1
|
Chen H, Zhang X, Wang Z, Luo J, Liu Y, Shao R. Activated kynurenine pathway metabolism by YKL-40 establishes an inhibitory immune microenvironment and drives glioblastoma development. Cell Mol Life Sci 2024; 82:11. [PMID: 39718635 DOI: 10.1007/s00018-024-05497-5] [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: 07/09/2024] [Revised: 10/11/2024] [Accepted: 10/28/2024] [Indexed: 12/25/2024]
Abstract
BACKGROUND Glioblastoma (GB) is the stage IV of glioma and mesenchymal GB represents the most common and malignant subtype characterized with elevated expression of a mesenchymal marker YKL-40 and resistance to immune drug therapy. Here, we determined if YKL-40 regulates kynurenine (Kyn) pathway (KP) metabolism that contributes to establishing an immune suppressive microenvironment in GB. METHODS Tumor cells expressing YKL-40 from GB patients were isolated and activated cellular metabolisms were identified via gene microarray analysis. KP metabolism was determined by LC/MS/MS system. Indoleamine 2,3-dioxygenase 1 (IDO1), tryptophan 2,3-dioxygenase (TDO2), their regulatory transcription factors AhR and SRF were evaluated using WB. AhR and SRF transactivity was measured by luciferase reporter gene assays with binding motif mutation, while m6A-mediated AhR and SRF mRNA stability was determined in the presence of an METTL3inhibitor. YKL-40 and Kyn-induced tumor cell migration and CD8+ cytotoxic T cell (CTL) apoptosis were measured in cultured cells. Tumors cells expressing YKL-40 were injected to mouse brains to establish orthotpic tumor models. In GB, YKL-40, IDO1 and TDO2 expression was analyzed for correlation with patient survival. RESULTS KP metabolism was activated in YKL-40-expressing tumor cells. YKL-40 divergently regulated IDO1 and TDO2 via induction of AhR and SRF, respectively. mRNA levels of AhR and SRF were stabilized by decreased METTL3 and YTHDF2. YKL-40 and Kyn secreted from tumor cells and infiltrating M2 macrophages cooperated to enhance tumor cell migration and inhibit CTL immunity. In xenografts, tumors expressing YKL-40 displayed the elevated KP metabolism and macrophage infiltration, but decreased CTLs. Treatment with an anti-PD-1 antibody Tislelizumab significantly increased YKL-40+ mouse survival. In GB, YKL-40 was positively correlated with IDO1 expression and both were associated with decreased survival, whereas IDO1 was negatively correlated with TDO2. CONCLUSION YKL-40 upregulates IDO1 or TDO2 to activate KP metabolism, and coordinates with Kyn to establish an inhibitory tumor immune microenvironment, leading to tumor immune evasion.
Collapse
Affiliation(s)
- Hui Chen
- Shanghai Key Laboratory of Biliary Tract Disease Research, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xuemei Zhang
- Department of Pathology, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, 200080, China
| | - Ziyi Wang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Department of Biliary-Pancreatic Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Jing Luo
- Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Yingbin Liu
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
- Department of Biliary-Pancreatic Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China.
| | - Rong Shao
- Shanghai Key Laboratory of Biliary Tract Disease Research, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China.
