1
|
Zhang D, Song S, Lin J, Ye T, Yang X, Jiang Q, Mi Y, Zhang M, Ge X, Shen Y, Du P, Shi Y, Zhang X, Li L, Zhang Y, Ding L, Liu J, Zhang Y, Gao S, Ye Q. Glutamine binds HSC70 to transduce signals inhibiting IFN-β-mediated immunogenic cell death. Dev Cell 2025:S1534-5807(25)00117-0. [PMID: 40086433 DOI: 10.1016/j.devcel.2025.02.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 09/20/2024] [Accepted: 02/19/2025] [Indexed: 03/16/2025]
Abstract
Glutamine plays a role in cell signaling that regulates gene expression and impacts tumorigenesis. However, it is still unclear how glutamine transduces signals in cells. Here, we show that glutamine binds to heat shock cognate protein 70 (HSC70) to stimulate the deubiquitinase otubain domain containing protein (OTUD4) independently of known glutamine metabolic or signaling pathways, resulting in lactate dehydrogenase A (LDHA) stabilization via the microautophagy-lysosome pathway, increased lactate production and decreased expression of interferon (IFN)-β and its targets, hallmarks of immunogenic cell death (ICD). In cancer cell lines and patient-derived organoids and xenografts, glutamine depletion or glutamine transport inhibition combined with ICD-inducing chemotherapeutic drugs synergistically activates IFN-β, promotes CD8+ T cell recruitment, and inhibits cancer cell growth via the OTUD4/LDHA axis. CD8 expression is negatively correlated with expression of the glutamine transporter alanine/serine/cysteine transporter 2 (ASCT2), OTUD4, and LDHA in cancer patients. Thus, we identify an intracellular glutamine signaling pathway, and targeting this pathway is a promising strategy for cancer treatment.
Collapse
Affiliation(s)
- Deyu Zhang
- Department of Cell Engineering, Beijing Institute of Biotechnology, Beijing 100850, China; Department of Gastroenterology, General Hospital of Northern Theater Command, Shenyang 110840, China
| | - Songze Song
- Department of Cell Engineering, Beijing Institute of Biotechnology, Beijing 100850, China; Department of Cell Biology, Key Laboratory of Cell Biology of National Health Commission of the PRC, China Medical University, Shenyang 110122, China
| | - Jing Lin
- Department of Cell Engineering, Beijing Institute of Biotechnology, Beijing 100850, China; Department of Laboratory Medicine, The Fourth Medical Center of Chinese PLA General Hospital, Beijing 100037, China
| | - Tianxing Ye
- Department of Cell Engineering, Beijing Institute of Biotechnology, Beijing 100850, China
| | - Xiao Yang
- Department of Cell Engineering, Beijing Institute of Biotechnology, Beijing 100850, China
| | - Qiwei Jiang
- Department of Cell Engineering, Beijing Institute of Biotechnology, Beijing 100850, China
| | - Yue Mi
- Department of Cell Engineering, Beijing Institute of Biotechnology, Beijing 100850, China
| | - Mengting Zhang
- Department of Cell Engineering, Beijing Institute of Biotechnology, Beijing 100850, China
| | - Xiangwei Ge
- Department of Oncology, The Fifth Medical Center, Chinese PLA General Hospital, Beijing 100071, China
| | - Yanjie Shen
- Department of Cell Engineering, Beijing Institute of Biotechnology, Beijing 100850, China
| | - Peizhe Du
- Department of Cell Engineering, Beijing Institute of Biotechnology, Beijing 100850, China
| | - Yanzhu Shi
- Department of Cell Engineering, Beijing Institute of Biotechnology, Beijing 100850, China
| | - Xiujuan Zhang
- Department of Cell Engineering, Beijing Institute of Biotechnology, Beijing 100850, China
| | - Ling Li
- Department of Cell Engineering, Beijing Institute of Biotechnology, Beijing 100850, China
| | - Yanan Zhang
- Department of Cell Engineering, Beijing Institute of Biotechnology, Beijing 100850, China
| | - Lihua Ding
- Department of Cell Engineering, Beijing Institute of Biotechnology, Beijing 100850, China
| | - Jie Liu
- Department of Cell Engineering, Beijing Institute of Biotechnology, Beijing 100850, China
| | - Youzhi Zhang
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Shan Gao
- Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Southeast University, Nanjing 210096, China
| | - Qinong Ye
- Department of Cell Engineering, Beijing Institute of Biotechnology, Beijing 100850, China.
| |
Collapse
|
2
|
Matsuda K, Ota Y, Uemachi H, Taoda R, Tsunashima Y, Ban H, Nagai Y. Examination of the potential clinical application of 5DEX-0509R, the tumor macrophage-targeting nanomedicine. Cytokine 2025; 186:156842. [PMID: 39721228 DOI: 10.1016/j.cyto.2024.156842] [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: 11/08/2024] [Revised: 12/02/2024] [Accepted: 12/18/2024] [Indexed: 12/28/2024]
Abstract
Toll-like receptors (TLRs) are crucial for the detection of infections and activation of downstream signaling pathways that lead to the production of pro-inflammatory cytokines and interferons. Because of their strong immunostimulatory activity, TLRs are thought to be a "double-edged sword" for systemic treatment, even in the cancer field. To solve this, we have developed dextran-based TAM targeting activator conjugate (D-TAC) technology which successfully uses tumor-associated macrophages (TAMs) to deliver the TLR7 agonist DSP-0509. We have demonstrated that the anti-tumor effect of our best drug candidate 5DEX-0509R is dependent on the abundance of TAMs, which is consistent with their mechanism of action. In this study, we compared the anti-tumor effects of EIK1001 and 5DEX-0509R, and analyzed its unique immune reaction against tumors to evaluate whether 5DEX-0509R is suitable for further clinical study. 5DEX-0509R showed superior anti-tumor activity compared to EIK1001, an R848 sulfate currently in phase 2 trials, with comparable systemic cytokine profiles. 5DEX-0509R elicited unique CD4 T cell and B cell-dependent anti-tumor effects. We also found that 5DEX-0509R synergistically suppresses tumors with oxaliplatin by changing M2 macrophages that cause oxaliplatin to become resistant to antitumor M1 macrophages. In addition, 5DEX-0509R caused a rapid but not sustained cytokine elevation in both rats and dogs. We believe 5DEX-0509R is worth pursuing for clinical trials.
Collapse
Affiliation(s)
- K Matsuda
- Cancer Research Unit, Sumitomo Pharma Co Ltd, Osaka, Japan
| | - Y Ota
- Cancer Research Unit, Sumitomo Pharma Co Ltd, Osaka, Japan.
| | - H Uemachi
- Modality Research Unit, Sumitomo Pharma Co Ltd, Osaka, Japan
| | - R Taoda
- Cancer Research Unit, Sumitomo Pharma Co Ltd, Osaka, Japan
| | - Y Tsunashima
- Cancer Research Unit, Sumitomo Pharma Co Ltd, Osaka, Japan
| | - H Ban
- Cancer Research Unit, Sumitomo Pharma Co Ltd, Osaka, Japan; Modality Research Unit, Sumitomo Pharma Co Ltd, Osaka, Japan
| | - Y Nagai
- Cancer Research Unit, Sumitomo Pharma Co Ltd, Osaka, Japan.
| |
Collapse
|
3
|
Wang L, Bi S, Li Z, Liao A, Li Y, Yang L, Zhou X, Gao Y, Liu X, Zou Y, Zhang X, Shi J, Yu S, Yu Z, Guo J. Napabucasin deactivates STAT3 and promotes mitoxantrone-mediated cGAS-STING activation for hepatocellular carcinoma chemo-immunotherapy. Biomaterials 2025; 313:122766. [PMID: 39180916 DOI: 10.1016/j.biomaterials.2024.122766] [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: 05/18/2024] [Revised: 08/15/2024] [Accepted: 08/19/2024] [Indexed: 08/27/2024]
Abstract
The immune resistance of tumor microenvironment (TME) causes immune checkpoint blockade therapy inefficient to hepatocellular carcinoma (HCC). Emerging strategies of using chemotherapy regimens to reverse the immune resistance provide the promise for promoting the efficiency of immune checkpoint inhibitors. The induction of cyclic guanosine monophosphate-adenosine monophosphate synthase (cGAS)-stimulator of interferon genes (STING) in tumor cells evokes the adaptive immunity and remodels the immunosuppressive TME. In this study, we report that mitoxantrone (MIT, a chemotherapeutic drug) activates the cGAS-STING signaling pathway of HCC cells. We provide an approach to augment the efficacy of MIT using a signal transducer and activator of transcription 3 (STAT3) inhibitor called napabucasin (NAP). We prepare an aminoethyl anisamide (AEAA)-targeted polyethylene glycol (PEG)-modified poly (lactic-co-glycolic acid) (PLGA)-based nanocarrier for co-delivery of MIT and NAP. The resultant co-nanoformulation can elicit the cGAS-STING-based immune responses to reshape the immunoresistant TME in the mice orthotopically grafted with HCC. Consequently, the resultant co-nanoformulation can promote anti-PD-1 antibody for suppressing HCC development, generating long-term survival, and inhibiting tumor recurrence. This study reveals the potential of MIT to activate the cGAS-STING signaling pathway, and confirms the feasibility of nano co-delivery for MIT and NAP on achieving HCC chemo-immunotherapy.
Collapse
Affiliation(s)
- Lingzhi Wang
- School of Pharmaceutical Sciences, Jilin University, Changchun, 130021, China
| | - Shengnan Bi
- Department of Pharmacy, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, 130021, China
| | - Zhuo Li
- Department of Pharmacy, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, 130021, China
| | - Anqi Liao
- School of Pharmaceutical Sciences, Jilin University, Changchun, 130021, China
| | - Yutong Li
- School of Pharmaceutical Sciences, Jilin University, Changchun, 130021, China
| | - Leilei Yang
- School of Pharmaceutical Sciences, Jilin University, Changchun, 130021, China
| | - Xinyi Zhou
- School of Pharmaceutical Sciences, Jilin University, Changchun, 130021, China
| | - Yuqiong Gao
- School of Pharmaceutical Sciences, Jilin University, Changchun, 130021, China
| | - Xiaobo Liu
- School of Pharmaceutical Sciences, Jilin University, Changchun, 130021, China
| | - Yifang Zou
- School of Pharmaceutical Sciences, Jilin University, Changchun, 130021, China
| | - Xuemei Zhang
- Department of Hepatopathy, Shuguang Hospital, Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Jia Shi
- Department of Hepatopathy, Shuguang Hospital, Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Shihan Yu
- Department of Hepatopathy, Shuguang Hospital, Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Zhuo Yu
- Department of Hepatopathy, Shuguang Hospital, Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Jianfeng Guo
- School of Pharmaceutical Sciences, Jilin University, Changchun, 130021, China.
| |
Collapse
|
4
|
Sun X, Li X, Zhao S, Li C, Lin Y, Shen Q, Ding J, Li T, Yin Y, Tao K. Which surrogate endpoint best predict survival in locally advanced gastric cancer patients undergoing neoadjuvant chemoimmunotherapy followed by surgery? A multicenter retrospective study. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2025; 51:109517. [PMID: 39662107 DOI: 10.1016/j.ejso.2024.109517] [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: 09/16/2024] [Revised: 11/18/2024] [Accepted: 12/04/2024] [Indexed: 12/13/2024]
Abstract
INTRODUCTION Recent clinical researches have reported that neoadjuvant chemoimmunotherapy (NCIT) significantly improve the pathological complete response (pCR) and major pathological response (MPR) rates. However, surrogate endpoints for survival remains controversy for locally advanced gastric cancer (LAGC) after NCIT. METHODS A retrospective analysis was performed on 84 patients with LAGC who had undergone NCIT following radical resection in three medical centers in China, between July 2020 and September 2023. Survival curves for event-free survival (EFS) and overall survival (OS) were estimated using the Kaplan-Meier method, and the log-rank test was used to compare survival outcomes. Univariate and multivariate analyses for prognostic factors were based on Cox regression analysis. RESULTS The rates of ypN0, pCR and MPR were 60.7 % (51/84), 26.2 % (22/84) and 39.3 %(33/84),respectively. Patients with ypN0 had better EFS and OS than those with ypN+ (all p < 0.05). Survival was equivalent between pCR and non-pCR group (all p > 0.05). while patients with MPR had better EFS than those with non-MPR (p = 0.028). Furthermore, a multivariate analysis revealed that the lymph nodes(LNs) status was an independent prognostic factor for the EFS (hazard ratio [HR] 5.533, 95 % confidence interval [CI] 1.186-25.804, p = 0.029) and OS (HR 5.116, 95 % CI 1.357-19.281, p = 0.016), but not pCR and MPR (all p > 0.05). Based on the status of pathologic LNs, ypN0 group showed lower depth of tumor invasion, and lower rate of perineural and vascular invasion (all p < 0.05). CONCLUSION These findings demonstrated that ypN0 may be important as a surrogate of favorable clinical outcome in LAGC patients who received NCIT plus curative surgery.
