1
|
Wang S, Liang X, Li H, Zou J, Xu L, Zhang Y, Lin J, Zeng J, Zhong X, Liu X, Liu Z, Zheng Y, Nie M, Yang L. The NET-DNA-CCDC25 inhibitor di-Pal-MTO suppresses tumor progression and promotes the innate immune response. Cell Mol Immunol 2025; 22:628-644. [PMID: 40229592 DOI: 10.1038/s41423-025-01286-7] [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: 08/02/2024] [Revised: 03/16/2025] [Accepted: 03/27/2025] [Indexed: 04/16/2025] Open
Abstract
The DNA component of neutrophil extracellular traps (NET-DNA) is associated with cancer metastasis and chemotherapy resistance. However, recent studies have suggested that NET-DNA contributes to the activation of dendritic cells (DCs) and promotes the innate immune response to anticancer immunity. Therefore, exploring therapeutic approaches to inhibit NET-mediated tumor progression while maintaining antitumor immunity is essential. Our groups recently identified CCDC25 as a specific NET-DNA sensor on the cytoplasmic membrane of cancer cells that promotes cancer metastasis. In this study, we performed small-molecule compound screening and revealed that mitoxantrone (MTO) could block the interaction between NET-DNA and CCDC25. Molecular docking results indicated that MTO competed with NET-DNA by binding with the amino acid residues Tyr24 (Y24), Glu25 (E25), and Asp28 (D28) of the crystal structure of CCDC25. More importantly, we conjugated MTO with palmitoleic acids such as di-Pal-MTO to increase its residence time on the cytoplasmic membrane, which increased its inhibitory efficiency and decreased its cytotoxicity. In addition, di-Pal-MTO markedly inhibited the RAC1-CDC42 cascade to alleviate the NET-induced cytoskeleton arrangement and chemotactic migration of cancer cells. In multiple mouse models, di-Pal-MTO can suppress breast cancer metastasis and have synergistic effects with chemotherapeutics. Moreover, di-Pal-MTO promotes NET-DNA-dependent DC activation, leading to the subsequent expression of various chemokines that facilitate the infiltration of CD8+ T cells. Overall, we successfully identified a small molecule inhibitor, di-Pal-MTO, with dual effects on tumor repression and the antitumor immune response, which provides a novel therapeutic strategy against breast cancer.
Collapse
Affiliation(s)
- Shun Wang
- Breast Tumor Center, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
- Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Xinyan Liang
- Breast Tumor Center, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
| | - Heliang Li
- Breast Tumor Center, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
| | - Junying Zou
- Breast Tumor Center, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
| | - Linxi Xu
- Breast Tumor Center, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
| | - Yetong Zhang
- Breast Tumor Center, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
| | - Jianghua Lin
- Breast Tumor Center, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
| | - Jiayi Zeng
- Breast Tumor Center, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
| | - Xiaoming Zhong
- Breast Tumor Center, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
| | - Xu Liu
- Breast Tumor Center, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
| | - Zhou Liu
- Breast Tumor Center, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
| | - Yue Zheng
- Breast Tumor Center, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China.
- Guangdong Province Key Laboratory of Pharmaceutical Bioactive Substances, School of Bioscience and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, 510006, P. R. China.
| | - Man Nie
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, People's Republic of China.
| | - Linbin Yang
- Breast Tumor Center, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China.
| |
Collapse
|
2
|
Xu L, Schaefer KG, King GM, Xie ZR, Bartlett MG. Insights into interactions between taxanes and P-glycoprotein using biophysical and in silico methods. J Pharm Sci 2025; 114:103708. [PMID: 40015511 DOI: 10.1016/j.xphs.2025.103708] [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: 11/11/2024] [Revised: 02/20/2025] [Accepted: 02/20/2025] [Indexed: 03/01/2025]
Abstract
Multidrug resistance mediated by P-glycoprotein (Pgp) is a significant obstacle to cancer chemotherapy. Taxane drugs, including paclitaxel, docetaxel, and cabazitaxel, are used to treat multiple types of cancer. All taxane drugs are Pgp substrates, but cabazitaxel is also a Pgp inhibitor, indicating potential differential interactions between Pgp and different taxanes. Here, we showed for the first time that cabazitaxel had a partial inhibitory effect on the ATPase activity at concentrations higher than 10 µM. We found the KD of paclitaxel, docetaxel, and cabazitaxel to Pgp are 0.85 µM, 40.59 µM, and 13.53 µM, respectively. Based on acrylamide quenching, paclitaxel induced Pgp into a wide inward-facing open conformation at a high concentration but a slightly occluded conformation at lower concentrations. Both docetaxel and cabazitaxel shifted Pgp towards occluded states, each drug resulting in a unique degree of occlusion. Furthermore, molecular docking and energy calculations revealed that cabazitaxel binds with the "access tunnel" and blocks the subsequent nucleotide-binding domain dimerization. Our results indicate that the preference of taxanes for different binding sites on Pgp leads to distinct transport mechanisms. These results provide valuable insight into the interaction between taxanes and Pgp, which will enhance future drug development.
Collapse
Affiliation(s)
- Longwen Xu
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia, Athens, GA 30602, USA
| | - Katherine G Schaefer
- Department of Physics and Astronomy, University of Missouri, Columbia, MO 65211, USA
| | - Gavin M King
- Department of Physics and Astronomy, University of Missouri, Columbia, MO 65211, USA; Joint with Department of Biochemistry, University of Missouri, Columbia, MO 65211, USA
| | - Zhong-Ru Xie
- Computational Drug Discovery Laboratory, School of Electrical and Computer Engineering, College of Engineering, University of Georgia, Athens, GA 30602, USA
| | - Michael G Bartlett
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia, Athens, GA 30602, USA.
| |
Collapse
|
3
|
Liu H, Wu C, Yu H. Co(II)-coordination Complex: Fluorescence Performances and Combined with Taxotere-Hydrogels on Breast Cancer Treatment and Clinical Care. J Fluoresc 2025; 35:2855-2864. [PMID: 38647959 DOI: 10.1007/s10895-024-03734-5] [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: 02/03/2024] [Accepted: 04/17/2024] [Indexed: 04/25/2024]
Abstract
In this study, a novel coordination polymer {Co2(Oaobtc)(bpe)(H2O)4]}n (1) was synthesized under hydrothermal conditions using a hybrid ligand synthesis method, where H4Oobtc represents 2,3,3'-tricarboxylate azobenzene, and bpe represents 1,2-bis(4-pyridyl)ethylene. The obtained CP1 was characterized by elemental analysis (EA), powder X-ray diffraction (PXRD), and thermal gravimetric analysis (TGA). Fluorescence testing confirmed the excellent photoluminescent performance of compound 1, indicating its potential as a cyan-emitting fluorescent material. Hyaluronic acid (HA) and carboxymethyl chitosan (CMCS) are natural polysaccharides known for their biocompatibility. HA/CMCS hydrogels were synthesized using a chemical synthesis method, featuring a three-dimensional network structure with interconnected pores, and an average pore size of 314.75 ± 11.25 μm. The characterization of the taxotere-loaded hydrogel was performed using infrared spectroscopy, confirming the effective encapsulation of the drug within the hydrogel. Utilizing taxotere as a model drug, a novel taxotere-loaded metal gel was synthesized, and its anticancer efficacy was evaluated. Furthermore, the influence of different pH levels on drug release rate was investigated. Finally, the encapsulation and release of taxotere in the hydrogel were studied using UV-visible spectroscopy.
Collapse
Affiliation(s)
- Hao Liu
- Department of Breast and Thyroid Surgery, Bishan Maternity and Child Hospital of Chongqing, Chongqing, China
| | - Chengyi Wu
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Haiying Yu
- Department of Oncology, Chongqing Hygeia Hospital, Chongqing, China.
| |
Collapse
|
4
|
Belmonte B, Spada S, Allavena P, Benelli M, Bronte V, Casorati G, D'Ambrosio L, Ferrara R, Mondino A, Nisticò P, Sommaggio R, Tazzari M, Tripodo C, Sica A, Ferrucci PF. Highlighting recent achievements to advance more effective cancer immunotherapy. J Exp Clin Cancer Res 2025; 44:57. [PMID: 39966867 PMCID: PMC11834592 DOI: 10.1186/s13046-025-03316-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Accepted: 02/04/2025] [Indexed: 02/20/2025] Open
Abstract
From 17 to 19th October 2024, the XXI Italian Network for Bio-Immunotherapy of Tumors Meeting (NIBIT) took place in Palermo, in the marvelous historical location of Teatro Politeama, under the auspices of the Italian Association of Medical Oncology (AIOM), Italian Association of Cancer Research (AIRC), Fondazione Pezcoller, Italian Alliance against Cancer (ACC), Italian Lymphoma Foundation (FIL), Grazia Focacci Foundation and Melagioco Foundation. The conference covered a spectrum of topics ranging from target discovery to therapeutic advances in immuno-oncology, bringing world-renowned experts to present groundbreaking innovations in basic, translational, and clinical cancer research. Six sessions focused on cellular therapies, digital pathology, vaccines, tertiary lymphoid structures, and microenvironment in order to get deep insights on how to personalize diagnosis and therapies in the clinical setting. Young investigators had the opportunity to meet and greet their mentors, promoting the synergy of the academic and industrial sectors within the national and international panorama, discussing the application of artificial intelligence on multi-specific antibodies, drug conjugates, and antibody fusion proteins that are advancing the efficacy of precision medicine and minimizing off-target effects.
Collapse
Affiliation(s)
- Beatrice Belmonte
- Tumor Immunology Unit, Departmentof Health Sciences, University of Palermo, Palermo, Italy
| | - Sheila Spada
- Tumor Immunology and Immunotherapy Unit, IRCCS-Regina Elena National Cancer Institute, Rome, Italy
| | | | | | | | - Giulia Casorati
- Experimental Immunology Unit, DivisionofImmunology,TransplantationandInfectiousDiseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | | | - Roberto Ferrara
- Department of Medical Oncology, ScientificInstituteforResearch,HospitalizationandHealthcare(IRCCS), San Raffaele Scientific Institute, Milan, Italy
| | - Anna Mondino
- Lymphocyte Activation Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Paola Nisticò
- Tumor Immunology and Immunotherapy Unit, IRCCS-Regina Elena National Cancer Institute, Rome, Italy
| | - Roberta Sommaggio
- Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padua, Italy
| | - Marcella Tazzari
- Advanced Cellular Therapies and Rare Tumors Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori" S.r.l., Meldola, Italy
| | - Claudio Tripodo
- IFOM ETS, The AIRC Institute of Molecular Oncology, Milan, Italy
- Department of Oncology and Heamato-Oncology, University of Milan, Milan, Italy
| | - Antonio Sica
- Department of Pharmaceutical Sciences, University of Piemonte Orientale "A. Avogadro", Novara, Italy.
- IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy.
| | | |
Collapse
|
5
|
Perużyńska M, Birger R, Kłos P, Kwiecień H, Struk Ł, Sośnicki JG, Lafanechère L, Droździk M. The Co-Administration of Paclitaxel with Novel Pyridine and Benzofuran Derivatives that Inhibit Tubulin Polymerisation: A Promising Anticancer Strategy. Pharmaceutics 2025; 17:223. [PMID: 40006590 PMCID: PMC11859455 DOI: 10.3390/pharmaceutics17020223] [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: 01/14/2025] [Revised: 02/04/2025] [Accepted: 02/06/2025] [Indexed: 02/27/2025] Open
Abstract
Background: Paclitaxel (PTX), a crucial microtubule-stabilising agent in cancer treatment, is limited by its adverse effects and hydrophobic nature, which necessitate the use of toxic solvents. This study proposes a novel approach combining PTX with new microtubule-destabilising compounds at low, safe doses that are ineffective when used individually. Objective: The aim was to evaluate the therapeutic efficacy of combining PTX with previously described pyridine (S1, S22) and benzofuran derivatives (13b, 14), which have demonstrated promising anticancer properties by inhibiting microtubule polymerisation. Methods: The PrestoBlue assay was used to determine the optimal concentrations of each compound, enabling synergistic interactions with a low dose of PTX in HeLa cervical cancer cells. The combined effects of the compounds and PTX on apoptosis, cell cycle distribution, and mitotic spindle formation were then evaluated. Results: The results showed that compounds 13b (1 µM), 14 (0.1 µM), S1 (2 µM), and S22 (2 µM) enhanced the proapoptotic and antimitotic effects of 1 nM PTX, which was ineffective alone. Notably, live-cell imaging revealed that the concurrent use of S1 and PTX produced effects similar to those of a higher PTX concentration (5 nM). Conclusions: These findings suggest that these compounds enhance the anticancer efficacy of low-dose PTX, potentially paving the way for more effective and safer cancer therapies.
Collapse
Affiliation(s)
- Magdalena Perużyńska
- Department of Experimental and Clinical Pharmacology, Pomeranian Medical University in Szczecin, Powstanców Wielkopolskich 72, 70-111 Szczecin, Poland; (R.B.); (M.D.)
| | - Radosław Birger
- Department of Experimental and Clinical Pharmacology, Pomeranian Medical University in Szczecin, Powstanców Wielkopolskich 72, 70-111 Szczecin, Poland; (R.B.); (M.D.)
| | - Patrycja Kłos
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstanców Wielkopolskich 72, 70-111 Szczecin, Poland;
| | - Halina Kwiecień
- Faculty of Chemical Technology and Engineering, West Pomeranian University of Technology in Szczecin, Piastów 42, 71-065 Szczecin, Poland;
| | - Łukasz Struk
- Department of Organic and Physical Chemistry, Faculty of Chemical Technology and Engineering, West Pomeranian University of Technology in Szczecin, Piastów 42, 71-065 Szczecin, Poland; (Ł.S.); (J.G.S.)
| | - Jacek G. Sośnicki
- Department of Organic and Physical Chemistry, Faculty of Chemical Technology and Engineering, West Pomeranian University of Technology in Szczecin, Piastów 42, 71-065 Szczecin, Poland; (Ł.S.); (J.G.S.)
| | - Laurence Lafanechère
- Institute for Advanced Biosciences, University Grenoble Alpes, CNRS UMR 5309, INSERM U1209, F-38700 Grenoble, France;
| | - Marek Droździk
- Department of Experimental and Clinical Pharmacology, Pomeranian Medical University in Szczecin, Powstanców Wielkopolskich 72, 70-111 Szczecin, Poland; (R.B.); (M.D.)
| |
Collapse
|
6
|
Xiong K, Pan B, Fang H, Tao Z. Single-cell sequencing analysis reveals cancer-associated pericyte subgroup in esophageal squamous cell carcinoma to predict prognosis. Front Immunol 2025; 15:1474673. [PMID: 39835116 PMCID: PMC11743493 DOI: 10.3389/fimmu.2024.1474673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 12/05/2024] [Indexed: 01/22/2025] Open
Abstract
Background The role of cancer-associated pericytes (CAPs) in tumor microenvironment (TME) suggests that they are potential targets for cancer treatment. The mechanism of CAP heterogeneity in esophageal squamous cell carcinoma (ESCC) remains unclear, which has limited the development of treatments for tumors through CAPs. Therefore, a comprehensive understanding of the classification, function, cellular communication and spatial distribution of CAP subpopulations in ESCC is urgently needed. Methods This study used large-sample single-cell transcriptome sequencing (scRNA-seq) data to investigate pericytes' subpopulation characteristics, functions, upstream and downstream regulation and interactions with other components of the TME in the ESCC, and analyzed prognostically in conjunction with Bulk RNA-seq data. In addition, pericyte subpopulations were validated and their spatial distribution in the ESCC TME was observed by multiplex immunofluorescence. Drug prediction and molecular docking was further used to validate the medicinal value of drug targets. Results CAPs in the ESCC TME were found to be highly heterogeneous, and we identified six pericyte subtypes: c1_ARHGDIB, c2_BCAM, c3_LUM, c4_SOD2, c5_TYMS, and c6_KRT17, which have commonality in a part of their functions, and each of them has a major function to play, by having different strengths of interaction with different components in the TME. In addition, we found that c4_SOD2 was negatively correlated with prognosis, conversely, c5_TYMS was positively correlated with prognosis. The drug with a better effect on c5_TYMS was docetaxel (binding energy = -8.1, -8.7 kcal/mol); raloxifene may be more effective against c4_SOD2, although raloxifene has a slightly lower binding energy to SOD2 (-6.4 kcal/mol), it has a higher binding energy to PDGFRβ (-8.1 kcal/mol). Conclusion The present study identified and discovered pericyte subpopulations that were significantly associated with prognosis, which provides new biomarkers for predicting patient prognosis and adds usable targets for immunotherapy, and it is also important for gaining insights into the composition of the TME in ESCC.
Collapse
Affiliation(s)
- Kai Xiong
- Department of Cardiothoracic Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Bing Pan
- Tianjia Genomes Tech Cor. Ltd., Hefei, China
| | - Hao Fang
- Department of Cardiothoracic Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Ziyou Tao
- Department of Thoracic Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
7
|
Song C, Tong T, Dai B, Zhu Y, Chen E, Zhang M, Zhang W. Osteoimmunology in bone malignancies: a symphony with evil. JOURNAL OF THE NATIONAL CANCER CENTER 2024; 4:354-368. [PMID: 39735445 PMCID: PMC11674455 DOI: 10.1016/j.jncc.2024.09.001] [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: 04/08/2024] [Revised: 09/11/2024] [Accepted: 09/11/2024] [Indexed: 12/31/2024] Open
Abstract
Bone marrow is pivotal for normal hematopoiesis and immune responses, yet it is often compromised by malignancies. The bone microenvironment (BME), composed of bone and immune cells, maintains skeletal integrity and blood production. The emergence of primary or metastatic tumors in the skeletal system results in severe complications and contributes significantly to cancer-related mortality. These tumors set off a series of interactions among cancer, bone, and immune cells, and disrupt the BME locally or distantly. However, the drivers, participants, and underlying molecules of these interactions are not fully understood. This review explores the crosstalk between bone metabolism and immune responses, synthesizing current knowledge on the intersection of cancer and osteoimmune biology. It outlines how bone marrow immune cells can either facilitate or hinder tumor progression by interacting with bone cells and pinpoints the molecules responsible for immunosuppression within bone tumors. Moreover, it discusses how primary tumors remotely alter the BME, leading to systemic immune suppression in cancer patients. This knowledge provides critical rationales for emerging immunotherapies in the treatment of bone-related tumors. Taken together, by summarizing the intricate relationship between tumor cells and the BME, this review aims to deepen the understanding of the diversity, complexity, and dynamics at play during bone tumor progression. Ultimately, it highlights the potential of targeting bone-tumor interactions to correct aberrant immune functions, thereby inhibiting tumor growth and metastasis.
Collapse
Affiliation(s)
- Churui Song
- Department of Breast Surgery and Oncology, Cancer Institute, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Tie Tong
- Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Biqi Dai
- Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Yue Zhu
- Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Elina Chen
- College of Natural Sciences, University of Texas at Austin, 110 Inner Campus Drive, Austin, USA
| | - Min Zhang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Weijie Zhang
- Zhejiang Provincial Key Laboratory of Cancer Molecular Cell Biology, Life Sciences Institute, and Department of Orthopaedic Surgery, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
8
|
Ma X, Mao M, Liu Z, Liang C, He J, Qu Y, Xu L, Cheng R, Zhuang W, Lei Y, Nie W, Yuan L, Pang DW, Xie HY. AND-Gate Logic Förster Resonance Energy Transfer/Magnetic Resonance Tuning Nanoprobe for Programmable Antitumor Immunity Imaging. J Am Chem Soc 2024; 146:31873-31884. [PMID: 39504515 DOI: 10.1021/jacs.4c11072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2024]
Abstract
Simultaneous detection of different biomarkers related to the spatiotemporally dynamic immune events is of particular importance for the accurate evaluation of antitumor immune effects. Here, we have developed an AND-gate logic dual resonance energy transfer nanoprobe (named DRET) for dynamic monitoring of programmed CD8+ T cell activation and tumor cell apoptosis. Immunotherapy-induced granzyme B secretion from CD8+ T cells and the subsequent caspase-3 release from apoptotic tumor cells individually activate one of the tiers of the "AND-gate" logic DRET. The resulting fluorescence recovery and magnetic resonance T1 enhancement can be used for precise immunomodulatory drug screening, early efficacy prediction, and immune stratification. Particularly, not only "Responders" can be distinguished from "Non-responders", but also "Acquired resistance" can be identified from "Maintain responders", providing a novel approach to put forward the accurate evaluation of antitumor immunity.