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
2
|
Zhang R, Jiang Q, Zhuang Z, Zeng H, Li Y. A bibliometric analysis of drug resistance in immunotherapy for breast cancer: trends, themes, and research focus. Front Immunol 2024; 15:1452303. [PMID: 39188717 PMCID: PMC11345160 DOI: 10.3389/fimmu.2024.1452303] [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: 06/20/2024] [Accepted: 07/24/2024] [Indexed: 08/28/2024] Open
Abstract
While breast cancer treatments have advanced significantly nowadays, yet metastatic, especially triple-negative breast cancer (TNBC), remains challenging with low survival. Cancer immunotherapy, a promising approach for HER2-positive and TNBC, still faces resistance hurdles. Recently, numerous studies have set their sights on the resistance of immunotherapy for breast cancer. Our study provides a thorough comprehension of the current research landscape, hotspots, and emerging breakthroughs in this critical area through a meticulous bibliometric analysis. As of March 26, 2024, a total of 1341 articles on immunology resistance in breast cancer have been gathered from Web of Science Core Collection, including 765 articles and 576 reviews. Bibliometrix, CiteSpace and VOSviewer software were utilized to examine publications and citations per year, prolific countries, contributive institutions, high-level journals and scholars, as well as highly cited articles, references and keywords. The research of immunotherapy resistance in breast cancer has witnessed a remarkable surge over the past seven years. The United States and China have made significant contributions, with Harvard Medical School being the most prolific institution and actively engaging in collaborations. The most contributive author is Curigliano, G from the European Institute of Oncology in Italy, while Wucherpfennig, K. W. from the Dana-Farber Cancer Institute in the USA, had the highest citations. Journals highly productive primarily focus on clinical, immunology and oncology research. Common keywords include "resistance", "expression", "tumor microenvironment", "cancer", "T cell", "therapy", "chemotherapy" and "cell". Current research endeavors to unravel the mechanisms of immune resistance in breast cancer through the integration of bioinformatics, basic experiments, and clinical trials. Efforts are underway to develop strategies that improve the effectiveness of immunotherapy, including the exploration of combination therapies and advancements in drug delivery systems. Additionally, there is a strong focus on identifying novel biomarkers that can predict patient response to immunology. This study will provide researchers with an up-to-date overview of the present knowledge in drug resistance of immunology for breast cancer, serving as a valuable resource for informed decision-making and further research on innovative approaches to address immunotherapy resistance.
Collapse
Affiliation(s)
- Rendong Zhang
- Department of Breast Surgery, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Qiongzhi Jiang
- Department of Radiation Oncology, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Zhemin Zhuang
- Engineering College, Shantou University, Shantou, Guangdong, China
| | - Huancheng Zeng
- Department of Breast Surgery, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Yaochen Li
- The Central Laboratory, Cancer Hospital of Shantou University Medical College, Shantou, China
| |
Collapse
|
3
|
Fan Y, Meng Y, Hu X, Liu J, Qin X. Uncovering novel mechanisms of chitinase-3-like protein 1 in driving inflammation-associated cancers. Cancer Cell Int 2024; 24:268. [PMID: 39068486 PMCID: PMC11282867 DOI: 10.1186/s12935-024-03425-y] [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: 02/25/2024] [Accepted: 07/01/2024] [Indexed: 07/30/2024] Open
Abstract
Chitinase-3-like protein 1 (CHI3L1) is a secreted glycoprotein that is induced and regulated by multiple factors during inflammation in enteritis, pneumonia, asthma, arthritis, and other diseases. It is associated with the deterioration of the inflammatory environment in tissues with chronic inflammation caused by microbial infection or autoimmune diseases. The expression of CHI3L1 expression is upregulated in several malignant tumors, underscoring the crucial role of chronic inflammation in the initiation and progression of cancer. While the precise mechanism connecting inflammation and cancer is unclear, the involvement of CHI3L1 is involved in chronic inflammation, suggesting its role as a contributing factor to in the link between inflammation and cancer. CHI3L1 can aggravate DNA oxidative damage, induce the cancerous phenotype, promote the development of a tumor inflammatory environment and angiogenesis, inhibit immune cells, and promote cancer cell growth, invasion, and migration. Furthermore, it participates in the initiation of cancer progression and metastasis by binding with transmembrane receptors to mediate intracellular signal transduction. Based on the current research on CHI3L1, we explore introduce the receptors that interact with CHI3L1 along with the signaling pathways that may be triggered during chronic inflammation to enhance tumorigenesis and progression. In the last section of the article, we provide a brief overview of anti-inflammatory therapies that target CHI3L1.
