1
|
Yan R, Cheng X, Song Y, Wang H, Zhang R, Jin Y, Li X, Chen Y, Xiang H. Cuproptosis nanoprodrug-initiated self-promoted cascade reactions for postoperative tumor therapy. Biomaterials 2025; 318:123176. [PMID: 39954313 DOI: 10.1016/j.biomaterials.2025.123176] [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: 12/12/2024] [Revised: 02/04/2025] [Accepted: 02/07/2025] [Indexed: 02/17/2025]
Abstract
Cancer metastasis and recurrence remain a regular cause of postoperative death in patients, implying that extra consolidation treatment strategies are needed. Here, a cuproptosis nanoprodrug, termed as Lipo@CP@DQ NPs, is developed to initiate self-promoted cascade reactions to achieve the combinational effect of cuproptosis, in situ chemotherapy, and oxidative stress amplification for effectively suppressing tumor recurrence and metastasis after postoperative treatment. Lipo@CP@DQ NPs are fabricated by loading copper peroxides (Cu2O2, CP) and hydrogen peroxide (H2O2)-repsonsive prodrug DQ into liposomal nanoparticles. Lipo@CP@DQ NPs rapidly dissociate in the acidic tumor microenvironment to release copper ions, H2O2, and prodrug DQ. Subsequently, the excessive accumulation of Cu ions induces cuproptosis and produces highly cytotoxic hydroxyl radicals (•OH). Meanwhile, the self-supplied H2O2 catalyzes the decomposition of DQ to diethyldithiocarbamate (DTC), which is chelated with self-supplied Cu ions to form the anticancer compound, Cu(DTC)2. The another decomposition product, quinone methide (QM), acts as a glutathione (GSH) scavenger for oxidative stress amplification. The synergistic effect of Lipo@CP@DQ NPs-mediated cuproptosis, in situ chemotherapy, and oxidative stress amplification effectively inhibits the growth and postoperative recurrence of triple-negative breast cancer. This work furnishes a strategy for developing cuproptosis-based nanomedicines for effective antitumor treatment after surgery.
Collapse
Affiliation(s)
- Ruiqi Yan
- School of Life Sciences, Shanghai University, Shanghai, 200444, China
| | - Xuan Cheng
- School of Life Sciences, Shanghai University, Shanghai, 200444, China
| | - Yujing Song
- School of Life Sciences, Shanghai University, Shanghai, 200444, China
| | - Haiyue Wang
- School of Life Sciences, Shanghai University, Shanghai, 200444, China
| | - Run Zhang
- School of Life Sciences, Shanghai University, Shanghai, 200444, China
| | - Yiqi Jin
- School of Life Sciences, Shanghai University, Shanghai, 200444, China
| | - Xingguang Li
- Shanghai Key Laboratory of Functional Materials Chemistry, Key Laboratory for Advanced Materials, School of Chemistry and Molecular Engineering, East China University of Science & Technology, Shanghai, 200237, China.
| | - Yu Chen
- School of Life Sciences, Shanghai University, Shanghai, 200444, China.
| | - Huijing Xiang
- School of Life Sciences, Shanghai University, Shanghai, 200444, China.
| |
Collapse
|
2
|
Liu D, Liu L, Zhao X, Zhang X, Chen X, Che X, Wu G. A comprehensive review on targeting diverse immune cells for anticancer therapy: Beyond immune checkpoint inhibitors. Crit Rev Oncol Hematol 2025; 210:104702. [PMID: 40122356 DOI: 10.1016/j.critrevonc.2025.104702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 03/02/2025] [Accepted: 03/07/2025] [Indexed: 03/25/2025] Open
Abstract
Although immune checkpoint inhibitors (ICIs) have revolutionized cancer treatment, primary resistance and acquired resistance continue to limit their efficacy for many patients. To address resistance and enhance the anti-tumor activity within the tumor immune microenvironment (TIME), numerous therapeutic strategies targeting both innate and adaptive immune cells have emerged. These include combination therapies with ICIs, chimeric antigen receptor T-cell (CAR-T), chimeric antigen receptor macrophages (CAR-Ms) or chimeric antigen receptor natural killer cell (CAR-NK) therapy, colony stimulating factor 1 receptor (CSF1R) inhibitors, dendritic cell (DC) vaccines, toll-like receptor (TLR) agonists, cytokine therapies, and chemokine inhibition. These approaches underscore the significant potential of the TIME in cancer treatment. This article provides a comprehensive and up-to-date review of the mechanisms of action of various innate and adaptive immune cells within the TIME, as well as the therapeutic strategies targeting each immune cell type, aiming to deepen the understanding of their therapeutic potential.
Collapse
Affiliation(s)
- Dequan Liu
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Lei Liu
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Xinming Zhao
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Xiaoman Zhang
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Xiaochi Chen
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian 116011, China.
| | - Xiangyu Che
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian 116011, China.
| | - Guangzhen Wu
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian 116011, China.
| |
Collapse
|
3
|
Wang Q, Yin X, Liu H, Wang Q, Zhang L, Wang Y, Lu H. Mitochondrial function changes in T cell subsets during radiotherapy for patients with nasopharyngeal carcinoma. Oncol Lett 2025; 29:273. [PMID: 40235681 PMCID: PMC11998068 DOI: 10.3892/ol.2025.15019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Accepted: 03/21/2025] [Indexed: 04/17/2025] Open
Abstract
Mitochondrial dysfunction-mediated T cell exhaustion is associated with the efficacy of tumor therapy; however, the effect of radiotherapy (RT) on the mitochondrial function of peripheral blood immune cells remains still unclear. Therefore, the current study aimed to determine mitochondrial function indicators in immune cells, in particular mitochondrial mass (MM) and mitochondrial membrane potential (MMP), to assess the dynamic changes of immune status in patients with nasopharyngeal carcinoma (NPC) during RT. Peripheral venous blood was collected from patients with locally advanced NPC at day 1 pre-RT, at the 10th fraction of RT and within 2 days after RT. Based on a novel immunofluorescence technique, flow cytometry was used to assess the proportion of lymphocytes and their subsets in peripheral blood and the mitochondrial indexes, MM and low MMP (MMPlow). Univariate and multivariate logistic regression analyses were performed to evaluate the clinical factors associated with the efficacy of RT. A total of 27 patients were enrolled. After RT, lymphocyte count (P<0.05) and the proportion of CD4+ T cells (P<0.05) demonstrated a downward trend. In addition, the proportion of CD4+ memory-effector T (Tem; P<0.05) cells and CD8+ Tem cells (P=0.005) significantly increased during RT. No significant changes were demonstrated for MM in CD4+ effector T (Te) cells, whilst MMPlow was significantly reduced (P=0.047). However, the mitochondrial function of CD8+ T cells did not significantly change. Multivariate logistic regression analysis revealed that lymphocyte count [odds ratio (OR), 47.317; 95% confidence interval (CI), 1.240-1806.065] and MMPlow in CD4+ Te cells (OR, 0.889; 95% CI, 0.792-0.997) were independent factors that could affect clinical efficacy. Receiver operating characteristic curve analysis demonstrated that the area under the curve values for MMPlow in CD4+ T cells, lymphocyte count and their combination were 0.72 (P=0.13), 0.69 (P=0.19) and 0.89 (P=0.0073), respectively. These findings suggest that RT could inhibit immune cells in peripheral blood. However, this treatment approach could activate the memory cell subsets of immune cells and enhance the MMP of effector CD4+ T cells. Therefore, the evaluation of mitochondrial function in lymphocytes could be used as a predictor of RT efficacy in patients with locally advanced NPC.
Collapse
Affiliation(s)
- Quan Wang
- Department of Radiation Oncology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266035, P.R. China
| | - Xiangzhi Yin
- Department of Orthopaedics, Affiliated Hospital of Qingdao University, Qingdao, Shandong 266035, P.R. China
| | - Hongbo Liu
- Department of Radiation Oncology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266035, P.R. China
| | - Qing Wang
- Department of Clinical Laboratory, Affiliated Hospital of Qingdao University, Qingdao, Shandong 266035, P.R. China
| | - Lu Zhang
- Department of Radiation Oncology, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250014, P.R. China
| | - Yifan Wang
- Department of Emergency, Laizhou People's Hospital, Yantai, Shandong 261400, P.R. China
| | - Haijun Lu
- Department of Radiation Oncology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266035, P.R. China
| |
Collapse
|
4
|
Liu D, Ling Y, Dong L, Zhang J, Li X, Chen X, Huang H, Deng J, Guo Y. Ultrasound-triggered drug-loaded nanobubbles for enhanced T cell recruitment in cancer chemoimmunotherapy. Biomaterials 2025; 317:123086. [PMID: 39805187 DOI: 10.1016/j.biomaterials.2025.123086] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 12/09/2024] [Accepted: 01/01/2025] [Indexed: 01/16/2025]
Abstract
Chemotherapy combined with immunotherapy is a highly promising approach for treating tumors. However, chemotherapeutic drugs often fail to accumulate effectively at the tumor site after systemic administration and they lack sufficient immunogenicity to activate adaptive immunity, making an effective T-cell immune response within the tumor microenvironment difficult to achieve. Here, this work developed drug-loaded nanobubbles (DTX-R837@NBs) that encapsulate the chemotherapy drug docetaxel and the immune adjuvant R837 via a thin-film hydration method. Ultrasound-targeted nanobubble destruction promoted drug accumulation within tumor tissues and damaged tumor cells through the cavitation effect, inducing immunogenic cell death and releasing damage-associated molecular patterns to augment dendritic cell maturation. Notably, DTX-R837@NBs exhibited excellent contrast-enhanced ultrasound imaging capabilities, enabling the seamless integration of diagnosis and treatment. In combination with immune checkpoint blockade targeting programmed cell death protein 1 (PD-1), the generated immunological responses attacked residual tumor cells and ameliorated the immunosuppressive tumor microenvironment, inhibiting distant tumor growth and metastasis. Moreover, this strategy exhibited robust immune memory effects, effectively protecting the host and preventing tumor recurrence upon rechallenge. Overall, ultrasound-mediated DTX-R837@NBs combined with anti-PD-1 immune checkpoint blockade therapy exhibits robust antitumor efficiency, represent a promising strategy for overcoming immunotherapy resistance in cold tumors, and warrant further investigation for clinical translation.
Collapse
Affiliation(s)
- Deng Liu
- Department of Ultrasound, Southwest Hospital, Army Medical University, Chongqing, 400038, China; Bioengineering College, Chongqing University, Chongqing, 400044, China
| | - Yi Ling
- Department of Ultrasound, Southwest Hospital, Army Medical University, Chongqing, 400038, China
| | - Li Dong
- Department of Ultrasound, Southwest Hospital, Army Medical University, Chongqing, 400038, China
| | - Jun Zhang
- Department of Ultrasound, Southwest Hospital, Army Medical University, Chongqing, 400038, China
| | - Xin Li
- Department of Ultrasound, Southwest Hospital, Army Medical University, Chongqing, 400038, China
| | - Xuemei Chen
- Department of Ultrasound, Southwest Hospital, Army Medical University, Chongqing, 400038, China
| | - Haiyun Huang
- Department of Ultrasound, Southwest Hospital, Army Medical University, Chongqing, 400038, China.
| | - Jun Deng
- Institute of Burn Research, Southwest Hospital, State Key Lab of Trauma and Chemical Poisoning, Army Medical University, Chongqing, 400038, China.
| | - Yanli Guo
- Department of Ultrasound, Southwest Hospital, Army Medical University, Chongqing, 400038, China.
| |
Collapse
|
5
|
Teng Y, Wang D, Yang Z, Wang R, Ning S, Zhang R, Yang H, Feng X, Liu J, Yang L, Tian Y. Bioorthogonal strategy-triggered In situ co-activation of aggregation-induced emission photosensitizers and chemotherapeutic prodrugs for boosting synergistic chemo-photodynamic-immunotherapy. Biomaterials 2025; 317:123092. [PMID: 39793168 DOI: 10.1016/j.biomaterials.2025.123092] [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: 11/25/2024] [Revised: 01/04/2025] [Accepted: 01/04/2025] [Indexed: 01/13/2025]
Abstract
In situ activation of prodrugs or photosensitizers is a promising strategy for specifically killing tumor cells while avoiding toxic side effects. Herein, we originally develop a bioorthogonally activatable prodrug and pro-photosensitizer system to synchronously yield an aggregation-induced emission (AIE) photosensitizer and a chemotherapeutic drug for synergistic chemo-photodynamic-immunotherapy of tumors. By employing molecular engineering strategy, we rationally design a family of tetrazine-functionalized tetraphenylene-based photosensitizers, one of which (named TzPS5) exhibits a high turn-on ratio, a NIR emission, a typical AIE character, and an excellent ROS generation efficiency upon bioorthogonal-activation. With the aid of integrin- or mitochondria-pretargeting, TzPS5 is successfully applied for highly effective PDT ablation of cancer cells both in vitro and in vivo. On this basis, tumor-targeting TzPS5 (TzPS5-cRGD) is constructed and used jointly with a bioorthogonal prodrug, DOX-TCO, and the two are mutually activated to induce cooperative and tumor-specific PDT and chemotherapy, resulting in amplified therapeutic outcomes and improved biosafeties. Moreover, this combination modality elicits robust immunogenic cell death, stimulates systemic antitumor immunity, thereby suppressing both primary and distant tumors, and blocking the pulmonary tumor metastasis. This work is expected to provide a useful guidance for the rational design of activatable phototheranostic agents, and offer a new strategy for co-activation of prodrugs/pro-photosensitizers to boost synergistic antitumor chemo-photodynamic-immunotherapy.
Collapse
Affiliation(s)
- Yu Teng
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Beijing Key Laboratory of Active Substances Discovery and Drugability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, PR China
| | - Dianyu Wang
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Tianjin Institutes of Health Science, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, PR China
| | - Ziyu Yang
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, PR China
| | - Ruxuan Wang
- Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100007, PR China
| | - Shuyi Ning
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, PR China
| | - Rongrong Zhang
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Beijing Key Laboratory of Active Substances Discovery and Drugability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, PR China
| | - Hong Yang
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Beijing Key Laboratory of Active Substances Discovery and Drugability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, PR China
| | - Xinchi Feng
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, PR China.
| | - Jianfeng Liu
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Tianjin Institutes of Health Science, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, PR China.
| | - Lijun Yang
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Tianjin Institutes of Health Science, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, PR China.
| | - Yulin Tian
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Beijing Key Laboratory of Active Substances Discovery and Drugability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, PR China.
| |
Collapse
|
6
|
Gao Y, Liu X, Lv J, Gu C, Tao T, Zhang C, Huang D, Jia R, Yu X, Su W. Ifosfamide alleviates autoimmune toxicity and enhances antitumor efficacy in melanoma immunotherapy. Biochem Pharmacol 2025; 236:116851. [PMID: 40081767 DOI: 10.1016/j.bcp.2025.116851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 01/21/2025] [Accepted: 03/03/2025] [Indexed: 03/16/2025]
Abstract
Autoimmune toxicity affects up to 60 % of patients receiving immune checkpoint inhibitor (ICI) therapy for cancer, presenting a notable clinical obstacle that constrains its wider application. Hence, there is an imperative demand to develop novel strategies to manage immune-related adverse events (irAEs). Ifosfamide (IFO) shares structural and functional resemblances with cyclophosphamide (CPA). Despite the acknowledged dual anti-tumor and immunomodulatory effects of CPA, the specific effect of IFO on autoimmune conditions remains elusive. Here, we evaluated the efficacy of IFO on experimental autoimmune uveitis (EAU) mouse models and explored the cell-specific effects of IFO under autoimmune conditions using single-cell RNA sequencing. Our data indicated that IFO effectively alleviated inflammatory infiltration and reversed pathological alterations of EAU. Subsequent single-cell data analysis and in vivo experiments suggested IFO exerted broad suppressive effects on autoimmune responses, concurrently restoring the balance between Th17 and Treg populations. In addition, we observed that IFO enhanced CD8+ T cell activation and its cytotoxic immune responses, highlighting the cell-type-specific immunomodulatory effects of IFO. Moreover, we constructed EAU models on tumor-bearing mice under ICI treatment, and found that ICI exacerbated EAU symptoms. IFO not only possessed anti-tumor effects as monotherapy, but also augmented ICI efficacy by promoting CD8+ T cell-mediated immunity. Furthermore, we found that IFO alleviated EAU symptoms exacerbated by ICI treatment and effectively restored Th17/Treg balance. Our results elucidated the immunomodulatory effects of IFO treatment, providing evidence for the application of IFO in managing autoimmune conditions and irAEs.
Collapse
Affiliation(s)
- Yuehan Gao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Xiuxing Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Jianjie Lv
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Chenyang Gu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Tianyu Tao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Chun Zhang
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Danping Huang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Renbing Jia
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Xinping Yu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Wenru Su
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China.
| |
Collapse
|
7
|
Yang X, Xu C, Zeng Y, Wang C, Gao Y, Ding J, Chen S, Pan Y, Zhang X, Mao Z, Shi S. Pyroptosis-Inducing Platinum(IV) Prodrugs via GSDME Pathway for Chemoimmunotherapy and Metastasis Inhibition in Triple-Negative Breast Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e05567. [PMID: 40432601 DOI: 10.1002/advs.202505567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2025] [Revised: 04/29/2025] [Indexed: 05/29/2025]
Abstract
Pyroptosis has attracted significant attention for its role in cancer chemotherapy and immunotherapy. However, few drugs have been reported to induce pyroptosis via the Caspase-3/gasdermin E (GSDME) pathway. Herein, three novel PtIV prodrugs, MRP, DRP, and HRP are rationally designed by conjugating DNA methyltransferase (DNMT) inhibitor (RG108) and/or histone deacetylase (HDAC) inhibitor (PhB) to the PtIV center. These prodrugs can be easily reduced to cisplatin (CDDP) due to the high glutathione (GSH) levels in tumors, liberating the coordinated ligands. Released RG108 reactivates the GSDME gene and reduces pyroptosis in low GSDME-expressing tumor cells. Meanwhile, PhB-induced chromatin loosening enhances CDDP-DNA binding, which not only increases Caspase-3 expression, but also upregulates GSDME. HRP demonstrates superior ability to suppress tumor growth and metastasis while reducing systemic toxicity compared with CDDP. By reactivating GSDME and loosening chromatin, HRP effectively boosts tumor cell pyroptosis and exhibits the most pronounced anticancer performance. These findings highlight HRP's potential as a therapeutic agent for triple-negative breast cancer (TNBC) and offer innovative strategies for combining chemotherapy with immunotherapy. To the best of current knowledge, this is the first report of platinum complexes inducing pyroptosis via the Caspase-3/GSDME pathway in low GSDME-expressing tumor cells.