Collapse
Affiliation(s)
- Xianbin Ma
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, P. R. China
| | - Mingchuan Mao
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, P. R. China
| | - Zhenya Liu
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Frontiers Science Center for New Organic Matter, Research Center for Analytical Sciences, College of Chemistry, Nankai University, Tianjin 300071, P. R. China
| | - Chao Liang
- School of Life Science, Beijing Institute of Technology, Beijing 100081, P. R. China
| | - Jiaqi He
- School of Life Science, Beijing Institute of Technology, Beijing 100081, P. R. China
| | - Yun Qu
- School of Life Science, Beijing Institute of Technology, Beijing 100081, P. R. China
| | - Luzheng Xu
- Medical and Health Analysis Center, Peking University, Beijing 100191, P. R. China
| | - Ran Cheng
- School of Life Science, Beijing Institute of Technology, Beijing 100081, P. R. China
| | - Wanru Zhuang
- State Key Laboratory of Natural and Biomimetic Drugs, Chemical Biology Center, School of Pharmaceutical Sciences, Peking University, Beijing 100191, P. R. China
| | - Yao Lei
- State Key Laboratory of Natural and Biomimetic Drugs, Chemical Biology Center, School of Pharmaceutical Sciences, Peking University, Beijing 100191, P. R. China
| | - Weidong Nie
- School of Life Science, Beijing Institute of Technology, Beijing 100081, P. R. China
| | - Lan Yuan
- Medical and Health Analysis Center, Peking University, Beijing 100191, P. R. China
| | - Dai-Wen Pang
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Frontiers Science Center for New Organic Matter, Research Center for Analytical Sciences, College of Chemistry, Nankai University, Tianjin 300071, P. R. China
| | - Hai-Yan Xie
- State Key Laboratory of Natural and Biomimetic Drugs, Chemical Biology Center, School of Pharmaceutical Sciences, Peking University, Beijing 100191, P. R. China
| |
Collapse
|
9
|
Cao C, Xu M, Wei Y, Peng T, Lin S, Liu X, Xu Y, Chu T, Liu S, Wu P, Hu B, Ding W, Li L, Ma D, Wu P. CXCR4 orchestrates the TOX-programmed exhausted phenotype of CD8 + T cells via JAK2/STAT3 pathway. CELL GENOMICS 2024; 4:100659. [PMID: 39317187 PMCID: PMC11602566 DOI: 10.1016/j.xgen.2024.100659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 06/17/2024] [Accepted: 08/22/2024] [Indexed: 09/26/2024]
Abstract
Evidence from clinical trials suggests that CXCR4 antagonists enhance immunotherapy effectiveness in several cancers. However, the specific mechanisms through which CXCR4 contributes to immune cell phenotypes are not fully understood. Here, we employed single-cell transcriptomic analysis and identified CXCR4 as a marker gene in T cells, with CD8+PD-1high exhausted T (Tex) cells exhibiting high CXCR4 expression. By blocking CXCR4, the Tex phenotype was attenuated in vivo. Mechanistically, CXCR4-blocking T cells mitigated the Tex phenotype by regulating the JAK2-STAT3 pathway. Single-cell RNA/TCR/ATAC-seq confirmed that Cxcr4-deficient CD8+ T cells epigenetically mitigated the transition from functional to exhausted phenotypes. Notably, clinical sample analysis revealed that CXCR4+CD8+ T cells showed higher expression in patients with a non-complete pathological response. Collectively, these findings demonstrate the mechanism by which CXCR4 orchestrates CD8+ Tex cells and provide a rationale for combining CXCR4 antagonists with immunotherapy in clinical trials.
Collapse
Affiliation(s)
- Canhui Cao
- Department of Gynecologic Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430199, China; Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Miaochun Xu
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China; Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430199, China; National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Ye Wei
- Department of Gynecologic Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430199, China; Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Ting Peng
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China; Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430199, China; National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Shitong Lin
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China; Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430199, China; National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Xiaojie Liu
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China; Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430199, China; National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Yashi Xu
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China; Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430199, China; National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Tian Chu
- Department of Gynecologic Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430199, China; Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Shiyi Liu
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China; Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430199, China; National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Ping Wu
- Department of Gynecologic Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430199, China; Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Bai Hu
- Department of Gynecologic Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430199, China; Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Wencheng Ding
- Department of Gynecologic Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430199, China; Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Li Li
- Department of Gynecologic Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430199, China; Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Ding Ma
- Department of Gynecologic Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430199, China; Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China.
| | - Peng Wu
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China; Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430199, China; National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China.
| |
Collapse
|
10
|
Yuan X, Hao X, Chan HL, Zhao N, Pedroza DA, Liu F, Le K, Smith AJ, Calderon SJ, Lieu N, Soth MJ, Jones P, Zhang XH, Rosen JM. CREB-binding protein/P300 bromodomain inhibition reduces neutrophil accumulation and activates antitumor immunity in triple-negative breast cancer. JCI Insight 2024; 9:e182621. [PMID: 39287984 PMCID: PMC11533985 DOI: 10.1172/jci.insight.182621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 09/10/2024] [Indexed: 09/19/2024] Open
Abstract
Tumor-associated neutrophils (TANs) have been shown to promote immunosuppression and tumor progression, and a high TAN frequency predicts poor prognosis in triple-negative breast cancer (TNBC). Dysregulation of CREB-binding protein (CBP)/P300 function has been observed with multiple cancer types. The bromodomain (BRD) of CBP/P300 has been shown to regulate its activity. In this study, we found that IACS-70654, a selective CBP/P300 BRD inhibitor, reduced TANs and inhibited the growth of neutrophil-enriched TNBC models. In the bone marrow, CBP/P300 BRD inhibition reduced the tumor-driven abnormal differentiation and proliferation of neutrophil progenitors. Inhibition of CBP/P300 BRD also stimulated the immune response by inducing an IFN response and MHCI expression in tumor cells and increasing tumor-infiltrated cytotoxic T cells. Moreover, IACS-70654 improved the response of a neutrophil-enriched TNBC model to docetaxel and immune checkpoint blockade. This provides a rationale for combining a CBP/P300 BRD inhibitor with standard-of-care therapies in future clinical trials for neutrophil-enriched TNBC.
Collapse
Affiliation(s)
- Xueying Yuan
- Department of Molecular and Cellular Biology and
| | - Xiaoxin Hao
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas, USA
| | - Hilda L. Chan
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas, USA
| | - Na Zhao
- Department of Molecular and Cellular Biology and
| | - Diego A. Pedroza
- Department of Molecular and Cellular Biology and
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas, USA
| | - Fengshuo Liu
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas, USA
| | - Kang Le
- Institute for Applied Cancer Science (IACS), University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | | | | | - Nadia Lieu
- Department of Molecular and Cellular Biology and
| | - Michael J. Soth
- Institute for Applied Cancer Science (IACS), University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Philip Jones
- Institute for Applied Cancer Science (IACS), University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Xiang H.F. Zhang
- Department of Molecular and Cellular Biology and
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas, USA
| | | |
Collapse
|
11
|
Contreras-Martínez OI, Angulo-Ortíz A, Santafé Patiño G, Sierra Martinez J, Berrio Soto R, de Almeida Rodolpho JM, de Godoy KF, de Freitas Aníbal F, de Lima Fragelli BD. Synergistic Antifungal Effect and In Vivo Toxicity of a Monoterpene Isoespintanol Obtained from Oxandra xylopioides Diels. Molecules 2024; 29:4417. [PMID: 39339412 PMCID: PMC11433975 DOI: 10.3390/molecules29184417] [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: 08/17/2024] [Revised: 09/07/2024] [Accepted: 09/14/2024] [Indexed: 09/30/2024] Open
Abstract
Candida sp. infections are a threat to global health, with high morbidity and mortality rates due to drug resistance, especially in immunocompromised people. For this reason, the search for new alternatives is urgent, and in recent years, a combined therapy with natural compounds has been proposed. Considering the biological potential of isoespintanol (ISO) and continuing its study, the objective of this research was to assess the effect of ISO in combination with the antifungals fluconazole (FLZ), amphotericin B (AFB) and caspofungin (CASP) against clinical isolates of C. tropicalis and to evaluate the cytotoxic effect of this compound in the acute phase (days 0 and 14) and chronic phase (days 0, 14, 28, 42, 56, 70 and 84) in female mice (Mus musculus) of the Balb/c lineage. The results show that ISO can potentiate the effect of FLZ, AFB and CASP, showing synergism with these antifungals. An evaluation of the mice via direct observation showed no behavioral changes or variations in weight during treatment; furthermore, an analysis of the cytokines IFN-γ and TNF in plasma, peritoneal cavity lavage (PCL) and bronchoalveolar lavage (BAL) indicated that there was no inflammation process. In addition, histopathological studies of the lungs, liver and kidneys showed no signs of toxicity caused by ISO. This was consistent with an analysis of oxaloacetic transaminases (GOT) and pyruvic transaminases (GPT), which remained in the standard range. These findings indicate that ISO does not have a cytotoxic effect at the doses evaluated, placing it as a monoterpene of interest in the search for compounds with pharmacological potential.