Collapse
Affiliation(s)
- Yan Fan
- Department of Laboratory Medicine, Liaoning Clinical Research Center for Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, 110122, China
- Liaoning Clinical Research Center for Laboratory Medicine, Shenyang, Liaoning Province, China
| | - Yuan Meng
- Department of Laboratory Medicine, Liaoning Clinical Research Center for Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, 110122, China
- Liaoning Clinical Research Center for Laboratory Medicine, Shenyang, Liaoning Province, China
| | - Xingwei Hu
- Department of Laboratory Medicine, Liaoning Clinical Research Center for Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, 110122, China
- Liaoning Clinical Research Center for Laboratory Medicine, Shenyang, Liaoning Province, China
| | - Jianhua Liu
- Department of Laboratory Medicine, Liaoning Clinical Research Center for Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, 110122, China
- Liaoning Clinical Research Center for Laboratory Medicine, Shenyang, Liaoning Province, China
| | - Xiaosong Qin
- Department of Laboratory Medicine, Liaoning Clinical Research Center for Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, 110122, China.
- Liaoning Clinical Research Center for Laboratory Medicine, Shenyang, Liaoning Province, China.
| |
Collapse
|
4
|
Tapia IJ, Perico D, Wolos VJ, Villaverde MS, Abrigo M, Di Silvestre D, Mauri P, De Palma A, Fiszman GL. Proteomic Characterization of a 3D HER2+ Breast Cancer Model Reveals the Role of Mitochondrial Complex I in Acquired Resistance to Trastuzumab. Int J Mol Sci 2024; 25:7397. [PMID: 39000504 PMCID: PMC11242363 DOI: 10.3390/ijms25137397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 07/02/2024] [Accepted: 07/02/2024] [Indexed: 07/16/2024] Open
Abstract
HER2-targeted therapies, such as Trastuzumab (Tz), have significantly improved the clinical outcomes for patients with HER2+ breast cancer (BC). However, treatment resistance remains a major obstacle. To elucidate functional and metabolic changes associated with acquired resistance, we characterized protein profiles of BC Tz-responder spheroids (RSs) and non-responder spheroids (nRSs) by a proteomic approach. Three-dimensional cultures were generated from the HER2+ human mammary adenocarcinoma cell line BT-474 and a derived resistant cell line. Before and after a 15-day Tz treatment, samples of each condition were collected and analyzed by liquid chromatography-mass spectrometry. The analysis of differentially expressed proteins exhibited the deregulation of energetic metabolism and mitochondrial pathways. A down-regulation of carbohydrate metabolism and up-regulation of mitochondria organization proteins, the tricarboxylic acid cycle, and oxidative phosphorylation, were observed in nRSs. Of note, Complex I-related proteins were increased in this condition and the inhibition by metformin highlighted that their activity is necessary for nRS survival. Furthermore, a correlation analysis showed that overexpression of Complex I proteins NDUFA10 and NDUFS2 was associated with high clinical risk and worse survival for HER2+ BC patients. In conclusion, the non-responder phenotype identified here provides a signature of proteins and related pathways that could lead to therapeutic biomarker investigation.
Collapse
Affiliation(s)
- Ivana J. Tapia
- Universidad de Buenos Aires, Instituto de Oncología Ángel H. Roffo, Área de Investigación, 5481 San Martín Av., Ciudad Autónoma de Buenos Aires C1417DTB, Argentina; (V.J.W.); (M.S.V.); (M.A.); (G.L.F.)
| | - Davide Perico
- Institute of Biomedical Technologies-National Research Council ITB-CNR, Via Fratelli Cervi 93, 20054 Segrate, Italy; (D.P.); (D.D.S.); (P.M.)
| | - Virginia J. Wolos
- Universidad de Buenos Aires, Instituto de Oncología Ángel H. Roffo, Área de Investigación, 5481 San Martín Av., Ciudad Autónoma de Buenos Aires C1417DTB, Argentina; (V.J.W.); (M.S.V.); (M.A.); (G.L.F.)
| | - Marcela S. Villaverde
- Universidad de Buenos Aires, Instituto de Oncología Ángel H. Roffo, Área de Investigación, 5481 San Martín Av., Ciudad Autónoma de Buenos Aires C1417DTB, Argentina; (V.J.W.); (M.S.V.); (M.A.); (G.L.F.)