Collapse
Affiliation(s)
- Xiong Sun
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xuanfei Li
- Department of Gastrointestinal Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Shijun Zhao
- Department of General Surgery, The Second Hospital of Shandong University, Jinan, China
| | - Chengguo Li
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yao Lin
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qian Shen
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jianing Ding
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tianhao Li
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuping Yin
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Kaixiong Tao
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
5
|
Zheng Z, Chen Y, Wang C, Li L, Tan B. Triptolide induces immunogenic cell death in cervical cancer cells via ER stress and redox modulation. Am J Cancer Res 2025; 15:69-83. [PMID: 39949945 PMCID: PMC11815369 DOI: 10.62347/xnvr3799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 12/26/2024] [Indexed: 02/16/2025] Open
Abstract
Cervical cancer remains a significant global health burden, particularly in developing countries, despite advances in screening and prevention. Novel therapeutic strategies are urgently needed to improve outcomes for patients with advanced or metastatic disease. Triptolide (TL), a key component of the Chinese herb Tripterygium wilfordii, has shown potent anticancer effects in various malignancies. In this study, we investigated the anticancer effects of TL on cervical cancer cells in vitro and in vivo, focusing on its ability to induce immunogenic cell death (ICD). TL exhibited potent cytotoxicity, inhibited proliferation, induced apoptosis, and suppressed tumor growth in cervical cancer models. Mechanistically, TL induced ICD in cervical cancer cells, as evidenced by the calreticulin (CRT) exposure on the cell surface and the release of HMGB1 and ATP. TL-induced CRT exposure was mediated by endoplasmic reticulum (ER) stress, as demonstrated by the upregulation of ATF3 and the activation of oxidative stress and immune pathways. Oral administration of TL significantly inhibited tumor growth in a cervical cancer xenograft model, without overt toxicities. These findings highlight the potential of TL as a novel immunotherapeutic agent for cervical cancer and warrant further investigation into its clinical translation. The combination of TL with immune checkpoint inhibitors or other immunotherapies may provide a promising strategy to enhance the efficacy of cervical cancer treatment while minimizing adverse effects.
Collapse
Affiliation(s)
- Ziwen Zheng
- The Second Affiliated Hospital, Jiangxi Medical College, Nanchang UniversityNanchang 330006, Jiangxi, China
- Department of Gynecological Oncology, Jiangxi Cancer Hospital, Jiangxi Cancer CenterNanchang 330029, Jiangxi, China
| | - Yamei Chen
- Department of Gynecological Oncology, Jiangxi Cancer Hospital, Jiangxi Cancer CenterNanchang 330029, Jiangxi, China
| | - Chao Wang
- Department of Gynecological Oncology, Jiangxi Cancer Hospital, Jiangxi Cancer CenterNanchang 330029, Jiangxi, China
| | - Ling Li
- Department of Gynecological Oncology, Jiangxi Maternal and Child Health HospitalNanchang 330006, Jiangxi, China
| | - Buzhen Tan
- The Second Affiliated Hospital, Jiangxi Medical College, Nanchang UniversityNanchang 330006, Jiangxi, China
| |
Collapse
|
6
|
Wu Y, Zhao Z, Ma M, Zhang W, Liu W, Liang X, Zhao T, Luo Y, Wang Y, Li M, Li T, Liu C, Luo X, Wang S, Li W, Zeng W, Wang H, Li W, Wu T, Ke Z, Luo F. Ultrasound-activated erythrocyte membrane-camouflaged Pt (II) layered double hydroxide enhances PD-1 inhibitor efficacy in triple-negative breast cancer through cGAS-STING pathway-mediated immunogenic cell death. Theranostics 2025; 15:1456-1477. [PMID: 39816689 PMCID: PMC11729553 DOI: 10.7150/thno.102284] [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: 08/13/2024] [Accepted: 12/09/2024] [Indexed: 01/18/2025] Open
Abstract
Rationale: Immunogenic cell death (ICD) offers a promising avenue for the treatment of triple-negative breast cancer (TNBC). However, optimizing immune responses remains a formidable challenge. This study presents the design of RBCm@Pt-CoNi layered double hydroxide (RmPLH), an innovative sonosensitizer for sonodynamic therapy (SDT), aimed at enhancing the efficacy of programmed cell death protein 1 (PD-1) inhibitors by inducing robust ICD responses. Methods: Pt-CoNi layered double hydroxide (LDH) nanocages were synthesized using a two-step method, followed by functionalization with red blood cell membranes to prepare RmPLH. The in vitro assessments included evaluations of cell toxicity, cellular uptake, and sonodynamic effects of RmPLH. Key mechanisms-such as oxidative stress, DNA damage, pyroptosis, cGAS/STING pathway activation, and inhibition of cellular migration and invasion-were explored under varying treatment conditions in 4T1 cells. Tumor-bearing mice were employed to evaluate tumor-targeting capabilities and the synergistic tumor-suppressive effects of RmPLH combined with PD-1 inhibitors. Comprehensive safety evaluations, including blood tests, biochemical analyses, and histopathological examinations, were also conducted. Results: The synthesized Pt-CoNi LDH exhibited a uniform rhombic dodecahedral nanocage morphology with an average particle size of approximately 231 nm. Encapsulation with red blood cell membranes conferred prolonged systemic circulation, enhanced tumor targeting, and reduced immune clearance for RmPLH. Upon ultrasound (US) stimulation, the LDH released substantial levels of reactive oxygen species (ROS) and platinum ions. The ROS effectively induced endoplasmic reticulum stress and ferroptosis, while platinum ions facilitated DNA crosslinking, triggering significant DNA damage. ROS-induced pyroptosis released inflammatory mediators and damage-associated molecular patterns (DAMPs), which activated the cGAS/STING pathway and reinforced ICD. Combining RmPLH with PD-1 inhibitors significantly enhanced therapeutic efficacy against TNBC. Furthermore, safety assessments confirmed the excellent biocompatibility and biosafety of RmPLH. Conclusion: The integration of RmPLH with PD-1 inhibitors substantially amplifies ICD, fostering robust antigen-specific T cell immunity and offering a promising therapeutic strategy for TNBC. This study represents a pioneering application of Pt (II)-based LDH nanocages in oncology, laying a foundation for future innovations in tumor immunotherapy.
Collapse
Affiliation(s)
- Yanjie Wu
- School of Science and Engineering, Shenzhen Key Laboratory of Innovative Drug Synthesis, The Chinese University of Hong Kong, Shenzhen, 518172, China
| | - Zhiyu Zhao
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen, 361102, China
- Thomas Lord Department of Mechanical Engineering and Materials Science, Duke University, Durham, NC, 27708, USA
| | - Mengli Ma
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen, 361102, China
| | - Weijin Zhang
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen, 361102, China
| | - Wei Liu
- The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangdong Pharmaceutical University, Guangzhou, 510026, China
| | - Xiaochen Liang
- Environmental Toxicology, University of California, Riverside, CA, 92507, USA
| | - Ting Zhao
- Environmental Toxicology, University of California, Riverside, CA, 92507, USA
| | - Yi Luo
- Institute of Immunotherapy, School of Basic Medicine, Fujian Medical University, Fuzhou, 350122, China
| | - Yunjie Wang
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen, 361102, China
| | - Mengqi Li
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen, 361102, China
| | - Tingting Li
- Department of Anesthesiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
| | - Cong Liu
- Department of Anesthesiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
| | - Xian Luo
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen, 361102, China
| | - Shengyu Wang
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen, 361102, China
| | - Wanyun Li
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen, 361102, China
| | - Wei Zeng
- Department of Gastroenterology, Department of Obstetrics and Gynecology, Affiliated Xiang'an Hospital, Medical Center, Xiamen University, 361000, China
| | - Hong Wang
- Department of Gastroenterology, Department of Obstetrics and Gynecology, Affiliated Xiang'an Hospital, Medical Center, Xiamen University, 361000, China
| | - Wengang Li
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen, 361102, China
| | - Ting Wu
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen, 361102, China
| | - Zhihai Ke
- School of Science and Engineering, Shenzhen Key Laboratory of Innovative Drug Synthesis, The Chinese University of Hong Kong, Shenzhen, 518172, China
| | - Fanghong Luo
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen, 361102, China
| |
Collapse
|
7
|
Hu CY, Yin YF, Xu DP, Xu Y, Yang JY, Xu YN, Hua R. Construction and validation of immunogenic cell death-related molecular clusters, signature, and immune landscape in pancreatic cancer. Clin Exp Med 2024; 25:19. [PMID: 39708151 DOI: 10.1007/s10238-024-01533-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 12/03/2024] [Indexed: 12/23/2024]
Abstract
Pancreatic cancer (PC) is a malignancy of the gastrointestinal tract that is characterized by a poor prognosis. This study investigates the roles of immunogenic cell death (ICD) genes in the prognosis and progression of PC. Expression data for PC patients were obtained from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) datasets, while ICD genes were sourced from published literature. We explored the expression patterns and identified two distinct clusters based on ICD genes. Kaplan-Meier analysis, differential expression analysis, tumor mutational burden analysis, and immune cell infiltration analysis were performed on these clusters. An ICD gene-based risk model was developed, categorizing samples from the TCGA and GEO datasets into low- and high-risk groups. Additionally, we investigated the expression levels of the genes included in the risk model within the TCGA cohort and our own samples. Finally, a loss-of-function assay was conducted to assess the role of MYD88 in PC. Two clusters of PC samples were identified, patients in the ICD-low cluster exhibited a higher degree of immune cell enrichment. The survival time of patients in the low-risk group was longer than that of those in the high-risk group. The genes included in the risk model (CASP1, MYD88, and PIK3CA) showed upregulated expression levels in tumor samples. Furthermore, the predictive accuracy of our risk model was validated using our own samples. Genetic inhibition of MYD88 led to significantly decreased proliferation and migration of PC cells in the loss-of-function assay. There were disparities in survival time and tumor immune microenvironment (TIME) between two ICD gene clusters. Additionally, we developed an ICD-related risk model that was validated as an independent prognostic indicator for patients with PC.
Collapse
Affiliation(s)
- Cheng-Yu Hu
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, People's Republic of China
| | - Yi-Fan Yin
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, People's Republic of China
| | - Da-Peng Xu
- Shanghai Key Laboratory for Cancer Systems Regulation and Clinical Translation, Department of General Surgery, Jiading District Central Hospital Affiliated Shanghai University of Medicine & Health Sciences, Shanghai, People's Republic of China.
| | - Yu Xu
- Department of Hepatopancreatobiliary Surgery, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China
| | - Jian-Yu Yang
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, People's Republic of China
| | - Yan-Nan Xu
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, People's Republic of China
| | - Rong Hua
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, People's Republic of China.
| |
Collapse
|
8
|
Hong S, Park J, Oh Y, Cho H, Kim K. Nanotechnology-Based Strategies for Safe and Effective Immunotherapy. Molecules 2024; 29:5855. [PMID: 39769944 PMCID: PMC11676242 DOI: 10.3390/molecules29245855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 12/03/2024] [Accepted: 12/10/2024] [Indexed: 01/11/2025] Open
Abstract
Cancer immunotherapy using immune checkpoint blockades has emerged as a promising therapeutic approach. However, immunotherapy faces challenges such as low response rates in solid tumors, necessitating strategies to remodel the immune-suppressive tumor microenvironment (TME) into an immune-activated state. One of the primary approaches to achieve this transformation is through the induction of immunogenic cell death (ICD). Herein, we discussed strategies to maximize ICD induction using nanoparticles. In particular, this review highlighted various studies integrating chemotherapy, radiation therapy (RT), photodynamic therapy (PDT), and photothermal therapy (PTT) with nanoparticle-based immunotherapy. The research covered in this review aims to provide valuable insights for future studies on nanoparticle-assisted immunotherapy.