Collapse
Affiliation(s)
| | - Alberto Angulo-Ortíz
- Chemistry Department, Faculty of Basic Sciences, University of Córdoba, Montería 230002, Colombia
| | - Gilmar Santafé Patiño
- Chemistry Department, Faculty of Basic Sciences, University of Córdoba, Montería 230002, Colombia
| | - Jesus Sierra Martinez
- Department of Genetics and Evolution, Federal University of São Carlos, São Carlos 13565-905, SP, Brazil
| | - Ricardo Berrio Soto
- Biology Department, Faculty of Basic Sciences, University of Córdoba, Montería 230002, Colombia
| | - Joice Margareth de Almeida Rodolpho
- Laboratory of Inflammation and Infectious Diseases, Department of Morphology and Pathology, Federal University of São Carlos, São Carlos 13565-905, SP, Brazil
| | - Krissia Franco de Godoy
- Laboratory of Inflammation and Infectious Diseases, Department of Morphology and Pathology, Federal University of São Carlos, São Carlos 13565-905, SP, Brazil
| | - Fernanda de Freitas Aníbal
- Laboratory of Inflammation and Infectious Diseases, Department of Morphology and Pathology, Federal University of São Carlos, São Carlos 13565-905, SP, Brazil
| | - Bruna Dias de Lima Fragelli
- Functional Materials Development Center, Interdisciplinary Laboratory of Electrochemistry and Ceramics, Department of Chemistry, Federal University of São Carlos, São Carlos 13565-905, SP, Brazil
| |
Collapse
|
12
|
De S, Ehrlich M. Arrest and Attack: Microtubule-Targeting Agents and Oncolytic Viruses Employ Complementary Mechanisms to Enhance Anti-Tumor Therapy Efficacy. Genes (Basel) 2024; 15:1193. [PMID: 39336785 PMCID: PMC11431212 DOI: 10.3390/genes15091193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 08/25/2024] [Accepted: 09/04/2024] [Indexed: 09/30/2024] Open
Abstract
Oncolytic viruses (OVs) are promising cancer immunotherapy agents that stimulate anti-tumor immunity through the preferential infection and killing of tumor cells. OVs are currently under limited clinical usage, due in part to their restricted efficacy as monotherapies. Current efforts for enhancement of the therapeutic potency of OVs involve their combination with other therapy modalities, aiming at the concomitant exploitation of complementary tumor weaknesses. In this context, microtubule-targeting agents (MTAs) pose as an enticing option, as they perturb microtubule dynamics and function, induce cell-cycle arrest, and cause mitotic cell death. MTAs induce therapeutic benefit through cancer-cell-autonomous and non-cell-autonomous mechanisms and are a main component of the standard of care for different malignancies. However, off-target effects and acquired resistance involving distinct cellular and molecular mechanisms may limit the overall efficacy of MTA-based therapy. When combined, OVs and MTAs may enhance therapeutic efficacy through increases in OV infection and immunogenic cell death and a decreased probability of acquired resistance. In this review, we introduce OVs and MTAs, describe molecular features of their activity in cancer cells, and discuss studies and clinical trials in which the combination has been tested.
Collapse
Affiliation(s)
| | - Marcelo Ehrlich
- Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 6997801, Israel;
| |
Collapse
|
13
|
Zhou Z, Luo W, Zheng C, Wang H, Hu R, Deng H, Shen J. Mitochondrial metabolism blockade nanoadjuvant reversed immune-resistance microenvironment to sensitize albumin-bound paclitaxel-based chemo-immunotherapy. Acta Pharm Sin B 2024; 14:4087-4101. [PMID: 39309498 PMCID: PMC11413680 DOI: 10.1016/j.apsb.2024.05.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Revised: 05/15/2024] [Accepted: 05/17/2024] [Indexed: 09/25/2024] Open
Abstract
Currently, the efficacy of albumin-bound paclitaxel (PTX@Alb) is still limited due to the impaired PTX@Alb accumulation in tumors partly mediated by the dense collagen distribution. Meanwhile, acquired immune resistance always occurs due to the enhanced programmed cell death-ligand 1 (PD-L1) expression after PTX@Alb treatment, which then leads to immune tolerance. To fill these gaps, we newly revealed that tamoxifen (TAM), a clinically widely used adjuvant therapy for breast cancer with mitochondrial metabolism blockade capacity, could also be used as a novel effective PD-L1 and TGF-β dual-inhibitor via inducing the phosphorylation of adenosine 5'-monophosphate-activated protein kinase (AMPK) protein. Following this, to obtain a more significant effect, TPP-TAM was prepared by conjugating mitochondria-targeted triphenylphosphine (TPP) with TAM, which then further self-assembled with albumin (Alb) to form TPP-TAM@Alb nanoparticles. By doing this, TPP-TAM@Alb nanoparticles effectively decreased the expression of collagen in vitro, which then led to the enhanced accumulation of PTX@Alb in 4T1 tumors. Besides, TPP-TAM@Alb also effectively decreased the expression of PD-L1 and TGF-β in tumors to better sensitize PTX@Alb-mediated chemo-immunotherapy by enhancing T cell infiltration. All in all, we newly put forward a novel mitochondrial metabolism blockade strategy to inhibit PTX@Alb-resistant tumors, further supporting its better clinical application.
Collapse
Affiliation(s)
- Zaigang Zhou
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Wenjuan Luo
- School & Hospital of Stomatology, Wenzhou Medical University, Wenzhou 325027, China
| | - Chunjuan Zheng
- School & Hospital of Stomatology, Wenzhou Medical University, Wenzhou 325027, China
| | - Haoxiang Wang
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Rui Hu
- Department of the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Hui Deng
- School & Hospital of Stomatology, Wenzhou Medical University, Wenzhou 325027, China
| | - Jianliang Shen
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325001, China
| |
Collapse
|
14
|
Wang S, Guo S, Guo J, Du Q, Wu C, Wu Y, Zhang Y. Cell death pathways: molecular mechanisms and therapeutic targets for cancer. MedComm (Beijing) 2024; 5:e693. [PMID: 39239068 PMCID: PMC11374700 DOI: 10.1002/mco2.693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 07/24/2024] [Accepted: 07/28/2024] [Indexed: 09/07/2024] Open
Abstract
Cell death regulation is essential for tissue homeostasis and its dysregulation often underlies cancer development. Understanding the different pathways of cell death can provide novel therapeutic strategies for battling cancer. This review explores several key cell death mechanisms of apoptosis, necroptosis, autophagic cell death, ferroptosis, and pyroptosis. The research gap addressed involves a thorough analysis of how these cell death pathways can be precisely targeted for cancer therapy, considering tumor heterogeneity and adaptation. It delves into genetic and epigenetic factors and signaling cascades like the phosphatidylinositol 3-kinase/protein kinase B/mammalian target of rapamycin (PI3K/AKT/mTOR) and mitogen-activated protein kinase/extracellular signal-regulated kinase (MAPK/ERK) pathways, which are critical for the regulation of cell death. Additionally, the interaction of the microenvironment with tumor cells, and particularly the influence of hypoxia, nutrient deprivation, and immune cellular interactions, are explored. Emphasizing therapeutic strategies, this review highlights emerging modulators and inducers such as B cell lymphoma 2 (BCL2) homology domain 3 (BH3) mimetics, tumour necrosis factor-related apoptosis-inducing ligand (TRAIL), chloroquine, and innovative approaches to induce ferroptosis and pyroptosis. This review provides insights into cancer therapy's future direction, focusing on multifaceted approaches to influence cell death pathways and circumvent drug resistance. This examination of evolving strategies underlines the considerable clinical potential and the continuous necessity for in-depth exploration within this scientific domain.
Collapse
Affiliation(s)
- Shaohui Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Ethnic Medicine Chengdu University of Traditional Chinese Medicine Chengdu China
| | - Sa Guo
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy Chengdu University of Traditional Chinese Medicine Chengdu China
| | - Jing Guo
- College of Clinical Medicine Hospital of Chengdu University of Traditional Chinese Medicine Chengdu China
| | - Qinyun Du
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy Chengdu University of Traditional Chinese Medicine Chengdu China
| | - Cen Wu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy Chengdu University of Traditional Chinese Medicine Chengdu China
| | - Yeke Wu
- College of Clinical Medicine Hospital of Chengdu University of Traditional Chinese Medicine Chengdu China
| | - Yi Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Ethnic Medicine Chengdu University of Traditional Chinese Medicine Chengdu China
| |
Collapse
|
15
|
Opdam M, van Rossum AGJ, Hoogstraat M, Bounova G, Horlings HM, van Werkhoven E, Mandjes IAM, van Leeuwen-Stok AE, Canisius S, van Tinteren H, Imholz ALT, Portielje JEA, Bos MEMM, Bakker S, Wesseling J, Kester L, van Rheenen J, Rutgers EJ, de Menezes RX, Wessels LFA, Kok M, Oosterkamp HM, Linn SC. Predictive gene expression profile for adjuvant taxane benefit in breast cancer in the MATADOR trial. iScience 2024; 27:110425. [PMID: 39206149 PMCID: PMC11357803 DOI: 10.1016/j.isci.2024.110425] [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: 04/05/2024] [Revised: 05/10/2024] [Accepted: 06/27/2024] [Indexed: 09/04/2024] Open
Abstract
The primary objective of the prospective, randomized, multicenter, phase 3 biomarker Microarray Analysis in breast cancer to Taylor Adjuvant Drugs Or Regimens trial (MATADOR: ISRCTN61893718) is to generate a gene expression profile that can predict benefit from either docetaxel, doxorubicin, and cyclophosphamide (TAC) or dose-dense scheduled doxorubicin and cyclophosphamide (ddAC). Patients with a pT1-3, pN0-3 tumor were randomized 1:1 between ddAC and TAC. The primary endpoint was a gene profile-treatment interaction for recurrence-free survival (RFS). We observed 117 RFS events in 664 patients with a median follow-up of 7 years. Hallmark gene set analyses showed significant association between enrichment in immune-related gene expression and favorable outcome after TAC in hormone receptor-negative, human epidermal growth factor receptor 2 (HER2)-negative breast cancer (BC) (triple-negative breast cancer [TNBC]). We validated this association in TNBC patients treated with TAC on H&E slides; stromal tumor-infiltrating lymphocytes (sTILs) ≥20% was associated with longer RFS (hazard ratio 0.18, p = 0.01), while in patients treated with ddAC no difference in RFS was seen (hazard ratio 0.92, p = 0.86, p interaction = 0.02).