Collapse
Affiliation(s)
- Xinda Yang
- School of Chemical Science and Engineering, Department of Laboratory Medicine, Shanghai Tenth People's Hospital of Tongji University, Tongji University, Shanghai, 200092, P. R. China
| | - Chuansheng Xu
- School of Chemical Science and Engineering, Department of Laboratory Medicine, Shanghai Tenth People's Hospital of Tongji University, Tongji University, Shanghai, 200092, P. R. China
| | - Youliang Zeng
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, Guangdong Basic Research Center of Excellence for Functional Molecular Engineering, GBRCE for Functional Molecular Engineering, School of Chemistry, Sun Yat-Sen University, Guangzhou, 510275, P. R. China
| | - Chunhui Wang
- School of Chemical Science and Engineering, Department of Laboratory Medicine, Shanghai Tenth People's Hospital of Tongji University, Tongji University, Shanghai, 200092, P. R. China
| | - Yan Gao
- School of Chemical Science and Engineering, Department of Laboratory Medicine, Shanghai Tenth People's Hospital of Tongji University, Tongji University, Shanghai, 200092, P. R. China
| | - Jie Ding
- School of Chemical Science and Engineering, Department of Laboratory Medicine, Shanghai Tenth People's Hospital of Tongji University, Tongji University, Shanghai, 200092, P. R. China
| | - Sirui Chen
- School of Chemical Science and Engineering, Department of Laboratory Medicine, Shanghai Tenth People's Hospital of Tongji University, Tongji University, Shanghai, 200092, P. R. China
| | - Yuheng Pan
- School of Chemical Science and Engineering, Department of Laboratory Medicine, Shanghai Tenth People's Hospital of Tongji University, Tongji University, Shanghai, 200092, P. R. China
| | - Xin Zhang
- School of Chemical Science and Engineering, Department of Laboratory Medicine, Shanghai Tenth People's Hospital of Tongji University, Tongji University, Shanghai, 200092, P. R. China
| | - Zongwan Mao
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, Guangdong Basic Research Center of Excellence for Functional Molecular Engineering, GBRCE for Functional Molecular Engineering, School of Chemistry, Sun Yat-Sen University, Guangzhou, 510275, P. R. China
| | - Shuo Shi
- School of Chemical Science and Engineering, Department of Laboratory Medicine, Shanghai Tenth People's Hospital of Tongji University, Tongji University, Shanghai, 200092, P. R. China
| |
Collapse
|
8
|
Zhang Y, Li W, Chen S, Zhang Y, Zhu Y, Lan F, Du H, Fan R, Zhu J, Pan W, Situ B, Zheng L, Luo S, Yan X. Layered-Responsive Multivalent Tetrahedral DNA Framework-Decorated CRISPR-Cas12a Nanocapsule Enables Precise and Enhanced Tumor Chemotherapy. ACS NANO 2025; 19:19274-19286. [PMID: 40366179 DOI: 10.1021/acsnano.5c01747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2025]
Abstract
The lack of selective tumor targeting and the high toxicity of conventional chemotherapy treatments remain major challenges in cancer therapy. Here, we develop a self-controlled DNA nanostructure-CRISPR-12a system, a triple-locked cascade tumor therapy nanocapsule (Tatna), for efficient and targeted tumor treatment. Tatna integrates structural DNA tetrahedrons (DTs) with high drug-loading capacity, Cas12a/crRNA ribonucleoprotein (Cas12a RNP), and doxorubicin (DOX) to enable multisite response for precise drug delivery and augmented tumor treatment. By incorporation of a nucleolin-targeting aptamer, Tatna achieves selective targeting and efficient tumor cell internalization. Encapsulation in pH-responsive poly l-lactic-co-glycolic acid (PLGA) nanocapsule ensures stable circulation and controlled release of both DOX and Cas12a until tumor-specific activation in the acidic microenvironment. The Cas12a RNP, triggered by APE1 mRNA overexpression in tumor cells, induces trans-cleavage of DTs, releasing DOX and Cas12a to transport into the nucleus and induce enhanced cell apoptosis. This self-regulating and multifunctional approach enhances the efficacy of chemotherapy while reducing off-target effects. Tatna's programmable, tumor-specific delivery system represents a powerful strategy for advancing precision medicine and personalized cancer treatment.
Collapse
Affiliation(s)
- Ye Zhang
- Department of Laboratory Medicine, Medical Research Center of Nanfang Hospital, Guangdong Provincial Key Laboratory of Precision Medical Diagnostics, Guangdong Engineering and Technology Research Center for Rapid Diagnostic Biosensors, Guangdong Provincial Key Laboratory of Single-cell and Extracellular Vesicles, Nanfang Hospital, Southern Medical University, Guangzhou 510515, P. R. China
| | - Wenbin Li
- Department of Laboratory Medicine, Medical Research Center of Nanfang Hospital, Guangdong Provincial Key Laboratory of Precision Medical Diagnostics, Guangdong Engineering and Technology Research Center for Rapid Diagnostic Biosensors, Guangdong Provincial Key Laboratory of Single-cell and Extracellular Vesicles, Nanfang Hospital, Southern Medical University, Guangzhou 510515, P. R. China
| | - Siting Chen
- School of Basic Medical Sciences, Medical Research Center of Nanfang Hospital, Department of Laboratory Medicine, Southern Medical University, Guangzhou 510515, P. R. China
- The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, Guangdong 511518, P. R. China
| | - Yu Zhang
- School of Basic Medical Sciences, Medical Research Center of Nanfang Hospital, Department of Laboratory Medicine, Southern Medical University, Guangzhou 510515, P. R. China
| | - Yitong Zhu
- School of Basic Medical Sciences, Medical Research Center of Nanfang Hospital, Department of Laboratory Medicine, Southern Medical University, Guangzhou 510515, P. R. China
| | - Fei Lan
- Department of Laboratory Medicine, Medical Research Center of Nanfang Hospital, Guangdong Provincial Key Laboratory of Precision Medical Diagnostics, Guangdong Engineering and Technology Research Center for Rapid Diagnostic Biosensors, Guangdong Provincial Key Laboratory of Single-cell and Extracellular Vesicles, Nanfang Hospital, Southern Medical University, Guangzhou 510515, P. R. China
| | - Huijun Du
- The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, Guangdong 511518, P. R. China
| | - Rui Fan
- Department of Laboratory Medicine, Medical Research Center of Nanfang Hospital, Guangdong Provincial Key Laboratory of Precision Medical Diagnostics, Guangdong Engineering and Technology Research Center for Rapid Diagnostic Biosensors, Guangdong Provincial Key Laboratory of Single-cell and Extracellular Vesicles, Nanfang Hospital, Southern Medical University, Guangzhou 510515, P. R. China
| | - Junfang Zhu
- Center for Clinical Laboratory Diagnosis and Research, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise 533000, Guangxi, P. R. China
- Key Laboratory of Research on Clinical Molecular Diagnosis for High Incidence Diseases in Western Guangxi of Guangxi Higher Education Institutions, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise 533000, Guangxi, P. R. China
| | - Weilun Pan
- Department of Laboratory Medicine, Medical Research Center of Nanfang Hospital, Guangdong Provincial Key Laboratory of Precision Medical Diagnostics, Guangdong Engineering and Technology Research Center for Rapid Diagnostic Biosensors, Guangdong Provincial Key Laboratory of Single-cell and Extracellular Vesicles, Nanfang Hospital, Southern Medical University, Guangzhou 510515, P. R. China
| | - Bo Situ
- Department of Laboratory Medicine, Medical Research Center of Nanfang Hospital, Guangdong Provincial Key Laboratory of Precision Medical Diagnostics, Guangdong Engineering and Technology Research Center for Rapid Diagnostic Biosensors, Guangdong Provincial Key Laboratory of Single-cell and Extracellular Vesicles, Nanfang Hospital, Southern Medical University, Guangzhou 510515, P. R. China
| | - Lei Zheng
- Department of Laboratory Medicine, Medical Research Center of Nanfang Hospital, Guangdong Provincial Key Laboratory of Precision Medical Diagnostics, Guangdong Engineering and Technology Research Center for Rapid Diagnostic Biosensors, Guangdong Provincial Key Laboratory of Single-cell and Extracellular Vesicles, Nanfang Hospital, Southern Medical University, Guangzhou 510515, P. R. China
| | - Shihua Luo
- Center for Clinical Laboratory Diagnosis and Research, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise 533000, Guangxi, P. R. China
- Key Laboratory of Research on Clinical Molecular Diagnosis for High Incidence Diseases in Western Guangxi of Guangxi Higher Education Institutions, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise 533000, Guangxi, P. R. China
| | - Xiaohui Yan
- Department of Laboratory Medicine, Medical Research Center of Nanfang Hospital, Guangdong Provincial Key Laboratory of Precision Medical Diagnostics, Guangdong Engineering and Technology Research Center for Rapid Diagnostic Biosensors, Guangdong Provincial Key Laboratory of Single-cell and Extracellular Vesicles, Nanfang Hospital, Southern Medical University, Guangzhou 510515, P. R. China
- School of Basic Medical Sciences, Medical Research Center of Nanfang Hospital, Department of Laboratory Medicine, Southern Medical University, Guangzhou 510515, P. R. China
| |
Collapse
|
9
|
Zhang M, Bai L, Chen J, Meng Q, Lu Y, Zhang D. Clinical benefit of continuation of PD-1 inhibitors after progression on first-line chemoimmunotherapy in metastatic gastric cancer and biomarker exploration. BMC Cancer 2025; 25:935. [PMID: 40413463 DOI: 10.1186/s12885-025-14286-7] [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: 02/08/2024] [Accepted: 05/07/2025] [Indexed: 05/27/2025] Open
Abstract
BACKGROUND Whether continuing immunotherapy after progression in second-line settings of metastatic gastric cancer (MGC) remains unclear. Herein, we explored the efficacy of PD-1 inhibitors for MGC after progression on previous chemoimmunotherapy. METHODS We retrospectively identified MGC patients who received oxaliplatin-based chemotherapy plus PD-1 inhibitor, with or without trastuzumab, as first-line treatment. The patients received treatment with or without PD-1 inhibitors beyond progression in patients with MGC were divided into treatment beyond progression (TBP) group and non-TBP (NTBP) group. The median progression free survival (PFS) and overall survival (OS) from the start of treatment after progression were assessed. RESULTS The mOS and mPFS in the TBP group was significantly longer than that in the NTBP group (mOS: 9.0 vs. 5.0 months, P = 0.011; mPFS: 4.3 vs. 2.7 months, P = 0.03). Moreover, TBP was an independent prognostic factor for both PFS and OS in multivariate analysis. In the subgroup analysis, patients who were male, had a favorable ECOG (0-1), classified into diffuse histologic subtype and achieved disease control in the prior chemoimmunotherapy, might be more likely to benefit from continuing immunotherapy compared to discontinuation beyond progression. CONCLUSION PD-1 inhibitors based therapeutic strategy may be a reasonable option in second-line setting for MGC who progressed on prior immunotherapy. Further larger prospective trials are warranted to validate these findings.
Collapse
Affiliation(s)
- Mengwei Zhang
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University, Guangzhou, 510060, P. R. China
- Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou, 510060, P. R. China
| | - Long Bai
- Department of VIP Region, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University, Guangzhou, 510060, P. R. China
| | - Jianwen Chen
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University, Guangzhou, 510060, P. R. China
- Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou, 510060, P. R. China
| | - Qi Meng
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University, Guangzhou, 510060, P. R. China
- Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou, 510060, P. R. China
| | - Yunxin Lu
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University, Guangzhou, 510060, P. R. China.
- Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou, 510060, P. R. China.
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Kaiyang Fifth Road, Guangzhou, Guangzhou, 510555, China.
| | - Dongsheng Zhang
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University, Guangzhou, 510060, P. R. China.
- Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou, 510060, P. R. China.
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Kaiyang Fifth Road, Guangzhou, Guangzhou, 510555, China.
| |
Collapse
|
10
|
Ran R, Chen X, Yang J, Xu B. Immunotherapy in breast cancer: current landscape and emerging trends. Exp Hematol Oncol 2025; 14:77. [PMID: 40405250 PMCID: PMC12096519 DOI: 10.1186/s40164-025-00667-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Accepted: 05/08/2025] [Indexed: 05/24/2025] Open
Abstract
Breast cancer remains one of the most prevalent malignancies worldwide, underscoring an urgent need for innovative therapeutic strategies. Immunotherapy has emerged as a transformative frontier in this context. In triple-negative breast cancer (TNBC), the combination of immunotherapy based on PD-1/PD-L1 immune checkpoint inhibitors (ICIs) with chemotherapy has proven efficacious in both early and advanced clinical trials. These encouraging results have led to the approval of ICIs for TNBC, opening up new therapeutic avenues for challenging-to-treat patient populations. Furthermore, a multitude of ongoing trials are actively investigating the efficacy of immunotherapy-based combinations, including ICIs in conjunction with chemotherapy, targeted therapy and radiation therapy, as well as other novel strategies such as bispecific antibodies, CAR-T cells and cancer vaccines across all breast cancer subtypes, including HR-positive/HER2-negative and HER2-positive disease. This review provides a comprehensive overview of current immunotherapeutic approaches in breast cancer, highlighting pivotal findings from recent clinical trials and the potential impact of these advancements on patient outcomes.
Collapse
Affiliation(s)
- Ran Ran
- Cancer Center, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Precision Medicine Center, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xi Chen
- Cancer Center, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Precision Medicine Center, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jin Yang
- Cancer Center, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
- Precision Medicine Center, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
| | - Binghe Xu
- Cancer Center, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
- National Cancer Center, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
11
|
Cui Y, Qiao Y, An R, Pan X, Tu J. The application of compressed sensing on tumor mutation burden calculation from overlapped pooling sequencing data. BMC Bioinformatics 2025; 26:129. [PMID: 40394464 PMCID: PMC12090583 DOI: 10.1186/s12859-025-06148-7] [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: 11/10/2023] [Accepted: 04/24/2025] [Indexed: 05/22/2025] Open
Abstract
BACKGROUND Tumor Mutation Burden (TMB) is commonly characterized as the number of non-synonymous somatic SNVs per megabase within the gene region identified through whole exon sequencing or targeted sequencing in a tumor sample. It has been statistically demonstrated that TMB was related to the ability of neoantigen production and used to predict the efficacy of immunotherapy for various types of cancers. However, screening for TMB in patients poses challenges due to the extensive labor and financial resources required for the preparation of large quantities of parallel sequencing libraries. RESULTS In this study, we employed compressed sensing (CS) to calculate TMB from overlapped pooling sequencing data, aiming to reduce the sequencing cost by minimizing the number of library builds. Over 90% SNPs could still be detected without a significant loss of mutation information even when the data is pooled from ten different samples. Based on this, the orthogonal matching pursuit (OMP) algorithm and the basic pursuit (BP) algorithm were used to reconstruct TMB from pooling sequencing data. The performance of these two algorithms was evaluated. The BP algorithm consistently performed well across all cases, albeit necessitating extended computational time. The OMP algorithm has been proved to be suitable for scenarios where the original matrix was sparse but it showed low overall performance. Based on an accurate calculation of TMB, we determined that the number of sequencing runs could be reduced to 0.6 times the total number of samples, resulting in a 40% reduction in sequencing cost. CONCLUSIONS In conclusion, we calculated TMB from overlapped pooling sequencing data utilizing compressed sensing strategy to reduce sequencing cost. Our findings confirm that the SNP calling from ten samples' pooling sequencing data is feasible. Additionally, we performed an assessment of the reconstruction efficiency of both the BP model and the OMP model.
Collapse
Affiliation(s)
- Yue Cui
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Yi Qiao
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Rongming An
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
- Monash University-Southeast University Joint Research Institute, Suzhou, 215123, China
| | - Xuan Pan
- Department of Medical Oncology, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, 210009, China.
| | - Jing Tu
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China.
| |
Collapse
|
12
|
Rahal Z, El Darzi R, Moghaddam SJ, Cascone T, Kadara H. Tumour and microenvironment crosstalk in NSCLC progression and response to therapy. Nat Rev Clin Oncol 2025:10.1038/s41571-025-01021-1. [PMID: 40379986 DOI: 10.1038/s41571-025-01021-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/16/2025] [Indexed: 05/19/2025]
Abstract
The treatment landscape of non-small-cell lung cancer (NSCLC) is evolving rapidly, driven by advances in the development of targeted agents and immunotherapies. Despite this progress, some patients have suboptimal responses to treatment, highlighting the need for new therapeutic strategies. In the past decade, the important role of the tumour microenvironment (TME) in NSCLC progression, metastatic dissemination and response to treatment has become increasingly evident. Understanding the complexity of the TME and its interactions with NSCLC can propel efforts to improve current treatment modalities, overcome resistance and develop new treatments, which will ultimately improve the outcomes of patients. In this Review, we provide a comprehensive view of the NSCLC TME, examining its components and highlighting distinct archetypes characterized by spatial niches within and surrounding tumour nests, which form complex neighbourhoods. Next, we explore the interactions within these components, focusing on how inflammation and immunosuppression shape the dynamics of the NSCLC TME. We also address the emerging influences of patient-related factors, such as ageing, sex and health disparities, on the NSCLC-TME crosstalk. Finally, we discuss how various therapeutic strategies interact with and are influenced by the TME in NSCLC. Overall, we emphasize the interconnectedness of these elements and how they influence therapeutic outcomes and tumour progression.
Collapse
Affiliation(s)
- Zahraa Rahal
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Roy El Darzi
- Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Seyed Javad Moghaddam
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Graduate School of Biomedical Sciences (GSBS), UTHealth Houston, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Tina Cascone
- Graduate School of Biomedical Sciences (GSBS), UTHealth Houston, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Thoracic-Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Humam Kadara
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Graduate School of Biomedical Sciences (GSBS), UTHealth Houston, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
13
|
Shi X, Shen T, Gu M, Guan Y, Aimaiti G, Yu W, Zhang X, Yuan WE, Su J. Development of a novel Cu-Mn hydroxide layered nanosheet-loaded drug modulating the tumour microenvironment and enhancing antitumor effects. J Colloid Interface Sci 2025; 696:137904. [PMID: 40393131 DOI: 10.1016/j.jcis.2025.137904] [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: 02/13/2025] [Revised: 05/13/2025] [Accepted: 05/15/2025] [Indexed: 05/22/2025]
Abstract
The tumor microenvironment (TME) impedes the effectiveness of therapeutic strategies such as chemodynamic therapy (CDT). This study presents a novel nanoscale drug delivery system designed for the precise release of the chemotherapeutic agent doxorubicin (DOX), aiming to overcome treatment limitations, reduce systemic toxicity, and enhance antitumor efficacy. Mn(III) serves as an immunomodulatory agent, while Cu(II) regulates the levels of glutathione (GSH). Layered double hydroxides (LDHs) were synthesized and efficiently loaded with DOX, followed by surface modification with hyaluronic acid (HA). The HA-coated LDH/DOX nanocarriers showed effective internalization by tumor cells and provided a pH-responsive release of DOX. In vitro, the LDH/HA/DOX complex exhibited strong catalytic activity in the Fenton reaction. In vivo studies using an H22 hepatocarcinoma model confirmed its potent antitumor activity and excellent biocompatibility. Immunohistochemical analyses revealed that treatment with LDH/HA/DOX significantly increased infiltration of M1-polarized tumor-associated macrophages (TAMs), CD4 + T cells and CD8 + T cells, while decreasing M2-polarized TAMs. This change in immune cell profile was associated with notable tumor growth inhibition.