Collapse
Affiliation(s)
| | | | | | | | - Kwangmeyung Kim
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul 03760, Republic of Korea; (S.H.); (J.P.); (Y.O.); (H.C.)
| |
Collapse
|
9
|
Vijayan Y, James S, Viswanathan A, Aparna JS, Bindu A, Namitha NN, Anantharaman D, Babu Lankadasari M, Harikumar KB. Targeting acid ceramidase enhances antitumor immune response in colorectal cancer. J Adv Res 2024; 65:73-87. [PMID: 38142035 PMCID: PMC11518951 DOI: 10.1016/j.jare.2023.12.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 12/14/2023] [Accepted: 12/16/2023] [Indexed: 12/25/2023] Open
Abstract
INTRODUCTION Acid ceramidase (hereafter referred as ASAH1) is an enzyme in sphingolipid metabolism that converts pro-survival ceramide into sphingosine. ASAH1 has been shown to be overexpressed in certain cancers. However, the role of ASAH1 in colorectal cancer still remain elusive. OBJECTIVE The present study is aimed to understand how ASAH1 regulates colorectal cancer (CRC) progression and resistance to checkpoint inhibitor therapy. METHODS Both pharmacological and genetic silencing of ASAH1 was used in the study. In vitro experiments were done on human and mouse CRC cell lines. The in vivo studies were conducted in NOD-SCID and BALB/c mice models. The combination of ASAH1 inhibitor and checkpoint inhibitor was tested using a syngeneic tumor model of CRC. Transcriptomic and metabolomic analyses were done to understand the effect of ASAH1 silencing. RESULTS ASAH1 is overexpressed in human CRC cases, and silencing the expression resulted in the induction of immunological cell death (ICD) and mitochondrial stress. The ASAH1 inhibitor (LCL-521), either as monotherapy or in combination with an anti-PD-1 antibody, resulted in reduction of tumors and, through induction of type I and II interferon response, activation of M1 macrophages and T cells, leading to enhanced infiltration of cytotoxic T cells. Our findings supported that the combination of LCL-521 and ICIs, which enhances the antitumor responses, and ASAH1 can be a druggable target in CRC.
Collapse
Affiliation(s)
- Yadu Vijayan
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram, 695014, India; Manipal Academy of Higher Education (MAHE), Manipal, 576104, India
| | - Shirley James
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram, 695014, India
| | - Arun Viswanathan
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram, 695014, India; Manipal Academy of Higher Education (MAHE), Manipal, 576104, India
| | - Jayasekharan S Aparna
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram, 695014, India
| | - Anu Bindu
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram, 695014, India
| | - Narayanan N Namitha
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram, 695014, India
| | - Devasena Anantharaman
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram, 695014, India
| | - Manendra Babu Lankadasari
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram, 695014, India
| | - Kuzhuvelil B Harikumar
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram, 695014, India.
| |
Collapse
|
10
|
Borges F, Laureano RS, Vanmeerbeek I, Sprooten J, Demeulenaere O, Govaerts J, Kinget L, Saraswat S, Beuselinck B, De Vleeschouwer S, Clement P, De Smet F, Sorg RV, Datsi A, Vigneron N, Naulaerts S, Garg AD. Trial watch: anticancer vaccination with dendritic cells. Oncoimmunology 2024; 13:2412876. [PMID: 39398476 PMCID: PMC11469433 DOI: 10.1080/2162402x.2024.2412876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 09/30/2024] [Accepted: 10/01/2024] [Indexed: 10/15/2024] Open
Abstract
Dendritic cells (DCs) are critical players at the intersection of innate and adaptive immunity, making them ideal candidates for anticancer vaccine development. DC-based immunotherapies typically involve isolating patient-derived DCs, pulsing them with tumor-associated antigens (TAAs) or tumor-specific antigens (TSAs), and utilizing maturation cocktails to ensure their effective activation. These matured DCs are then reinfused to elicit tumor-specific T-cell responses. While this approach has demonstrated the ability to generate potent immune responses, its clinical efficacy has been limited due to the immunosuppressive tumor microenvironment. Recent efforts have focused on enhancing the immunogenicity of DC-based vaccines, particularly through combination therapies with T cell-targeting immunotherapies. This Trial Watch summarizes recent advances in DC-based cancer treatments, including the development of new preclinical and clinical strategies, and discusses the future potential of DC-based vaccines in the evolving landscape of immuno-oncology.
Collapse
Affiliation(s)
- Francisca Borges
- Cell Stress & Immunity, Department of Cellular & Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Raquel S. Laureano
- Cell Stress & Immunity, Department of Cellular & Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Isaure Vanmeerbeek
- Cell Stress & Immunity, Department of Cellular & Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Jenny Sprooten
- Cell Stress & Immunity, Department of Cellular & Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Octavie Demeulenaere
- Cell Stress & Immunity, Department of Cellular & Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Jannes Govaerts
- Cell Stress & Immunity, Department of Cellular & Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Lisa Kinget
- Cell Stress & Immunity, Department of Cellular & Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Saurabh Saraswat
- Cell Stress & Immunity, Department of Cellular & Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Benoit Beuselinck
- Department of Medical Oncology, University Hospitals Leuven, KU Leuven, Leuven, Belgium
| | - Steven De Vleeschouwer
- Research Group Experimental Neurosurgery and Neuroanatomy, Department of Neurosciences, KU Leuven, Leuven, Belgium
- Department of Neurosurgery, University Hospitals Leuven, Leuven, Belgium
- Leuven Brain Institute (LBI), KU Leuven, Leuven, Belgium
| | - Paul Clement
- Department of Oncology, KU Leuven, Leuven, Belgium
| | - Frederik De Smet
- Laboratory for Precision Cancer Medicine, Translational Cell and Tissue Unit, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
- Leuven Institute for Single-Cell Omics (LISCO), KU Leuven, Leuven, Belgium
- Leuven Cancer Institute, KU Leuven, Leuven, Belgium
| | - Rüdiger V. Sorg
- Institute for Transplantation Diagnostics and Cell Therapeutics, Medical Faculty, Heinrich Heine University Hospital, Düsseldorf, Germany
| | - Angeliki Datsi
- Institute for Transplantation Diagnostics and Cell Therapeutics, Medical Faculty, Heinrich Heine University Hospital, Düsseldorf, Germany
| | - Nathalie Vigneron
- Ludwig Institute for Cancer Research and Cellular Genetics Unit, Université de Louvain, Brussels, Belgium
| | - Stefan Naulaerts
- Cell Stress & Immunity, Department of Cellular & Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Abhishek D. Garg
- Cell Stress & Immunity, Department of Cellular & Molecular Medicine, KU Leuven, Leuven, Belgium
| |
Collapse
|
11
|
Rencinai A, Tollapi E, Marianantoni G, Brunetti J, Henriquez T, Pini A, Bracci L, Falciani C. Branched oncolytic peptides target HSPGs, inhibit metastasis, and trigger the release of molecular determinants of immunogenic cell death in pancreatic cancer. Front Mol Biosci 2024; 11:1429163. [PMID: 39417004 PMCID: PMC11479992 DOI: 10.3389/fmolb.2024.1429163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 09/17/2024] [Indexed: 10/19/2024] Open
Abstract
Immunogenic cell death (ICD) can be exploited to treat non-immunoreactive tumors that do not respond to current standard and innovative therapies. Not all chemotherapeutics trigger ICD, among those that do exert this effect, there are anthracyclines, irinotecan, some platinum derivatives and oncolytic peptides. We studied two new branched oncolytic peptides, BOP7 and BOP9 that proved to elicit the release of damage-associated molecular patterns DAMPS, mediators of ICD, in pancreatic cancer cells. The two BOPs selectively bound and killed tumor cells, particularly PANC-1 and Mia PaCa-2, but not cells of non-tumor origin such as RAW 264.7, CHO-K1 and pgsA-745. The cancer selectivity of the two BOPs may be attributed to their repeated cationic sequences, which enable multivalent binding to heparan sulfate glycosaminoglycans (HSPGs), bearing multiple anionic sulfation patterns on cancer cells. This interaction of BOPs with HSPGs not only fosters an anti-metastatic effect in vitro, as demonstrated by reduced adhesion and migration of PANC-1 cancer cells, but also shows promising tumor-specific cytotoxicity and low hemolytic activity. Remarkably, the cytotoxicity induced by BOPs triggers the release of DAMPs, particularly HMGB1, IFN-β and ATP, by dying cells, persisting longer than the cytotoxicity of conventional chemotherapeutic agents such as irinotecan and daunorubicin. An in vivo assay in nude mice showed an encouraging 20% inhibition of tumor grafting and growth in a pancreatic cancer model by BOP9.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Chiara Falciani
- Department of Medical Biotechnology, University of Siena, Siena, Italy
| |
Collapse
|
12
|
Fan J, Gillespie KP, Mesaros C, Blair IA. HMGB2-induced calreticulin translocation required for immunogenic cell death and ferroptosis of cancer cells are controlled by the nuclear exporter XPO1. Commun Biol 2024; 7:1234. [PMID: 39354146 PMCID: PMC11445383 DOI: 10.1038/s42003-024-06930-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: 03/26/2024] [Accepted: 09/20/2024] [Indexed: 10/03/2024] Open
Abstract
Cisplatin and oxaliplatin cause the secretion of high mobility group box 1 (HMGB1) protein from cancer cells, which is necessary for initiation of immunogenic cell death (ICD). Calreticulin (CRT) translocation from the endoplasmic reticulum to the plasma membrane is also required; oxaliplatin induces this translocation but cisplatin does not. We have discovered that oxaliplatin causes the secretion of both HMGB1 and HMGB2 from the cell nucleus into the extracellular milieu. We previously showed that cisplatin-mediated secretion of HMGB1 is controlled by the nuclear exporter XPO1 (chromosomal maintenance 1; CRM1). We now find that XPO1 regulates oxaliplatin-mediated secretion of both HMGB1 and HMGB2. XPO1 inhibition causes nuclear accumulation of both proteins, inhibition of oxaliplatin-mediated ferroptosis of colon cancer cells, and inhibition of CRT translocation to the plasma membrane of lung and colon cancer cells. Incubation of cancer cells with cell targeted (CT)-HMGB2 confirmed that HMGB2 is required for the CRT translocation. Furthermore, CT-HMGB2 is three orders of magnitude more potent at inducing CRT translocation than oxaliplatin.
Collapse
Affiliation(s)
- Jingqi Fan
- Penn/CHOP Center of Excellence in Friedreich's Ataxia, Center of Excellence in Environmental Toxicology, and Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kevin P Gillespie
- Penn/CHOP Center of Excellence in Friedreich's Ataxia, Center of Excellence in Environmental Toxicology, and Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Clementina Mesaros
- Penn/CHOP Center of Excellence in Friedreich's Ataxia, Center of Excellence in Environmental Toxicology, and Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ian A Blair
- Penn/CHOP Center of Excellence in Friedreich's Ataxia, Center of Excellence in Environmental Toxicology, and Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
13
|
Larionova I, Iamshchikov P, Kazakova A, Rakina M, Menyalo M, Enikeeva K, Rafikova G, Sharifyanova Y, Pavlov V, Villert A, Kolomiets L, Kzhyshkowska J. Platinum-based chemotherapy promotes antigen presenting potential in monocytes of patients with high-grade serous ovarian carcinoma. Front Immunol 2024; 15:1414716. [PMID: 39315092 PMCID: PMC11417001 DOI: 10.3389/fimmu.2024.1414716] [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: 04/09/2024] [Accepted: 08/20/2024] [Indexed: 09/25/2024] Open
Abstract
Ovarian cancer (OC) is the most lethal gynecologic malignancy worldwide. The major clinical challenge includes the asymptomatic state of the disease, making diagnosis possible only at advanced stages. Another OC complication is the high relapse rate and poor prognosis following the standard first-line treatment with platinum-based chemotherapy. At present, numerous clinical trials are being conducted focusing on immunotherapy in OC; nevertheless, there are still no FDA-approved indications. Personalized decision regarding the immunotherapy, including immune checkpoint blockade and immune cell-based immunotherapies, can depend on the effective antigen presentation required for the cytotoxic immune response. The major aim of our study was to uncover tumor-specific transcriptional and epigenetic changes in peripheral blood monocytes in patients with high-grade serous ovarian cancer (HGSOC). Another key point was to elucidate how chemotherapy can reprogram monocytes and how that relates to changes in other immune subpopulations in the blood. To this end, we performed single-cell RNA sequencing of peripheral blood mononuclear cells (PBMCs) from patients with HGSOC who underwent neoadjuvant chemotherapeutic treatment (NACT) and in treatment-naïve patients. Monocyte cluster was significantly affected by tumor-derived factors as well as by chemotherapeutic treatment. Bioinformatical analysis revealed three distinct monocyte subpopulations within PBMCs based on feature gene expression - CD14.Mn.S100A8.9hi, CD14.Mn.MHC2hi and CD16.Mn subsets. The intriguing result was that NACT induced antigen presentation in monocytes by the transcriptional upregulation of MHC class II molecules, but not by epigenetic changes. Increased MHC class II gene expression was a feature observed across all three monocyte subpopulations after chemotherapy. Our data also demonstrated that chemotherapy inhibited interferon-dependent signaling pathways, but activated some TGFb-related genes. Our results can enable personalized decision regarding the necessity to systemically re-educate immune cells to prime ovarian cancer to respond to anti-cancer therapy or to improve personalized prescription of existing immunotherapy in either combination with chemotherapy or a monotherapy regimen.