Collapse
Affiliation(s)
- Mark Opdam
- Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Annelot G J van Rossum
- Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Marlous Hoogstraat
- Division of Molecular Carcinogenesis, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Gergana Bounova
- Division of Molecular Carcinogenesis, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Hugo M Horlings
- Department of Pathology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Erik van Werkhoven
- Biometrics department, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Ingrid A M Mandjes
- Data center, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | | | - Sander Canisius
- Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Division of Molecular Carcinogenesis, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Harm van Tinteren
- Biometrics department, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Alex L T Imholz
- Department of Medical Oncology, Deventer Ziekenhuis, Deventer, the Netherlands
| | - Johanneke E A Portielje
- Department of Medical Oncology, HagaZiekenhuis, The Hague, the Netherlands
- Department of Medical Oncology, Leiden University Medical Center, Leiden, the Netherlands
| | - Monique E M M Bos
- Department of Internal Oncology, Reinier de Graaf Gasthuis, Delft, the Netherlands
| | - Sandra Bakker
- Department of Medical Oncology, Zaans Medisch Centrum, Zaandam, the Netherlands
| | - Jelle Wesseling
- Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Department of Pathology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Department of Pathology, Leiden University Medical Center, Leiden, the Netherlands
| | - Lennart Kester
- Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Jacco van Rheenen
- Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Emiel J Rutgers
- Department of Surgical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Renee X de Menezes
- Biostatistics Centre, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Department of Psychosocial Research and Epidemiology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Lodewyk F A Wessels
- Division of Molecular Carcinogenesis, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Faculty of Electrical Engineering, Mathematics and Computer Science, Delft University of Technology, Delft, the Netherlands
| | - Marleen Kok
- Division of Tumor biology & Immunology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Hendrika M Oosterkamp
- Department of Medical Oncology, Haaglanden Medisch Centrum, The Hague, the Netherlands
| | - Sabine C Linn
- Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Department of Pathology, University Medical Center, Utrecht, the Netherlands
| |
Collapse
|
16
|
Sun HK, Jiang WL, Zhang SL, Xu PC, Wei LM, Liu JB. Predictive value of tumor-infiltrating lymphocytes for neoadjuvant therapy response in triple-negative breast cancer: A systematic review and meta-analysis. World J Clin Oncol 2024; 15:920-935. [PMID: 39071463 PMCID: PMC11271722 DOI: 10.5306/wjco.v15.i7.920] [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: 03/09/2024] [Revised: 05/21/2024] [Accepted: 06/06/2024] [Indexed: 07/16/2024] Open
Abstract
BACKGROUND The association between tumor-infiltrating lymphocyte (TIL) levels and the response to neoadjuvant therapy (NAT) in patients with triple-negative breast cancer (TNBC) remains unclear. AIM To investigate the predictive potential of TIL levels for the response to NAT in TNBC patients. METHODS A systematic search of the National Center for Biotechnology Information PubMed database was performed to collect relevant published literature prior to August 31, 2023. The correlation between TIL levels and the NAT pathologic complete response (pCR) in TNBC patients was assessed using a systematic review and meta-analysis. Subgroup analysis, sensitivity analysis, and publication bias analysis were also conducted. RESULTS A total of 32 studies were included in this meta-analysis. The overall meta-analysis results indicated that the pCR rate after NAT treatment in TNBC patients in the high TIL subgroup was significantly greater than that in patients in the low TIL subgroup (48.0% vs 27.7%) (risk ratio 2.01; 95% confidence interval 1.77-2.29; P < 0.001, I 2 = 56%). Subgroup analysis revealed that the between-study heterogeneity originated from differences in study design, TIL level cutoffs, and study populations. Publication bias could have existed in the included studies. The meta-analysis based on different NAT protocols revealed that all TNBC patients with high levels of TILs had a greater rate of pCR after NAT treatment in all protocols (all P ≤ 0.01), and there was no significant between-protocol difference in the statistics among the different NAT protocols (P = 0.29). Additionally, sensitivity analysis demonstrated that the overall results of the meta-analysis remained consistent when the included studies were individually excluded. CONCLUSION TILs can serve as a predictor of the response to NAT treatment in TNBC patients. TNBC patients with high levels of TILs exhibit a greater NAT pCR rate than those with low levels of TILs, and this predictive capability is consistent across different NAT regimens.
Collapse
Affiliation(s)
- Hai-Kuan Sun
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang 471000, Henan Province, China
| | - Wen-Long Jiang
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang 471000, Henan Province, China
| | - Shi-Lei Zhang
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang 471000, Henan Province, China
| | - Peng-Cheng Xu
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang 471000, Henan Province, China
| | - Li-Min Wei
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang 471000, Henan Province, China
| | - Jiang-Bo Liu
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang 471000, Henan Province, China
| |
Collapse
|
17
|
Hu Y, Lou X, Zhang K, Pan L, Bai Y, Wang L, Wang M, Yan Y, Wan J, Yao X, Duan X, Ni C, Qin Z. Tumor necrosis factor receptor 2 promotes endothelial cell-mediated suppression of CD8+ T cells through tuning glycolysis in chemoresistance of breast cancer. J Transl Med 2024; 22:672. [PMID: 39033271 PMCID: PMC11265105 DOI: 10.1186/s12967-024-05472-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 07/03/2024] [Indexed: 07/23/2024] Open
Abstract
BACKGROUND T cells play a pivotal role in chemotherapy-triggered anti-tumor effects. Emerging evidence underscores the link between impaired anti-tumor immune responses and resistance to paclitaxel therapy in triple-negative breast cancer (TNBC). Tumor-related endothelial cells (ECs) have potential immunoregulatory activity. However, how ECs regulate T cell activity during TNBC chemotherapy remains poorly understood. METHODS Single-cell analysis of ECs in patients with TNBC receiving paclitaxel therapy was performed using an accessible single-cell RNA sequencing (scRNA-seq) dataset to identify key EC subtypes and their immune characteristics. An integrated analysis of a tumor-bearing mouse model, immunofluorescence, and a spatial transcriptome dataset revealed the spatial relationship between ECs, especially Tumor necrosis factor receptor (TNFR) 2+ ECs, and CD8+ T cells. RNA sequencing, CD8+ T cell proliferation assays, flow cytometry, and bioinformatic analyses were performed to explore the immunosuppressive function of TNFR2 in ECs. The downstream metabolic mechanism of TNFR2 was further investigated using RNA sequencing, cellular glycolysis assays, and western blotting. RESULTS In this study, we identified an immunoregulatory EC subtype, characterized by enhanced TNFR2 expression in non-responders. By a mouse model of TNBC, we revealed a dynamic reduction in the proportion of the CD8+ T cell-contacting tumor vessels that could co-localize spatially with CD8+ T cells during chemotherapy and an increased expression of TNFR2 by ECs. TNFR2 suppresses glycolytic activity in ECs by activating NF-κB signaling in vitro. Tuning endothelial glycolysis enhances programmed death-ligand (PD-L) 1-dependent inhibitory capacity, thereby inducing CD8+ T cell suppression. In addition, TNFR2+ ECs showed a greater spatial affinity for exhausted CD8+ T cells than for non-exhausted CD8+ T cells. TNFR2 blockade restores impaired anti-tumor immunity in vivo, leading to the loss of PD-L1 expression by ECs and enhancement of CD8+ T cell infiltration into the tumors. CONCLUSIONS These findings reveal the suppression of CD8+ T cells by ECs in chemoresistance and indicate the critical role of TNFR2 in driving the immunosuppressive capacity of ECs via tuning glycolysis. Targeting endothelial TNFR2 may serve as a potent strategy for treating TNBC with paclitaxel.
Collapse
Affiliation(s)
- Yu Hu
- Henan China-Germany International Joint Laboratory of Tumor Immune Microenvironment and Disease, Medical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Xiaohan Lou
- Henan China-Germany International Joint Laboratory of Tumor Immune Microenvironment and Disease, Medical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Kaili Zhang
- Henan China-Germany International Joint Laboratory of Tumor Immune Microenvironment and Disease, Medical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Longze Pan
- Henan China-Germany International Joint Laboratory of Tumor Immune Microenvironment and Disease, Medical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
- Department of Medicine, Luohe Medical College, Luohe, 462000, China
| | - Yueyue Bai
- Henan China-Germany International Joint Laboratory of Tumor Immune Microenvironment and Disease, Medical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
- Shangqiu Hospital, The First Affiliated Hospital of Henan University of Chinese Medicine, Shangqiu, 476000, China
| | - Linlin Wang
- Henan China-Germany International Joint Laboratory of Tumor Immune Microenvironment and Disease, Medical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Ming Wang
- Henan China-Germany International Joint Laboratory of Tumor Immune Microenvironment and Disease, Medical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Yan Yan
- Henan China-Germany International Joint Laboratory of Tumor Immune Microenvironment and Disease, Medical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Jiajia Wan
- Henan China-Germany International Joint Laboratory of Tumor Immune Microenvironment and Disease, Medical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Xiaohan Yao
- Henan China-Germany International Joint Laboratory of Tumor Immune Microenvironment and Disease, Medical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Xixi Duan
- Henan China-Germany International Joint Laboratory of Tumor Immune Microenvironment and Disease, Medical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Chen Ni
- Henan China-Germany International Joint Laboratory of Tumor Immune Microenvironment and Disease, Medical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China.
| | - Zhihai Qin
- Henan China-Germany International Joint Laboratory of Tumor Immune Microenvironment and Disease, Medical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China.
| |
Collapse
|
18
|
Peng Y, Shi R, Yang S, Zhu J. Cuproptosis-related gene DLAT is a biomarker of the prognosis and immune microenvironment of gastric cancer and affects the invasion and migration of cells. Cancer Med 2024; 13:e70012. [PMID: 39031012 PMCID: PMC11258438 DOI: 10.1002/cam4.70012] [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: 02/16/2024] [Revised: 06/10/2024] [Accepted: 07/03/2024] [Indexed: 07/22/2024] Open
Abstract
OBJECTIVE Cuproptosis is a novel cell death dependent on mitochondrial respiration and regulated by copper. This study aimed to investigate the cuproptosis-related gene DLAT potential value in gastric cancer (GC). METHODS Bioinformatics was used to analyze DLAT expression. DLAT expression in GC cell lines was detected using qRT-PCR. Cell proliferation ability was assessed using CCK8 and cell cycle assay. Cell migration and invasion were assessed using wound healing and transwell assay. A prognostic assessment was performed through survival and Cox regression analysis. DLAT protein expression was analyzed through HPA immunohistochemistry. Biological functions and processes were analyzed through GO and KEGG enrichment analysis and PPI. Correlation with immune cell infiltration and immune checkpoint genes was analyzed for DLAT. RESULTS DLAT expression was upregulated in GC tissues and cells and correlated with shorter survival for patients. Age, gender, histological typing, lymph node metastasis, and distant metastasis were identified as independent prognostic factors affecting OS in GC. DLAT protein was upregulated in GC. The biological functions and pathways enriched in DLAT were mainly linked to mitochondrial respiration and the TCA cycle. The expression of DLAT was found to be positively correlated with the infiltration of Th and Th2 immune cells and only positively correlated with the expression of the BTN2A1 immune checkpoint gene. CONCLUSION DLAT has the potential to serve as a prognostic assessment factor in GC. The expression of DLAT was correlated with immune infiltration and tumor immune escape, providing a new target for immunotherapy of GC.
