activated Ferroptosis Immunotherapy. ACS NANO 2023; 17:25419-25438. [PMID: 38055636 DOI: 10.1021/acsnano.3c08907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/08/2023]
Abstract
Low-dose radiotherapy (LDR) has shown significant implications for inflaming the immunosuppressive tumor microenvironment (TME). Surprisingly, we identify that FABP-dependent lipid droplet biogenesis in tumor cells is a key determinant of LDR-evoked cytotoxic and immunostimulatory effects and developed a nanointegrated strategy to promote the antitumor efficacy of LDR through cooperative ferroptosis immunotherapy. Specifically, TCPP-TK-PEG-PAMAM-FA, a nanoscale multicomponent functional polymer with self-assembly capability, was synthesized for cooperatively entrapping hafnium ions (Hf4+) and HIF-1α-inhibiting siRNAs (siHIF-1α). The TCPP@Hf-TK-PEG-PAMAM-FA@siHIF-1α nanoassemblies are specifically taken in by folate receptor-overexpressing tumor cells and activated by the elevated cellular ROS stress. siHIF-1α could readily inhibit the FABP3/7 expression in tumor cells via HIF-1α-FABP3/7 signaling and abolish lipid droplet biogenesis for enhancing the peroxidation susceptibility of membrane lipids, which synergizes with the elevated ROS stress in the context of Hf4+-enhanced radiation exposure and evokes pronounced ferroptotic damage in vital membrane structures. Interestingly, TCPP@Hf-TK-PEG-PAMAM-FA@siHIF-1α-enhanced ferroptotic biomembrane damage also facilitates the exposure of tumor-associated antigens (TAAs) to promote post-LDR immunotherapeutic effects, leading to robust tumor regression in vivo. This study offers a nanointegrative approach to boost the antitumor effects of LDR through the utilization of tumor-intrinsic lipid metabolism.
Collapse
Affiliation(s)
- Qiqi Zhang
- School of Life Science, Chongqing University, Chongqing 400044, P. R. China
| | - Xuan Wang
- School of Life Science, Chongqing University, Chongqing 400044, P. R. China
| | - Yuanyuan Zhao
- School of Life Science, Chongqing University, Chongqing 400044, P. R. China
| | - Zhuo Cheng
- Chongqing Institute of Advanced Pathology, Jinfeng Laboratory, Chongqing 401329, P. R. China
| | - De Fang
- School of Life Science, Chongqing University, Chongqing 400044, P. R. China
| | - Yingqi Liu
- School of Life Science, Chongqing University, Chongqing 400044, P. R. China
| | - Gan Tian
- Chongqing Institute of Advanced Pathology, Jinfeng Laboratory, Chongqing 401329, P. R. China
| | - Menghuan Li
- School of Life Science, Chongqing University, Chongqing 400044, P. R. China
| | - Zhong Luo
- School of Life Science, Chongqing University, Chongqing 400044, P. R. China
| |
Collapse
|
28
|
Chen SF, Kau M, Wang YC, Chen MH, Tung FI, Chen MH, Liu TY. Synergistically Enhancing Immunotherapy Efficacy in Glioblastoma with Gold-Core Silica-Shell Nanoparticles and Radiation. Int J Nanomedicine 2023; 18:7677-7693. [PMID: 38111846 PMCID: PMC10726961 DOI: 10.2147/ijn.s440405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Accepted: 12/12/2023] [Indexed: 12/20/2023] Open
Abstract
Purpose Glioblastoma is a highly aggressive brain tumor with universally poor outcomes. Recent progress in immune checkpoint inhibitors has led to increased interest in their application in glioblastoma. Nonetheless, the unique immune milieu in the brain has posed remarkable challenges to the efficacy of immunotherapy. We aimed to leverage the radiation-induced immunogenic cell death to overcome the immunosuppressive network in glioblastoma. Methods We developed a novel approach using the gold-core silica-shell nanoparticles (Au@SiO2 NPs) in combination with low-dose radiation to enhance the therapeutic efficacy of the immune checkpoint inhibitor (atezolizumab) in brain tumors. The biocompatibility, immune cell recruitment, and antitumor ability of the combinatorial strategy were determined using in vitro assays and in vivo models. Results Our approach successfully induced the migration of macrophages towards brain tumors and promoted cancer cell apoptosis. Subcutaneous tumor models demonstrated favorable safety profiles and significantly enhanced anticancer effects. In orthotopic brain tumor models, the multimodal therapy yielded substantial prognostic benefits over any individual modalities, achieving an impressive 40% survival rate. Conclusion In summary, the combination of Au@SiO2 NPs and low-dose radiation holds the potential to improve the clinical efficacy of immune checkpoint inhibitors. The synergetic strategy modulates tumor microenvironments and enhances systemic antitumor immunity, paving a novel way for glioblastoma treatment.