Collapse
Affiliation(s)
- Irina Larionova
- Laboratory of Translational Cellular and Molecular Biomedicine, National Research Tomsk State University, Tomsk, Russia
- Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| | - Pavel Iamshchikov
- Laboratory of Translational Cellular and Molecular Biomedicine, National Research Tomsk State University, Tomsk, Russia
- Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| | - Anna Kazakova
- Laboratory of Translational Cellular and Molecular Biomedicine, National Research Tomsk State University, Tomsk, Russia
- Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| | - Militsa Rakina
- Laboratory of Translational Cellular and Molecular Biomedicine, National Research Tomsk State University, Tomsk, Russia
| | - Maxim Menyalo
- Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| | - Kadriia Enikeeva
- Institute of Urology and Clinical Oncology, Bashkir State Medical University of the Ministry of Health of Russia, Ufa, Russia
| | - Guzel Rafikova
- Institute of Urology and Clinical Oncology, Bashkir State Medical University of the Ministry of Health of Russia, Ufa, Russia
| | - Yuliya Sharifyanova
- Institute of Urology and Clinical Oncology, Bashkir State Medical University of the Ministry of Health of Russia, Ufa, Russia
| | - Valentin Pavlov
- Institute of Urology and Clinical Oncology, Bashkir State Medical University of the Ministry of Health of Russia, Ufa, Russia
| | - Alisa Villert
- Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| | - Larisa Kolomiets
- Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| | - Julia Kzhyshkowska
- Laboratory of Translational Cellular and Molecular Biomedicine, National Research Tomsk State University, Tomsk, Russia
- Institute of Urology and Clinical Oncology, Bashkir State Medical University of the Ministry of Health of Russia, Ufa, Russia
- Institute of Transfusion Medicine and Immunology, Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
- German Red Cross Blood Service Baden-Württemberg – Hessen, Mannheim, Germany
| |
Collapse
|
14
|
Zhou Z, Mai Y, Zhang G, Wang Y, Sun P, Jing Z, Li Z, Xu Y, Han B, Liu J. Emerging role of immunogenic cell death in cancer immunotherapy: Advancing next-generation CAR-T cell immunotherapy by combination. Cancer Lett 2024; 598:217079. [PMID: 38936505 DOI: 10.1016/j.canlet.2024.217079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 06/11/2024] [Accepted: 06/18/2024] [Indexed: 06/29/2024]
Abstract
Immunogenic cell death (ICD) is a stress-driven form of regulated cell death (RCD) in which dying tumor cells' specific signaling pathways are activated to release damage-associated molecular patterns (DAMPs), leading to the robust anti-tumor immune response as well as a reversal of the tumor immune microenvironment from "cold" to "hot". Chimeric antigen receptor (CAR)-T cell therapy, as a landmark in anti-tumor immunotherapy, plays a formidable role in hematologic malignancies but falls short in solid tumors. The Gordian knot of CAR-T cells for solid tumors includes but is not limited to, tumor antigen heterogeneity or absence, physical and immune barriers of tumors. The combination of ICD induction therapy and CAR-T cell immunotherapy is expected to promote the intensive use of CAR-T cell in solid tumors. In this review, we summarize the characteristics of ICD, stress-responsive mechanism, and the synergistic effect of various ICD-based therapies with CAR-T cells to effectively improve anti-tumor capacity.
Collapse
Affiliation(s)
- Zhaokai Zhou
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Yumiao Mai
- Department of Pediatrics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Ge Zhang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Henan Province Key Laboratory of Cardiac Injury and Repair, Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, Henan, 450052, China
| | - Yingjie Wang
- Department of Pediatrics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Pan Sun
- Department of Pediatrics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Zhaohe Jing
- Department of Pediatrics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Zhengrui Li
- Department of Oral and Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yudi Xu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Bo Han
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Jian Liu
- Department of Pediatrics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China.
| |
Collapse
|
15
|
Beltrán-Visiedo M, Serrano-Del Valle A, Jiménez-Aldúan N, Soler-Agesta R, Naval J, Galluzzi L, Marzo I. Cytofluorometric assessment of calreticulin exposure on CD38 + plasma cells from the human bone marrow. Methods Cell Biol 2024; 189:189-206. [PMID: 39393883 DOI: 10.1016/bs.mcb.2024.05.009] [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] [Indexed: 10/13/2024]
Abstract
Exposure of the endoplasmic reticulum chaperone calreticulin (CALR) on the surface of stressed and dying cells is paramount for their effective engulfment by professional antigen-presenting cells such as dendritic cells (DCs). Importantly, this is required (but not sufficient) for DCs to initiate an adaptive immune response that culminates with an effector phase as well as with the establishment of immunological memory. Conversely, the early exposure of phosphatidylserine (PS) on the outer layer of the plasma membrane is generally associated with the rapid engulfment of stressed and dying cells by tolerogenic macrophages. Supporting the clinical relevance of the CALR exposure pathway, the spontaneous or therapy-driven translocation of CALR to the surface of malignant cells, as well as intracellular biomarkers thereof, have been associated with improved disease outcome in patients affected by a variety of neoplasms, with the notable exception of multiple myeloma (MM). Here, we describe an optimized protocol for the flow cytometry-assisted quantification of surface-exposed CALR and PS on CD38+ plasma cells from the bone marrow of patients with MM. With some variations, we expect this method to be straightforwardly adaptable to the detection of CALR and PS on the surface of cancer cells isolated from patients with neoplasms other than MM.
Collapse
Affiliation(s)
- Manuel Beltrán-Visiedo
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, United States
| | | | - Nelia Jiménez-Aldúan
- Apoptosis, Immunity & Cancer Group, IIS Aragón, University of Zaragoza, Zaragoza, Spain
| | - Ruth Soler-Agesta
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, United States; Apoptosis, Immunity & Cancer Group, IIS Aragón, University of Zaragoza, Zaragoza, Spain
| | - Javier Naval
- Apoptosis, Immunity & Cancer Group, IIS Aragón, University of Zaragoza, Zaragoza, Spain
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, United States; Sandra and Edward Meyer Cancer Center, New York, NY, United States; Caryl and Israel Englander Institute for Precision Medicine, New York, NY, United States.
| | - Isabel Marzo
- Apoptosis, Immunity & Cancer Group, IIS Aragón, University of Zaragoza, Zaragoza, Spain.
| |
Collapse
|
16
|
Soler-Agesta R, Ripollés-Yuba C, Marco-Brualla J, Moreno-Loshuertos R, Sato A, Beltrán-Visiedo M, Galluzzi L, Anel A. Generation of transmitochondrial cybrids in cancer cells. Methods Cell Biol 2024; 189:23-40. [PMID: 39393884 DOI: 10.1016/bs.mcb.2024.05.010] [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] [Indexed: 10/13/2024]
Abstract
At odds with historical views suggesting that mitochondrial functions are largely dispensable for cancer cells, it is now clear that mitochondria have a major impact on malignant transformation, tumor progression and response to treatment. Mitochondria are indeed critical for neoplastic cells not only as an abundant source of ATP and other metabolic intermediates, but also as gatekeepers of apoptotic cell death and inflammation. Interestingly, while mitochondrial components are mostly encoded by nuclear genes, mitochondria contain a small, circular genome that codes for a few mitochondrial proteins, ribosomal RNAs and transfer RNAs. Here, we describe a straightforward method to generate transmitochondrial cybrids, i.e., cancer cells depleted of their mitochondrial DNA and reconstituted with intact mitochondria from another cellular source. Once established, transmitochondrial cybrids can be stably propagated and are valuable to dissect the specific impact of the mitochondrial genome on cancer cell functions.
Collapse
Affiliation(s)
- Ruth Soler-Agesta
- University of Zaragoza/Aragón Health Research Institute, Biochemistry and Molecular and Cell Biology, Zaragoza, Spain; Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, United States
| | - Cristina Ripollés-Yuba
- University of Zaragoza/Aragón Health Research Institute, Biochemistry and Molecular and Cell Biology, Zaragoza, Spain
| | - Joaquín Marco-Brualla
- University of Zaragoza/Aragón Health Research Institute, Biochemistry and Molecular and Cell Biology, Zaragoza, Spain
| | - Raquel Moreno-Loshuertos
- University of Zaragoza/Aragón Health Research Institute, Biochemistry and Molecular and Cell Biology, Zaragoza, Spain
| | - Ai Sato
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, United States
| | - Manuel Beltrán-Visiedo
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, United States
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, United States; Sandra and Edward Meyer Cancer Center, New York, NY, United States; Caryl and Israel Englander Institute for Precision Medicine, New York, NY, United States.
| | - Alberto Anel
- University of Zaragoza/Aragón Health Research Institute, Biochemistry and Molecular and Cell Biology, Zaragoza, Spain.
| |
Collapse
|
17
|
Liu W, He C, Li C, Ye S, Zhao J, Zhu C, Wang X, Ma Q, Li B. Natural compound Alternol actives multiple endoplasmic reticulum stress-responding pathways contributing to cell death. Front Pharmacol 2024; 15:1397116. [PMID: 38831880 PMCID: PMC11144888 DOI: 10.3389/fphar.2024.1397116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 05/02/2024] [Indexed: 06/05/2024] Open
Abstract
Background: Alternol is a small molecular compound isolated from the fermentation of a mutant fungus obtained from Taxus brevifolia bark. Our previous studies showed that Alternol treatment induced reactive oxygen species (ROS)-dependent immunogenic cell death. This study conducted a comprehensive investigation to explore the mechanisms involved in Alternol-induced immunogenic cell death. Methods: Prostate cancer PC-3, C4-2, and 22RV1 were used in this study. Alternol interaction with heat shock proteins (HSP) was determined using CETSA assay. Alternol-regulated ER stress proteins were assessed with Western blot assay. Extracellular adenosine triphosphate (ATP) was measured using ATPlite Luminescence Assay System. Results: Our results showed that Alternol interacted with multiple cellular chaperone proteins and increased their expression levels, including endoplasmic reticulum (ER) chaperone hypoxia up-regulated 1 (HYOU1) and heat shock protein 90 alpha family class B member 1 (HSP90AB1), as well as cytosolic chaperone heat shock protein family A member 8 (HSPA8). These data represented a potential cause of unfolded protein response (UPR) after Alternol treatment. Further investigation revealed that Alternol treatment triggered ROS-dependent (ER) stress responses via R-like ER kinase (PERK), inositol-requiring enzyme 1α (IRE1α). The double-stranded RNA-dependent protein kinase (PKR) but not activating transcription factor 6 (ATF6) cascades, leading to ATF-3/ATF-4 activation, C/EBP-homologous protein (CHOP) overexpression, and X-box binding protein XBP1 splicing induction. In addition, inhibition of these ER stress responses cascades blunted Alternol-induced extracellular adenosine triphosphate (ATP) release, one of the classical hallmarks of immunogenic cell death. Conclusion: Taken together, our data demonstrate that Alternol treatment triggered multiple ER stress cascades, leading to immunogenic cell death.
Collapse
Affiliation(s)
- Wang Liu
- Department of Urology, The University of Kansas Medical Center, Kansas City, KS, United States
| | - Chenchen He
- Department of Radiation Oncology, The First Affiliated Hospital of Xi’an Jiaotong University School of Medicine, Xi’an, China
| | - Changlin Li
- Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Shazhou Ye
- Translational Research Laboratory for Urology, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Jiang Zhao
- Department of Urology, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Cunle Zhu
- Department of Urology, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Xiangwei Wang
- Department of Urology, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Qi Ma
- Translational Research Laboratory for Urology, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Benyi Li
- Department of Urology, The University of Kansas Medical Center, Kansas City, KS, United States
| |
Collapse
|
18
|
An G, Zheng H, Guo L, Huang J, Yang C, Bai Z, Wang N, Yang W, Zhu Y. A metal-organic framework (MOF) built on surface-modified Cu nanoparticles eliminates tumors via multiple cascading synergistic therapeutic effects. J Colloid Interface Sci 2024; 662:298-312. [PMID: 38354557 DOI: 10.1016/j.jcis.2024.02.055] [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: 12/06/2023] [Revised: 02/01/2024] [Accepted: 02/05/2024] [Indexed: 02/16/2024]
Abstract
Tumors produce a hypoxic environment that greatly influences cancer treatment, and conventional chemotherapeutic drugs cannot selectively accumulate in the tumor region because of the lack of a tumor targeting mechanism, causing increased systemic toxicities and side effects. Hence, designing and developing new nanoplatforms that combine multimodal therapeutic regimens is essential to improve tumor therapeutic efficacy. Herein, we report the synthesis of ultrafine Cu nanoparticles loaded with a drug combination of cisplatin (Pt) and 1-methyl-d-tryptophan (1-MT) and externally coated with 5,10,15,20-tetrakis(4-carboxyphenyl)porphyrin (TCPP) photosensitizer, polydopamine (PDA) and CaO2 of MIL-101(Fe) as a new nanoplatform (Cu@MIL-101@PMTPC). The nanoplatform synergistically combined chemodynamic therapy (CDT), photodynamic therapy (PDT), and immunochemotherapy. The Fe3+ in MIL-101(Fe) and the surface Cu nanoparticles exhibited strong ability to consume intracellular glutathione (GSH), thereby generating a Fenton-like response in the tumor microenvironment (TME) with substantial peroxidase (POD)-like and superoxide dismutase (SOD)-like activities. In this design, we used the indoleamine 2,3-dioxygenase (IDO) inhibitor 1-MT to overcome chemotherapy-induced immune escape phenomena including enhanced CD8+ and CD4+ T cell expression, interferon-gamma (IFN-γ) and tumor necrosis factor-alpha (TNF-α) production, and accelerated immunogenic cell death. The targeted release of cisplatin loaded into Cu@MIL-101@PMTPC also reduced toxic side effects of chemotherapy. TCPP generated a large amount of singlet oxygen (1O2) upon specific laser irradiation to effectively kill tumor cells. CaO2 on the outer layer generated oxygen (O2) and hydrogen peroxide (H2O2) to ameliorate hypoxia in the tumor microenvironment, enhance the PDT effect, and provide a continuous supply of H2O2 for the Fenton-like reaction. Thus, this nanocarrier platform exhibited a powerful chemodynamic, photodynamic, and immunochemotherapeutic cascade, providing a new strategy for cancer treatment.