Collapse
Affiliation(s)
- Xiaoying Shi
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China; Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, and School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China; National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, Shanghai 200240, China; Inner Mongolia Research Institute of Shanghai Jiao Tong University, China
| | - Tianyi Shen
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China; Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, and School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China; National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, Shanghai 200240, China; Inner Mongolia Research Institute of Shanghai Jiao Tong University, China
| | - Muge Gu
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China; Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, and School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China; National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, Shanghai 200240, China; Inner Mongolia Research Institute of Shanghai Jiao Tong University, China
| | - Yuanye Guan
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China; Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, and School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China; National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, Shanghai 200240, China; Inner Mongolia Research Institute of Shanghai Jiao Tong University, China
| | - Gulizeba Aimaiti
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China; Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, and School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China; National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, Shanghai 200240, China; Inner Mongolia Research Institute of Shanghai Jiao Tong University, China
| | - Wei Yu
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China; Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, and School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China; National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, Shanghai 200240, China; Inner Mongolia Research Institute of Shanghai Jiao Tong University, China
| | - Xiangqi Zhang
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China; Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, and School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China; National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, Shanghai 200240, China; Inner Mongolia Research Institute of Shanghai Jiao Tong University, China
| | - Wei-En Yuan
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China; Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, and School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China; National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, Shanghai 200240, China; Inner Mongolia Research Institute of Shanghai Jiao Tong University, China.
| | - Jing Su
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China; Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, and School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China; National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, Shanghai 200240, China; Inner Mongolia Research Institute of Shanghai Jiao Tong University, China.
| |
Collapse
|
14
|
He S, Yu J, Cheng P, Liu J, Zhang C, Xu C, Pu K, Zhang Y. Differential Optical Imaging of Antigen Presentation Machinery Using Molecular Optical Reporters. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025:e2420393. [PMID: 40370186 DOI: 10.1002/adma.202420393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Revised: 04/15/2025] [Indexed: 05/16/2025]
Abstract
Detection of antigen presentation is central to understanding immunological processes and developing therapeutics for cancer, infectious diseases, and allergies. However, methods with the ability to dynamically and noninvasively distinguish between major histocompatibility complex class I (MHC-I) and MHC-II antigen presentations remain lacking. Herein, we develop activatable molecular optical reporters (MORs) for real-time differential imaging of antigen presentations in lymph nodes (LNs). These MORs are engineered to passively target LNs and activated through proteolytic cleavage by key enzymes in the MHC-I and MHC-II pathways, the immunoproteasome (iP) and cathepsin S (CTSS), respectively, triggering their chemiluminescent or fluorescent signals. Coupled with minimized signal crosstalk and high sensitivity, MORs delineate the subtle differences in the antigen presentation machinery across various disease models, including cancer and bacterial or viral infection, a feat unattainable for existing imaging methods. After systemic administration, MORs also allow real-time visualization of antigen presentation in the tumor microenvironment. Besides, MORs are validated to have potential for preclinical application in immunotherapeutics screening and clinical application in tissue biopsy. Thus, our study not only presents the first example of real-time, in vivo differential imaging of antigen presentation pathways but also opens new avenues for optical probes in immune contexture analysis.
Collapse
Affiliation(s)
- Shasha He
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 70 Nanyang Drive, Singapore, 637457, Singapore
- College of Chemistry and Chemical Engineering, Innovation Laboratory for Sciences and Technologies of Energy Materials of Fujian Province (IKKEM), Xiamen University, Xiamen, 361005, P. R. China
| | - Jie Yu
- National Engineering Research Centre for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, P. R. China
| | - Penghui Cheng
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 70 Nanyang Drive, Singapore, 637457, Singapore
| | - Jing Liu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 70 Nanyang Drive, Singapore, 637457, Singapore
| | - Chi Zhang
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 70 Nanyang Drive, Singapore, 637457, Singapore
| | - Cheng Xu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 70 Nanyang Drive, Singapore, 637457, Singapore
| | - Kanyi Pu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 70 Nanyang Drive, Singapore, 637457, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, 59 Nanyang Drive, Singapore, 636921, Singapore
| | - Yan Zhang
- National Engineering Research Centre for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, P. R. China
| |
Collapse
|
15
|
Wang L, Gao X, Zuo X, Wang T, Shi X. Prognostic value of circadian rhythm-associated genes in breast cancer. World J Surg Oncol 2025; 23:186. [PMID: 40369575 PMCID: PMC12077051 DOI: 10.1186/s12957-025-03829-8] [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: 01/07/2025] [Accepted: 04/28/2025] [Indexed: 05/16/2025] Open
Abstract
OBJECTIVE Breast cancer (BC) remains the most prevalent malignancy among women. Clinical evidence indicates that genetic variations related to circadian rhythms, as well as the timing of therapeutic interventions, influence the response to radiation therapy and the toxicity of pharmacological treatments in women with BC. This study aimed to identify key circadian rhythm-related genes (CRGs) using bioinformatics and machine learning, and construct a prognostic model to predict clinical outcomes. METHODS Transcriptome data for BC were retrieved from The Cancer Genome Atlas database. Univariate Cox regression and least absolute shrinkage and selection operator regression analyses were used to develop a prognostic model based on CRGs. The predictive performance of the risk score model was evaluated. Univariate and multivariate Cox regression analyses were applied to construct the prognostic model and stratify patients into high-risk and low-risk groups. Additionally, differences in immune microenvironment, immunotherapy efficacy, and tumor mutation burden were assessed between risk groups. RESULTS A prognostic risk score model comprising 17 CRGs was developed. The areas under the receiver operating characteristic curve for overall survival at 1, 3, 5, and 7 years exceeded 0.6, indicating acceptable predictive performance. Calibration plots and decision curve analyses demonstrated the use of the model in prognostic prediction. Significant differences in immune microenvironment, immunotherapy efficacy, and tumor mutation burden were identified between the low-risk and high-risk groups. CONCLUSION The circadian rhythm-based gene model, effectively predicted the prognosis of individuals with BC, highlighting its potential to inform personalized therapeutic strategies and improve patient outcomes.
Collapse
Affiliation(s)
- Ling Wang
- Department of Breast Surgery, Dongzhimen Hospital, Beijing University of Chinese Medicine, Haiyuncang 5th, Dongcheng District, Beijing, 100700, China
| | - Xiang Gao
- Department of Breast Surgery, Dongzhimen Hospital, Beijing University of Chinese Medicine, Haiyuncang 5th, Dongcheng District, Beijing, 100700, China
| | - Ximeng Zuo
- Department of Breast Surgery, Dongzhimen Hospital, Beijing University of Chinese Medicine, Haiyuncang 5th, Dongcheng District, Beijing, 100700, China
| | - Tangshun Wang
- Department of Breast Surgery, Dongzhimen Hospital, Beijing University of Chinese Medicine, Haiyuncang 5th, Dongcheng District, Beijing, 100700, China
| | - Xiaoguang Shi
- Department of Breast Surgery, Dongzhimen Hospital, Beijing University of Chinese Medicine, Haiyuncang 5th, Dongcheng District, Beijing, 100700, China.
| |
Collapse
|
16
|
Xu T, Zhang H, Yang BB, Qadir J, Yuan H, Ye T. Tumor-infiltrating immune cells state-implications for various breast cancer subtypes. Front Immunol 2025; 16:1550003. [PMID: 40438111 PMCID: PMC12116345 DOI: 10.3389/fimmu.2025.1550003] [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: 12/22/2024] [Accepted: 04/24/2025] [Indexed: 06/01/2025] Open
Abstract
Breast cancer presents a variety of subtypes due to its cellular and molecular heterogeneity. The capacity of cancer cells to proliferate, invade, and metastasize depends not only on their intrinsic characters but also on their dynamic interaction with the host tumor microenvironment (TME), which includes immune cells. Meanwhile, the infiltration of immune cells in the TME severely affects the occurrence, development, treatment, and prognosis of breast cancer. Therefore, this review aims to explore the immune invasive tumor microenvironment in different intrinsic subtypes of breast cancer. Additionally, it highlights the mechanistic influence of the infiltrating immune cells on stage-wise dynamics of breast tumorigenesis. Moreover, the present review also attempts to discern the regulatory relationship between tumor infiltrating immune cells and immune microenvironment in different molecular subtypes of breast cancer, thus, spotlighting its clinical significance.
Collapse
Affiliation(s)
- Tianshuang Xu
- Department of Immunology, Mudanjiang Medical University, Mudanjiang, Heilongjiang, China
| | - Hongjun Zhang
- Department of Immunology, Mudanjiang Medical University, Mudanjiang, Heilongjiang, China
| | - Burton B. Yang
- Sunnybrook Research Institute and the Department of Laboratory Medicine and Pathobiology at the University of Toronto, Toronto, ON, Canada
| | - Javeria Qadir
- Department of Biosciences, COMSATS University Islamabad, Islamabad, Pakistan
| | - Hui Yuan
- School of Stomatology and School of Basic Medical Sciences, Mudanjiang Medical University, Mudanjiang, Heilongjiang, China
| | - Ting Ye
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
17
|
Zhao J, Liu M, Zhu C, Li Z, Liu Z, Abulizi D, Liu S, Wang X, Yang H, Hou X. Cancer-associated fibroblasts and metabolic reprogramming predict pathologic response to neoadjuvant PD-1 blockade in resected non-small cell lung cancer. Cell Oncol (Dordr) 2025:10.1007/s13402-025-01067-4. [PMID: 40358847 DOI: 10.1007/s13402-025-01067-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Accepted: 04/27/2025] [Indexed: 05/15/2025] Open
Abstract
PURPOSE Immunotherapy has transformed the neoadjuvant treatment landscape for patients with resectable locally advanced non-small cell lung cancer (NSCLC). However, a population of patients cannot obtain major pathologic response (MPR) and thus benefit less from neoadjuvant immunotherapy, highlighting the need to uncover the underlying mechanisms driving resistance to immunotherapy. METHODS Two published single-cell RNA sequencing (scRNA-seq) datasets were used to analyze the subsets of cancer-associated fibroblasts (CAFs) and T cells and functional alterations after neoadjuvant immunotherapy. The stromal signature predicting ICI response was identified and validated using our local cohort with stage III NSCLC receiving neoadjuvant immunotherapy and other 4 public ICI transcriptomic cohorts. RESULTS Non-MPR tumors showed higher enrichment of CAFs and increased extracellular matrix deposition than MPR tumors, as suggested by bioinformatic analysis. Further, CAF-mediated immune suppression may involve reciprocal interactions with T cells in addition to a physical barrier mechanism. In contrast, MPR tumors demonstrated therapy-induced activation of memory CD8+ T cells into an effector phenotype. Additionally, neoadjuvant immunotherapy resulted in expansion of precursor exhausted T (Texp) cells, which were remodeled into an anti-tumor phenotype. Notably, we identified metabolic heterogeneity within distinct T cell clusters during immunotherapy. Methionine recycling emerged as a predictive factor for T-cell differentiation and a favorable pathological response. The stromal signature was associated with ICI response, and this association was validated in five independent ICI transcriptomic cohorts. CONCLUSION These discoveries underscore the distinct tumor microenvironments in MPR and non-MPR patients and may elucidate resistance mechanisms to immunotherapy in NSCLC.
Collapse
Affiliation(s)
- Jiaqi Zhao
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, No. 651, Dongfeng East Road, Guangzhou City, Guangdong Province, 510060, PR China
| | - Maolin Liu
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, No. 651, Dongfeng East Road, Guangzhou City, Guangdong Province, 510060, PR China
| | - Chongmei Zhu
- Department of Pathology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| | - Zhuolin Li
- Guangzhou BioScript Biotechnology Co., Ltd, Guangzhou, PR China
| | - Zuhui Liu
- The Department of Breast Disease, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, PR China
| | - Dilimulati Abulizi
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, No. 651, Dongfeng East Road, Guangzhou City, Guangdong Province, 510060, PR China
| | - Siqing Liu
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, No. 651, Dongfeng East Road, Guangzhou City, Guangdong Province, 510060, PR China
| | - Xin Wang
- Department of Thoracic Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, No. 651, Dongfeng East Road, Guangzhou City, Guangdong Province, 510060, PR China
| | - Haoxian Yang
- Department of Thoracic Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, No. 651, Dongfeng East Road, Guangzhou City, Guangdong Province, 510060, PR China.
| | - Xue Hou
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, No. 651, Dongfeng East Road, Guangzhou City, Guangdong Province, 510060, PR China.
| |
Collapse
|
18
|
Giram P, Md Mahabubur Rahman K, Aqel O, You Y. In Situ Cancer Vaccines: Redefining Immune Activation in the Tumor Microenvironment. ACS Biomater Sci Eng 2025; 11:2550-2583. [PMID: 40223683 DOI: 10.1021/acsbiomaterials.5c00121] [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: 04/15/2025]
Abstract
Cancer is one of the leading causes of mortality worldwide. Nanomedicines have significantly improved life expectancy and survival rates for cancer patients in current standard care. However, recurrence of cancer due to metastasis remains a significant challenge. Vaccines can provide long-term protection and are ideal for preventing bacterial and viral infections. Cancer vaccines, however, have shown limited therapeutic efficacy and raised safety concerns despite extensive research. Cancer vaccines target and stimulate responses against tumor-specific antigens and have demonstrated great potential for cancer treatment in preclinical studies. However, tumor-associated immunosuppression and immune tolerance driven by immunoediting pose significant challenges for vaccine design. In situ vaccination represents an alternative approach to traditional cancer vaccines. This strategy involves the intratumoral administration of immunostimulants to modulate the growth and differentiation of innate immune cells, such as dendritic cells, macrophages, and neutrophils, and restore T-cell activity. Currently approved in situ vaccines, such as T-VEC, have demonstrated clinical promise, while ongoing clinical trials continue to explore novel strategies for broader efficacy. Despite these advancements, failures in vaccine research highlight the need to address tumor-associated immune suppression and immune escape mechanisms. In situ vaccination strategies combine innate and adaptive immune stimulation, leveraging tumor-associated antigens to activate dendritic cells and cross-prime CD8+ T cells. Various vaccine modalities, such as nucleotide-based vaccines (e.g., RNA and DNA vaccines), peptide-based vaccines, and cell-based vaccines (including dendritic, T-cell, and B-cell approaches), show significant potential. Plant-based viral approaches, including cowpea mosaic virus and Newcastle disease virus, further expand the toolkit for in situ vaccination. Therapeutic modalities such as chemotherapy, radiation, photodynamic therapy, photothermal therapy, and Checkpoint blockade inhibitors contribute to enhanced antigen presentation and immune activation. Adjuvants like CpG-ODN and PRR agonists further enhance immune modulation and vaccine efficacy. The advantages of in situ vaccination include patient specificity, personalization, minimized antigen immune escape, and reduced logistical costs. However, significant barriers such as tumor heterogeneity, immune evasion, and logistical challenges remain. This review explores strategies for developing potent cancer vaccines, examines ongoing clinical trials, evaluates immune stimulation methods, and discusses prospects for advancing in situ cancer vaccination.
Collapse
Affiliation(s)
- Prabhanjan Giram
- Department of Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, New York 14214, United States
| | - Kazi Md Mahabubur Rahman
- Department of Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, New York 14214, United States
| | - Osama Aqel
- Department of Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, New York 14214, United States
| | - Youngjae You
- Department of Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, New York 14214, United States
| |
Collapse
|
19
|
Zhao L, Zeng J, Wen J, Li Z, Xu J, Wang J, Tang X, Hou L. Global trends and research hotspots in autophagy and tumor drug resistance: a bibliometric analysis. Discov Oncol 2025; 16:734. [PMID: 40354002 PMCID: PMC12069191 DOI: 10.1007/s12672-025-02379-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Accepted: 04/11/2025] [Indexed: 05/14/2025] Open
Abstract
Autophagy plays a crucial role in tumor drug resistance by enabling cancer cells to survive under stress conditions, including chemotherapy. It helps tumor cells maintain homeostasis, resist cell death, and contribute to therapy failure. This study analyzed the literature related to autophagy and tumor drug resistance based on the Web of Science Core Collection (WoSCC) database. The results revealed that there are 9284 relevant articles published to date, covering 103 countries and regions, with contributions from 5964 institutions and 37,240 researchers. The annual number of publications has steadily increased since 2004, especially after 2019, indicating the growing importance of autophagy in tumor drug resistance research. China leads globally in terms of publication output, accounting for nearly 50% of the total publications. Additionally, international collaboration and cross-country research have become increasingly prominent, particularly collaborations between China and countries like South Korea and Japan. Journal analysis showed that the International Journal of Molecular Sciences and Oncotarget are the most productive journals, while Autophagy stands out with a higher impact factor. Author, citation, and keyword analyses revealed research hotspots and future trends in the field of autophagy and tumor drug resistance, including chemotherapy resistance, cell death mechanisms, and immunotherapy. This study provides a systematic academic perspective for future research in the field of autophagy and tumor drug resistance and emphasizes the importance of strengthening international cooperation.
Collapse
Affiliation(s)
- Long Zhao
- Department of Neurosurgery, Affiliated Hospital of North Sichuan Medical College, No. 1 South Maoyuan Road, Shunqing District, Nanchong, 637000, Sichuan, People's Republic of China
| | - Jiao Zeng
- Department of Clinical Medicine, North Sichuan Medical College, Nanchong, 637000, People's Republic of China
| | - Junfeng Wen
- Department of Clinical Medicine, North Sichuan Medical College, Nanchong, 637000, People's Republic of China
| | - Zhaoyang Li
- Department of Clinical Medicine, North Sichuan Medical College, Nanchong, 637000, People's Republic of China
| | - Jianguo Xu
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, 610041, People's Republic of China
| | - Jinxiang Wang
- Department of Urology, Kidney and Urology Center, Pelvic Floor Disorders Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, Guangdong, People's Republic of China.
| | - Xiaoping Tang
- Department of Neurosurgery, Affiliated Hospital of North Sichuan Medical College, No. 1 South Maoyuan Road, Shunqing District, Nanchong, 637000, Sichuan, People's Republic of China.
| | - Lingmi Hou
- Department of Breast Surgery, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, No. 55, Section 4, Renmin South Road, Wuhou District, Chengdu, 610041, Sichuan, People's Republic of China.
| |
Collapse
|
20
|
Miyatsu M, Sukhbaatar A, Mishra R, Dorai A, Mori S, Kodama T. Optimization of lymphatic drug delivery system with carboplatin for metastatic lymph nodes. Sci Rep 2025; 15:16037. [PMID: 40341825 PMCID: PMC12062337 DOI: 10.1038/s41598-025-99602-8] [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: 02/20/2025] [Accepted: 04/21/2025] [Indexed: 05/11/2025] Open
Abstract
Systemic chemotherapy is a common method for treatment of metastatic lymph nodes (LNs), but it has low tissue selectivity and high toxicity. Lymphatic drug delivery system (LDDS) is a novel approach to treat and prevent LN metastases. In a previous study, it was found that the increase of osmotic pressure with varied viscosity of the drug reagent enhances drug retention in the LNs. Here, we optimized the administration conditions to achieve a long-term therapeutic response by varying the dosages and injection rate, using the optimized osmotic pressure and varied viscosity of drug reagent for LDDS. A metastatic LN mouse model was created with MXH10/Mo/lpr mice. Luciferase labelled FM3A mouse mammary carcinoma cells were inoculated in subiliac LN (SiLN) to induce metastasis to the proper axillary LN (PALN). 4 days post tumor cell inoculation, carboplatin (CBDCA) was injected into the tumor-bearing SiLN under different administration conditions. Superior drug retention was observed in the group that received two-doses of CBDCA solution adjusted to an osmotic pressure and viscosity of 1897 kPa and 12 mPa·s, at an injection rate of 10 µL/min. Furthermore, this effect persisted for 42 days. This effect was accompanied by an upregulated expression of CD8, IL-12a, and IFN-γ in the spleen. These results suggest that dual-dose administration at 10 µL/min with hyper-osmotic and high viscosity formulation is optimal and can improve the long-term therapeutic efficacy of LN metastasis.