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Ciudad Autónoma de Buenos Aires C1425FQB, Argentina
| | - Marianela Abrigo
- Universidad de Buenos Aires, Instituto de Oncología Ángel H. Roffo, Área de Investigación, 5481 San Martín Av., Ciudad Autónoma de Buenos Aires C1417DTB, Argentina; (V.J.W.); (M.S.V.); (M.A.); (G.L.F.)
| | - Dario Di Silvestre
- Institute of Biomedical Technologies-National Research Council ITB-CNR, Via Fratelli Cervi 93, 20054 Segrate, Italy; (D.P.); (D.D.S.); (P.M.)
| | - Pierluigi Mauri
- Institute of Biomedical Technologies-National Research Council ITB-CNR, Via Fratelli Cervi 93, 20054 Segrate, Italy; (D.P.); (D.D.S.); (P.M.)
- Institute of Life Sciences, Sant’Anna School of Advanced Study, 56127 Pisa, Italy
| | - Antonella De Palma
- Institute of Biomedical Technologies-National Research Council ITB-CNR, Via Fratelli Cervi 93, 20054 Segrate, Italy; (D.P.); (D.D.S.); (P.M.)
| | - Gabriel L. Fiszman
- Universidad de Buenos Aires, Instituto de Oncología Ángel H. Roffo, Área de Investigación, 5481 San Martín Av., Ciudad Autónoma de Buenos Aires C1417DTB, Argentina; (V.J.W.); (M.S.V.); (M.A.); (G.L.F.)
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Ciudad Autónoma de Buenos Aires C1425FQB, Argentina
| |
Collapse
|
5
|
Salembier R, De Haes C, Bellemans J, Demeyere K, Van Den Broeck W, Sanders NN, Van Laere S, Lyons TR, Meyer E, Steenbrugge J. Chitin-mediated blockade of chitinase-like proteins reduces tumor immunosuppression, inhibits lymphatic metastasis and enhances anti-PD-1 efficacy in complementary TNBC models. Breast Cancer Res 2024; 26:63. [PMID: 38605414 PMCID: PMC11007917 DOI: 10.1186/s13058-024-01815-8] [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: 08/21/2023] [Accepted: 03/23/2024] [Indexed: 04/13/2024] Open
Abstract
BACKGROUND Chitinase-like proteins (CLPs) play a key role in immunosuppression under inflammatory conditions such as cancer. CLPs are enzymatically inactive and become neutralized upon binding of their natural ligand chitin, potentially reducing CLP-driven immunosuppression. We investigated the efficacy of chitin treatment in the context of triple-negative breast cancer (TNBC) using complementary mouse models. We also evaluated the immunomodulatory influence of chitin on immune checkpoint blockade (ICB) and compared its efficacy as general CLP blocker with blockade of a single CLP, i.e. chitinase 3-like 1 (CHI3L1). METHODS Female BALB/c mice were intraductally injected with luciferase-expressing 4T1 or 66cl4 cells and systemically treated with chitin in combination with or without anti-programmed death (PD)-1 ICB. For single CLP blockade, tumor-bearing mice were treated with anti-CHI3L1 antibodies. Metastatic progression was monitored through bioluminescence imaging. Immune cell changes in primary tumors and lymphoid organs (i.e. axillary lymph nodes and spleen) were investigated through flow cytometry, immunohistochemistry, cytokine profiling and RNA-sequencing. CHI3L1-stimulated RAW264.7 macrophages were subjected to 2D lymphatic endothelial cell adhesion and 3D lymphatic integration in vitro assays for studying macrophage-mediated lymphatic remodeling. RESULTS Chitin significantly reduced primary tumor progression in the 4T1-based model by decreasing the high production of CLPs that originate from tumor-associated neutrophils (TANs) and Stat3 signaling, prominently affecting the CHI3L1 and CHI3L3 primary tumor levels. It reduced immunosuppressive cell types and increased anti-tumorigenic T-cells in primary tumors as well as axillary lymph nodes. Chitin also significantly reduced CHI3L3 primary tumor levels and immunosuppression in the 66cl4-based model. Compared to anti-CHI3L1, chitin enhanced primary tumor growth reduction and anti-tumorigenicity. Both treatments equally inhibited lymphatic adhesion and integration of macrophages, thereby hampering lymphatic tumor cell spreading. Upon ICB combination therapy, chitin alleviated anti-PD-1 resistance in both TNBC models, providing a significant add-on reduction in primary tumor and lung metastatic growth compared to chitin monotherapy. These add-on effects occurred through additional increase in CD8α+ T-cell infiltration and activation in primary tumor and lymphoid organs. CONCLUSIONS Chitin, as a general CLP blocker, reduces CLP production, enhances anti-tumor immunity as well as ICB responses, supporting its potential clinical relevance in immunosuppressed TNBC patients.