Collapse
Affiliation(s)
- Guanghui An
- State Key Laboratory of Featured Metal Materials and Life-cycle Safety for Composite Structures, MOE Key Laboratory of New Processing Technology for Nonferrous Metals and Materials, and School of Resources, Environment and Materials, Guangxi University, Nanning 530004, China
| | - Heming Zheng
- State Key Laboratory of Featured Metal Materials and Life-cycle Safety for Composite Structures, MOE Key Laboratory of New Processing Technology for Nonferrous Metals and Materials, and School of Resources, Environment and Materials, Guangxi University, Nanning 530004, China
| | - Lianshan Guo
- Department of Nephrology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530007, China
| | - Jingmei Huang
- Department of Nephrology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530007, China
| | - Congling Yang
- State Key Laboratory of Featured Metal Materials and Life-cycle Safety for Composite Structures, MOE Key Laboratory of New Processing Technology for Nonferrous Metals and Materials, and School of Resources, Environment and Materials, Guangxi University, Nanning 530004, China
| | - Zhihao Bai
- State Key Laboratory of Featured Metal Materials and Life-cycle Safety for Composite Structures, MOE Key Laboratory of New Processing Technology for Nonferrous Metals and Materials, and School of Resources, Environment and Materials, Guangxi University, Nanning 530004, China
| | - Nannan Wang
- State Key Laboratory of Featured Metal Materials and Life-cycle Safety for Composite Structures, MOE Key Laboratory of New Processing Technology for Nonferrous Metals and Materials, and School of Resources, Environment and Materials, Guangxi University, Nanning 530004, China.
| | - Wenhui Yang
- Department of Medical Laboratory, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi Zhuang 530021, China.
| | - Yanqiu Zhu
- State Key Laboratory of Featured Metal Materials and Life-cycle Safety for Composite Structures, MOE Key Laboratory of New Processing Technology for Nonferrous Metals and Materials, and School of Resources, Environment and Materials, Guangxi University, Nanning 530004, China; College of Engineering, Mathematics and Physical Sciences, University of Exeter, Exeter EX4 4QF, UK.
| |
Collapse
|
19
|
Arimoto KI, Miyauchi S, Liu M, Zhang DE. Emerging role of immunogenic cell death in cancer immunotherapy. Front Immunol 2024; 15:1390263. [PMID: 38799433 PMCID: PMC11116615 DOI: 10.3389/fimmu.2024.1390263] [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: 02/23/2024] [Accepted: 04/26/2024] [Indexed: 05/29/2024] Open
Abstract
Cancer immunotherapy, such as immune checkpoint blockade (ICB), has emerged as a groundbreaking approach for effective cancer treatment. Despite its considerable potential, clinical studies have indicated that the current response rate to cancer immunotherapy is suboptimal, primarily attributed to low immunogenicity in certain types of malignant tumors. Immunogenic cell death (ICD) represents a form of regulated cell death (RCD) capable of enhancing tumor immunogenicity and activating tumor-specific innate and adaptive immune responses in immunocompetent hosts. Therefore, gaining a deeper understanding of ICD and its evolution is crucial for developing more effective cancer therapeutic strategies. This review focuses exclusively on both historical and recent discoveries related to ICD modes and their mechanistic insights, particularly within the context of cancer immunotherapy. Our recent findings are also highlighted, revealing a mode of ICD induction facilitated by atypical interferon (IFN)-stimulated genes (ISGs), including polo-like kinase 2 (PLK2), during hyperactive type I IFN signaling. The review concludes by discussing the therapeutic potential of ICD, with special attention to its relevance in both preclinical and clinical settings within the field of cancer immunotherapy.
Collapse
Affiliation(s)
- Kei-ichiro Arimoto
- Moores Cancer Center, University of California San Diego, La Jolla, CA, United States
| | - Sayuri Miyauchi
- Moores Cancer Center, University of California San Diego, La Jolla, CA, United States
| | - Mengdan Liu
- Moores Cancer Center, University of California San Diego, La Jolla, CA, United States
- School of Biological Sciences, University of California San Diego, La Jolla, CA, United States
| | - Dong-Er Zhang
- Moores Cancer Center, University of California San Diego, La Jolla, CA, United States
- School of Biological Sciences, University of California San Diego, La Jolla, CA, United States
- Department of Pathology, University of California San Diego, La Jolla, CA, United States
| |
Collapse
|
20
|
Catanzaro E, Demuynck R, Naessens F, Galluzzi L, Krysko DV. Immunogenicity of ferroptosis in cancer: a matter of context? Trends Cancer 2024; 10:407-416. [PMID: 38368244 DOI: 10.1016/j.trecan.2024.01.013] [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: 12/12/2023] [Revised: 01/24/2024] [Accepted: 01/26/2024] [Indexed: 02/19/2024]
Abstract
Ferroptosis is a variant of regulated cell death (RCD) elicited by an imbalance of cellular redox homeostasis that culminates with extensive lipid peroxidation and rapid plasma membrane breakdown. Since other necrotic forms of RCD, such as necroptosis, are highly immunogenic, ferroptosis inducers have attracted considerable attention as potential tools to selectively kill malignant cells while eliciting therapeutically relevant tumor-targeting immune responses. However, rather than being consistently immunogenic, ferroptosis mediates context-dependent effects on anticancer immunity. The inability of ferroptotic cancer cells to elicit adaptive immune responses may arise from contextual deficiencies in intrinsic aspects of the process, such as adjuvanticity and antigenicity, or from microenvironmental defects imposed by ferroptotic cancer cells themselves or elicited by the induction of ferroptosis in immune cells.
Collapse
Affiliation(s)
- Elena Catanzaro
- Cell Death Investigation and Therapy (CDIT) Laboratory, Anatomy and Embryology Unit, Department of Human Structure and Repair, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium; Cancer Research Institute Ghent, Ghent University, Ghent, Belgium
| | - Robin Demuynck
- Cell Death Investigation and Therapy (CDIT) Laboratory, Anatomy and Embryology Unit, Department of Human Structure and Repair, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium; Cancer Research Institute Ghent, Ghent University, Ghent, Belgium
| | - Faye Naessens
- Cell Death Investigation and Therapy (CDIT) Laboratory, Anatomy and Embryology Unit, Department of Human Structure and Repair, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium; Cancer Research Institute Ghent, Ghent University, Ghent, Belgium
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA; Sandra and Edward Meyer Cancer Center, New York, NY, USA; Caryl and Israel Englander Institute for Precision Medicine, New York, NY, USA
| | - Dmitri V Krysko
- Cell Death Investigation and Therapy (CDIT) Laboratory, Anatomy and Embryology Unit, Department of Human Structure and Repair, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium; Cancer Research Institute Ghent, Ghent University, Ghent, Belgium.
| |
Collapse
|
21
|
Werner W, Kuzminskaya M, Lurje I, Tacke F, Hammerich L. Overcoming Resistance to Immune Checkpoint Blockade in Liver Cancer with Combination Therapy: Stronger Together? Semin Liver Dis 2024; 44:159-179. [PMID: 38806159 PMCID: PMC11245330 DOI: 10.1055/a-2334-8311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/30/2024]
Abstract
Primary liver cancer, represented mainly by hepatocellular carcinoma (HCC) and intrahepatic cholangiocarcinoma (CCA), is one of the most common and deadliest tumors worldwide. While surgical resection or liver transplantation are the best option in early disease stages, these tumors often present in advanced stages and systemic treatment is required to improve survival time. The emergence of immune checkpoint inhibitor (ICI) therapy has had a positive impact especially on the treatment of advanced cancers, thereby establishing immunotherapy as part of first-line treatment in HCC and CCA. Nevertheless, low response rates reflect on the usually cold or immunosuppressed tumor microenvironment of primary liver cancer. In this review, we aim to summarize mechanisms of resistance leading to tumor immune escape with a special focus on the composition of tumor microenvironment in both HCC and CCA, also reflecting on recent important developments in ICI combination therapy. Furthermore, we discuss how combination of ICIs with established primary liver cancer treatments (e.g. multikinase inhibitors and chemotherapy) as well as more complex combinations with state-of-the-art therapeutic concepts may reshape the tumor microenvironment, leading to higher response rates and long-lasting antitumor immunity for primary liver cancer patients.
Collapse
Affiliation(s)
- Wiebke Werner
- Department of Hepatology and Gastroenterology, Charité Universitaetsmedizin Berlin, Berlin, Germany
| | - Maria Kuzminskaya
- Department of Hepatology and Gastroenterology, Charité Universitaetsmedizin Berlin, Berlin, Germany
| | - Isabella Lurje
- Department of Hepatology and Gastroenterology, Charité Universitaetsmedizin Berlin, Berlin, Germany
| | - Frank Tacke
- Department of Hepatology and Gastroenterology, Charité Universitaetsmedizin Berlin, Berlin, Germany
| | - Linda Hammerich
- Department of Hepatology and Gastroenterology, Charité Universitaetsmedizin Berlin, Berlin, Germany
| |
Collapse
|
22
|
Blair I, Fan J, Gillespie K, Mesaros C. Ferroptosis and HMGB2 induced calreticulin translocation required for immunogenic cell death are controlled by the nuclear exporter XPO1. RESEARCH SQUARE 2024:rs.3.rs-4009459. [PMID: 38496553 PMCID: PMC10942558 DOI: 10.21203/rs.3.rs-4009459/v2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/09/2024]
Abstract
Cisplatin and oxaliplatin cause the secretion of high mobility group box 1 (HMGB1) from cancer cells, which is necessary for initiation of immunogenic cell death (ICD). Calreticulin (CRT) translocation from the endoplasmic reticulum to the plasma membrane is also required; oxaliplatin induces this translocation but cisplatin does not. We have discovered that oxaliplatin causes the secretion of both HMGB1 and HMGB2 from the nucleus into the extracellular milieu. We previously showed that cisplatin mediated secretion of HMGB1 is controlled by the nuclear exporter XPO1 (chromosomal maintenance 1; CRM1). We now find that XPO1 regulates oxaliplatin mediated secretion of both HMGB1 and HMGB2. XPO1 inhibition causes nuclear accumulation of both proteins, inhibition of oxaliplatin-mediated ferroptosis of colon cancer cells, and inhibition of CRT translocation to the plasma membrane of lung and colon cancer cells. Incubation of cancer cells with cell targeted (CT)-HMGB2 confirmed that HMGB2 is responsible for translocation of CRT to the plasma membrane. CT-HMGB2 is three orders of magnitude more potent than oxaliplatin at inducing CRT translocation. Inhibition of HMGB1 and HMGB2 secretion and/or their activation of nuclear factor-kappa B (NF-kB) has potential utility for treating cardiovascular, and neurodegenerative diseases; whereas CT-HMGB2 could augment therapeutic approaches to cancer treatment.
Collapse
|
23
|
Blair I, Fan J, Gillespie K, Mesaros C. Ferroptosis and HMGB2 induced calreticulin translocation required for immunogenic cell death are controlled by the nuclear exporter XPO1. RESEARCH SQUARE 2024:rs.3.rs-4009459. [PMID: 38496553 PMCID: PMC10942558 DOI: 10.21203/rs.3.rs-4009459/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Cisplatin and oxaliplatin cause the secretion of high mobility group box 1 (HMGB1) from cancer cells, which is necessary for initiation of immunogenic cell death (ICD). Calreticulin (CRT) translocation from the endoplasmic reticulum to the plasma membrane is also required; oxaliplatin induces this translocation but cisplatin does not. We have discovered that oxaliplatin causes the secretion of both HMGB1 and HMGB2 from the nucleus into the extracellular milieu. We previously showed that cisplatin mediated secretion of HMGB1 is controlled by the nuclear exporter XPO1 (chromosomal maintenance 1; CRM1). We now find that XPO1 regulates oxaliplatin mediated secretion of both HMGB1 and HMGB2. XPO1 inhibition causes nuclear accumulation of both proteins, inhibition of oxaliplatin-mediated ferroptosis of colon cancer cells, and inhibition of CRT translocation to the plasma membrane of lung and colon cancer cells. Incubation of cancer cells with cell targeted (CT)-HMGB2 confirmed that HMGB2 is responsible for translocation of CRT to the plasma membrane. CT-HMGB2 is three orders of magnitude more potent than oxaliplatin at inducing CRT translocation. Inhibition of HMGB1 and HMGB2 secretion and/or their activation of nuclear factor-kappa B (NF-kB) has potential utility for treating cardiovascular, and neurodegenerative diseases; whereas CT-HMGB2 could augment therapeutic approaches to cancer treatment.