Collapse
Affiliation(s)
- Miriu Miyatsu
- Laboratory of Biomedical Engineering for Cancer, Graduate School of Biomedical Engineering, Tohoku University, 4‑1 Seiryo, Aoba, Sendai, Miyagi, 980‑8575, Japan
| | - Ariunbuyan Sukhbaatar
- Laboratory of Biomedical Engineering for Cancer, Graduate School of Biomedical Engineering, Tohoku University, 4‑1 Seiryo, Aoba, Sendai, Miyagi, 980‑8575, Japan
- Division of Oral and Maxillofacial Oncology and Surgical Sciences, Graduate School of Dentistry, Tohoku University, 4‑1 Seiryo, Aoba, Sendai, Miyagi, 980‑8575, Japan
- Biomedical Engineering Cancer Research Center, Graduate School of Biomedical Engineering, Tohoku University, 4‑1 Seiryo, Aoba, Sendai, Miyagi, 980‑8575, Japan
| | - Radhika Mishra
- Laboratory of Biomedical Engineering for Cancer, Graduate School of Biomedical Engineering, Tohoku University, 4‑1 Seiryo, Aoba, Sendai, Miyagi, 980‑8575, Japan
| | - Arunkumar Dorai
- Institute of Multidisciplinary Research for Advanced Materials, Tohoku University, 2‑1-1 Katahira, Aoba, Sendai, Miyagi, 980‑8577, Japan
| | - Shiro Mori
- Laboratory of Biomedical Engineering for Cancer, Graduate School of Biomedical Engineering, Tohoku University, 4‑1 Seiryo, Aoba, Sendai, Miyagi, 980‑8575, Japan
- Division of Oral and Maxillofacial Oncology and Surgical Sciences, Graduate School of Dentistry, Tohoku University, 4‑1 Seiryo, Aoba, Sendai, Miyagi, 980‑8575, Japan
- Biomedical Engineering Cancer Research Center, Graduate School of Biomedical Engineering, Tohoku University, 4‑1 Seiryo, Aoba, Sendai, Miyagi, 980‑8575, Japan
| | - Tetsuya Kodama
- Laboratory of Biomedical Engineering for Cancer, Graduate School of Biomedical Engineering, Tohoku University, 4‑1 Seiryo, Aoba, Sendai, Miyagi, 980‑8575, Japan.
- Biomedical Engineering Cancer Research Center, Graduate School of Biomedical Engineering, Tohoku University, 4‑1 Seiryo, Aoba, Sendai, Miyagi, 980‑8575, Japan.
| |
Collapse
|
21
|
Carnevale MG, Colciago RR, De Santis MC, Cortesi L, De Marco C, Marra A, Vingiani A, Nolè F, Curigliano G, Pruneri G, Llombart-Cussac A, Di Cosimo S, Cortes J. Advancing breast cancer therapy in the era of molecular diagnostics. Breast 2025; 82:104488. [PMID: 40424679 DOI: 10.1016/j.breast.2025.104488] [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: 12/04/2024] [Revised: 03/20/2025] [Accepted: 05/05/2025] [Indexed: 05/29/2025] Open
Abstract
Advances in cancer biology and drug development now enable treatments tailored to individual tumor profile. Targeting specific molecular alterations marked a significant step forward in cancer care, including breast cancer. Access to these therapies is improving thanks to the implementation of molecular tumor boards and efforts to provide molecular diagnostics at sustainable costs for all. In this context, we highlight recent progress in breast cancer therapy, focusing on biomarker-driven approaches, immunotherapy, and precision medicine paving the way for increasingly personalized and effective options.
Collapse
Affiliation(s)
| | | | | | - Laura Cortesi
- Azienda USL IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Cinzia De Marco
- Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | | | - Andrea Vingiani
- Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - Franco Nolè
- European Institute of Oncology IRCCS, Milano, Italy
| | - Giuseppe Curigliano
- European Institute of Oncology IRCCS, Milano, Italy; University "La Statale", Milano, Italy
| | - Giancarlo Pruneri
- Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy; University "La Statale", Milano, Italy
| | | | | | - Javier Cortes
- International Breast Cancer Center, Quironsalud Group, Barcelona, Spain; Universidad Europea de Madrid, Madrid, Spain
| |
Collapse
|
22
|
Shalaby SM, Shawky SA, Ashour H, Sarhan W. The interplay between COX-2, chemotherapeutic drugs, and chemoresistance in colon cancer. Sci Rep 2025; 15:15837. [PMID: 40328989 PMCID: PMC12056169 DOI: 10.1038/s41598-025-98451-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Accepted: 04/11/2025] [Indexed: 05/08/2025] Open
Abstract
Chemoresistance and tumor relapse remain major clinical problems. Evidence indicates that COX2/PGE2/EP axis has a critical role in tumorogenesis and chemoresistance. This study assessed the relation of the COX-2 gene expression with chemoresistance in colon cancer (CC) patients. Also, it explored the effect of chemotherapy on COX-2 expression. The study included 24 patients with CC without chemotherapeutic treatment and 24 chemoresistant CC patients. Tumor and adjacent non-neoplastic colon tissue samples were collected and COX-2 mRNA expression was measured. Also, COX-2 and its related genes; TROP2 and DUSP4 expression were analysed in 5 flurouracil and Oxalliplatin treated Caco-2 and SW-620 cells. The results indicated significant upregulation of COX-2 expression in tissues of chemoresistant CC patients when compared with that in CC tissues without chemotherapy (p < 0.001). There was a relation between COX-2 expression with lymph nodes, metastases and staging in both groups. Concerning in-vitro experiments, there was a dose dependent significant increase of COX-2, TROP2 and DUSP4 mRNA and protein expression levels in 5flurouracil and Oxalliplatin treated cells. These findings demonstrated that overexpression of COX-2 in the chemoresistant CC patients. Both 5 flurouracil and Oxalliplatin induced COX-2 overexpression and in turn COX-2 upregulation may decrease the response of cancer to chemotherapy.
Collapse
Affiliation(s)
- Sally M Shalaby
- Medical Biochemistry Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt.
| | - Salma A Shawky
- Medical Biochemistry Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Hassan Ashour
- Surgery Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Walaa Sarhan
- Medical Biochemistry Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| |
Collapse
|
23
|
Xin J, Xiang Y, Jiang J, Jiang Z, Yi B. Next-generation probiotics Alistipes onderdonkii enhances the efficacy of anti-PD-1 therapy in colorectal cancer. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167891. [PMID: 40328413 DOI: 10.1016/j.bbadis.2025.167891] [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: 05/09/2024] [Revised: 01/22/2025] [Accepted: 05/02/2025] [Indexed: 05/08/2025]
Abstract
Despite significant breakthroughs of therapeutic antibodies targeting PD-1 in cancer treatment, most colorectal cancer (CRC) patients respond poorly to anti-PD-1 immunotherapy. The combination therapy strategies are used to overcome the limitations mentioned above. Alistipes (A.) onderdonkii has anti-cancer effects. This study aimed to examine A. onderdonkii's effects and the related mechanisms in the anti-PD-1 treatment for CRC. In this study, anti-PD-1 therapy notably affected gut microbiota community composition in mice, particularly upregulating A. onderdonkii abundance. A. onderdonkii supplementation markedly promoted anti-PD-1 therapy's effectiveness on CRC treatment, as manifested by the elevated CD8+ T cell infiltration, promoted intestinal barrier integrity, and affected serum metabolomics. A. onderdonkii showed similar effects to the identified next-generation probiotics (NGP) Akkermansia (A.) muciniphila. Additionally, the combination therapy of anti-PD-1 and probiotics supplementation reduced PD-1 level and elevated IL-2 level, indicating that anti-PD-1 combined with A. onderdonkii efficiently restored T cell functions by inhibiting suppressive checkpoints. In conclusion, anti-PD-1 plus A. onderdonkii supplementation could account for a potential method for CRC therapy. These results provide strong evidence for A. onderdonkii as a potential NGP.
Collapse
Affiliation(s)
- Jiaxuan Xin
- Department of Gastrointestinal Surgery, the Third Xiangya Hospital, Central South University, Changsha 410013, Hunan, China
| | - Yandong Xiang
- Department of Gastrointestinal Surgery, the Third Xiangya Hospital, Central South University, Changsha 410013, Hunan, China
| | - Juan Jiang
- Department of Nephrology, the Third Xiangya Hospital, Central South University, Changsha 410013, Hunan, China
| | - Zhengqi Jiang
- Department of Gastrointestinal Surgery, the Third Xiangya Hospital, Central South University, Changsha 410013, Hunan, China
| | - Bo Yi
- Department of Gastrointestinal Surgery, the Third Xiangya Hospital, Central South University, Changsha 410013, Hunan, China.
| |
Collapse
|
24
|
Wang J, Cheng W, Yang R. Nervous system-gut microbiota-immune system axis: future directions for preventing tumor. Front Immunol 2025; 16:1535955. [PMID: 40376000 PMCID: PMC12078214 DOI: 10.3389/fimmu.2025.1535955] [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: 11/28/2024] [Accepted: 04/01/2025] [Indexed: 05/18/2025] Open
Abstract
Tumor is one of the leading causes of death worldwide. The occurrence and development of tumors are related to multiple systems and factors such as the immune system, gut microbiota, and nervous system. The immune system plays a critical role in tumor development. Studies have also found that the gut microbiota can directly or indirectly affect tumorigenesis and tumor development. With increasing attention on the tumor microenvironment in recent years, the nervous system has emerged as a novel regulator of tumor development. Some tumor therapies based on the nervous system have also been tested in clinical trials. However, the nervous system can not only directly interact with tumor cells but also indirectly affect tumor development through the gut microbiota. The nervous system-mediated gut microbiota can regulate tumorigenesis, growth, invasion, and metastasis through the immune system. Here, we mainly explore the potential effects of the nervous system-gut microbiota-immune system axis on tumorigenesis and tumor development. The effects of the nervous system-gut microbiota-immune system axis on tumors involve the nervous system regulating immune cells through the gut microbiota, which can prevent tumor development. Meanwhile, the direct effects of the gut microbiota on tumors and the regulation of the immune system by the nervous system, which can affect tumor development, are also reviewed.
Collapse
Affiliation(s)
- Juanjuan Wang
- Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin, China
| | - Wenyue Cheng
- Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin, China
| | - Rongcun Yang
- Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin, China
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, China
- Translational Medicine Institute, Affiliated Tianjin Union Medical Center of Nankai University, Nankai University, Tianjin, China
| |
Collapse
|
25
|
Tulsian K, Thakker D, Vyas VK. Overcoming chimeric antigen receptor-T (CAR-T) resistance with checkpoint inhibitors: Existing methods, challenges, clinical success, and future prospects : A comprehensive review. Int J Biol Macromol 2025; 306:141364. [PMID: 39988153 DOI: 10.1016/j.ijbiomac.2025.141364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 11/20/2024] [Accepted: 02/19/2025] [Indexed: 02/25/2025]
Abstract
Immune checkpoint blockade is, as of today, the most successful form of cancer immunotherapy, with more than 43 % of cancer patients in the US eligible to receive it; however, only up to 12.5 % of patients respond to it. Similarly, adoptive cell therapy using bioengineered chimeric antigen receptorT (CAR-T) cells and T-cell receptor (TCR) cells has provided excellent responses against liquid tumours, but both forms of immunotherapy have encountered challenges within a tumour microenvironment that is both lacking in tumour-specific T-cells and is strongly immunosuppressive toward externally administered CAR-T and TCR cells. This review focuses on understanding approved checkpoint blockade and adoptive cell therapy at both biological and clinical levels before delving into how and why their combination holds significant promise in overcoming their individual shortcomings. The advent of next-generation checkpoint inhibitors has further strengthened the immune checkpoint field, and a special section explores how these inhibitors can address existing hurdles in combining checkpoint blockade with adoptive cell therapy and homing in on our cancer target for long-term immunity.
Collapse
Affiliation(s)
- Kartik Tulsian
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Nirma University, Ahmedabad, 382481, Gujarat, India
| | - Dhinal Thakker
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Nirma University, Ahmedabad, 382481, Gujarat, India
| | - Vivek K Vyas
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Nirma University, Ahmedabad, 382481, Gujarat, India.
| |
Collapse
|
26
|
Xing L, Wu S, Xue S, Li X. A Novel Neutrophil Extracellular Trap Signature Predicts Patient Chemotherapy Resistance and Prognosis in Lung Adenocarcinoma. Mol Biotechnol 2025; 67:1939-1957. [PMID: 38734842 DOI: 10.1007/s12033-024-01170-1] [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: 01/09/2024] [Accepted: 04/02/2024] [Indexed: 05/13/2024]
Abstract
Chemoresistance is a key obstacle in the long-term survival of patients with locally and advanced lung adenocarcinoma (LUAD). This study used bioinformatic analysis to reveal the chemoresistance of gene-neutrophil extracellular traps (NETs) associated with LUAD. RNA sequencing data and LUAD expression patterns were obtained from the Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases, respectively. The GeneCards database was used to identify NETosis-related genes (NRGs). To identify hub genes with significant and consistent expression, differential analysis was performed using the TCGA-LUAD and GEO datasets. LUAD subtypes were determined based on these hub genes, followed by prognostic analysis. Immunological scoring and infiltration analysis were conducted using NETosis scores (N-scores) derived from the TCGA-LUAD dataset. A clinical prognostic model was established and analyzed, and its clinical applications explored. Twenty-two hub genes were identified, and consensus clustering was used to identify two subgroups based on their expression levels. The Kaplan-Meier (KM) curves demonstrated statistically significant differences in prognosis between the two LUAD subtypes. Based on the median score, patients were further divided into high and low N-score groups, and KM curves showed that the N-scores were more precise at predicting the prognosis of patients with LUAD for overall survival (OS). Immunological infiltration analysis revealed significant differences in the abundances of 10 immune cell infiltrates between the high and low N-score groups. Risk scores indicated significant differences in prognosis between the two extreme score groups. The risk scores for the prognostic model also indicated significant differences between the two groups. The results provide new insights into NETosis-related differentially expressed genes (NRDEGs) associated with chemotherapy resistance in patients with LUAD. The established prognostic model is promising and could help with clinical applications to evaluate patient survival and therapeutic efficiency.
Collapse
Affiliation(s)
- Long Xing
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe East Road, Zhengzhou, 450000, Henan, China
- Department of Oncology, Affiliated Hospital of Qingdao Binhai University, Qingdao, Shandong, China
| | - Shuangli Wu
- Department of Special Examination, Affiliated Hospital of Qingdao Binhai University, Qingdao, Shandong, China
| | - Shiyue Xue
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, Henan, China
| | - Xingya Li
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe East Road, Zhengzhou, 450000, Henan, China.
| |
Collapse
|
27
|
Yang X, Cao X, Zhu Q, Wu H. Pan-cancer analysis of GJB5 as a novel prognostic and immunological biomarker. Sci Rep 2025; 15:14879. [PMID: 40295550 PMCID: PMC12038054 DOI: 10.1038/s41598-025-96389-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Accepted: 03/27/2025] [Indexed: 04/30/2025] Open
Abstract
Gap junction protein B5 (GJB5, also known as Connexin 31.1) has recently been reported to be downregulated in several cancer types, where it functions primarily as a tumor suppressor in cancers such as melanoma and non-small cell lung cancer (NSCLC). However, there no reports describing its prognostic and immunological roles in pan-cancer. This study evaluated the association of GJB5 in various cancer types by a comprehensive pan-cancer analysis. The differential GJB5 expression in tumor and adjacent tissues acquired from The Cancer Genome Atlas (TCGA) databases was compared. Furthermore, univariate Cox regression and Kaplan-Meier survival analyses were performed to assess the influence of GJB5 on the disease-specific survival (DSS), disease-free interval (DFI), clinical stage, progression-free interval (PFI), and overall survival (OS) in various cancers. Moreover, the levels of GJB5 and its activity in the tumor microenvironment were assessed via the Tumor Immune Single-cell Hub (TISCH). In addition, the biological importance of GJB5 levels in various cancers was further assessed via Gene Set Enrichment Analysis. Tumor-Immune System Interactions Database (TISIDB) and Tumor Immune Estimation Resource Database 2.0 (TIMER2.0) tools indicated that GJB5 affected the tumor's immune infiltration potential. This research also evaluated the association of GJB5 with immune features: immune modulatory genes, tumor mutational burden (TMB), and microsatellite instability (MSI). The data indicated that enhanced GJB5 level was linked to worse DFI, OS, PFI, and DSS in some cancers. Additionally, GJB5 level was positively related to immune modulatory genes, TMB, immune cell infiltration, immunological checkpoints, and MSI in malignancies. Furthermore, our study demonstrated that GJB5 was upregulated in colorectal cancer tissues compared to normal tissues. We also assessed GJB5 expression across various pancreatic cell lines. Notably, GJB5 was highly expressed in pancreatic cancer cells relative to normal pancreatic epithelial cells. Additionally, GJB5 knockdown in pancreatic cancer cells resulted in a significant reduction in cell proliferation. In summary, the findings indicated the potential of GJB5 as a prospective prognostic indicator and immunological biomarker.
Collapse
Affiliation(s)
- Xiaojuan Yang
- Liver Digital Transformation Research Laboratory, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, 610041, Sichuan, People's Republic of China
| | - Xunjie Cao
- Abdominal Oncology Ward, Cancer Center, West China Hospital of Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
| | - Qing Zhu
- Abdominal Oncology Ward, Cancer Center, West China Hospital of Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
| | - Hong Wu
- Liver Digital Transformation Research Laboratory, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, 610041, Sichuan, People's Republic of China.
| |
Collapse
|
28
|
Huang X, Ji M, Shang X, Zhang H, Zhang X, Zhou J, Yin T. Smart on-demand drug release strategies for cancer combination therapy. J Control Release 2025; 383:113782. [PMID: 40294796 DOI: 10.1016/j.jconrel.2025.113782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 04/06/2025] [Accepted: 04/24/2025] [Indexed: 04/30/2025]
Abstract
In cancer therapy, enhancing therapeutic indices and patient compliance has been a central focus in recent drug delivery technology development. However, achieving a delicate balance between improving anti-tumor efficacy and minimizing toxicity to normal tissues remains a significant challenge. With the advent of smart on-demand drug release strategies, new opportunities have emerged. These strategies represent a promising approach to drug delivery, enabling precise control over the release of therapeutic agents in a programmed and spatiotemporal manner. Recent studies have focused on designing delivery systems capable of releasing multiple therapeutic agents sequentially, while achieving spatial resolution in vivo. Smart on-demand drug release strategies have demonstrated considerable potential in tumor combination therapy for achieving precision drug delivery and controlled release by responding to specific physiological signals or external physical stimuli in the tumor microenvironment. These strategies not only improve tumor targeting and reduce toxicity to healthy tissues but also enable sequential release in combination therapy, allowing multiple drugs to be released in a specific spatiotemporal order to enhance synergistic treatment effects. In this paper, we systematically reviewed the current research progress of smart on-demand drug release drug delivery strategies in anti-tumor combination therapy. We highlighted representative integrated drug delivery systems and discussed the challenges associated with their clinical application. Additionally, potential future research directions are proposed to further advance this promising field.