Collapse
Affiliation(s)
- Robbe Salembier
- Laboratory of Biochemistry, Department of Veterinary and Biosciences, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Caro De Haes
- Laboratory of Biochemistry, Department of Veterinary and Biosciences, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Julie Bellemans
- Laboratory of Biochemistry, Department of Veterinary and Biosciences, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Kristel Demeyere
- Laboratory of Biochemistry, Department of Veterinary and Biosciences, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Wim Van Den Broeck
- Department of Morphology, Imaging, Orthopedics, Rehabilitation and Nutrition, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Niek N Sanders
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
- Laboratory of Gene Therapy, Department of Veterinary and Biosciences, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Steven Van Laere
- Center for Oncological Research (CORE), Faculty of Medicine and Health Sciences, University of Antwerp, Wilrijk, Belgium
| | - Traci R Lyons
- Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- University of Colorado Cancer Center Young Women's Breast Cancer Translational Program, Aurora, CO, USA
| | - Evelyne Meyer
- Laboratory of Biochemistry, Department of Veterinary and Biosciences, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Jonas Steenbrugge
- Laboratory of Biochemistry, Department of Veterinary and Biosciences, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium.
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium.
- Center for Oncological Research (CORE), Faculty of Medicine and Health Sciences, University of Antwerp, Wilrijk, Belgium.
| |
Collapse
|
6
|
Akinsipe T, Mohamedelhassan R, Akinpelu A, Pondugula SR, Mistriotis P, Avila LA, Suryawanshi A. Cellular interactions in tumor microenvironment during breast cancer progression: new frontiers and implications for novel therapeutics. Front Immunol 2024; 15:1302587. [PMID: 38533507 PMCID: PMC10963559 DOI: 10.3389/fimmu.2024.1302587] [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: 09/26/2023] [Accepted: 02/16/2024] [Indexed: 03/28/2024] Open
Abstract
The breast cancer tumor microenvironment (TME) is dynamic, with various immune and non-immune cells interacting to regulate tumor progression and anti-tumor immunity. It is now evident that the cells within the TME significantly contribute to breast cancer progression and resistance to various conventional and newly developed anti-tumor therapies. Both immune and non-immune cells in the TME play critical roles in tumor onset, uncontrolled proliferation, metastasis, immune evasion, and resistance to anti-tumor therapies. Consequently, molecular and cellular components of breast TME have emerged as promising therapeutic targets for developing novel treatments. The breast TME primarily comprises cancer cells, stromal cells, vasculature, and infiltrating immune cells. Currently, numerous clinical trials targeting specific TME components of breast cancer are underway. However, the complexity of the TME and its impact on the evasion of anti-tumor immunity necessitate further research to develop novel and improved breast cancer therapies. The multifaceted nature of breast TME cells arises from their phenotypic and functional plasticity, which endows them with both pro and anti-tumor roles during tumor progression. In this review, we discuss current understanding and recent advances in the pro and anti-tumoral functions of TME cells and their implications for developing safe and effective therapies to control breast cancer progress.