Collapse
Affiliation(s)
- Yanyu Peng
- Department of Histology and EmbryologyShenyang Medical CollegeShenyangLiaoningChina
| | - Ruimeng Shi
- Shenyang Medical CollegeShenyangLiaoningChina
| | - Siwen Yang
- Shenyang Medical CollegeShenyangLiaoningChina
| | - Jiayi Zhu
- Shenyang Medical CollegeShenyangLiaoningChina
| |
Collapse
|
19
|
Tan RX, Chen XY. Uncovering the baccatin III biosynthetic pathway for sustainable taxoid supply. Sci Bull (Beijing) 2024; 69:1182-1184. [PMID: 38514298 DOI: 10.1016/j.scib.2024.03.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/23/2024]
Affiliation(s)
- Ren Xiang Tan
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Functional Biomolecules, School of Life Sciences, Nanjing University, Nanjing 210023, China.
| | - Xiao-Ya Chen
- State Key Laboratory of Plant Molecular Genetics, CAS Center for Excellence in Molecular Plant Sciences, Chinese Academy of Sciences, Shanghai 200032, China.
| |
Collapse
|
20
|
Schramm WC, Bala N, Arekar T, Malik Z, Chacko KM, Lewis RL, Denslow ND, Scindia Y, Alli AA. Enrichment of Bioactive Lipids in Urinary Extracellular Vesicles and Evidence of Apoptosis in Kidneys of Hypertensive Diabetic Cathepsin B Knockout Mice after Streptozotocin Treatment. Biomedicines 2024; 12:1038. [PMID: 38791000 PMCID: PMC11117475 DOI: 10.3390/biomedicines12051038] [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: 02/01/2024] [Revised: 04/15/2024] [Accepted: 04/18/2024] [Indexed: 05/26/2024] Open
Abstract
Cathepsin B (CtsB) is a ubiquitously expressed cysteine protease that plays important roles in health and disease. Urinary extracellular vesicles (uEVs) are released from cells associated with urinary organs. The antibiotic streptozotocin (STZ) is known to induce pancreatic islet beta cell destruction, diabetic nephropathy, and hypertension. We hypothesized that streptozotocin-induced diabetic kidney disease and hypertension result in the release of bioactive lipids from kidney cells that induce oxidative stress and renal cell death. Lipidomics was performed on uEVs isolated from CtsB knockout mice treated with or without STZ, and their kidneys were used to investigate changes in proteins associated with cell death. Lysophosphatidylethanolamine (LPE) (18:1), lysophosphatidylserine (LPS) (22:6), and lysophosphatidylglycerol (LPG) (22:5) were among the bioactive lipids enriched in uEVs from CtsB knockout mice treated with STZ compared to untreated CtsB mice (n = 3 uEV preparations per group). Anti-oxidant programming was activated in the kidneys of the CtsB knockout mice treated with STZ, as indicated by increased expression of glutathione peroxidase 4 (GPX4) and the cystine/glutamate antiporter SLC7A11 (XCT) (n = 4 mice per group), which was supported by a higher reactivity to 4-hydroxy-2-nonenal (4-HNE), a marker for oxidative stress (n = 3 mice per group). Apoptosis but not ferroptosis was the ongoing form of cell death in these kidneys as cleaved caspase-3 levels were significantly elevated in the STZ-treated CtsB knockout mice (n = 4 mice per group). There were no appreciable differences in the pro-ferroptosis enzyme acyl-CoA synthetase long-chain family member 4 (ACSL4) or the inflammatory marker CD93 in the kidneys (n = 3 mice per group), which further supports apoptosis as the prevalent mechanism of pathology. These data suggest that STZ treatment leads to oxidative stress, inducing apoptotic injury in the kidneys during the development of diabetic kidney disease and hypertension.
Collapse
Affiliation(s)
- Whitney C. Schramm
- Department of Medicine, Division of Nephrology, Hypertension, and Renal Transplantation, College of Medicine, University of Florida, Gainesville, FL 32610, USA; (W.C.S.); (N.B.); (T.A.); (Z.M.); (K.M.C.); (Y.S.)
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Niharika Bala
- Department of Medicine, Division of Nephrology, Hypertension, and Renal Transplantation, College of Medicine, University of Florida, Gainesville, FL 32610, USA; (W.C.S.); (N.B.); (T.A.); (Z.M.); (K.M.C.); (Y.S.)
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Tanmay Arekar
- Department of Medicine, Division of Nephrology, Hypertension, and Renal Transplantation, College of Medicine, University of Florida, Gainesville, FL 32610, USA; (W.C.S.); (N.B.); (T.A.); (Z.M.); (K.M.C.); (Y.S.)
| | - Zeeshan Malik
- Department of Medicine, Division of Nephrology, Hypertension, and Renal Transplantation, College of Medicine, University of Florida, Gainesville, FL 32610, USA; (W.C.S.); (N.B.); (T.A.); (Z.M.); (K.M.C.); (Y.S.)
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Kevin M. Chacko
- Department of Medicine, Division of Nephrology, Hypertension, and Renal Transplantation, College of Medicine, University of Florida, Gainesville, FL 32610, USA; (W.C.S.); (N.B.); (T.A.); (Z.M.); (K.M.C.); (Y.S.)
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Russell L. Lewis
- Department of Physiological Sciences, Center for Environmental and Human Toxicology, University of Florida, Gainesville, FL 32608, USA; (R.L.L.); (N.D.D.)
| | - Nancy D. Denslow
- Department of Physiological Sciences, Center for Environmental and Human Toxicology, University of Florida, Gainesville, FL 32608, USA; (R.L.L.); (N.D.D.)
| | - Yogesh Scindia
- Department of Medicine, Division of Nephrology, Hypertension, and Renal Transplantation, College of Medicine, University of Florida, Gainesville, FL 32610, USA; (W.C.S.); (N.B.); (T.A.); (Z.M.); (K.M.C.); (Y.S.)
- Department of Molecular Genetics and Microbiology, College of Medicine, University of Florida, Gainesville, FL 32608, USA
| | - Abdel A. Alli
- Department of Medicine, Division of Nephrology, Hypertension, and Renal Transplantation, College of Medicine, University of Florida, Gainesville, FL 32610, USA; (W.C.S.); (N.B.); (T.A.); (Z.M.); (K.M.C.); (Y.S.)
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| |
Collapse
|
21
|
Yuan X, Hao X, Chan HL, Zhao N, Pedroza DA, Liu F, Le K, Smith AJ, Calderon SJ, Lieu N, Soth MJ, Jones P, Zhang XHF, Rosen JM. CBP/P300 BRD Inhibition Reduces Neutrophil Accumulation and Activates Antitumor Immunity in TNBC. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.25.590983. [PMID: 38712292 PMCID: PMC11071628 DOI: 10.1101/2024.04.25.590983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Tumor-associated neutrophils (TANs) have been shown to promote immunosuppression and tumor progression, and a high TAN frequency predicts poor prognosis in triple-negative breast cancer (TNBC). Dysregulation of CREB binding protein (CBP)/P300 function has been observed with multiple cancer types. The bromodomain (BRD) of CBP/P300 has been shown to regulate its activity. In this study, we found that IACS-70654, a novel and selective CBP/P300 BRD inhibitor, reduced TANs and inhibited the growth of neutrophil-enriched TNBC models. In the bone marrow, CBP/P300 BRD inhibition reduced the tumor-driven abnormal differentiation and proliferation of neutrophil progenitors. Inhibition of CBP/P300 BRD also stimulated the immune response by inducing an IFN response and MHCI expression in tumor cells and increasing tumor-infiltrated CTLs. Moreover, IACS-70654 improved the response of a neutrophil-enriched TNBC model to docetaxel and immune checkpoint blockade. This provides a rationale for combining a CBP/P300 BRD inhibitor with standard-of-care therapies in future clinical trials for neutrophil-enriched TNBC.