Collapse
|
24
|
Apetoh L. Chemoimmunotherapy combinations: translating basic knowledge into clinical successes. Genes Immun 2024; 25:99-101. [PMID: 38480829 DOI: 10.1038/s41435-024-00264-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/19/2024]
Affiliation(s)
- Lionel Apetoh
- Brown Center for Immunotherapy, Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
| |
Collapse
|
25
|
Yi Q, He S, Liao K, Yue Z, Mei L. Nanoparticles integrated with mild photothermal therapy and oxaliplatin for tumor chemotherapy and immunotherapy. Nanomedicine (Lond) 2024; 19:841-854. [PMID: 38436253 DOI: 10.2217/nnm-2023-0335] [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] [Indexed: 03/05/2024] Open
Abstract
Aims: Preparation and evaluation of nanoparticles for tumor chemotherapy and immunotherapy mild photothermal therapy and oxaliplatin. Methods: The double emulsion method was used for nanoparticle preparations. Polydopamine was deposited on the surface, which was further modified with folic acid. Cytotoxicity assays were carried out by cell counting kit-8. In vivo antitumor assays were carried out on 4T1 tumor-bearing mice. Results: The nanoparticles exhibited a 190 nm-diameter pomegranate-like sphere, which could increase temperature to 43-46°C. In vivo distribution showed enhanced accumulation. The nanoparticles generated stronger immunogenic cell death effects. By stimulating the maturation of dendritic cells, mild photothermal therapy combined with oxaliplatin significantly increased the antitumor effect by a direct killing effect and activation of immunotherapy. Conclusion: This study provided a promising strategy of combination therapy for tumors.
Collapse
Affiliation(s)
- Qiong Yi
- Traditional Chinese Medicine Hospital of Meishan, Meishan, 620010, China
| | - Shumin He
- Affiliated Meishan Hospital of Chengdu University of TCM, Meishan, 620010, China
| | - Kai Liao
- School of Pharmacy, Chengdu University, Chengdu, 610106, China
| | - Zongxiang Yue
- Traditional Chinese Medicine Hospital of Meishan, Meishan, 620010, China
| | - Ling Mei
- School of Pharmacy, Chengdu University, Chengdu, 610106, China
| |
Collapse
|
26
|
Carnet Le Provost K, Kepp O, Kroemer G, Bezu L. Trial watch: local anesthetics in cancer therapy. Oncoimmunology 2024; 13:2308940. [PMID: 38504848 PMCID: PMC10950281 DOI: 10.1080/2162402x.2024.2308940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2024] Open
Abstract
Preclinical evidence indicates potent antitumor properties of local anesthetics. Numerous underlying mechanisms explaining such anticancer effects have been identified, suggesting direct cytotoxic as well as indirect immunemediated effects that together reduce the proliferative, invasive and migratory potential of malignant cells. Although some retrospective and correlative studies support these findings, prospective randomized controlled trials have not yet fully confirmed the antineoplastic activity of local anesthetics, likely due to the intricate methodology required for mitigating confounding factors. This trial watch aims at compiling all published preclinical and clinical research, along with completed and ongoing trials, that explore the potential antitumor effects of local anesthetics.
Collapse
Affiliation(s)
- Killian Carnet Le Provost
- Equipe Labellisée Par La Ligue Contre Le Cancer, Université de Paris, Sorbonne Université, Centre de Recherche des Cordeliers, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy, Université Paris Saclay, Villejuif, France
| | - Oliver Kepp
- Equipe Labellisée Par La Ligue Contre Le Cancer, Université de Paris, Sorbonne Université, Centre de Recherche des Cordeliers, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy, Université Paris Saclay, Villejuif, France
| | - Guido Kroemer
- Equipe Labellisée Par La Ligue Contre Le Cancer, Université de Paris, Sorbonne Université, Centre de Recherche des Cordeliers, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy, Université Paris Saclay, Villejuif, France
- Pôle de Biologie, Hôpital européen Georges Pompidou, AP-HP, Paris, France
| | - Lucillia Bezu
- Equipe Labellisée Par La Ligue Contre Le Cancer, Université de Paris, Sorbonne Université, Centre de Recherche des Cordeliers, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy, Université Paris Saclay, Villejuif, France
- Gustave Roussy, Département Anesthésie, Chirurgie et Interventionnel, Villejuif, France
| |
Collapse
|
27
|
Pan H, Liu P, Zhao L, Pan Y, Mao M, Kroemer G, Kepp O. Immunogenic cell stress and death in the treatment of cancer. Semin Cell Dev Biol 2024; 156:11-21. [PMID: 37977108 DOI: 10.1016/j.semcdb.2023.10.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 10/30/2023] [Accepted: 10/30/2023] [Indexed: 11/19/2023]
Abstract
The successful treatment of oncological malignancies which results in long-term disease control or the complete eradication of cancerous cells necessitates the onset of adaptive immune responses targeting tumor-specific antigens. Such desirable anticancer immunity can be triggered via the induction of immunogenic cell death (ICD) of cancer cells, thus converting malignant cells into an in situ vaccine that elicits T cell mediated adaptive immune responses and establishes durable immunological memory. The exploration of ICD for cancer treatment has been subject to extensive research. However, functional heterogeneity among ICD activating therapies in many cases requires specific co-medications to achieve full-blown efficacy. Here, we described the hallmarks of ICD and classify ICD activators into three distinct functional categories namely, according to their mode of action: (i) ICD inducers, which increase the immunogenicity of malignant cells, (ii) ICD sensitizers, which prime cellular circuitries for ICD induction by conventional cytotoxic agents, and (iii) ICD enhancers, which improve the perception of ICD signals by antigen presenting dendritic cells. Altogether, ICD induction, sensitization and enhancement offer the possibility to convert well-established conventional anticancer therapies into immunotherapeutic approaches that activate T cell-mediated anticancer immunity.
Collapse
Affiliation(s)
- Hui Pan
- Centre de Recherche des Cordeliers, Equipe Labellisée par la Ligue Contre le Cancer, Université de Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, 75006 Paris, France; Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, 94800 Villejuif, France
| | - Peng Liu
- Centre de Recherche des Cordeliers, Equipe Labellisée par la Ligue Contre le Cancer, Université de Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, 75006 Paris, France; Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, 94800 Villejuif, France
| | - Liwei Zhao
- Centre de Recherche des Cordeliers, Equipe Labellisée par la Ligue Contre le Cancer, Université de Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, 75006 Paris, France; Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, 94800 Villejuif, France
| | - Yuhong Pan
- Centre de Recherche des Cordeliers, Equipe Labellisée par la Ligue Contre le Cancer, Université de Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, 75006 Paris, France; Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, 94800 Villejuif, France
| | - Misha Mao
- Centre de Recherche des Cordeliers, Equipe Labellisée par la Ligue Contre le Cancer, Université de Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, 75006 Paris, France; Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, 94800 Villejuif, France
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Equipe Labellisée par la Ligue Contre le Cancer, Université de Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, 75006 Paris, France; Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, 94800 Villejuif, France; Department of Biology, Institut du Cancer Paris CARPEM, Hôpital Européen Georges Pompidou, AP-HP, 75015 Paris, France.
| | - Oliver Kepp
- Centre de Recherche des Cordeliers, Equipe Labellisée par la Ligue Contre le Cancer, Université de Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, 75006 Paris, France; Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, 94800 Villejuif, France.
| |
Collapse
|
28
|
Sabatelle RC, Chu NQ, Blessing W, Kharroubi H, Bressler E, Tsai L, Shih A, Grinstaff MW, Colson Y. Decreased Lung Metastasis in Triple Negative Breast Cancer Following Locally Delivered Supratherapeutic Paclitaxel-Loaded Polyglycerol Carbonate Nanoparticle Therapy. Biomacromolecules 2024; 25:1800-1809. [PMID: 38380618 PMCID: PMC11331584 DOI: 10.1021/acs.biomac.3c01258] [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] [Indexed: 02/22/2024]
Abstract
Breast cancer is among the most prevalent malignancies, accounting for 685,000 deaths worldwide in 2020, largely due to its high metastatic potential. Depending on the stage and tumor characteristics, treatment involves surgery, chemotherapy, targeted biologics, and/or radiation therapy. However, current treatments are insufficient for treating or preventing metastatic disease. Herein, we describe supratherapeutic paclitaxel-loaded nanoparticles (81 wt % paclitaxel) to treat the primary tumor and reduce the risk of subsequent metastatic lesions in the lungs. Primary tumor volume and lung metastasis are reduced by day 30, compared to the paclitaxel clinical standard treatment. The ultrahigh levels of paclitaxel afford an immunotherapeutic effect, increasing natural killer cell activation and decreasing NETosis in the lung, which limits the formation of metastatic lesions.
Collapse
Affiliation(s)
- Robert C. Sabatelle
- Boston University, Departments of Chemistry and Biomedical Engineering, Boston, MA 02215
| | - Ngoc-Quynh Chu
- Beth Israel Deaconess Medical Center, Department of Surgery, Boston, MA 02215
- Massachusetts General Hospital, Department of Surgery, Boston, MA 02114
| | - William Blessing
- Massachusetts General Hospital, Department of Surgery, Boston, MA 02114
| | - Hussein Kharroubi
- Massachusetts General Hospital, Department of Surgery, Boston, MA 02114
| | - Eric Bressler
- Boston University, Departments of Chemistry and Biomedical Engineering, Boston, MA 02215
| | - Lillian Tsai
- Beth Israel Deaconess Medical Center, Department of Surgery, Boston, MA 02215
| | - Angela Shih
- Massachusetts General Hospital, Department of Pathology, Boston, MA 02114
| | - Mark W. Grinstaff
- Boston University, Departments of Chemistry and Biomedical Engineering, Boston, MA 02215
| | - Yolonda Colson
- Beth Israel Deaconess Medical Center, Department of Surgery, Boston, MA 02215
- Massachusetts General Hospital, Department of Surgery, Boston, MA 02114
| |
Collapse
|
29
|
Cassanello G, Luna de Abia A, Falchi L. Trial watch: bispecific antibodies for the treatment of relapsed or refractory large B-cell lymphoma. Oncoimmunology 2024; 13:2321648. [PMID: 38445082 PMCID: PMC10913711 DOI: 10.1080/2162402x.2024.2321648] [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: 01/17/2024] [Revised: 02/15/2024] [Accepted: 02/16/2024] [Indexed: 03/07/2024] Open
Abstract
Immunotherapy has shaped the treatment approach to diffuse large B-cell lymphoma (DLBCL), with rituximab leading to remarkable improvements in outcomes for both relapsed and treatment-naïve patients. Recently, groundbreaking immunotherapies like chimeric antigen receptor T-cells have entered the treatment arena for relapsed/refractory (R/R) DLBCL and gained regulatory approval in several countries. The concept of harnessing a patient's own T-cells to combat cancer has been further explored through the development of bispecific antibodies (BsAbs), a class of engineered antibody products designed to simultaneously target two different antigens. These novel drugs have demonstrated impressive single-agent activity and manageable toxicity in patients with heavily pretreated B-cell non-Hodgkin lymphoma. In this review, we provide an up-to-date overview of recently completed or ongoing BsAbs trials in patients with R/R DLBCL, including single-agent results, emerging combination data, and novel constructs.