Collapse
Affiliation(s)
- Xiaolin Huang
- Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China
| | - Mengfei Ji
- Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China
| | - Xinyu Shang
- Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China
| | - Hengchuan Zhang
- Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China
| | - Xin Zhang
- Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China
| | - Jianping Zhou
- Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China.
| | - Tingjie Yin
- Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China.
| |
Collapse
|
29
|
Fisher JG, Bartlett LG, Kashyap T, Walker CJ, Khakoo SI, Blunt MD. Modulation of anti-tumour immunity by XPO1 inhibitors. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2025; 6:1002310. [PMID: 40291981 PMCID: PMC12022495 DOI: 10.37349/etat.2025.1002310] [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: 01/13/2025] [Accepted: 03/24/2025] [Indexed: 04/30/2025] Open
Abstract
Exportin-1 (XPO1) is a nuclear export protein that, when overexpressed, can facilitate cancer cell proliferation and survival and is frequently overexpressed or mutated in cancer patients. As such, selective inhibitors of XPO1 (XPO1i) function have been developed to inhibit cancer cell proliferation and induce apoptosis. This review outlines the evidence for the immunomodulatory properties of XPO1 inhibition and discusses the potential for combining and sequencing XPO1i with immunotherapy to improve the treatment of patients with cancer. Selinexor is a first-in-class XPO1i that is FDA-approved for the treatment of patients with relapsed and refractory (RR) multiple myeloma and RR diffuse large B cell lymphoma. In addition to the cancer cell intrinsic pro-apoptotic activity, increasing evidence suggests that XPO1 inhibition has immunomodulatory properties. In this review, we describe how XPO1i can lead to a skewing of macrophage polarisation, inhibition of neutrophil extracellular traps, modulation of immune checkpoint expression, blockade of myeloid-derived suppressor cells (MDSCs) and sensitisation of cancer cells to T cell and NK (natural killer) cell immunosurveillance. As such, there is an opportunity for selinexor to enhance immunotherapy efficacy and thus a need for clinical trials assessing selinexor in combination with immunotherapies such as immune checkpoint inhibitors, direct targeting monoclonal antibodies, chimeric antigen receptor (CAR)-T cells and cereblon E3 ligase modulators (CELMoDs).
Collapse
Affiliation(s)
- Jack G. Fisher
- Clinical and Experimental Sciences, University of Southampton, SO16 7YD Southampton, UK
| | - Laura G. Bartlett
- Clinical and Experimental Sciences, University of Southampton, SO16 7YD Southampton, UK
| | | | | | - Salim I. Khakoo
- Clinical and Experimental Sciences, University of Southampton, SO16 7YD Southampton, UK
| | - Matthew D. Blunt
- Clinical and Experimental Sciences, University of Southampton, SO16 7YD Southampton, UK
| |
Collapse
|
30
|
Zhao S, Sun D, Yu H, Wang M, Xu B, Wang Y, Hu F, Wang X, Zhang J, Wang Y, Chai J. Oxaliplatin accelerates immunogenic cell death by activating the cGAS/STING/TBK1/IRF5 pathway in gastric cancer. FEBS J 2025. [PMID: 40260556 DOI: 10.1111/febs.70102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 11/11/2024] [Accepted: 04/07/2025] [Indexed: 04/23/2025]
Abstract
Immunogenic cell death is a tumor cell death involving both innate and adaptive immune responses. Given the published findings that oxaliplatin causes the secretion of high mobility group box 1 (HMGB1) from cancer cells, which is necessary for the initiation of immunogenic cell death, we investigated whether oxaliplatin plays an anticancer role in gastric cancer by inducing immunogenic cell death and further explored its mechanism. We found that oxaliplatin inhibited viability and induced pyroptosis, immunogenic cell death, the production of reactive oxygen species, mitochondrial permeability transition pore (mPTP) opening, and cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) axis activation in gastric cancer cells. Suppressing mPTP opening (cyclosporine treatment), depleting mitochondrial DNA (mtDNA; ethidium bromide treatment), or STING downregulation (H151 or si-STING treatment) reversed cGAS/STING pathway activation and the increased immunogenic cell death induced by oxaliplatin in MKN-45 and AGS cells. Moreover, oxaliplatin induced immunogenic cell death via activating the cGAS/STING/TANK-binding kinase 1 (TBK1; also known as serine/threonine-protein kinase TBK1)/interferon regulatory factor 5 (IRF5) pathway. In conclusion, oxaliplatin treatment could induce immunogenic cell death and mPTP opening and activate the cGAS/STING/TBK1/IRF5 pathway in gastric cancer cells.
Collapse
Affiliation(s)
- Siwei Zhao
- Department of Gastrointestinal Surgery, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Dong Sun
- Department of Gastrointestinal Surgery, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Hang Yu
- Department of Gastrointestinal Surgery, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Menglin Wang
- Department of Plastic Surgery, The First Affiliated Hospital, Dalian Medical University, China
| | - Botao Xu
- Department of Gastrointestinal Surgery, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Yufei Wang
- Department of Gastrointestinal Surgery, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Fangqi Hu
- Department of Gastrointestinal Surgery, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Xiaofeng Wang
- Department of Gastrointestinal Surgery, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Jiazi Zhang
- Department of Gastrointestinal Surgery, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Yongsheng Wang
- Department of Breast Surgery, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Jie Chai
- Department of Gastrointestinal Surgery, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| |
Collapse
|
31
|
Sun Y, Deng Z, Sun H, Wei X, Wang L, Wang S, Gao A, Sun Y, Li J. Prognostic impact of the timing of immunotherapy in first-line immunochemotherapy for patients with advanced lung adenocarcinoma: A propensity score-matched analysis. Int J Cancer 2025. [PMID: 40259531 DOI: 10.1002/ijc.35447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 03/20/2025] [Accepted: 03/26/2025] [Indexed: 04/23/2025]
Abstract
Immunochemotherapy combinations have been the standard first-line therapy for advanced lung adenocarcinoma (LUAD) without driver mutations, wherein concurrent chemotherapy and immunotherapy are conventionally anchored in the established dosing regimen. A few studies have suggested that the timing of immunotherapy in combinations may have a significant impact on the efficacy. However, this issue has not been addressed in an advanced LUAD cohort. We aimed to investigate the prognostic significance of the timing of immunotherapy in first-line immunochemotherapy combinations for patients with advanced LUAD. We retrospectively analyzed 508 patients with advanced LUAD without driver mutations who received immunochemotherapy as initial systemic treatment. The patients were divided into two groups-the induction and non-induction groups-with induction defined as receiving chemotherapy alone before concurrent immunochemotherapy. The bias between different groups was minimized using propensity score matching (PSM). We found both the PFS and OS of the patients in the induction group were significantly longer than those in the non-induction group before (PFS: p < 0.0001, OS: p < 0.0001) and after PSM (PFS: p = 0.0045, OS: p = 0.00073). After adjusting for confounders, induction chemotherapy was still a significant favorable factor for both PFS (p = 0.001) and OS (p = 0.001). In subsequent analyses, we found that both ≥2-cycles induction (PFS: p = 0.000, OS: p = 0.000) and 1-cycle induction (PFS: p = 0.013, OS: p = 0.002) were superior to non-induction and these differences were still significant after PSM. Our findings highlight the notable benefits of induction chemotherapy for patients with advanced LUAD treated with first-line immunochemotherapy combinations.
Collapse
Affiliation(s)
- Yanxin Sun
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong, China
- Phase I Clinical Trail Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Zhenzhen Deng
- Phase I Clinical Trail Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Haifeng Sun
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong, China
- Phase I Clinical Trail Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Xiaojuan Wei
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Leirong Wang
- Phase I Clinical Research Center, Shandong University Cancer Center, Jinan, Shandong, China
| | - Shuyun Wang
- Phase I Clinical Trail Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Aiqin Gao
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Yuping Sun
- Phase I Clinical Trail Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Juan Li
- Phase I Clinical Trail Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| |
Collapse
|
32
|
Zhao S, Xu W, Zeng W, He S, Luo X, Ge D, Duan Y, Han R, Chen CY, Yang Y, Hu Y, Zhang C. Engineered bacterial outer membrane vesicles co-delivering Angio-3 and doxorubicin to enhance tumor therapy. Colloids Surf B Biointerfaces 2025; 253:114707. [PMID: 40262304 DOI: 10.1016/j.colsurfb.2025.114707] [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: 03/01/2025] [Revised: 04/10/2025] [Accepted: 04/13/2025] [Indexed: 04/24/2025]
Abstract
Bacterial outer membrane vesicles (OMVs) have emerged as versatile nanomaterial-based drug delivery systems that can stimulate systemic immune responses and facilitate precise co-delivery of multiple therapeutic agents. This study introduces a bioengineering approach that enables the co-delivery of the angiogenesis inhibitor Angio-3 and the chemotherapeutic agent doxorubicin (DOX) within OMVs, creating a potent antitumor therapeutic platform. Angio-3 displayed on the surface of OMVs inhibited angiogenesis and decreased vascular permeability, which in turn impeded the supply of nutrients necessary for tumor growth. Moreover, intrinsic properties of OMVs triggered a systemic immune response. Both in vitro and in vivo studies, including a CT26 tumor-bearing mouse model, have demonstrated that the OMV@A&D-based therapeutic regimen, which integrates antiangiogenesis, chemotherapy, and immune activation, significantly suppresses tumor proliferation. This study highlights the potential of bioengineered OMVs in revolutionizing cancer therapy by offering a multifaceted and synergistic platform that enhances therapeutic outcomes.
Collapse
Affiliation(s)
- Shuai Zhao
- State Key Laboratory of Biocatalysis and Enzyme Engineering, College of Life Sciences, Hubei University, Wuhan 430062, PR China; Hubei Key Laboratory of Industrial Biotechnology, College of Life Sciences, Hubei University, Wuhan 430062, PR China
| | - Wenxuan Xu
- State Key Laboratory of Biocatalysis and Enzyme Engineering, College of Life Sciences, Hubei University, Wuhan 430062, PR China; Hubei Key Laboratory of Industrial Biotechnology, College of Life Sciences, Hubei University, Wuhan 430062, PR China
| | - Wanting Zeng
- State Key Laboratory of Biocatalysis and Enzyme Engineering, College of Life Sciences, Hubei University, Wuhan 430062, PR China; Hubei Key Laboratory of Industrial Biotechnology, College of Life Sciences, Hubei University, Wuhan 430062, PR China
| | - Shuailin He
- State Key Laboratory of Biocatalysis and Enzyme Engineering, College of Life Sciences, Hubei University, Wuhan 430062, PR China; Hubei Key Laboratory of Industrial Biotechnology, College of Life Sciences, Hubei University, Wuhan 430062, PR China
| | - Xuan Luo
- State Key Laboratory of Biocatalysis and Enzyme Engineering, College of Life Sciences, Hubei University, Wuhan 430062, PR China; Hubei Key Laboratory of Industrial Biotechnology, College of Life Sciences, Hubei University, Wuhan 430062, PR China
| | - Dejie Ge
- State Key Laboratory of Biocatalysis and Enzyme Engineering, College of Life Sciences, Hubei University, Wuhan 430062, PR China; Hubei Key Laboratory of Industrial Biotechnology, College of Life Sciences, Hubei University, Wuhan 430062, PR China
| | - Yamin Duan
- State Key Laboratory of Biocatalysis and Enzyme Engineering, College of Life Sciences, Hubei University, Wuhan 430062, PR China; Hubei Key Laboratory of Industrial Biotechnology, College of Life Sciences, Hubei University, Wuhan 430062, PR China
| | - Rui Han
- State Key Laboratory of Biocatalysis and Enzyme Engineering, College of Life Sciences, Hubei University, Wuhan 430062, PR China; Hubei Key Laboratory of Industrial Biotechnology, College of Life Sciences, Hubei University, Wuhan 430062, PR China
| | - Chin-Yu Chen
- State Key Laboratory of Biocatalysis and Enzyme Engineering, College of Life Sciences, Hubei University, Wuhan 430062, PR China; Hubei Key Laboratory of Industrial Biotechnology, College of Life Sciences, Hubei University, Wuhan 430062, PR China
| | - Yong Yang
- State Key Laboratory of Biocatalysis and Enzyme Engineering, College of Life Sciences, Hubei University, Wuhan 430062, PR China; Hubei Key Laboratory of Industrial Biotechnology, College of Life Sciences, Hubei University, Wuhan 430062, PR China
| | - Yunhong Hu
- State Key Laboratory of Biocatalysis and Enzyme Engineering, College of Life Sciences, Hubei University, Wuhan 430062, PR China; Hubei Key Laboratory of Industrial Biotechnology, College of Life Sciences, Hubei University, Wuhan 430062, PR China.
| | - Cheng Zhang
- State Key Laboratory of Biocatalysis and Enzyme Engineering, College of Life Sciences, Hubei University, Wuhan 430062, PR China; Hubei Key Laboratory of Industrial Biotechnology, College of Life Sciences, Hubei University, Wuhan 430062, PR China.
| |
Collapse
|
33
|
Xu J, Zhou H, Liu Z, Huang Y, Zhang Z, Zou H, Wang Y. PDT-regulated immune gene prognostic model reveals tumor microenvironment in colorectal cancer liver metastases. Sci Rep 2025; 15:13129. [PMID: 40240471 PMCID: PMC12003684 DOI: 10.1038/s41598-025-97667-z] [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: 07/03/2024] [Accepted: 04/07/2025] [Indexed: 04/18/2025] Open
Abstract
Liver metastasis is the most common site of metastasis in colorectal cancer, and the prognosis of colorectal cancer patients with liver metastasis is extremely poor. Revealing the key genes of CLM and implementing targeted interventions is of great significance for colorectal cancer patients. By using the weighted gene co-expression network analysis (WGCNA) algorithm, key gene modules related to metastasis in colorectal cancer were identified. Subsequently, immune-regulating and prognostic-influencing key gene sets were identified from these modules to construct a prognostic model related to colorectal cancer metastasis. Genetic background differences underlying this model were analyzed using colorectal cancer methylation and mutation data, followed by Gene Ontology (GO) analysis and Gene Set Enrichment Analysis (GSEA) analysis of the relevant biological processes associated with the model. The value of predicting tumor drug response through the model was assessed using drug half maximal inhibitory concentration (IC50) data from colorectal cancer cell lines. Subsequently, utilizing single-cell sequencing data about liver metastasis, the colorectal cancer immune microenvironment reflected in the predictive model was analyzed, and a key gene set of the model was identified. Lastly, experimental validation was conducted to investigate the regulatory effects of photodynamic therapy (PDT) on the key genes of the model, and the cytotoxic effect of PDT on colorectal cancer was confirmed. An immune-related gene prognostic model regulating CLM was constructed, consisting of HSPA1A, ULBP2, RBP7, OXT, SLC11A1, INHBB, and ICOS. This model can predict the clinical response of colorectal cancer patients to Oxaliplatin, Cisplatin, Irinotecan, and 5-Fluorouracil. Single-cell sequencing results demonstrate that the model is associated with an immunosuppressive microenvironment in CLM. The higher the model's riskscore, the weaker the MHC-I, MHC-II, and various tumor immune signaling pathway networks in the colorectal cancer microenvironment. Causal analysis reveals that SLC11A1, ICOS, and HSPA1A play key roles in this model. PDT can kill colorectal cancer cells, inhibit colorectal cancer cell metastasis, significantly influence the expression of genes such as SLC11A1, ICOS, and HSPA1A in these processes, and suppress the infiltration of macrophages in the colorectal microenvironment, inhibiting the immune escape process of PD-1/PD-L1. A prognostic model based on immunity regulated by PDT has been established for assessing the prognosis of CLM patients, as well as clinical responses to chemotherapy drugs and immunotherapy.
Collapse
Affiliation(s)
- Jiachi Xu
- Department of General Surgery, The Second Xiangya Hospital of Central South University, Changsha, 410011, China
| | - Hui Zhou
- Department of General Surgery, The Second Xiangya Hospital of Central South University, Changsha, 410011, China
| | - Zhongtao Liu
- Department of General Surgery, The Second Xiangya Hospital of Central South University, Changsha, 410011, China
| | - Yunpeng Huang
- Department of General Surgery, The Second Xiangya Hospital of Central South University, Changsha, 410011, China
| | - Zijian Zhang
- Department of General Surgery, The Second Xiangya Hospital of Central South University, Changsha, 410011, China
| | - Heng Zou
- Department of General Surgery, The Second Xiangya Hospital of Central South University, Changsha, 410011, China.
| | - Yongxiang Wang
- Department of General Surgery, The Second Xiangya Hospital of Central South University, Changsha, 410011, China.
| |
Collapse
|
34
|
Xiao X, Wang QW, Zhou ZY, Wang LS, Huang P. Precision treatment for human epidermal growth factor receptor 2-amplified advanced rectal cancer: A case report. World J Gastrointest Oncol 2025; 17:102690. [PMID: 40235909 PMCID: PMC11995321 DOI: 10.4251/wjgo.v17.i4.102690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 01/23/2025] [Accepted: 02/17/2025] [Indexed: 03/25/2025] Open
Abstract
BACKGROUND Although targeted therapy provides survival benefits for patients with metastatic colorectal cancer, some patients develop resistance to these treatments. Human epidermal growth factor receptor 2 (HER2) is overexpressed in a subset of patients with colorectal cancer and has been established as a therapeutic target. CASE SUMMARY This case report describes a Chinese patient with HER2-amplified advanced rectal cancer who showed no response to chemotherapy and targeted therapies against epidermal growth factor receptor and vascular endothelial growth factor but achieved a remarkable response following treatment with immune checkpoint inhibitors (ICIs) in combination with pyrotinib. The combination of oxaliplatin and ICIs with pyrotinib demonstrates synergistic effects after late-stage disease progression. CONCLUSION ICIs and pyrotinib may be effective in treating HER2-amplified advanced rectal cancer. Chemotherapy following disease progression could enhance efficacy synergistically.