Collapse
Affiliation(s)
- Tosin Akinsipe
- Department of Biological Sciences, College of Science and Mathematics, Auburn University, Auburn, AL, United States
| | - Rania Mohamedelhassan
- Department of Chemical Engineering, College of Engineering, Auburn University, Auburn, AL, United States
| | - Ayuba Akinpelu
- Department of Anatomy, Physiology, and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL, United States
| | - Satyanarayana R. Pondugula
- Department of Chemical Engineering, College of Engineering, Auburn University, Auburn, AL, United States
| | - Panagiotis Mistriotis
- Department of Anatomy, Physiology, and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL, United States
| | - L. Adriana Avila
- Department of Biological Sciences, College of Science and Mathematics, Auburn University, Auburn, AL, United States
| | - Amol Suryawanshi
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL, United States
| |
Collapse
|
7
|
Boje AS, Pekar L, Koep K, Lipinski B, Rabinovich B, Evers A, Gehlert CL, Krohn S, Xiao Y, Krah S, Zaynagetdinov R, Toleikis L, Poetzsch S, Peipp M, Zielonka S, Klausz K. Impact of antibody architecture and paratope valency on effector functions of bispecific NKp30 x EGFR natural killer cell engagers. MAbs 2024; 16:2315640. [PMID: 38372053 PMCID: PMC10877975 DOI: 10.1080/19420862.2024.2315640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 02/02/2024] [Indexed: 02/20/2024] Open
Abstract
Natural killer (NK) cells emerged as a promising effector population that can be harnessed for anti-tumor therapy. In this work, we constructed NK cell engagers (NKCEs) based on NKp30-targeting single domain antibodies (sdAbs) that redirect the cytotoxic potential of NK cells toward epidermal growth factor receptor (EGFR)-expressing tumor cells. We investigated the impact of crucial parameters such as sdAb location, binding valencies, the targeted epitope on NKp30, and the overall antibody architecture on the redirection capacity. Our study exploited two NKp30-specific sdAbs, one of which binds a similar epitope on NKp30 as its natural ligand B7-H6, while the other sdAb addresses a non-competing epitope. For EGFR-positive tumor targeting, humanized antigen-binding domains of therapeutic antibody cetuximab were used. We demonstrate that NKCEs bivalently targeting EGFR and bivalently engaging NKp30 are superior to monovalent NKCEs in promoting NK cell-mediated tumor cell lysis and that the architecture of the NKCE can substantially influence killing capacities depending on the NKp30-targeting sdAb utilized. While having a pronounced impact on NK cell killing efficacy, the capabilities of triggering antibody-dependent cellular phagocytosis or complement-dependent cytotoxicity were not significantly affected comparing the bivalent IgG-like NKCEs with cetuximab. However, the fusion of sdAbs can have a slight impact on the NK cell release of immunomodulatory cytokines, as well as on the pharmacokinetic profile of the NKCE due to unfavorable spatial orientation within the molecule architecture. Ultimately, our findings reveal novel insights for the engineering of potent NKCEs triggering the NKp30 axis.