Collapse
Affiliation(s)
- Giulio Cassanello
- Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, USA
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Alejandro Luna de Abia
- Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, USA
- Hematology Service, Hospital Universitario Ramon y Cajal, Madrid, Spain
| | - Lorenzo Falchi
- Department of Medicine, Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, USA
| |
Collapse
|
30
|
Kroemer G, Chan TA, Eggermont AMM, Galluzzi L. Immunosurveillance in clinical cancer management. CA Cancer J Clin 2024; 74:187-202. [PMID: 37880100 PMCID: PMC10939974 DOI: 10.3322/caac.21818] [Citation(s) in RCA: 45] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 09/27/2023] [Accepted: 10/03/2023] [Indexed: 10/27/2023] Open
Abstract
The progression of cancer involves a critical step in which malignant cells escape from control by the immune system. Antineoplastic agents are particularly efficient when they succeed in restoring such control (immunosurveillance) or at least establish an equilibrium state that slows down disease progression. This is true not only for immunotherapies, such as immune checkpoint inhibitors (ICIs), but also for conventional chemotherapy, targeted anticancer agents, and radiation therapy. Thus, therapeutics that stress and kill cancer cells while provoking a tumor-targeting immune response, referred to as immunogenic cell death, are particularly useful in combination with ICIs. Modern oncology regimens are increasingly using such combinations, which are referred to as chemoimmunotherapy, as well as combinations of multiple ICIs. However, the latter are generally associated with severe side effects compared with single-agent ICIs. Of note, the success of these combinatorial strategies against locally advanced or metastatic cancers is now spurring successful attempts to move them past the postoperative (adjuvant) setting to the preoperative (neoadjuvant) setting, even for patients with operable cancers. Here, the authors critically discuss the importance of immunosurveillance in modern clinical cancer management.
Collapse
Affiliation(s)
- Guido Kroemer
- Centre de Recherche des Cordeliers, Equipe Labellisée par la Ligue Contre le Cancer, Inserm U1138, Université Paris Cité, Sorbonne Université, Institut Universitaire de France, Paris, France; Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Villejuif, France; Institut du Cancer Paris Carpem, Department of Biology, Hôpital Européen Georges Pompidou, AP-HP, Paris, France
| | - Timothy A. Chan
- Department of Radiation Oncology, Taussig Cancer Center, Cleveland Clinic, Cleveland, OH, USA; Center for Immunotherapy and Precision Immuno-Oncology, Cleveland Clinic, Cleveland, OH, USA; National Center for Regenerative Medicine, Cleveland, OH, USA; Case Comprehensive Cancer Center, Cleveland, OH, USA
| | - Alexander M. M. Eggermont
- University Medical Center Utrecht & Princess Maxima Center, Utrecht, the Netherlands; Comprehensive Cancer Center München, Technical University München & Ludwig Maximilian University, München, Germany
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA; Sandra and Edward Meyer Cancer Center, New York, NY, USA; Caryl and Israel Englander Institute for Precision Medicine, New York, NY, USA
| |
Collapse
|
31
|
Phan TT, Scott KS, Chelette B, Phillip West A, Dantzer R. The fatigue-inducing effects of cancer and its therapy are characterized by decreased physical activity in the absence of any motivational deficit. Brain Behav Immun 2024; 117:205-214. [PMID: 38244945 PMCID: PMC11633841 DOI: 10.1016/j.bbi.2024.01.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Revised: 01/09/2024] [Accepted: 01/14/2024] [Indexed: 01/22/2024] Open
Abstract
Although cancer and its therapy are well known to be associated with fatigue, the exact nature of cancer-related fatigue remains ill-defined. We previously reported that fatigue-like behavior induced independently by tumor growth and by the chemotherapeutic agent cisplatin is characterized by reduced voluntary wheel running and an intact motivation to expand effort for food rewards. The present set of experiments was initiated to characterize the functional consequences of fatigue induced by chemoradiotherapy in tumor-bearing mice and relate them to changes in the expression of genes coding for inflammation, mitochondria dynamics and metabolism. Two syngeneic murine models of cancer were selected for this purpose, a model of human papilloma virus-related head and neck cancer and a model of lung cancer. In both models, tumor-bearing mice were submitted to chemoradiotherapy to limit tumor progression. Two dimensions of fatigue were assessed, the physical dimension by changes in physical activity in mice trained to run in wheels and the motivational dimension by changes in the performance of mice trained to nose poke to obtain a food reward in a progressive ratio schedule of food reinforcement. Chemoradiotherapy reliably decreased wheel running activity but had no effect on performance in the progressive ratio in both murine models of cancer. These effects were the same for the two murine models of cancer and did not differ according to sex. Livers and brains were collected at the end of the experiments for qRT-PCR analysis of expression of genes coding for inflammation, mitochondria dynamics, and metabolism. The observed changes were mainly apparent in the liver and typical of activation of type I interferon and NF-κB-dependent signaling, with alterations in mitochondrial dynamics and a shift toward glycolysis. Although the importance of these alterations for the pathophysiology of cancer-related fatigue remains to be explored, the present findings indicate that fatigue brought on by cancer therapy in tumor-bearing mice is more physical than motivational.
Collapse
Affiliation(s)
- Thien T Phan
- Department of Symptom Research, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Current address: Department of Medical Physiology, School of Medicine, Texas A&M University, Bryan, TX 77807, USA
| | - Kiersten S Scott
- Department of Symptom Research, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Present address: Department of Neurology, McGovern School of Medicine, UT Health Houston, TX 77030, USA
| | - Brandon Chelette
- Department of Symptom Research, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - A Phillip West
- Department of Microbial Pathogenesis and Immunology, School of Medicine, Texas A&M University, Bryan, TX 77087, USA; Present address: The Jackson Laboratory, Bar Harbor, ME 04609, USA
| | - Robert Dantzer
- Department of Symptom Research, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
32
|
Pang G, Wei S, Zhao J, Wang FJ. Improving nanochemoimmunotherapy efficacy by boosting "eat-me" signaling and downregulating "don't-eat-me" signaling with Ganoderma lucidum polysaccharide-based drug delivery. J Mater Chem B 2023; 11:11562-11577. [PMID: 37982298 DOI: 10.1039/d3tb02118a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2023]
Abstract
To address the challenges posed by low immunogenicity and immune checkpoints during cancer treatment, we propose an alternative strategy that combines immunogenic cell death (ICD) effects with CD47/SIRPα blockade to reactivate phagocytosis of tumor cells by macrophages with polysaccharide-based drug delivery. In this study, the EGFR inhibitor gefitinib was identified as a novel CD47 modulator, which promoted the translocation of CD47 molecules from the cell membrane to endosomes through the EGFR-Rab5 pathway, leading to reduced cell surface CD47 levels and limiting interaction with SIRPα. Based on this finding, we developed prophagocytic mixed nanodrugs to enhance macrophage phagocytosis by encapsulating ICD inducer doxorubicin and CD47 inhibitor gefitinib with immunostimulatory polysaccharides from Ganoderma lucidum. This approach downregulated cell surface CD47 expression to attenuate "don't-eat-me" signaling, while increasing doxorubicin accumulation in tumors by inhibiting drug-resistance proteins, leading to more exposure of calreticulin and amplifying the "eat-me" signaling. In vivo experiments demonstrated that this approach significantly suppressed intraperitoneal tumor dissemination, reversed doxorubicin-induced weight loss, and effectively induced macrophage polarization, dendritic cell maturation, and CD8+ T cell activation. These findings highlighted the significant potential of our macrophage-centered therapeutic strategy using polysaccharide-based nanocarriers and provided new perspectives for chemoimmunotherapy.
Collapse
Affiliation(s)
- Guibin Pang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, P. R. China.
| | - Siqi Wei
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, P. R. China.
| | - Jian Zhao
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, P. R. China.
- ECUST-FONOW Joint Research Center for Innovative Medicines, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, P. R. China
| | - Fu-Jun Wang
- New Drug R&D Center, Zhejiang Fonow Medicine Co., Ltd., 209 West Hulian Road, Dongyang 322100, Zhejiang, P. R. China
- ECUST-FONOW Joint Research Center for Innovative Medicines, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, P. R. China
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai 201203, P. R. China.
| |
Collapse
|
33
|
Wimmer K, Sachet M, Ramos C, Frantal S, Birnleitner H, Brostjan C, Exner R, Filipits M, Bago-Horvath Z, Rudas M, Bartsch R, Gnant M, Singer CF, Balic M, Egle D, Oehler R, Fitzal F. Differential immunomodulatory effects of epirubicin/cyclophosphamide and docetaxel in breast cancer patients. J Exp Clin Cancer Res 2023; 42:300. [PMID: 37957750 PMCID: PMC10644559 DOI: 10.1186/s13046-023-02876-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 10/29/2023] [Indexed: 11/15/2023] Open
Abstract
BACKGROUND Epirubicin/cyclophosphamide (EC) and docetaxel (D) are commonly used in a sequential regimen in the neoadjuvant treatment of early, high-risk or locally advanced breast cancer (BC). Novel approaches to increase the response rate combine this treatment with immunotherapies such as PD-1 inhibition. However, the expected stimulatory effect on lymphocytes may depend on the chemotherapy backbone. Therefore, we separately compared the immunomodulatory effects of EC and D in the setting of a randomized clinical trial. METHODS Tumor and blood samples of 154 patients from the ABCSG-34 trial were available (76 patients received four cycles of EC followed by four cycles of D; 78 patients get the reverse treatment sequence). Tumor-infiltrating lymphocytes, circulating lymphocytes and 14 soluble immune mediators were determined at baseline and at drug change. Furthermore, six BC cell lines were treated with E, C or D and co-cultured with immune cells. RESULTS Initial treatment with four cycles of EC reduced circulating B and T cells by 94% and 45%, respectively. In contrast, no comparable effects on lymphocytes were observed in patients treated with initial four cycles of D. Most immune mediators decreased under EC whereas D-treatment resulted in elevated levels of CXCL10, urokinase-type plasminogen activator (uPA) and its soluble receptor (suPAR). Accordingly, only the exposure of BC cell lines to D induced similar increases as compared to E. While treatment of BC cells with E was associated with cell shrinkage and apoptosis, D induced cell swelling and accumulation of cells in G2 phase. CONCLUSION The deleterious effect of EC on lymphocytes indicates strong immunosuppressive properties of this combination therapy. D, in contrast, has no effect on lymphocytes, but triggers the secretion of stimulatory proteins in vivo and in vitro, indicating a supportive effect on the immune system. Underlying differences in the induced cell death might be causal. These divergent immunomodulatory effects of epirubicin/cyclophosphamide and docetaxel should be considered when planning future combinations with immunotherapies in breast cancer.
Collapse
Affiliation(s)
- Kerstin Wimmer
- Department of General Surgery, Division of Visceral Surgery and Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, A-1090, Vienna, Austria
- Austrian Breast & Colorectal Cancer Study Group (ABCSG), Vienna, Austria
| | - Monika Sachet
- Department of General Surgery, Division of Visceral Surgery and Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, A-1090, Vienna, Austria
| | - Cristiano Ramos
- Department of General Surgery, Division of Visceral Surgery and Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, A-1090, Vienna, Austria
| | - Sophie Frantal
- Austrian Breast & Colorectal Cancer Study Group (ABCSG), Vienna, Austria
| | - Hanna Birnleitner
- Department of General Surgery, Division of Visceral Surgery and Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, A-1090, Vienna, Austria
| | - Christine Brostjan
- Department of General Surgery, Division of Vascular Surgery, Medical University of Vienna, 1090, Vienna, Austria
| | - Ruth Exner
- Department of General Surgery, Division of Visceral Surgery and Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, A-1090, Vienna, Austria
| | - Martin Filipits
- Austrian Breast & Colorectal Cancer Study Group (ABCSG), Vienna, Austria
- Center for Cancer Research, Medical University of Vienna, 1090, Vienna, Austria
| | - Zsuzsanna Bago-Horvath
- Austrian Breast & Colorectal Cancer Study Group (ABCSG), Vienna, Austria
- Department of Pathology, Medical University of Vienna, 1090, Vienna, Austria
- Comprehensive Cancer Center, Medical University of Vienna, Spitalgasse 23, 1090, Vienna, Austria
| | - Margaretha Rudas
- Department of Pathology, Medical University of Vienna, 1090, Vienna, Austria
- Comprehensive Cancer Center, Medical University of Vienna, Spitalgasse 23, 1090, Vienna, Austria
| | - Rupert Bartsch
- Austrian Breast & Colorectal Cancer Study Group (ABCSG), Vienna, Austria
- Department of Medicine 1, Division of Oncology, Medical University of Vienna, 1090, Vienna, Austria
| | - Michael Gnant
- Austrian Breast & Colorectal Cancer Study Group (ABCSG), Vienna, Austria
- Comprehensive Cancer Center, Medical University of Vienna, Spitalgasse 23, 1090, Vienna, Austria
| | - Christian F Singer
- Austrian Breast & Colorectal Cancer Study Group (ABCSG), Vienna, Austria
- Comprehensive Cancer Center, Medical University of Vienna, Spitalgasse 23, 1090, Vienna, Austria
- Department of Gynecology, Medical University of Vienna, 1090, Vienna, Austria
| | - Marija Balic
- Austrian Breast & Colorectal Cancer Study Group (ABCSG), Vienna, Austria
- Department of Oncology, Medical University of Graz, Graz, Austria
| | - Daniel Egle
- Austrian Breast & Colorectal Cancer Study Group (ABCSG), Vienna, Austria
- Department of Gynecology, Medical University Innsbruck, Innsbruck, Austria
| | - Rudolf Oehler
- Department of General Surgery, Division of Visceral Surgery and Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, A-1090, Vienna, Austria.