Collapse
Affiliation(s)
- Xueying Yuan
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Xiaoxin Hao
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA
| | - Hilda L Chan
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA
| | - Na Zhao
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Diego A Pedroza
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA
| | - Fengshuo Liu
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA
| | - Kang Le
- Institute for Applied Cancer Science (IACS), the University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Alex J Smith
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Sebastian J Calderon
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Nadia Lieu
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Michael J Soth
- Institute for Applied Cancer Science (IACS), the University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Philip Jones
- Institute for Applied Cancer Science (IACS), the University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Xiang H-F Zhang
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA
| | - Jeffrey M Rosen
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
22
|
Hu X, Li J, Yu L, Ifejola J, Guo Y, Zhang D, Khosravi Z, Zhang K, Cui H. Screening of anti-melanoma compounds from Morus alba L.: Sanggenon C promotes melanoma cell apoptosis by disrupting intracellular Ca 2+ homeostasis. JOURNAL OF ETHNOPHARMACOLOGY 2024; 324:117759. [PMID: 38219884 DOI: 10.1016/j.jep.2024.117759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 12/18/2023] [Accepted: 01/11/2024] [Indexed: 01/16/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Morus alba L. is a widespread plant that has long been considered to have remarkable medical values, including anti-inflammation in Traditional Chinese Medicine (TCM). The components of Morus Alba L. constituents have been extensively studied and have been shown to have high prospects for cancer therapy. However, limited investigations have been done on the bioactive compounds in Morus alba L. AIM OF THE STUDY This study aimed to systematically examine the anticancer properties of 28 commercially available compounds from Morus alba L. against melanoma cells in vitro. Additionally, the anticancer mechanisms of the bioactive compound exhibiting the most significant potential were further studied. MATERIALS AND METHODS The anti-proliferative effects of Morus alba L.-derived compounds on melanoma cells were determined by colony formation assays. Their effects on cell viability and apoptosis were determined using the CCK8 assay and flow cytometry, respectively. The binding affinity of identified Morus alba L. compounds with anticancer activities towards melanoma targets was analyzed via molecular docking. The molecular mechanism of Sanggenon C was explored using soft agar assays, EdU incorporation assays, flow cytometry, western blotting, transcriptome analysis, and xenograft assays. RESULTS Based on colony formation assays, 11 compounds at 20 μM significantly inhibited colony growth on a panel of melanoma cells. These compounds displayed IC50 values (half maximal inhibitory concentrations) ranging from 5 μM to 30 μM. Importantly, six compounds were identified as novel anti-melanoma agents, including Sanggenon C, 3'-Geranyl-3-prenyl-2',4',5,7-tetrahydroxyflavone, Moracin P, Moracin O, Kuwanon A, and Kuwanon E. Among them, Sanggenon C showed the most potent effects, with an IC50 of about 5 μM, significantly reducing proliferation and inducing apoptosis in melanoma cells. Based on the xenograft model assay, Sanggenon C significantly inhibited melanoma cell proliferation in vivo. Sanggenon C triggered ER stress in a dose-dependent manner, which further disrupted cellular calcium ion (Ca2+) homeostasis. The Ca2+ chelator BAPTA partially restored cell apoptosis induced by Sanggenon C, confirming that Ca2+ signaling contributed to the anticancer activity of Sanggenon C against melanoma. CONCLUSIONS In our study, 11 compounds demonstrated anti-melanoma properties. Notably, Sanggenon C was found to promote apoptosis by disrupting the intracellular calcium homeostasis in melanoma cells. This study provides valuable information for the future development of novel cancer therapeutic agents from Morus alba L.
Collapse
Affiliation(s)
- Xin Hu
- State Key Laboratory of Resource Insects, Medical Research Institute, Southwest University, Chongqing, 400715, China; Jinfeng Laboratory, Chongqing, 401329, China; Chongqing Engineering and Technology Research Center for Silk Biomaterials and Regenerative Medicine, Chongqing, 400716, China.
| | - Jing Li
- State Key Laboratory of Resource Insects, Medical Research Institute, Southwest University, Chongqing, 400715, China; Jinfeng Laboratory, Chongqing, 401329, China; Chongqing Engineering and Technology Research Center for Silk Biomaterials and Regenerative Medicine, Chongqing, 400716, China.
| | - Lang Yu
- State Key Laboratory of Resource Insects, Medical Research Institute, Southwest University, Chongqing, 400715, China; Jinfeng Laboratory, Chongqing, 401329, China; Chongqing Engineering and Technology Research Center for Silk Biomaterials and Regenerative Medicine, Chongqing, 400716, China.
| | - Jemirade Ifejola
- State Key Laboratory of Resource Insects, Medical Research Institute, Southwest University, Chongqing, 400715, China; Chongqing Engineering and Technology Research Center for Silk Biomaterials and Regenerative Medicine, Chongqing, 400716, China.
| | - Yan Guo
- State Key Laboratory of Resource Insects, Medical Research Institute, Southwest University, Chongqing, 400715, China; Jinfeng Laboratory, Chongqing, 401329, China.
| | - Dandan Zhang
- State Key Laboratory of Resource Insects, Medical Research Institute, Southwest University, Chongqing, 400715, China; Jinfeng Laboratory, Chongqing, 401329, China; Chongqing Engineering and Technology Research Center for Silk Biomaterials and Regenerative Medicine, Chongqing, 400716, China.
| | - Zahra Khosravi
- State Key Laboratory of Resource Insects, Medical Research Institute, Southwest University, Chongqing, 400715, China; Chongqing Engineering and Technology Research Center for Silk Biomaterials and Regenerative Medicine, Chongqing, 400716, China.
| | - Kui Zhang
- State Key Laboratory of Resource Insects, Medical Research Institute, Southwest University, Chongqing, 400715, China; Jinfeng Laboratory, Chongqing, 401329, China; Chongqing Engineering and Technology Research Center for Silk Biomaterials and Regenerative Medicine, Chongqing, 400716, China.
| | - Hongjuan Cui
- State Key Laboratory of Resource Insects, Medical Research Institute, Southwest University, Chongqing, 400715, China; Jinfeng Laboratory, Chongqing, 401329, China; Chongqing Engineering and Technology Research Center for Silk Biomaterials and Regenerative Medicine, Chongqing, 400716, China.
| |
Collapse
|
23
|
Yuan Q, Yin L, He J, Zeng Q, Liang Y, Shen Y, Zu X. Metabolism of asparagine in the physiological state and cancer. Cell Commun Signal 2024; 22:163. [PMID: 38448969 PMCID: PMC10916255 DOI: 10.1186/s12964-024-01540-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 02/22/2024] [Indexed: 03/08/2024] Open
Abstract
Asparagine, an important amino acid in mammals, is produced in several organs and is widely used for the production of other nutrients such as glucose, proteins, lipids, and nucleotides. Asparagine has also been reported to play a vital role in the development of cancer cells. Although several types of cancer cells can synthesise asparagine alone, their synthesis levels are insufficient to meet their requirements. These cells must rely on the supply of exogenous asparagine, which is why asparagine is considered a semi-essential amino acid. Therefore, nutritional inhibition by targeting asparagine is often considered as an anti-cancer strategy and has shown success in the treatment of leukaemia. However, asparagine limitation alone does not achieve an ideal therapeutic effect because of stress responses that upregulate asparagine synthase (ASNS) to meet the requirements for asparagine in cancer cells. Various cancer cells initiate different reprogramming processes in response to the deficiency of asparagine. Therefore, it is necessary to comprehensively understand the asparagine metabolism in cancers. This review primarily discusses the physiological role of asparagine and the current progress in the field of cancer research.
Collapse
Affiliation(s)
- Qiong Yuan
- Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Liyang Yin
- Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China
| | - Jun He
- Department of Spine Surgery, The Nanhua Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Qiting Zeng
- Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Yuxin Liang
- Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Yingying Shen
- Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China.
| | - Xuyu Zu
- Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China.
| |
Collapse
|
24
|
Liu G, Chen T, Zhang X, Hu B, Yu J. Nomogram for predicting pathologic complete response to neoadjuvant chemoradiotherapy in patients with esophageal squamous cell carcinoma. Cancer Med 2024; 13:e7075. [PMID: 38477511 DOI: 10.1002/cam4.7075] [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: 10/10/2023] [Revised: 02/12/2024] [Accepted: 02/20/2024] [Indexed: 03/14/2024] Open
Abstract
PURPOSE A pathologic complete response (pCR) to neoadjuvant chemoradiotherapy (nCRT) is seen in up to 40% of the patients with esophageal squamous cell carcinoma (ESCC). No nomogram has been constructed for the prediction of pCR for patients whose primary chemotherapy was a taxane-based regimen. The aim is to identify characteristics associated with a pCR through analyzing multiple pre- and post-nCRT variables and to develop a nomogram for the prediction of pCR for these patients by integrating clinicopathological characteristics and hematological biomarkers. MATERIALS AND METHODS We analyzed 293 patients with ESCC who underwent nCRT followed by esophagectomy. Clinicopathological factors, hematological parameters before nCRT, and hematotoxicity during nCRT were collected. Univariate and multivariate logistic regression analyses were performed to identify predictive factors for pCR. A nomogram model was built and evaluated for both discrimination and calibration. RESULTS After surgery, 37.88% of the study patients achieved pCR. Six variables were included in the nomogram: sex, cN stage, chemotherapy regimen, duration of nCRT, pre-nCRT neutrophil-to-lymphocyte ratio (NLR), and pre-nCRT platelet-to-lymphocyte ratio (PLR). The nomogram indicated good accuracy and consistency in predicting pCR, with a C-index of 0.743 (95% confidence interval: 0.686, 0.800) and a p value of 0.600 (>0.05) in the Hosmer-Lemeshow goodness-of-fit test. CONCLUSIONS Female, earlier cN stage, duration of nCRT (< 62 days), chemotherapy regimen of taxane plus platinum, pre-nCRT NLR (≥2.199), and pre-nCRT PLR (≥99.302) were significantly associated with a higher pCR in ESCC patients whose primary chemotherapy was a taxane-based regimen for nCRT. A nomogram was developed and internally validated, showing good accuracy and consistency.
Collapse
Affiliation(s)
- Guihong Liu
- Department of Radiotherapy, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Tao Chen
- Department of Cardiology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xin Zhang
- Department of Radiotherapy, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Binbin Hu
- Department of Radiotherapy, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jiayun Yu
- Department of Radiotherapy, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
25
|
Carrato A, Pazo-Cid R, Macarulla T, Gallego J, Jiménez-Fonseca P, Rivera F, Cano MT, Rodriguez-Garrote M, Pericay C, Alés I, Layos L, Graña B, Iranzo V, Gallego I, Garcia-Carbonero R, de Mena IR, Guillén-Ponce C, Aranda E. Nab-Paclitaxel plus Gemcitabine and FOLFOX in Metastatic Pancreatic Cancer. NEJM EVIDENCE 2024; 3:EVIDoa2300144. [PMID: 38320486 DOI: 10.1056/evidoa2300144] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2024]
Abstract
BACKGROUND: Sequential nab-paclitaxel plus gemcitabine followed by modified FOLFOX-6 (oxaliplatin, leucovorin, and 5-fluorouracil) (nab-P/Gem-mFOLFOX) showed a good safety and clinical profile in metastatic pancreatic ductal adenocarcinoma (mPDAC) in the phase I SEQUENCE trial. METHODS: The safety and efficacy of sequential nab-P/Gem-mFOLFOX was compared with standard nab-paclitaxel plus gemcitabine (nab-P/Gem) as first-line treatment in a multi-institutional, randomized, open-label, phase II trial in patients with untreated mPDAC. We randomly assigned patients in a 1:1 ratio to receive nab-P/Gem on days 1, 8, and 15 followed by mFOLFOX on day 29 of a 6-week cycle (experimental group) or nab-P/Gem on days 1, 8, and 15 of a 4-week cycle (control group). The primary end point was the 12-month overall survival rate. RESULTS: A total of 157 patients were randomly assigned: 78 to nab-P/Gem-mFOLFOX and 79 to nab-P/Gem. Patients receiving nab-P/Gem-mFOLFOX had a 12-month overall survival of 55.3% (95% confidence interval [CI], 44.2 to 66.5) versus 35.4% (95% CI, 24.9 to 46) in the control group (P=0.02). Similarly, the 24-month survival was 22.4% (95% CI, 13 to 31.8) with nab-P/Gem-mFOLFOX versus 7.6% (95% CI, 1.8 to 13.4) with control treatment. The median overall survival was 13.2 months (95% CI, 10.1 to 16.2) with nab-P/Gem-mFOLFOX and 9.7 months (95% CI, 7.5 to 12) with nab-P/Gem (hazard ratio for death, 0.68; 95% CI, 0.48 to 0.95). The safety profile showed a higher incidence of grade 3 or higher neutropenia (35 of 76 vs. 19 of 79 patients, P=0.004), grade 3 or higher thrombocytopenia (18 of 78 vs. 6 of 79 patients, P=0.007), and two treatment-related deaths (2.6%) with nab-P/Gem-mFOLFOX compared with none with control treatment. CONCLUSIONS: Sequential nab-P/Gem-mFOLFOX showed a significantly higher 12-month survival when compared with the standard nab-P/Gem treatment; this came with greater treatment toxicity. (Funded by Celgene; EuCT number, 2014-005350-19; ClinicalTrials.gov number, NCT02504333.)