Collapse
Affiliation(s)
- Ammelie Svea Boje
- Division of Antibody-Based Immunotherapy, Department of Internal Medicine II, University Medical Center Schleswig-Holstein and University of Kiel, Kiel, Germany
| | - Lukas Pekar
- Antibody Discovery & Protein Engineering, Merck Healthcare KGaA, Darmstadt, Germany
| | - Katharina Koep
- Drug Metabolism and Pharmacokinetics, Merck Healthcare KGaA, Darmstadt, Germany
| | - Britta Lipinski
- Antibody Discovery & Protein Engineering, Merck Healthcare KGaA, Darmstadt, Germany
| | - Brian Rabinovich
- Department of Oncology and Immuno-Oncology, EMD Serono Research & Development Institute Inc, 45A Middlesex Turnpike, Billerica, MA, USA
| | - Andreas Evers
- Antibody Discovery & Protein Engineering, Merck Healthcare KGaA, Darmstadt, Germany
| | - Carina Lynn Gehlert
- Division of Antibody-Based Immunotherapy, Department of Internal Medicine II, University Medical Center Schleswig-Holstein and University of Kiel, Kiel, Germany
| | - Steffen Krohn
- Division of Antibody-Based Immunotherapy, Department of Internal Medicine II, University Medical Center Schleswig-Holstein and University of Kiel, Kiel, Germany
| | - Yanping Xiao
- Department of Oncology and Immuno-Oncology, EMD Serono Research & Development Institute Inc, 45A Middlesex Turnpike, Billerica, MA, USA
| | - Simon Krah
- Antibody Discovery & Protein Engineering, Merck Healthcare KGaA, Darmstadt, Germany
| | - Rinat Zaynagetdinov
- Department of Oncology and Immuno-Oncology, EMD Serono Research & Development Institute Inc, 45A Middlesex Turnpike, Billerica, MA, USA
| | - Lars Toleikis
- Early Protein Supply & Characterization, Merck Healthcare KGaA, Darmstadt, Germany
| | - Sven Poetzsch
- Strategic Innovation, Merck Healthcare KGaA, Darmstadt, Germany
| | - Matthias Peipp
- Division of Antibody-Based Immunotherapy, Department of Internal Medicine II, University Medical Center Schleswig-Holstein and University of Kiel, Kiel, Germany
| | - Stefan Zielonka
- Antibody Discovery & Protein Engineering, Merck Healthcare KGaA, Darmstadt, Germany
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Darmstadt, Germany
| | - Katja Klausz
- Division of Antibody-Based Immunotherapy, Department of Internal Medicine II, University Medical Center Schleswig-Holstein and University of Kiel, Kiel, Germany
| |
Collapse
|
8
|
Díaz-Basabe A, Lattanzi G, Perillo F, Amoroso C, Baeri A, Farini A, Torrente Y, Penna G, Rescigno M, Ghidini M, Cassinotti E, Baldari L, Boni L, Vecchi M, Caprioli F, Facciotti F, Strati F. Porphyromonas gingivalis fuels colorectal cancer through CHI3L1-mediated iNKT cell-driven immune evasion. Gut Microbes 2024; 16:2388801. [PMID: 39132842 PMCID: PMC11321422 DOI: 10.1080/19490976.2024.2388801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 07/19/2024] [Accepted: 07/31/2024] [Indexed: 08/13/2024] Open
Abstract
The interaction between the gut microbiota and invariant Natural Killer T (iNKT) cells plays a pivotal role in colorectal cancer (CRC). The pathobiont Fusobacterium nucleatum influences the anti-tumor functions of CRC-infiltrating iNKT cells. However, the impact of other bacteria associated with CRC, like Porphyromonas gingivalis, on their activation status remains unexplored. In this study, we demonstrate that mucosa-associated P. gingivalis induces a protumour phenotype in iNKT cells, subsequently influencing the composition of mononuclear-phagocyte cells within the tumor microenvironment. Mechanistically, in vivo and in vitro experiments showed that P. gingivalis reduces the cytotoxic functions of iNKT cells, hampering the iNKT cell lytic machinery through increased expression of chitinase 3-like-1 protein (CHI3L1). Neutralization of CHI3L1 effectively restores iNKT cell cytotoxic functions suggesting a therapeutic potential to reactivate iNKT cell-mediated antitumour immunity. In conclusion, our data demonstrate how P. gingivalis accelerates CRC progression by inducing the upregulation of CHI3L1 in iNKT cells, thus impairing their cytotoxic functions and promoting host tumor immune evasion.