- Austrian Breast & Colorectal Cancer Study Group (ABCSG), Vienna, Austria.
| | - Florian Fitzal
- Department of General Surgery, Division of Visceral Surgery and Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, A-1090, Vienna, Austria
- Austrian Breast & Colorectal Cancer Study Group (ABCSG), Vienna, Austria
| |
Collapse
|
34
|
Gabizon A, Shmeeda H, Draper B, Parente-Pereira A, Maher J, Carrascal-Miniño A, de Rosales RTM, La-Beck NM. Harnessing Nanomedicine to Potentiate the Chemo-Immunotherapeutic Effects of Doxorubicin and Alendronate Co-Encapsulated in Pegylated Liposomes. Pharmaceutics 2023; 15:2606. [PMID: 38004584 PMCID: PMC10675201 DOI: 10.3390/pharmaceutics15112606] [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: 10/06/2023] [Revised: 10/31/2023] [Accepted: 11/03/2023] [Indexed: 11/26/2023] Open
Abstract
Encapsulation of Doxorubicin (Dox), a potent cytotoxic agent and immunogenic cell death inducer, in pegylated (Stealth) liposomes, is well known to have major pharmacologic advantages over treatment with free Dox. Reformulation of alendronate (Ald), a potent amino-bisphosphonate, by encapsulation in pegylated liposomes, results in significant immune modulatory effects through interaction with tumor-associated macrophages and activation of a subset of gamma-delta T lymphocytes. We present here recent findings of our research work with a formulation of Dox and Ald co-encapsulated in pegylated liposomes (PLAD) and discuss its pharmacological properties vis-à-vis free Dox and the current clinical formulation of pegylated liposomal Dox. PLAD is a robust formulation with high and reproducible remote loading of Dox and high stability in plasma. Results of biodistribution studies, imaging with radionuclide-labeled liposomes, and therapeutic studies as a single agent and in combination with immune checkpoint inhibitors or gamma-delta T lymphocytes suggest that PLAD is a unique product with distinct tumor microenvironmental interactions and distinct pharmacologic properties when compared with free Dox and the clinical formulation of pegylated liposomal Dox. These results underscore the potential added value of PLAD for chemo-immunotherapy of cancer and the relevance of the co-encapsulation approach in nanomedicine.
Collapse
Affiliation(s)
- Alberto Gabizon
- Nano-Oncology Research Center, Oncology Institute, Shaare Zedek Medical Center, Jerusalem 9103102, Israel;
- Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112102, Israel
| | - Hilary Shmeeda
- Nano-Oncology Research Center, Oncology Institute, Shaare Zedek Medical Center, Jerusalem 9103102, Israel;
| | - Benjamin Draper
- King’s College London, School of Cancer and Pharmaceutical Sciences, Guy’s Cancer Centre, Great Maze Pond, London SE1 9RT, UK; (B.D.); (A.P.-P.); (J.M.)
| | - Ana Parente-Pereira
- King’s College London, School of Cancer and Pharmaceutical Sciences, Guy’s Cancer Centre, Great Maze Pond, London SE1 9RT, UK; (B.D.); (A.P.-P.); (J.M.)
| | - John Maher
- King’s College London, School of Cancer and Pharmaceutical Sciences, Guy’s Cancer Centre, Great Maze Pond, London SE1 9RT, UK; (B.D.); (A.P.-P.); (J.M.)
| | - Amaia Carrascal-Miniño
- King’s College London, School of Biomedical Engineering & Imaging Sciences, St. Thomas’ Hospital, London SE1 7EH, UK; (A.C.-M.); (R.T.M.d.R.)
| | - Rafael T. M. de Rosales
- King’s College London, School of Biomedical Engineering & Imaging Sciences, St. Thomas’ Hospital, London SE1 7EH, UK; (A.C.-M.); (R.T.M.d.R.)
| | - Ninh M. La-Beck
- Department of Immunotherapeutics and Biotechnology, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX 79601, USA;
| |
Collapse
|
35
|
Redkin TS, Sleptsova EE, Turubanova VD, Saviuk MO, Lermontova SA, Klapshina LG, Peskova NN, Balalaeva IV, Krysko O, Mishchenko TA, Vedunova MV, Krysko DV. Dendritic Cells Pulsed with Tumor Lysates Induced by Tetracyanotetra(aryl)porphyrazines-Based Photodynamic Therapy Effectively Trigger Anti-Tumor Immunity in an Orthotopic Mouse Glioma Model. Pharmaceutics 2023; 15:2430. [PMID: 37896190 PMCID: PMC10610423 DOI: 10.3390/pharmaceutics15102430] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 09/26/2023] [Accepted: 10/03/2023] [Indexed: 10/29/2023] Open
Abstract
Research in the past decade on immunogenic cell death (ICD) has shown that the immunogenicity of dying tumor cells is crucial for effective anticancer therapy. ICD induction leads to the emission of specific damage-associated molecular patterns (DAMPs), which act as danger signals and as adjuvants to activate specific anti-tumor immune responses, leading to the elimination of tumor cells and the formation of long-term immunological memory. ICD can be triggered by many anticancer treatment modalities, including photodynamic therapy (PDT). However, due to the variety of photosensitizers used and the lack of a universally adopted PDT protocol, there is a need to develop novel PDT with a proven ICD capability. In the present study, we characterized the abilities of two photoactive dyes to induce ICD in experimental glioma in vitro and in vivo. One dye was from the tetracyanotetra(aryl)porphyrazine group with 9-phenanthrenyl (pz I), and the other was from the 4-(4-fluorobenzyoxy)phenyl (pz III) group in the aryl frame of the macrocycle. We showed that after the photosensitizers penetrated into murine glioma GL261 cells, they localized predominantly in the Golgi apparatus and partially in the endoplasmic reticulum, providing efficient phototoxic activity against glioma GL261 cells upon light irradiation at a dose of 20 J/cm2 (λex 630 nm; 20 mW/cm2). We demonstrated that pz I-PDT and pz III-PDT can act as efficient ICD inducers when applied to glioma GL261 cells, facilitating the release of two crucial DAMPs (ATP and HMGB1). Moreover, glioma GL261 cells stimulated with pz I-PDT or pz III-PDT provided strong protection against tumor growth in a prophylactic subcutaneous glioma vaccination model. Finally, we showed that dendritic cell (DC) vaccines pulsed with the lysates of glioma GL261 cells pre-treated with pz-I-PDT or pz-III-PDT could act as effective inducers of adaptive anti-tumor immunity in an intracranial orthotopic glioma mouse model.
Collapse
Affiliation(s)
- Tikhon S. Redkin
- Institute of Neurosciences, National Research Lobachevsky State University of Nizhny Novgorod, 23 Gagarin Ave., 603022 Nizhny Novgorod, Russia; (T.S.R.); (E.E.S.); (M.O.S.)
| | - Ekaterina E. Sleptsova
- Institute of Neurosciences, National Research Lobachevsky State University of Nizhny Novgorod, 23 Gagarin Ave., 603022 Nizhny Novgorod, Russia; (T.S.R.); (E.E.S.); (M.O.S.)
| | - Victoria D. Turubanova
- Institute of Neurosciences, National Research Lobachevsky State University of Nizhny Novgorod, 23 Gagarin Ave., 603022 Nizhny Novgorod, Russia; (T.S.R.); (E.E.S.); (M.O.S.)
| | - Mariia O. Saviuk
- Institute of Neurosciences, National Research Lobachevsky State University of Nizhny Novgorod, 23 Gagarin Ave., 603022 Nizhny Novgorod, Russia; (T.S.R.); (E.E.S.); (M.O.S.)
- Cell Death Investigation and Therapy Laboratory, Anatomy and Embryology Unit, Department of Human Structure and Repair, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium;
- Cancer Research Institute Ghent, 9000 Ghent, Belgium
| | - Svetlana A. Lermontova
- Sector of Chromophors for Medicine, G.A. Razuvaev Institute of Organometallic Chemistry of the Russian Academy of Sciences, 49 Tropinin St., 603137 Nizhny Novgorod, Russia; (S.A.L.); (L.G.K.)
| | - Larisa G. Klapshina
- Sector of Chromophors for Medicine, G.A. Razuvaev Institute of Organometallic Chemistry of the Russian Academy of Sciences, 49 Tropinin St., 603137 Nizhny Novgorod, Russia; (S.A.L.); (L.G.K.)
| | - Nina N. Peskova
- Institute of Biology and Biomedicine, National Research Lobachevsky State University of Nizhny Novgorod, 23 Gagarin Ave., 603022 Nizhny Novgorod, Russia; (N.N.P.); (I.V.B.); (T.A.M.); (M.V.V.)
| | - Irina V. Balalaeva
- Institute of Biology and Biomedicine, National Research Lobachevsky State University of Nizhny Novgorod, 23 Gagarin Ave., 603022 Nizhny Novgorod, Russia; (N.N.P.); (I.V.B.); (T.A.M.); (M.V.V.)
| | - Olga Krysko
- Cell Death Investigation and Therapy Laboratory, Anatomy and Embryology Unit, Department of Human Structure and Repair, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium;
| | - Tatiana A. Mishchenko
- Institute of Biology and Biomedicine, National Research Lobachevsky State University of Nizhny Novgorod, 23 Gagarin Ave., 603022 Nizhny Novgorod, Russia; (N.N.P.); (I.V.B.); (T.A.M.); (M.V.V.)
| | - Maria V. Vedunova
- Institute of Biology and Biomedicine, National Research Lobachevsky State University of Nizhny Novgorod, 23 Gagarin Ave., 603022 Nizhny Novgorod, Russia; (N.N.P.); (I.V.B.); (T.A.M.); (M.V.V.)
| | - Dmitri V. Krysko
- Cell Death Investigation and Therapy Laboratory, Anatomy and Embryology Unit, Department of Human Structure and Repair, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium;
- Cancer Research Institute Ghent, 9000 Ghent, Belgium
- Department of Pathophysiology, Sechenov First Moscow State Medical University (Sechenov University), 125009 Moscow, Russia
| |
Collapse
|
36
|
Yang Y, Shao Y, Wang J, Cheng Q, Yang H, Li Y, Liu J, Zhou Y, Zhou Z, Wang M, Ji B, Yao J. Development and validation of novel immune-inflammation-based clinical predictive nomograms in HER2-negative advanced gastric cancer. Front Oncol 2023; 13:1185240. [PMID: 37746295 PMCID: PMC10516559 DOI: 10.3389/fonc.2023.1185240] [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: 03/15/2023] [Accepted: 08/07/2023] [Indexed: 09/26/2023] Open
Abstract
Purpose To explore the predictive value of multiple immune-inflammatory biomarkers including serum VEGFA and systemic immune-inflammation index (SII) in HER2-negative advanced gastric cancer (AGC) and establish nomograms for predicting the first-line chemotherapeutic efficacy, progression-free survival (PFS) and overall survival (OS) of patients with this fatal disease. Methods From November 2017 to April 2022, 102 and 34 patients with a diagnosis of HER2-negative AGC at the First Affiliated Hospital of Bengbu Medical College were enrolled as development and validation cohorts, respectively. Univariate and multivariate analyses were performed to evaluate the clinical value of the candidate indicators. The variables were screened using LASSO regression analysis. Predictive models were developed using significant predictors and are displayed as nomograms. Results Baseline VEGFA expression was significantly higher in HER2-negative AGC patients than in nonneoplastic patients and was associated with malignant serous effusion and therapeutic efficacy (all p<0.001). Multivariate analysis indicated that VEGFA was an independent predictor for first-line therapeutic efficacy and PFS (both p<0.01) and SII was an independent predictor for first-line PFS and OS (both p<0.05) in HER2-negative AGC patients. The therapeutic efficacy model had an R2 of 0.37, a Brier score of 0.15, and a Harrell's C-index of 0.82 in the development cohort and 0.90 in the validation cohort. The decision curve analysis indicated that the model added more net benefits than VEGFA assessment alone. The PFS/OS models had Harrell's C-indexes of 0.71/0.69 in the development cohort and 0.71/0.62 in the validation cohort. Conclusion The established nomograms integrating serum VEGFA/SII and commonly available baseline characteristics provided satisfactory performance in predicting the therapeutic efficacy and prognosis of HER2-negative AGC patients.
Collapse
Affiliation(s)
- Yan Yang
- Department of Medical Oncology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Yu Shao
- Department of Medical Oncology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Junjun Wang
- Department of Radiation Oncology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Qianqian Cheng
- Department of Medical Oncology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Hanqi Yang
- Department of Clinical Laboratory, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Yulong Li
- Department of Surgical Oncology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Jing Liu
- Department of Medical Oncology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Yangyang Zhou
- Department of Medical Oncology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Zhengguang Zhou
- Department of Medical Oncology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Mingxi Wang
- Department of Medical Oncology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Baoan Ji
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, United States
| | - Jinghao Yao
- Department of Medical Oncology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|