Collapse
Affiliation(s)
- Alfredo Carrato
- Department of Medical Oncology, Alcalá University, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Ramon y Cajal University Hospital, Madrid
- Pancreatic Cancer Europe, Brussels
| | - Roberto Pazo-Cid
- Department of Medical Oncology, Aragon Institute of Biomedical Research, Miguel Servet University Hospital, Zaragoza, Spain
| | - Teresa Macarulla
- Vall d'Hebrón Institute of Oncology, Vall d'Hebrón University Hospital, Barcelona
| | - Javier Gallego
- Department of Medical Oncology, Elche University Hospital, Alicante, Spain
| | - Paula Jiménez-Fonseca
- Department of Medical Oncology, Asturias Central University Hospital, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| | - Fernando Rivera
- Department of Medical Oncology, Marques de Valdecilla University Hospital, Instituto de Investigación Valdecilla (IDIVAL), Santander, Spain
| | - Maria Teresa Cano
- Department of Medical Oncology, Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Cordoba University, Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Reina Sofia University Hospital, Cordoba, Spain
| | - Mercedes Rodriguez-Garrote
- Department of Medical Oncology, University, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Ramon y Cajal University Hospital, Madrid
| | - Carles Pericay
- Department of Medical Oncology, Sabadell University Hospital, Parc Tauli, Sabadell, Spain
| | - Inmaculada Alés
- Unidad de Gestión Clínica Intercentros (UGCI) Medical Oncology, University Regional and Virgen Victoria Hospital, Instituto de Investigación Biomédica de Málaga (IBIMA), Malaga, Spain
| | - Laura Layos
- Medical Oncology Department, Catalan Institute of Oncology (ICO), Badalona Applied Research Group in Oncology (B-ARGO), Germans Trias i Pujol University Hospital, Badalona, Spain
| | - Begoña Graña
- Department of Medical Oncology, A Coruña University Hospital, Instituto de Investigación Biomédica de A Coruña (INIBIC), A Coruña, Spain
| | - Vega Iranzo
- Department of Medical Oncology, University General Hospital Valencia, Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Valencia University, Valencia, Spain
| | - Inmaculada Gallego
- Department of Medical Oncology, Virgen del Rocio University Hospital, Instituto de Biomedicina de Sevilla (IBIS), Sevilla, Spain
| | - Rocio Garcia-Carbonero
- Department of Medical Oncology, Instituto de Investigación Sanitaria Hospital 12 de Octubre (Imas12), Universidad Complutense Madrid (UCM), Madrid
| | | | | | - Enrique Aranda
- Department of Medical Oncology, Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Cordoba University, Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Reina Sofia University Hospital, Cordoba, Spain
| |
Collapse
|
26
|
Gregorczyk M, Parkes EE. Targeting mitotic regulators in cancer as a strategy to enhance immune recognition. DNA Repair (Amst) 2023; 132:103583. [PMID: 37871511 DOI: 10.1016/j.dnarep.2023.103583] [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: 07/26/2023] [Revised: 10/06/2023] [Accepted: 10/12/2023] [Indexed: 10/25/2023]
Abstract
Eukaryotic DNA has evolved to be enclosed within the nucleus to protect the cellular genome from autoinflammatory responses driven by the immunogenic nature of cytoplasmic DNA. Cyclic GMP-AMP Synthase (cGAS) is the cytoplasmic dsDNA sensor, which upon activation of Stimulator of Interferon Genes (STING), mediates production of pro-inflammatory interferons (IFNs) and interferon stimulated genes (ISGs). However, although this pathway is crucial in detection of viral and microbial genetic material, cytoplasmic DNA is not always of foreign origin. It is now recognised that specifically in genomic instability, a hallmark of cancer, extranuclear material in the form of micronuclei (MN) can be generated as a result of unresolved DNA lesions during mitosis. Activation of cGAS-STING in cancer has been shown to regulate numerous tumour-immune interactions such as acquisition of 'immunologically hot' phenotype which stimulates immune-mediated elimination of transformed cells. Nonetheless, a significant percentage of poorly prognostic cancers is 'immunologically cold'. As this state has been linked with low proportion of tumour-infiltrating lymphocytes (TILs), improving immunogenicity of cold tumours could be clinically relevant by exhibiting synergy with immunotherapy. This review aims to present how inhibition of vital mitotic regulators could provoke cGAS-STING response in cancer and improve the efficacy of current immunotherapy regimens.
Collapse
Affiliation(s)
- Mateusz Gregorczyk
- Oxford Centre for Immuno-Oncology, Department of Oncology, University of Oxford, Oxford OX3 7DQ, United Kingdom
| | - Eileen E Parkes
- Oxford Centre for Immuno-Oncology, Department of Oncology, University of Oxford, Oxford OX3 7DQ, United Kingdom.
| |
Collapse
|
27
|
Wang Y, Song Y, He Y, Wang Y, Maurer J, Kiessling F, Lammers T, Wang F, Shi Y. Direct immunoactivation by chemotherapeutic drugs in cancer treatment. ADVANCED THERAPEUTICS 2023; 6:2300209. [PMID: 38249990 PMCID: PMC7615547 DOI: 10.1002/adtp.202300209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Indexed: 01/23/2024]
Abstract
The immune system plays a crucial role in recognizing and eliminating pathogenic substances and malignant cells in the body. For cancer treatment, immunotherapy is becoming the standard treatment for many types of cancer and is often combined with chemotherapy. Although chemotherapeutic agents are often reported to have adverse effects, including immunosuppression, they can also play a positive role in immunotherapy by directly stimulating the immune system. This has been demonstrated in preclinical and clinical studies in the past decades. Chemotherapeutics can activate immune cells through different immune receptors and signaling pathways depending on their chemical structure and formulation. In this review, we summarize and discuss the direct immunoactivation effects of chemotherapeutics and possible mechanisms behind these effects. Finally, we prospect chemo-immunotherapeutic combinations for the more effective and safer treatment of cancer.
Collapse
Affiliation(s)
- Yurui Wang
- Department of Polymer Therapeutics, Institute for Experimental Molecular Imaging, Uniklinik RWTH Aachen and Helmholtz Institute for Biomedical Engineering, Faculty of Medicine, RWTH Aachen University, Aachen 52074, Germany
| | - Yiran Song
- Department of Gastroenterology, Shanghai 10th People's Hospital, School of Medicine, Tongji University, Shanghai 200040, PR China
| | - Yazhi He
- Department of Gastroenterology, Shanghai 10th People's Hospital, School of Medicine, Tongji University, Shanghai 200040, PR China
| | - Yang Wang
- Department of Gastroenterology, Shanghai 10th People's Hospital, School of Medicine, Tongji University, Shanghai 200040, PR China
| | - Jochen Maurer
- Department of Gynecology and Obstetrics, Uniklinik RWTH Aachen, Aachen 52074, Germany
| | - Fabian Kiessling
- Institute for Experimental Molecular Imaging, Uniklinik RWTH Aachen and Helmholtz Institute for Biomedical Engineering, Faculty of Medicine, RWTH Aachen University, Aachen 52074, Germany
| | - Twan Lammers
- Institute for Experimental Molecular Imaging, Uniklinik RWTH Aachen and Helmholtz Institute for Biomedical Engineering, Faculty of Medicine, RWTH Aachen University, Aachen 52074, Germany
| | - Feng Wang
- Department of Gastroenterology, Huadong Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, PR China
| | - Yang Shi
- Department of Polymer Therapeutics, Institute for Experimental Molecular Imaging, Uniklinik RWTH Aachen and Helmholtz Institute for Biomedical Engineering, Faculty of Medicine, RWTH Aachen University, Aachen 52074, Germany
| |
Collapse
|
28
|
Xu Y, Xiao Y, Shao ZM. Taxanes directly induce T cell cytotoxic extracellular vesicles to eradicate tumor cells. Cancer Cell 2023; 41:1011-1013. [PMID: 37311412 DOI: 10.1016/j.ccell.2023.05.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 05/16/2023] [Accepted: 05/16/2023] [Indexed: 06/15/2023]
Abstract
Taxanes are widely used in cancer therapy, yet their mitotic-independent mechanisms in vivo remain enigmatic. Vennin et al. illuminate a mode of action whereby taxanes directly stimulate T cells to release cytotoxic extracellular vesicles to eradicate tumor cells. Taxanes-pre-treated T cells may enhance anti-tumor effects while circumventing systemic toxicity.
Collapse
Affiliation(s)
- Ying Xu
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, P.R. China
| | - Yi Xiao
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, P.R. China
| | - Zhi-Ming Shao
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, P.R. China.
| |
Collapse
|