Collapse
Affiliation(s)
- Xia Xiao
- Department of Oncology, Wuxi No. 2 People’s Hospital, Jiangnan University Medical Center, Wuxi 214002, Jiangsu Province, China
| | - Qing-Wen Wang
- Wuxi Medical College, Jiangnan University, Wuxi 214122, Jiangsu Province, China
| | - Zheng-Yang Zhou
- Wuxi Medical College, Jiangnan University, Wuxi 214122, Jiangsu Province, China
| | - Lei-Sheng Wang
- Wuxi Medical College, Jiangnan University, Wuxi 214122, Jiangsu Province, China
| | - Pei Huang
- Department of Oncology, Wuxi No. 2 People’s Hospital, Jiangnan University Medical Center, Wuxi 214002, Jiangsu Province, China
| |
Collapse
|
35
|
Martin SD, Thornton S, Chow C, Milne K, de Barros JS, Morris KA, Leung S, Jamieson A, Nelson BH, Cochrane DR, Huntsman DG, Gilks CB, Hoang L, McAlpine JN, Zhang AW. Activated immune infiltrates expand opportunities for targeted therapy in p53-abnormal endometrial carcinoma. J Pathol 2025. [PMID: 40223796 DOI: 10.1002/path.6429] [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: 10/18/2024] [Revised: 02/12/2025] [Accepted: 03/17/2025] [Indexed: 04/15/2025]
Abstract
Tumor protein p53 mutated/abnormal (p53abn) endometrial carcinomas account for over 50% of deaths but comprise only 15% of all endometrial carcinomas. Most patients show limited response to standard-of-care chemotherapy with or without radiotherapy, and only a minority of cases are amenable to targeted therapies like poly-ADP ribose polymerase (PARP) inhibitors and HER2-directed therapies. Recent immunotherapy clinical trials have demonstrated remarkable efficacy, not only in mismatch repair deficient (MMRd) tumors but also in a subset of mismatch repair-proficient (MMRp) tumors. However, the immune microenvironment and its relationship to other therapeutic targets in MMRp endometrial carcinoma remains poorly understood. Here, we characterize the immune microenvironment of p53abn endometrial carcinoma, the most clinically aggressive subtype of MMRp endometrial carcinoma, and correlate antitumor immune signatures with other targetable alterations. We accrued 256 treatment-naïve p53abn endometrial carcinomas and systemically profiled T-cell, B-cell, myeloid, and tumor-cell populations with multiplex immunofluorescence to assess the tissue localization and functional status of immune cells. Shallow whole-genome sequencing was performed on a subset of 126 cases. Patterns of immune infiltration were compared to survival outcomes and mutational signatures. Mixture modeling divided p53abn endometrial carcinoma into tumor-infiltrating lymphocyte (TIL)-rich and TIL-poor subsets. Over 50% of tumors were TIL-rich. TIL-rich cases overexpressed targetable immune evasion molecules and were associated with longer overall and disease-specific survival in multivariate analysis. This effect was particularly pronounced in advanced stage disease and in patients who did not receive adjuvant chemotherapy. TIL did not associate with homologous recombination deficient mutational signatures or HER2 amplification. Our findings demonstrate a biological rationale for immunotherapy in a substantial subset of patients with p53abn endometrial cancer and may help inform combination therapies with immune checkpoint inhibition, PARP inhibitors, and anti-HER2 agents. © 2025 The Author(s). The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Spencer D Martin
- Department of Pathology and Laboratory Medicine, Faculty of Medicine, The University of British Columbia, Vancouver, Canada
| | - Shelby Thornton
- Molecular and Advanced Pathology Core (MAPcore), The University of British Columbia, Vancouver, Canada
| | - Christine Chow
- Molecular and Advanced Pathology Core (MAPcore), The University of British Columbia, Vancouver, Canada
| | - Katy Milne
- Trev and Joyce Deeley Research Centre, British Columbia Cancer Agency, Victoria, Canada
| | - Juliana Sobral de Barros
- Department of Molecular Oncology, British Columbia Cancer Agency, The University of British Columbia, Vancouver, Canada
| | - Kayleigh A Morris
- Trev and Joyce Deeley Research Centre, British Columbia Cancer Agency, Victoria, Canada
| | - Samuel Leung
- Department of Molecular Oncology, British Columbia Cancer Agency, The University of British Columbia, Vancouver, Canada
| | - Amy Jamieson
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, The University of British Columbia, Vancouver, Canada
| | - Brad H Nelson
- Trev and Joyce Deeley Research Centre, British Columbia Cancer Agency, Victoria, Canada
- Department of Medical Genetics, Faculty of Medicine, The University of British Columbia, Vancouver, Canada
| | - Dawn R Cochrane
- Department of Molecular Oncology, British Columbia Cancer Agency, The University of British Columbia, Vancouver, Canada
| | - David G Huntsman
- Department of Pathology and Laboratory Medicine, Faculty of Medicine, The University of British Columbia, Vancouver, Canada
| | - C Blake Gilks
- Department of Pathology and Laboratory Medicine, Faculty of Medicine, The University of British Columbia, Vancouver, Canada
| | - Lien Hoang
- Department of Pathology and Laboratory Medicine, Faculty of Medicine, The University of British Columbia, Vancouver, Canada
| | - Jessica N McAlpine
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, The University of British Columbia, Vancouver, Canada
| | - Allen W Zhang
- Department of Pathology and Laboratory Medicine, Faculty of Medicine, The University of British Columbia, Vancouver, Canada
| |
Collapse
|
36
|
Aronson SL, Thijssen B, Lopez-Yurda M, Koole SN, van der Leest P, León-Castillo A, Harkes R, Seignette IM, Sanders J, Alkemade M, Kemper I, Holtkamp MJ, Mandjes IAM, Broeks A, Lahaye MJ, Rijlaarsdam MA, van den Broek D, Wessels LFA, Horlings HM, van Driel WJ, Sonke GS. Neo-adjuvant pembrolizumab in stage IV high-grade serous ovarian cancer: the phase II Neo-Pembro trial. Nat Commun 2025; 16:3520. [PMID: 40229272 PMCID: PMC11997049 DOI: 10.1038/s41467-025-58440-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: 10/22/2024] [Accepted: 03/24/2025] [Indexed: 04/16/2025] Open
Abstract
While immune checkpoint inhibitors (ICIs) have revolutionized cancer treatment, their efficacy in high-grade serous ovarian cancer (HGSOC) remains limited. Some patients, however, achieve lasting responses, emphasizing the need to understand how tumor microenvironment and molecular characteristics influence ICI response. The phase 2 Neo-Pembro study (NCT03126812) included 33 untreated stage IV HGSOC patients, who were scheduled for 6 cycles of carboplatin-paclitaxel and interval cytoreductive surgery. Pembrolizumab (pembro) was added from cycle two and continued for one year. The primary objective was to assess intratumoral immune activation using multiplexed immunofluorescence and immune-related gene expression. Our findings show immune activation, evidenced by an increase in CD3 + , CD8 + , CD8 + /FOXP3+ ratio, TNF-α and interferon-γ signaling. Treatment was well-tolerated. We observed major pathologic responses in 9/33 patients (27%, 95%CI 14-46), with pathologic response strongly associated with immune activation and OS. At a median follow-up of 52.8 months, 8/9 major responders were alive, with 6 patients recurrence-free. In contrast, 4/24 minor responders survived, including one recurrence-free. ctDNA clearance was observed in all major responders and was associated with prolonged PFS and OS. PD-L1 expression and homologous recombination deficiency were predictive of major response and may serve as biomarkers, warranting further exploration. These results suggest major responders may benefit from neo-adjuvant pembro.
Collapse
Affiliation(s)
- S L Aronson
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Center for Gynecologic Oncology Amsterdam, Department of Gynecologic Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - B Thijssen
- Division of Molecular Carcinogenesis, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Oncode Institute, Utrecht, Netherlands
| | - M Lopez-Yurda
- Department of Biometrics, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - S N Koole
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Center for Gynecologic Oncology Amsterdam, Department of Gynecologic Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - P van der Leest
- Department of Laboratory Medicine, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - A León-Castillo
- Department of Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - R Harkes
- Department of Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - I M Seignette
- Department of Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - J Sanders
- Department of Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - M Alkemade
- Core Facility Molecular Pathology & Biobanking, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - I Kemper
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - M J Holtkamp
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - I A M Mandjes
- Department of Biometrics, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - A Broeks
- Core Facility Molecular Pathology & Biobanking, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - M J Lahaye
- Department of Radiology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- GROW Research Institute for Oncology and Reproduction, Maastricht University, Maastricht, The Netherlands
| | - M A Rijlaarsdam
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - D van den Broek
- Department of Laboratory Medicine, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - L F A Wessels
- Division of Molecular Carcinogenesis, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Oncode Institute, Utrecht, Netherlands
| | - H M Horlings
- Department of Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - W J van Driel
- Center for Gynecologic Oncology Amsterdam, Department of Gynecologic Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - G S Sonke
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands.
| |
Collapse
|
37
|
Gao X, Xu Y, Hu X, Chen J, Zhang D, Xu X. Comprehensive analysis of mitochondrial solute carrier family 25 (SLC25) identifies member 19 (SLC25A19) as a regulatory factor in hepatocellular carcinoma. Gene 2025; 944:149299. [PMID: 39892835 DOI: 10.1016/j.gene.2025.149299] [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: 11/01/2024] [Revised: 01/08/2025] [Accepted: 01/28/2025] [Indexed: 02/04/2025]
Abstract
BACKGROUND The mitochondrial solute carrier family 25 (SLC25) is known to play a pivotal role in oncogenesis, yet its specific involvement in hepatocellular carcinoma (HCC) remains poorly elucidated. METHODS In this study, we performed a clustering analysis of HCC patients in the Cancer Genome Atlas database based on the expression levels of SLC25 members, and conducted clinical feature analysis for each patient within the clusters. Subsequently, we developed a prognostic model using a Lasso regression approach with SLC25A19, SLC25A49, and SLC25A51 as features, and generated a risk score for each HCC patient. We then identified SLC25A19 as a potential prognostic marker for HCC through single-cell analysis, and validated this finding using in vitro and in vivo experiments. RESULTS Our results revealed significant differences in the expression of most SLC25 family members in HCC patients, enabling the stratification of patients into three clusters, with those in cluster 1 exhibiting the most favorable prognosis and showing a correlation with enhanced immune infiltration. The risk scores derived from the features SLC25A19, SLC25A49, and SLC25A51 effectively predicted the prognosis of HCC patients, with area under the curve (AUC) values exceeding 0.7 in the test group. Single-cell analysis further demonstrated h eightened expression of SLC25A19 in the immune microenvironment of HCC, and in vitro experiments indicated that SLC25A19 may regulate the proliferation, migration, invasion, cycle, and apoptosis of liver cancer cells through the Wnt pathway. In the HepG2 animal model, overexpression of SLC25A19 significantly promotes tumor growth, while knockdown inhibits tumor growth. Analysis of patient tumor tissues shows that SLC25A19 is highly expressed in liver cancer tissues and is associated with CD8+ T cell infiltration. CONCLUSIONS In conclusion, our comprehensive analysis of the role of SLC25 in HCC unveiled SLC25A19 as a potential regulatory factor in HCC.
Collapse
Affiliation(s)
- Xueke Gao
- Renmin Hospital of Wuhan University, Wuhan, Hubei, China 430060
| | - Yangtao Xu
- Renmin Hospital of Wuhan University, Wuhan, Hubei, China 430060
| | - Xinyao Hu
- Renmin Hospital of Wuhan University, Wuhan, Hubei, China 430060
| | - Jiayu Chen
- Renmin Hospital of Wuhan University, Wuhan, Hubei, China 430060
| | - Daoming Zhang
- Renmin Hospital of Wuhan University, Wuhan, Hubei, China 430060
| | - Ximing Xu
- Renmin Hospital of Wuhan University, Wuhan, Hubei, China 430060.
| |
Collapse
|
38
|
Zafar M, Sweis N, Kapoor H, Gantt G. Advances and Challenges in the Treatment of HPV-Associated Lower Genital Tract Cancers by Immune Checkpoint Blockers: Insights from Basic and Clinical Science. Cancers (Basel) 2025; 17:1260. [PMID: 40282436 PMCID: PMC12026392 DOI: 10.3390/cancers17081260] [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: 02/26/2025] [Revised: 03/26/2025] [Accepted: 03/27/2025] [Indexed: 04/29/2025] Open
Abstract
Human papillomavirus (HPV)-related lower genital cancers, including cervical cancer, anal squamous cell carcinoma (SCC), vaginal cancer, vulvar cancer, and penile cancer, pose a significant health burden, with approximately 45,000 new cases diagnosed annually. Current effective treatment modalities include chemoradiotherapy, systemic chemotherapy, and immune checkpoint inhibitors (ICIs). The tumor microenvironment in HPV-related cancers is characterized by immune evasion mechanisms, including the modulation of immune checkpoints such as PD-L1/PD-1. HPV oncoproteins E5, E6, and E7 play crucial roles in this process, altering the expression of immune inhibitory molecules and the recruitment of immune cells. ICIs, such as programmed cell death protein 1 (PD-1) inhibitors, have shown efficacy in enhancing the immune response against HPV-associated tumors by blocking proteins that allow cancer cells to evade immune surveillance. Recent studies have demonstrated that HPV-positive tumors exhibit a more favorable response to ICI-based therapies compared to HPV-negative tumors. The integration of ICIs into treatment regimens for HPV-related cancers has been supported by several clinical trials. The inclusion of ICIs in the treatment approach for HPV-related lower genital cancers presents a promising opportunity for improving patient outcomes. Ongoing research and clinical trials are advancing our understanding of the immune microenvironment and the therapeutic potential of immunotherapy for these cancers.
Collapse
|
39
|
Su Q, Fang L, Li C, Yue L, Yun Z, Zhang H, Liu Q, Ma R, Zhong P, Liu H, Lou Z, Chen Z, Tan Y, Hao X, Wu C. Multi-omics insights into the roles of CCNB1, PLK1, and HPSE in breast cancer progression: implications for prognosis and immunotherapy. Discov Oncol 2025; 16:471. [PMID: 40186712 PMCID: PMC11972280 DOI: 10.1007/s12672-025-02282-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/01/2025] [Accepted: 04/01/2025] [Indexed: 04/07/2025] Open
Abstract
BACKGROUND This study examines the roles of Cyclin B1 (CCNB1), Polo-Like Kinase 1 (PLK1), and Heparanase (HPSE) in breast cancer progression using a multi-omics approach. These genes are known for their involvement in various cancer-related processes, but their precise contributions to breast cancer remain unclear. METHODS We employed an integrative analysis combining transcriptomics, proteomics, DNA methylation profiling, immune infiltration analysis, and single-cell RNA sequencing to investigate the expression patterns, regulatory mechanisms, and functional impacts of CCNB1, PLK1, and HPSE in breast cancer. Functional assays using si-RNA knockdown of CCNB1 and PLK1 were performed to assess their roles in cell proliferation. RESULTS CCNB1, PLK1, and HPSE are upregulated in breast tumors at the mRNA and protein levels. CCNB1 and PLK1 promote tumor growth and metastasis, while HPSE is linked to immune pathways. DNA methylation in BRCA correlates with prognosis, with PLK1 alterations protective for recurrence-free survival. High expression of these genes worsens prognosis, with CCNB1 as a risk factor for overall survival. Immune infiltration analysis associates these genes with tumor-infiltrating immune cells, highlighting HPSE's immunotherapeutic potential. Single-cell RNA sequencing confirms CCNB1 and PLK1 drive malignant proliferation and an immunosuppressive environment. Functional assays demonstrated that silencing CCNB1 and PLK1 significantly reduced breast cancer cell proliferation, indicating regulatory interactions among PLK1, CCNB1, and MKI67. CONCLUSIONS This study provides evidence that CCNB1, PLK1, and HPSE are key players in breast cancer progression and potential biomarkers for prognosis. Furthermore, their roles in immune regulation suggest they could be promising targets for immunotherapy.
Collapse
Affiliation(s)
- Qisheng Su
- Key Laboratory of Clinical Laboratory Medicine of Guangxi Department of Education, Department of Clinical Laboratory, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Leiming Fang
- Faculty of Medicine, Dalian University of Technology, Dalian, China
- Department of Stem Cell and Regenerative Medicine, Institute of Health Service and Transfusion Medicine, Beijing, China
| | - Chaofan Li
- Graduate School of Hebei North University, Zhangjiakou, China
- Department of Tuberculosis Medicine, The Eighth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Liang Yue
- Department of Stem Cell and Regenerative Medicine, Institute of Health Service and Transfusion Medicine, Beijing, China
| | - Zhimin Yun
- Department of Stem Cell and Regenerative Medicine, Institute of Health Service and Transfusion Medicine, Beijing, China
| | - Huiqiang Zhang
- Breast Cancer Department of Oncology Institute, The Fifth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Qi Liu
- Faculty of Medicine, Dalian University of Technology, Dalian, China
- Department of Stem Cell and Regenerative Medicine, Institute of Health Service and Transfusion Medicine, Beijing, China
| | - Ruilin Ma
- Faculty of Medicine, Dalian University of Technology, Dalian, China
- Department of Stem Cell and Regenerative Medicine, Institute of Health Service and Transfusion Medicine, Beijing, China
| | - Pengfei Zhong
- Department of Stem Cell and Regenerative Medicine, Institute of Health Service and Transfusion Medicine, Beijing, China
| | - He Liu
- Faculty of Medicine, Dalian University of Technology, Dalian, China
- Department of Stem Cell and Regenerative Medicine, Institute of Health Service and Transfusion Medicine, Beijing, China
| | - Zhangrong Lou
- Faculty of Medicine, Dalian University of Technology, Dalian, China
| | - Zhi Chen
- Department of Tuberculosis Medicine, The Eighth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Yingxia Tan
- Department of Stem Cell and Regenerative Medicine, Institute of Health Service and Transfusion Medicine, Beijing, China.
| | - Xiaopeng Hao
- Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing, China.
- Breast Cancer Department of Oncology Institute, The Fifth Medical Center, Chinese PLA General Hospital, Beijing, China.
| | - Chengjun Wu
- School of Health and Life Sciences, Qingdao Central Hospital, University of Health and Rehabilitation Sciences, Qinadao, 266113, China.
| |
Collapse
|
40
|
Ding Y, Yu Y. Therapeutic potential of flavonoids in gastrointestinal cancer: Focus on signaling pathways and improvement strategies (Review). Mol Med Rep 2025; 31:109. [PMID: 40017144 PMCID: PMC11884236 DOI: 10.3892/mmr.2025.13474] [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: 09/18/2024] [Accepted: 01/30/2025] [Indexed: 03/01/2025] Open
Abstract
Flavonoids are a group of polyphenolic compounds distributed in vegetables, fruits and other plants, which have considerable antioxidant, anti‑tumor and anti‑inflammatory activities. Several types of gastrointestinal (GI) cancer are the most common malignant tumors in the world. A large number of studies have shown that flavonoids have inhibitory effects on cancer, and they are recognized as a class of potential anti‑tumor drugs. Therefore, the present review investigated the molecular mechanisms of flavonoids in the treatment of different types of GI cancer and summarized the drug delivery systems commonly used to improve their bioavailability. First, the classification of flavonoids and the therapeutic effects of various flavonoids on human diseases were briefly introduced. Then, to clarify the mechanism of action of flavonoids on different types of GI cancer in the human body, the metabolic process of flavonoids in the human body and the associated signaling pathways causing five common types of GI cancer were discussed, as well as the corresponding therapeutic targets of flavonoids. Finally, in clinical settings, flavonoids have poor water solubility, low permeability and inferior stability, which lead to low absorption efficiency in vivo. Therefore, the three most widely used drug delivery systems were summarized. Suggestions for improving the bioavailability of flavonoids and the focus of the next stage of research were also put forward.
Collapse
Affiliation(s)
- Ye Ding
- Henan Key Laboratory of Helicobacter Pylori and Microbiota and Gastrointestinal Cancer, Marshall Medical Research Center, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
- Department of Gastroenterology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Yong Yu
- Henan Key Laboratory of Helicobacter Pylori and Microbiota and Gastrointestinal Cancer, Marshall Medical Research Center, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
- Department of Gastroenterology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| |
Collapse
|
41
|
Tredicine M, Mucci M, Recchiuti A, Mattoscio D. Immunoregulatory mechanisms of the arachidonic acid pathway in cancer. FEBS Lett 2025; 599:927-951. [PMID: 39973474 PMCID: PMC11995684 DOI: 10.1002/1873-3468.70013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 01/10/2025] [Accepted: 01/27/2025] [Indexed: 02/21/2025]
Abstract
The arachidonic acid (AA) pathway promotes tumor progression by modulating the complex interactions between cancer and immune cells within the microenvironment. In this Review, we summarize the knowledge acquired thus far concerning the intricate mechanisms through which eicosanoids either promote or suppress the antitumor immune response. In addition, we will discuss the impact of eicosanoids on immune cells and how they affect responsiveness to immunotherapy, as well as potential strategies for manipulating the AA pathway to improve anticancer immunotherapy. Understanding the molecular pathways and mechanisms underlying the role played by AA and its metabolites in tumor progression may contribute to the development of more effective anticancer immunotherapies.