Collapse
Affiliation(s)
- Shuo-Fu Chen
- Department of Heavy Particles & Radiation Oncology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Min Kau
- Department of Biomedical Engineering, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yu-Chi Wang
- Department of Biomedical Engineering, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Ming-Hong Chen
- Division of Neurosurgery, Department of Surgery, Far Eastern Memorial Hospital, New Taipei City, Taiwan
| | - Fu-I Tung
- Department of Orthopedics, Yang-Ming Branch, Taipei City Hospital, Taipei, Taiwan
- Department of Health and Welfare, College of City Management, University of Taipei, Taipei, Taiwan
| | - Mei-Hsiu Chen
- Department of Internal Medicine, Far Eastern Memorial Hospital, New Taipei City, Taiwan
| | - Tse-Ying Liu
- Department of Biomedical Engineering, National Yang Ming Chiao Tung University, Taipei, Taiwan
| |
Collapse
|
29
|
Wang L, Katipally RR, Liang HL, Yang K, Pitroda SP, He C, Weichselbaum RR. RNA m 6A methylation and MDSCs: Roles and therapeutic implications for radiotherapy. MED 2023; 4:863-874. [PMID: 38070481 PMCID: PMC10751059 DOI: 10.1016/j.medj.2023.09.001] [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: 07/13/2023] [Revised: 08/29/2023] [Accepted: 09/13/2023] [Indexed: 12/18/2023]
Abstract
Emerging evidence suggests that local tumor radiotherapy reshapes the repertoire of circulating myeloid-derived suppressor cells (MDSCs) and leads to their infiltration into the tumor microenvironment, which poses a major obstacle for radiotherapy efficacy. Recent findings have identified RNA m6A modification at the nexus of both anti-tumor immunity and radiation response. Here, we examine the mechanisms by which this RNA modification modulates the immune milieu of the radiation-remodeled tumor microenvironment. We discuss potential therapeutic interventions targeting m6A machinery to improve radiotherapy response.
Collapse
Affiliation(s)
- Liangliang Wang
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, IL 60637, USA; Ludwig Center for Metastasis Research, University of Chicago, Chicago, IL 60637, USA.
| | - Rohan R Katipally
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, IL 60637, USA
| | - Hua Laura Liang
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, IL 60637, USA; Ludwig Center for Metastasis Research, University of Chicago, Chicago, IL 60637, USA
| | - Kaiting Yang
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, IL 60637, USA; Ludwig Center for Metastasis Research, University of Chicago, Chicago, IL 60637, USA
| | - Sean P Pitroda
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, IL 60637, USA; Ludwig Center for Metastasis Research, University of Chicago, Chicago, IL 60637, USA
| | - Chuan He
- Department of Chemistry, Department of Biochemistry and Molecular Biology, and Institute for Biophysical Dynamics, The University of Chicago, Chicago, IL 60637, USA; Howard Hughes Medical Institute, University of Chicago, Chicago, IL 60637, USA
| | - Ralph R Weichselbaum
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, IL 60637, USA; Ludwig Center for Metastasis Research, University of Chicago, Chicago, IL 60637, USA.
| |
Collapse
|
30
|
Luo H, Ma W, Chen Q, Yang Z, Dai Y. Radiotherapy-activated tumor immune microenvironment: Realizing radiotherapy-immunity combination therapy strategies. NANO TODAY 2023; 53:102042. [DOI: 10.1016/j.nantod.2023.102042] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
31
|
Hu C, Liu M, Li Y, Zhao Y, Sharma A, Liu H, Schmidt-Wolf IGH. Recent advances and future perspectives of CAR-T cell therapy in head and neck cancer. Front Immunol 2023; 14:1213716. [PMID: 37457699 PMCID: PMC10346844 DOI: 10.3389/fimmu.2023.1213716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 06/12/2023] [Indexed: 07/18/2023] Open
Abstract
Head and neck cancer (HNC) ranks as the sixth most prevalent type of cancer globally and accounts for about 4% of all types of cancer. Among all HNC, most are head and neck squamous cell carcinoma (HNSCC) with clinical therapies that include surgery, radiation therapy, chemotherapy, immunotherapy, targeted therapy, and multimodal treatments. In recent years, chimeric antigen receptor (CAR)-T cell immunotherapy has significantly transformed the therapeutic approaches for leukemia and lymphoma and has garnered increased attention as a potential treatment for a wide range of cancers. However, CAR-T immunotherapy in solid tumors, especially HNSCCs, lags significantly behind due to the paucity of tumor-specific antigens, high levels of tumor heterogeneity, immunosuppressive tumor microenvironment, the risk of treatment-related toxicities and off-target adverse events in HNSCCs. The objective of this review is to explore the advancement of CAR-T cell therapy in the treatment of HNSCCs. We aim to outline the targeted antigens in HNSCCs, highlight the challenges and potential solutions, and discuss the relevant combination therapies. Our review presents a comprehensive overview of the recent developments in CAR-T cell therapy for HNSCCs, and provides valuable insights into future research avenues.
Collapse
Affiliation(s)
- Chunmei Hu
- Department of Otolaryngology-Head and Neck Surgery, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Min Liu
- Department of Otolaryngology-Head and Neck Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Yutao Li
- Department of Integrated Oncology, Center for Integrated Oncology (CIO), University Hospital Bonn, Bonn, Germany
| | - Yi Zhao
- Department of Otolaryngology-Head and Neck Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Amit Sharma
- Department of Integrated Oncology, Center for Integrated Oncology (CIO), University Hospital Bonn, Bonn, Germany
- Department of Neurosurgery, University Hospital Bonn, Bonn, Germany
| | - Haotian Liu
- Department of Otolaryngology-Head and Neck Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Ingo G. H. Schmidt-Wolf
- Department of Integrated Oncology, Center for Integrated Oncology (CIO), University Hospital Bonn, Bonn, Germany
| |
Collapse
|