Collapse
Affiliation(s)
- Maria Tredicine
- Department of Medical, Oral and Biotechnological SciencesUniversity of Chieti‐PescaraItaly
- Center for Advanced Studies and TechnologyUniversity of Chieti‐PescaraItaly
| | - Matteo Mucci
- Department of Medical, Oral and Biotechnological SciencesUniversity of Chieti‐PescaraItaly
- Center for Advanced Studies and TechnologyUniversity of Chieti‐PescaraItaly
| | - Antonio Recchiuti
- Department of Medical, Oral and Biotechnological SciencesUniversity of Chieti‐PescaraItaly
- Center for Advanced Studies and TechnologyUniversity of Chieti‐PescaraItaly
| | - Domenico Mattoscio
- Department of Medical, Oral and Biotechnological SciencesUniversity of Chieti‐PescaraItaly
- Center for Advanced Studies and TechnologyUniversity of Chieti‐PescaraItaly
| |
Collapse
|
42
|
Ruan X, Wu L, Tang Z, Li Y, Wang J, Jiang H, Zhang L, Wang S, Chen Z, Yuan C, Xia Y, Pan Y, Gao J, Zhao X. Two chemotherapeutic agents expand stem-like CD62L +CD8 + T cells in antitumor immune responses. Front Immunol 2025; 16:1533857. [PMID: 40236705 PMCID: PMC11996895 DOI: 10.3389/fimmu.2025.1533857] [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: 11/25/2024] [Accepted: 03/14/2025] [Indexed: 04/17/2025] Open
Abstract
Introduction Recent findings reveal that the precursors of exhausted CD8+ T (CD8+ Tpex) cells possess stem-like signatures in tumor immunity, which originate from tumor draining lymph node (TdLN)-derived tumor-specific memory (CD8+ TTSM) cells. Both of these T subsets can be collectively referred to as stem-like CD8+ T cells, which demonstrate robust self-renewal ability and can proliferate and differentiate into transitory effector-like exhausted T cells (Texint). There are reports that chemotherapeutic drugs can promote the antitumor immune responses of patients by increasing the number of CD8+ T cells; however, whether chemotherapeutic drugs increase these two stem-like CD8+ T cells remain further exploration. Methods Tpex cell-associated subpopulations in human colorectal tumors were analyzed by using single-cell sequencing data. CT26 and B16 tumor models of wild type and Eomes conditional knockout mice were constructed, and the changes of TTSM, Tpex and Tex subsets in mice were dissected by flow cytometry after treatment with decitabine (DAC), doxorubicin (DOX) and 5-Fluorouracil (5-FU). Results In this study, we demonstrated that DAC and 5-FU expanded CD8+ TTSM cells in TdLNs. At the same time, we validated that DAC and 5-FU substantially promoted the expansion of CD62L+CD8+ Tpex cells and subsequently increased effector function of CX3CR1+ CD8+ Texint cells. In addition, the conditional knockout of transcription factor Eomes in CD8+ T cells partially eliminated DAC-amplified CD62L+ CD8+ Tpex cells, but had no effect on such CD8+ T subset expanded by 5-FU. Conclusion The present study demonstrated that both DAC and 5-FU promoted the differentiation of stem-like CD8+ TTSM cells in TdLNs and significantly enhanced the differentiation and expansion of stem-like CD62L+ CD8+ Tpex and CX3CR1+ Texint cells in tumor microenvironment. The knockout of Eomes partially influenced the role of DAC in promoting the differentiation and expansion of stem-like CD8+ T cells.
Collapse
Affiliation(s)
- Xiaokang Ruan
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
- Department of General Surgery, People's Hospital of Dongxihu District, Wuhan, China
| | - Linwei Wu
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Zijian Tang
- The Affiliated Infectious Diseases Hospital, Suzhou Medical College of Soochow University, Suzhou, China
| | - Yao Li
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jin Wang
- Department of General Surgery, The Fourth Affiliated Hospital of Soochow University, Suzhou, China
| | - Haolin Jiang
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Li Zhang
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Shengjia Wang
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou, China
- Jiangsu Key Laboratory of Gastrointestinal Tumor Immunology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Zhaoqiang Chen
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Chenlei Yuan
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yujian Xia
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yan Pan
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jianling Gao
- Department of Critical Care Medicine, The Fourth Affiliated Hospital of Soochow University, Suzhou, China
| | - Xin Zhao
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou, China
- Jiangsu Key Laboratory of Gastrointestinal Tumor Immunology, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
43
|
Dong Y, Li J, Dai Y, Zhang X, Wang T, Zhao B, Liu W, Chen L, Yang S, Du P, Jiao Z. Redox-responsive metal-organic framework nanocapsules enhance tumor chemo-immunotherapy by modulating tumor metabolic reprogramming. Mater Today Bio 2025; 31:101487. [PMID: 39896279 PMCID: PMC11786678 DOI: 10.1016/j.mtbio.2025.101487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Revised: 01/04/2025] [Accepted: 01/11/2025] [Indexed: 02/04/2025] Open
Abstract
Immunotherapy, particularly immune checkpoint blockade (ICB), has opened the era of modern oncology, offering significant promise for modern oncology. However, the efficacy of immunotherapy is frequently curtailed by the immunosuppressive tumor microenvironment (ITM), a milieu shaped by tumor metabolic reprogramming. Herein, a novel tumor microenvironment-responsive nanocapsules (DNMCs) were developed that simultaneously modulate tumor metabolism and the ITM to enhance the effectiveness of chemo-immunotherapy. DNMCs consist of an acidic and redox-sensitive metal-organic framework (MOF) encapsulating Doxorubicin (DOX) and the indoleamine-2,3-dioxygenase1 (IDO1) inhibitor NLG919. In the tumor microenvironment, DNMCs degrade, rapidly releasing DOX and NLG919. DOX induces immunogenic cell death (ICD), while NLG919 regulates amino acid metabolism by modulating IDO1 activity, thereby reversing the immunosuppressive of ITM. Consequently, DNMCs elicit effective anti-tumor immune responses, characterized by an increased density of tumor-infiltrating CD8+ cytotoxic T cells as well as depletion of immunosuppressive regulatory T cells (Tregs), thus effectively suppressing pancreatic cancer growth in KPC mice through combined chemo-immunotherapy. Overall, DNMCs exhibit significant tumor growth inhibition in pancreatic cancer patient-derived organoids (PDOs) and mouse models. This study presents a promising approach to enhancing chemo-immunotherapy by targeting tumor metabolic reprogramming and augmenting immune response against malignant tumors.
Collapse
Affiliation(s)
- Yuman Dong
- Cuiying Biomedical Research Center, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, 730030, China
| | - Jieru Li
- The Second Clinical Medical College, Lanzhou University, Lanzhou, 730030, China
| | - Yiwei Dai
- The Second Clinical Medical College, Lanzhou University, Lanzhou, 730030, China
| | - Xinyu Zhang
- The Second Clinical Medical College, Lanzhou University, Lanzhou, 730030, China
| | - Tao Wang
- The Second Clinical Medical College, Lanzhou University, Lanzhou, 730030, China
| | - Bin Zhao
- The Second Clinical Medical College, Lanzhou University, Lanzhou, 730030, China
| | - Wenbo Liu
- The Second Clinical Medical College, Lanzhou University, Lanzhou, 730030, China
| | - Li Chen
- Department of Orthopaedics, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, 730030, China
| | - Shaopei Yang
- College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, 730000, China
| | - Pengcheng Du
- College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, 730000, China
| | - Zuoyi Jiao
- Department of General Surgery, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, 730030, China
| |
Collapse
|
44
|
Xu RC, Zhang YW, Liu CC, Xu YY, Shao ZM, Yu KD. Immunotherapy and its racial specificity for breast cancer treatment in Asia: a narrative review. THE LANCET REGIONAL HEALTH. WESTERN PACIFIC 2025; 57:101180. [PMID: 40443538 PMCID: PMC12121431 DOI: 10.1016/j.lanwpc.2024.101180] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 07/23/2024] [Accepted: 08/13/2024] [Indexed: 06/02/2025]
Abstract
Immunotherapy, including immune checkpoint inhibitors, adoptive cell therapy, cancer vaccines, and other modalities, represents a significant advancement in cancer treatment. Breast cancer, traditionally considered less amenable to immunotherapy, has demonstrated responsiveness to immunotherapy when combined with conventional treatment options. These integrative strategies enhance the effectiveness of immunotherapy, bringing hope to patients. Furthermore, precision therapies guided by predictive biomarkers refine the scope of breast cancer immunotherapy and broaden its advantages. Notably, it is essential to recognise the differences in breast cancer epidemiology, clinical outcomes, and molecular signatures between Asian populations and those in Europe and North America. These include a higher proportion of premenopausal patients and variation in subtype distribution and gene mutation profiles, underscoring the importance of considering racial specificity in immunotherapy. Clinical efforts in Asia, supported by ethnicity-specific studies, indigenous immunotherapeutic agents, and precision medicine informed by predictive biomarkers, provide tailored treatment options. This review aims to present an overview of breast cancer immunotherapy while address the racial specificity to inform its application for Asian patients.
Collapse
Affiliation(s)
- Rui-Chen Xu
- Department of Breast Surgery, Fudan University Shanghai Cancer Center and Cancer Institute, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, PR China
- Key Laboratory of Breast Cancer in Shanghai, Shanghai, 200032, PR China
| | - Yan-Wu Zhang
- Department of Breast Surgery, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, PR China
| | - Cui-Cui Liu
- Department of Breast Surgery, Fudan University Shanghai Cancer Center and Cancer Institute, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, PR China
- Key Laboratory of Breast Cancer in Shanghai, Shanghai, 200032, PR China
| | - Ying-Ying Xu
- Department of Breast Surgery, The First Affiliated Hospital of China Medical University, Shenyang, 110001, PR China
| | - Zhi-Ming Shao
- Department of Breast Surgery, Fudan University Shanghai Cancer Center and Cancer Institute, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, PR China
- Key Laboratory of Breast Cancer in Shanghai, Shanghai, 200032, PR China
| | - Ke-Da Yu
- Department of Breast Surgery, Fudan University Shanghai Cancer Center and Cancer Institute, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, PR China
- Key Laboratory of Breast Cancer in Shanghai, Shanghai, 200032, PR China
| |
Collapse
|
45
|
Li J, Lv W, Han Z, Li Y, Deng J, Huang Y, Wan S, Sun J, Dai B. Mitoxantrone-Encapsulated ZIF-8 Enhances Chemo-Immunotherapy via Amplified Immunogenic Cell Death. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2501542. [PMID: 39950857 PMCID: PMC11984868 DOI: 10.1002/advs.202501542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Indexed: 04/12/2025]
Abstract
Chemo-immunotherapy, combining systemic chemotherapeutic drugs and immune checkpoint blockers, is a promising paradigm in cancer treatment. However, challenges such as limited induction of immune responses and systemic immune toxicity have hindered its clinical applications. Here, a zeolite imidazolate framework-8 (ZIF-8) that encapsulates mitoxantrone (MIT), an immune cell death (ICD)-inducing chemotherapeutic agent (MIT@ZIF-8), is synthesized using a one-pot aqueous-phase process. ZIF-8 serves as a dual-functional nanomaterial for chemo-immunotherapy: a carrier to enhance tumor uptake of MIT for improved chemotherapy efficacy, and a pyroptosis inducer to amplify MIT-induced ICD for augmented anti-tumor immune responses. As a result, in vivo administration of MIT@ZIF-8 markedly inhibits tumor growth in both immunologically "hot" colon cancer and immunologically "cold" prostate cancer. Moreover, MIT@ZIF-8 treatment increases the abundance of cytotoxic CD8+ T cells and reduces the amount of immunosuppressive regulatory T cells in tumors, thereby enhancing anti-tumor immunity and sensitizing prostate cancer to anti-CTLA-4 immunotherapy. In summary, MIT@ZIF-8 offers a highly translational approach for chemo-immunotherapy.
Collapse
Affiliation(s)
- Junhong Li
- Department of UrologyFudan University Shanghai Cancer CenterShanghai200032P. R. China
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032P. R. China
- Beijing Engineering Research Center for BioNanotechnologyCAS Key Laboratory of Standardization and Measurement for NanotechnologyNational Center for Nanoscience and TechnologyBeijing100190P. R. China
| | - Wenxing Lv
- Beijing Engineering Research Center for BioNanotechnologyCAS Key Laboratory of Standardization and Measurement for NanotechnologyNational Center for Nanoscience and TechnologyBeijing100190P. R. China
- School of Future TechnologyUniversity of Chinese Academy of SciencesBeijing100049P. R. China
| | - Ziwei Han
- Beijing Engineering Research Center for BioNanotechnologyCAS Key Laboratory of Standardization and Measurement for NanotechnologyNational Center for Nanoscience and TechnologyBeijing100190P. R. China
- School of Future TechnologyUniversity of Chinese Academy of SciencesBeijing100049P. R. China
| | - Yike Li
- Beijing Engineering Research Center for BioNanotechnologyCAS Key Laboratory of Standardization and Measurement for NanotechnologyNational Center for Nanoscience and TechnologyBeijing100190P. R. China
| | - Jinqi Deng
- Beijing Engineering Research Center for BioNanotechnologyCAS Key Laboratory of Standardization and Measurement for NanotechnologyNational Center for Nanoscience and TechnologyBeijing100190P. R. China
- School of Future TechnologyUniversity of Chinese Academy of SciencesBeijing100049P. R. China
| | - Yanjuan Huang
- Beijing Engineering Research Center for BioNanotechnologyCAS Key Laboratory of Standardization and Measurement for NanotechnologyNational Center for Nanoscience and TechnologyBeijing100190P. R. China
- School of Future TechnologyUniversity of Chinese Academy of SciencesBeijing100049P. R. China
| | - Shuo Wan
- Foundation for Applied Molecular EvolutionAlachuaFlorida32615US
| | - Jiashu Sun
- Beijing Engineering Research Center for BioNanotechnologyCAS Key Laboratory of Standardization and Measurement for NanotechnologyNational Center for Nanoscience and TechnologyBeijing100190P. R. China
- School of Future TechnologyUniversity of Chinese Academy of SciencesBeijing100049P. R. China
| | - Bo Dai
- Department of UrologyFudan University Shanghai Cancer CenterShanghai200032P. R. China
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032P. R. China
| |
Collapse
|
46
|
Chen YC, Dolladille C, Rao A, Palaskas NL, Deswal A, Lehmann L, Cautela J, Courand PY, Hayek S, Zhu H, Cheng RK, Alexandre J, Baldassarre LA, Roubille F, Laufer-Perl M, Asnani A, Ederhy S, Tamura Y, Francis S, Gaughan EM, Johnson DB, Flint DL, Rainer PP, Bailly G, Ewer SM, Aras MA, Arangalage D, Cariou E, Florido R, Peretto G, Itzhaki Ben Zadok O, Akhter N, Narezkina A, Levenson JE, Liu Y, Crusz SM, Issa N, Piriou N, Leong D, Sandhu S, Turker I, Moliner P, Obeid M, Heinzerling L, Chang WT, Stewart A, Venkatesh V, Du Z, Yadavalli A, Kim D, Chandra A, Zhang KW, Power JR, Moslehi J, Salem JE, Zaha VG. Immune Checkpoint Inhibitor Myocarditis and Left Ventricular Systolic Dysfunction. JACC CardioOncol 2025; 7:234-248. [PMID: 40246381 PMCID: PMC12046861 DOI: 10.1016/j.jaccao.2025.01.020] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 01/20/2025] [Accepted: 01/21/2025] [Indexed: 04/19/2025] Open
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) have transformed cancer treatment, but ICI myocarditis (ICI-M) remains a potentially fatal complication. The clinical implications and predictors of left ventricular ejection fraction (LVEF) <50% in ICI-M are not well understood. OBJECTIVES The aim of this study was to identify factors associated with LVEF <50% vs ≥50% at the time of hospitalization for ICI-M. A secondary objective was to evaluate the relationship between LVEF and 30-day all-cause mortality. METHODS The International ICI-Myocarditis Registry, a retrospective, international, multicenter database, included 757 patients hospitalized with ICI-M. Patients were stratified by LVEF as reduced LVEF (<50%) or preserved LVEF (≥50%) on admission. Cox proportional hazards models were used to assess the associations between LVEF and clinical events, and multivariable logistic regression was conducted to examine factors linked to LVEF. RESULTS Of 757 patients, 707 had documented LVEFs on admission: 244 (35%) with LVEF <50% and 463 (65%) with LVEF ≥50%. Compared with patients with LVEF ≥50%, those with LVEF <50% were younger (<70 years), had a body mass index of <25 kg/m2, and were more likely to have received chest radiation (24.2% vs 13.5%; P < 0.001). Multivariable analysis identified predictors of LVEF <50%, including exposure to v-raf murine sarcoma viral oncogene homolog B1/mitogen-activated protein kinase inhibitors, pre-existing heart failure, dyspnea at presentation, and at least 40 days from ICI initiation to ICI-M onset. Conversely, myositis symptoms were associated with LVEF ≥50%. LVEF <50% was marginally associated with 30-day all-cause mortality (unadjusted log-rank P = 0.062; adjusted for age, cancer types, and ICI therapy, HR: 1.50; 95% CI: 1.02-2.20). CONCLUSIONS Dyspnea, time from ICI initiation, a history of heart failure, and prior cardiotoxic therapy may be predictors of an initial LVEF <50% in patients with ICI-M.
Collapse
Affiliation(s)
- Yen-Chou Chen
- Division of Cardiology, University of California-San Francisco, San Francisco, California, USA; Division of Cardiology and Cardiovascular Research Center, Taipei Medical University Hospital, Taipei, Taiwan; Taipei Heart Institute, Taipei Medical University, Taipei, Taiwan.
| | - Charles Dolladille
- Normandie Université, UNICAEN, INSERM U1086 ANTICIPE, Caen, France; Caen-Normandy University Hospital, PICARO Cardio-Oncology Program, Department of Pharmacology, Caen, France; Department of Pharmacology, Sorbonne University, INSERM, CIC-1901, Hôpital Pitié-Salpétrière, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Anjali Rao
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA; Division of Cardiology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Nicolas L Palaskas
- Department of Cardiology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Anita Deswal
- Department of Cardiology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Lorenz Lehmann
- Department of Cardiology, Angiology, and Pneumology, University Hospital of Heidelberg, Heidelberg, Germany; German Centre for Cardiovascular Research, partner site Heidelberg/Mannheim, Mannheim, Germany; German Cancer Research Center, Heidelberg, Germany
| | - Jennifer Cautela
- Department of Cardiology, University Mediterranean Centre of CardioOncology, Nord Hospital, Assistance Publique-Hôpitaux de Marseille, Aix-Marseille University, INSERM, INRAE, C2VN, Marseille, France
| | - Pierre-Yves Courand
- Fédération de Cardiologie, Hôpital de la Croix-Rousse et Hôpital Lyon Sud, Hospices Civils de Lyon, Lyon, France; Université de Lyon, Université Claude Bernard, Lyon, France
| | - Salim Hayek
- Department of Cardiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Han Zhu
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, California; Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, California, USA
| | - Richard K Cheng
- Division of Cardiology, University of Washington, Seattle, Washington, USA
| | - Joachim Alexandre
- Normandie Université, UNICAEN, INSERM U1086 ANTICIPE, Caen, France; Caen-Normandy University Hospital, PICARO Cardio-Oncology Program, Department of Pharmacology, Caen, France
| | | | - François Roubille
- Department of Cardiology, INI-CRT, CHU de Montpellier, PhyMedExp, Université de Montpellier, Inserm, CNRS, Montpellier, France
| | - Michal Laufer-Perl
- Department of Cardiology, Tel Aviv Sourasky Medical Center, affiliated to the Tel Aviv University Faculty of Medicine, Tel Aviv-Yafo, Israel
| | - Aarti Asnani
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Stephane Ederhy
- Cardiology Department, Hospital Saint Antoine, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Yuichi Tamura
- Cardiovascular Center, International University of Health and Welfare, Mita Hospital, Tokyo, Japan
| | - Sanjeev Francis
- Cardiovascular Service Line, Maine Medical Center, Portland, Maine, USA
| | - Elizabeth M Gaughan
- Division of Hematology and Oncology, University of Virginia, Charlottesville, Virginia, USA
| | - Douglas B Johnson
- Division of Hematology and Oncology, Vanderbilt University, Nashville, Tennessee, USA
| | - Danette L Flint
- Heart and Vascular Center, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire, USA
| | - Peter P Rainer
- Division of Cardiology, Medical University of Graz, Graz Austria; BioTechMed Graz, Graz, Austria; St. Johann in Tirol General Hospital, St. Johann in Tirol, Austria
| | - Guillaume Bailly
- Assistance Publique-Hôpitaux de Paris Hôpital Lariboisière, Paris, France
| | - Steven M Ewer
- Division of Cardiovascular Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Mandar A Aras
- Division of Cardiology, University of California-San Francisco, San Francisco, California, USA
| | - Dimitri Arangalage
- Department of Cardiology, Bichat Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France; Université de Paris, UMRS1148, INSERM, Paris, France; Université de Paris, Paris, France
| | - Eve Cariou
- Department of Cardiology, Rangueil University Hospital, Toulouse, France
| | - Roberta Florido
- Division of Cardiology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Giovanni Peretto
- Disease Unit for Myocarditis and Arrhythmogenic Cardiomyopathies, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | | | - Nausheen Akhter
- Division of Cardiology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Anna Narezkina
- Division of Cardiovascular Medicine, University of California-San Diego, San Diego, California, USA
| | - Joshua E Levenson
- Heart and Vascular Institute, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Yan Liu
- Division of Cardiology, Department of Internal Medicine, Dell Medical School, University of Texas at Austin, Austin, Texas, USA
| | - Shanthini M Crusz
- Barts Health NHS Trust, University College London Hospital, London, United Kingdom
| | - Nahema Issa
- Bordeaux University Hospital, Bordeaux, France
| | - Nicolas Piriou
- Nantes Univesrité, CHU Nantes, Centre de Reference Cardiomyopathies, l'Institut du Thorax, Nantes, France
| | - Darryl Leong
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Shahneen Sandhu
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Isik Turker
- Department of Cardiology, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - Pedro Moliner
- Bellvitge University Hospital, Catalan Institute of Oncology, Cardiology Department, Bio-Heart Cardiovascular Diseases Research Group, Bellvitge Biomedical Research Institute, CIBER CV, l'Hospitalet de Llobregat, Barcelona, Spain
| | - Michel Obeid
- Centre Hospitalier Universitaire Vaudois, University of Lausanne, LCIT Center, Immunology and Allergy Service, Lausanne, Switzerland
| | - Lucie Heinzerling
- Department of Dermatology and Allergy, University Hospital, LMU Munich, Germany
| | | | - Andrew Stewart
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Vishnu Venkatesh
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Zoe Du
- University of Texas at Dallas, Dallas, Texas, USA
| | | | - Dohyeong Kim
- University of Texas at Dallas, Dallas, Texas, USA
| | - Alvin Chandra
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Kathleen W Zhang
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - John R Power
- Division of Cardiology, University of California-San Francisco, San Francisco, California, USA; Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, California
| | - Javid Moslehi
- Division of Cardiology, University of California-San Francisco, San Francisco, California, USA
| | - Joe-Elie Salem
- Department of Pharmacology, Sorbonne University, INSERM, CIC-1901, Hôpital Pitié-Salpétrière, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Vlad G Zaha
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA.
| |
Collapse
|
47
|
Liu G, Quan Q, Pan L, Duan H, Zhang G, Li K, Zhu X, Zhang D, Li P, Zhao J. Retinoic acid enhances γδ T cell cytotoxicity in nasopharyngeal carcinoma by reversing immune exhaustion. Cell Commun Signal 2025; 23:156. [PMID: 40158172 PMCID: PMC11955114 DOI: 10.1186/s12964-025-02161-8] [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: 11/10/2024] [Accepted: 03/19/2025] [Indexed: 04/01/2025] Open
Abstract
Recent studies have shown that the antitumor immunity of adaptive immune cells is regulated by Vitamin A (retinoic acid, RA). However, it remains unclear whether RA and retinoic acid receptor (RAR) signaling can modulate antitumor immunity by reversing immune exhaustion of innate-like γδ T cells in human nasopharyngeal carcinoma (NPC). Periphery blood samples from patients with NPC were prospectively collected, and phenotypic and functional analyses of γδ T cells were performed using flow cytometry. Tumor-bearing models and RAR inhibitor approaches were utilized to investigate RA/RAR-mediated regulation of T cell immunoglobulin domain and mucin domain 3 (Tim-3) and the antitumor activity of γδ T cells. Here, our findings indicate that immune exhaustion markers are highly expressed on peripheral αβ and γδ T cells in NPC patients. Serum RA levels are negatively correlated with the abundance of Tim-3 on circulating Vδ2 T cells. Mechanistic studies have demonstrated that RA/RAR signaling directly targets Vδ2 T cells, repressing Tim-3 expression, promoting NF-κB activation, and enhancing the production of antitumor-related cytokines. Notably, RA supplementation improved the efficacy of Vδ2 T cell-mediated immunotherapy in human NPC by suppressing Tim-3 expression. Collectively, these findings suggest that RA/RAR signaling plays a crucial role in reversing immune exhaustion and represents a promising target for γδ T cell antitumor immunotherapy.
Collapse
Affiliation(s)
- Guichao Liu
- The First Affiliated Hospital, Jinan University, Guangzhou, China
- Department of Radiation Oncology, The First People'S Hospital of Foshan City, Foshan, China
- Department of Oncology, Research Center of Cancer Diagnosis and Therapy, The First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Qiang Quan
- The First Affiliated Hospital, Jinan University, Guangzhou, China
- Department of Oncology, Research Center of Cancer Diagnosis and Therapy, The First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Lanhong Pan
- The First Affiliated Hospital, Jinan University, Guangzhou, China
- Department of Oncology, Research Center of Cancer Diagnosis and Therapy, The First Affiliated Hospital, Jinan University, Guangzhou, China
- Department of Ultrasound Medicine, The First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Haibo Duan
- The First Affiliated Hospital, Jinan University, Guangzhou, China
- Department of Oncology, Research Center of Cancer Diagnosis and Therapy, The First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Guojun Zhang
- The First Affiliated Hospital, Jinan University, Guangzhou, China
- Department of Oncology, Research Center of Cancer Diagnosis and Therapy, The First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Ke Li
- Department of Geriatrics, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Xinhai Zhu
- The First Affiliated Hospital, Jinan University, Guangzhou, China.
- Department of Oncology, Research Center of Cancer Diagnosis and Therapy, The First Affiliated Hospital, Jinan University, Guangzhou, China.
| | - Dongdong Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital, Jinan University, Guangzhou, China.
| | - Peng Li
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People'S Hospital Affiliated with Jinan University, Jinan University, Zhuhai, China.
- Faculty of Medical Science, The Biomedical Translational Research Institute, Jinan University, Guangzhou, China.
| | - Jianfu Zhao
- The First Affiliated Hospital, Jinan University, Guangzhou, China.
- Department of Oncology, Research Center of Cancer Diagnosis and Therapy, The First Affiliated Hospital, Jinan University, Guangzhou, China.
| |
Collapse
|
48
|
Ren W, Zhang H, Li Y, Sun W, Peng H, Guo H, Hou T, Wang M, Hu Z, Wu T, Liu B. Efficacy and safety of PD-1/PD-L1 inhibitors as first-line treatment for esophageal squamous cell carcinoma: a systematic review and meta-analysis. Front Immunol 2025; 16:1563300. [PMID: 40207226 PMCID: PMC11979238 DOI: 10.3389/fimmu.2025.1563300] [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: 01/19/2025] [Accepted: 03/07/2025] [Indexed: 04/11/2025] Open
Abstract
Purpose This study aims to investigate the efficacy and safety of PD-1/PD-L1 inhibitors in the first-line treatment of esophageal squamous cell carcinoma (ESCC) and identify factors influencing efficacy through a meta-analysis of multiple phase 3 randomized controlled trials (RCTs). Methods A systematic literature search was conducted in Cochrane, PubMed, and Embase databases. Two researchers independently extracted trial data, including efficacy-related outcomes such as overall survival (OS), progression-free survival (PFS), objective response rate (ORR), and duration of response (DoR), along with their subgroup data and safety-related indicators. The overall hazard ratio (HR) and 95% confidence interval (CI) were calculated for OS and PFS, while the overall odds ratio (OR) and 95% CI were computed for ORR to compare the classification and predictive abilities of combined positive score (CPS) and tumor proportion score (TPS) for PD-L1 status. Additionally, survival outcomes across different subgroups were evaluated to explore the potential influencing factors for the efficacy of PD-1/PD-L1 inhibitors in ESCC. Results This meta-analysis included eight phase 3 RCTs encompassing 4,479 participants. PD-1/PD-L1 inhibitors combined with chemotherapy significantly improved OS (HR: 0.68, 95% CI: 0.63-0.74) and PFS (HR: 0.62, 95% CI: 0.58-0.67) in ESCC patients compared to non-combination therapy. Patients with higher PD-L1 expression (CPS>1 or TPS>1) demonstrated superior responses to PD-1/PD-L1 inhibitions, with CPS identified as a stronger predictor of therapeutic benefit, particularly at a threshold of CPS =10. Subgroup analysis revealed that male, Asian, smoking, and liver metastasis patients exhibited a greater trend toward improved disease control with PD-1/PD-L1 inhibitors. However, there was no significant difference in treatment efficacy between immune therapy combined with TP (taxol [paclitaxel] + cisplatin) and FP (5-fluorouracil [5-FU] + cisplatin) regimens (POS =0.51, PPFS =0.11). Finally, PD-1/PD-L1 inhibition was associated with a higher incidence of grade ≥3 adverse events compared to chemotherapy alone (HR: 1.21, 95% CI: 1.07-1.37). Conclusions This study confirms that the combination of PD-1/PD-L1 inhibitors and chemotherapy provides significant clinical benefits in ESCC. CPS =10 serves as a key threshold for predicting treatment response. There is a trend suggesting that male, Asian, smoking, and liver metastasis patients may experience better survival benefits, while no significant difference was observed between TP- and FP-based regimens. Systematic Review Registration https://www.crd.york.ac.uk/prospero, identifier CRD42024536221.
Collapse
Affiliation(s)
- Wei Ren
- The Comprehensive Cancer Center of Drum Tower Hospital, Medical School of Nanjing University and Clinical Cancer Institute of Nanjing University, Nanjing, China
| | - Hanyu Zhang
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
| | - Yixin Li
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
| | - Wu Sun
- The Comprehensive Cancer Center of Drum Tower Hospital, Medical School of Nanjing University and Clinical Cancer Institute of Nanjing University, Nanjing, China
| | - Hexiang Peng
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
| | - Huangda Guo
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
| | - Tianjiao Hou
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
| | - Mengying Wang
- Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing, China
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing, China
| | - Zhendong Hu
- Department of Esophageal Surgery, Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Tao Wu
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
- Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing, China
| | - Baorui Liu
- The Comprehensive Cancer Center of Drum Tower Hospital, Medical School of Nanjing University and Clinical Cancer Institute of Nanjing University, Nanjing, China
| |
Collapse
|
49
|
Ren X, Guo A, Geng J, Chen Y, Wang X, Zhou L, Shi L. Pan-cancer analysis of co-inhibitory molecules revealing their potential prognostic and clinical values in immunotherapy. Front Immunol 2025; 16:1544104. [PMID: 40196117 PMCID: PMC11973099 DOI: 10.3389/fimmu.2025.1544104] [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: 12/12/2024] [Accepted: 03/03/2025] [Indexed: 04/09/2025] Open
Abstract
Background The widespread use of immune checkpoint inhibitors (anti-CTLA4 or PD-1) has opened a new chapter in tumor immunotherapy by providing long-term remission for patients. Unfortunately, however, these agents are not universally available and only a minority of patients respond to them. Therefore, there is an urgent need to develop novel therapeutic strategies targeting other co-inhibitory molecules. However, comprehensive information on the expression and prognostic value of co-inhibitory molecules, including co-inhibitory receptors and their ligands, in different cancers is not yet available. Methods We investigated the expression, correlation, and prognostic value of co-inhibitory molecules in different cancer types based on TCGA, UCSC Xena, TIMER, CellMiner datasets. We also examined the associations between the expression of these molecules and the extent of immune cell infiltration. Besides, we conducted a more in-depth study of VISTA. Result The results of differential expression analysis, correlation analysis, and drug sensitivity analysis suggest that CTLA4, PD-1, TIGIT, LAG3, TIM3, NRP1, VISTA, CD80, CD86, PD-L1, PD-L2, PVR, PVRL2, FGL1, LGALS9, HMGB1, SEMA4A, and VEGFA are associated with tumor prognosis and immune cell infiltration. Therefore, we believe that they are hopefully to serve as prognostic biomarkers for certain cancers. In addition, our analysis indicates that VISTA plays a complex role and its expression is related to TMB, MSI, cancer cell stemness, DNA/RNA methylation, and drug sensitivity. Conclusions These co-inhibitory molecules have the potential to serve as prognostic biomarkers and therapeutic targets for a broad spectrum of cancers, given their strong associations with key clinical metrics. Furthermore, the analysis results indicate that VISTA may represent a promising target for cancer therapy.
Collapse
Affiliation(s)
- Xiaoyu Ren
- School of Life Sciences, Chongqing University, Chongqing, China
| | - Anjie Guo
- School of Life Sciences, Chongqing University, Chongqing, China
| | - Jiahui Geng
- School of Life Sciences, Chongqing University, Chongqing, China
| | - Yuling Chen
- School of Life Sciences, Chongqing University, Chongqing, China
| | - Xue Wang
- School of Life Sciences, Chongqing University, Chongqing, China
| | - Lian Zhou
- Department of Head&Neck Cancer Center, Chongqing University Cancer Hospital, Chongqing, China
| | - Lei Shi
- School of Life Sciences, Chongqing University, Chongqing, China
| |
Collapse
|
50
|
Cannet F, Sequera C, Veloso PM, El Kaoutari A, Methia M, Richelme S, Kaya M, Cherni A, Dupont M, Borg JP, Morel C, Boursier Y, Maina F. Tracing specificity of immune landscape remodeling associated with distinct anticancer treatments. iScience 2025; 28:112071. [PMID: 40124507 PMCID: PMC11930375 DOI: 10.1016/j.isci.2025.112071] [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: 03/03/2024] [Revised: 07/18/2024] [Accepted: 02/10/2025] [Indexed: 03/25/2025] Open
Abstract
Immune cells within the tumor microenvironment impact cancer progression, resistance, response to treatments. Despite remarkable outcomes for some cancer patients, immunotherapies remain unsatisfactory for others. Here, we designed an experimental setting using the Alb-R26 Met "inside-out" mouse model, faithfully recapitulating molecular features of liver cancer patients, to explore the effects of distinct anticancer targeted therapies on the tumor immune landscape. Using two treatments in clinical trials for different cancer types, Decitabine and MEK+BCL-XL blockage, we show their capability to trigger tumor regression in Alb-R26 Met mice and to superimpose distinct profiles of immune cell types and immune-checkpoints, impacting immunotherapy response. A machine learning approach processing tumor imaging and immune profile data identified a putative signature predicting tumor treatment response in mice and patients. Outcomes exemplify how the tumor immune microenvironment is differentially reshaped by distinct anticancer agents and highlight the importance of measuring its modulation during treatment to optimize oncotherapy and immunotherapy combinations.
Collapse
Affiliation(s)
- Floriane Cannet
- Aix Marseille Univ, CNRS/IN2P3, CPPM, 13009 Marseille, France
- Aix Marseille Univ, CNRS, Inserm, Institut Paoli-Calmettes, Centre de Recherche en Cancérologie de Marseille (CRCM), 13009 Marseille, France
- Aix Marseille Univ, CNRS, Developmental Biology Institute of Marseille (IBDM), Turing Center for Living Systems, 13009 Marseille, France
| | - Célia Sequera
- Aix Marseille Univ, CNRS, Inserm, Institut Paoli-Calmettes, Centre de Recherche en Cancérologie de Marseille (CRCM), 13009 Marseille, France
- Aix Marseille Univ, CNRS, Developmental Biology Institute of Marseille (IBDM), Turing Center for Living Systems, 13009 Marseille, France
| | - Paula Michea Veloso
- Aix Marseille Univ, CNRS, Inserm, Institut Paoli-Calmettes, Centre de Recherche en Cancérologie de Marseille (CRCM), 13009 Marseille, France
| | - Abdessamad El Kaoutari
- Aix Marseille Univ, CNRS, Inserm, Institut Paoli-Calmettes, Centre de Recherche en Cancérologie de Marseille (CRCM), 13009 Marseille, France
| | - Melissa Methia
- Aix Marseille Univ, CNRS, Inserm, Institut Paoli-Calmettes, Centre de Recherche en Cancérologie de Marseille (CRCM), 13009 Marseille, France
| | - Sylvie Richelme
- Aix Marseille Univ, CNRS, Developmental Biology Institute of Marseille (IBDM), Turing Center for Living Systems, 13009 Marseille, France
| | - Muge Kaya
- Aix Marseille Univ, CNRS, Inserm, Institut Paoli-Calmettes, Centre de Recherche en Cancérologie de Marseille (CRCM), 13009 Marseille, France
| | - Afef Cherni
- Aix Marseille Univ, CNRS/IN2P3, CPPM, 13009 Marseille, France
| | - Mathieu Dupont
- Aix Marseille Univ, CNRS/IN2P3, CPPM, 13009 Marseille, France
| | - Jean-Paul Borg
- Aix Marseille Univ, CNRS, Inserm, Institut Paoli-Calmettes, Centre de Recherche en Cancérologie de Marseille (CRCM), 13009 Marseille, France
- Institut Universitaire de France, Paris, France
| | - Christian Morel
- Aix Marseille Univ, CNRS/IN2P3, CPPM, 13009 Marseille, France
| | | | - Flavio Maina
- Aix Marseille Univ, CNRS, Inserm, Institut Paoli-Calmettes, Centre de Recherche en Cancérologie de Marseille (CRCM), 13009 Marseille, France
- Aix Marseille Univ, CNRS, Developmental Biology Institute of Marseille (IBDM), Turing Center for Living Systems, 13009 Marseille, France
| |
Collapse
|