Collapse
Affiliation(s)
- Angélica Díaz-Basabe
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, Milan, Italy
| | - Georgia Lattanzi
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy
- Gastroenterology and Endoscopy Unit, Fondazione IRCCS Cà Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Federica Perillo
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, Milan, Italy
| | - Chiara Amoroso
- Gastroenterology and Endoscopy Unit, Fondazione IRCCS Cà Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Alberto Baeri
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milan, Italy
| | - Andrea Farini
- Neurology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Yvan Torrente
- Neurology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
- Centro Dino Ferrari, Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milano, Italy
| | - Giuseppe Penna
- IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Maria Rescigno
- IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Michele Ghidini
- Medical Oncology, Fondazione IRCCS Ca’ Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Elisa Cassinotti
- Department of General and Minimally Invasive Surgery, Fondazione IRCCS Ca’ Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Ludovica Baldari
- Department of General and Minimally Invasive Surgery, Fondazione IRCCS Ca’ Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Luigi Boni
- Department of General and Minimally Invasive Surgery, Fondazione IRCCS Ca’ Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Maurizio Vecchi
- Gastroenterology and Endoscopy Unit, Fondazione IRCCS Cà Granda, Ospedale Maggiore Policlinico, Milan, Italy
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy
| | - Flavio Caprioli
- Gastroenterology and Endoscopy Unit, Fondazione IRCCS Cà Granda, Ospedale Maggiore Policlinico, Milan, Italy
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy
| | - Federica Facciotti
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milan, Italy
| | - Francesco Strati
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milan, Italy
| |
Collapse
|
9
|
Ji S, Yu H, Zhou D, Fan X, Duan Y, Tan Y, Lang M, Shao G. Cancer stem cell-derived CHI3L1 activates the MAF/CTLA4 signaling pathway to promote immune escape in triple-negative breast cancer. J Transl Med 2023; 21:721. [PMID: 37838657 PMCID: PMC10576881 DOI: 10.1186/s12967-023-04532-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Accepted: 09/17/2023] [Indexed: 10/16/2023] Open
Abstract
BACKGROUND Triple-negative breast cancer (TNBC) development may be associated with tumor immune escape. This study explores whether the CHI3L1/MAF/CTLA4/S100A4 axis affects immune escape in TNBC through interplay with triple-negative breast cancer stem cells (TN-BCSCs). OBJECTIVE The aim of this study is to utilize single-cell transcriptome sequencing (scRNA-seq) to uncover the molecular mechanisms by which the CHI3L1/MAF/CTLA4 signaling pathway may mediate immune evasion in triple-negative breast cancer through the interaction between tumor stem cells (CSCs) and immune cells. METHODS Cell subsets in TNBC tissues were obtained through scRNA-seq, followed by screening differentially expressed genes in TN-BCSCs and B.C.s (CD44+ and CD24-) and predicting the transcription factor regulated by CHI3L1. Effect of CHI3L1 on the stemness phenotype of TNBC cells investigated. Effects of BCSCs-231-derived CHI3L1 on CTLA4 expression in T cells were explored after co-culture of BCSCs-231 cells obtained from microsphere culture of TN-BCSCs with T cells. BCSCs-231-treated T cells were co-cultured with CD8+ T cells to explore the resultant effect on T cell cytotoxicity. An orthotopic B.C. transplanted tumor model in mice with humanized immune systems was constructed, in which the Role of CHI3L1/MAF/CTLA4 in the immune escape of TNBC was explored. RESULTS Eight cell subsets were found in the TNBC tissues, and the existence of TN-BCSCs was observed in the epithelial cell subset. CHI3L1 was related to the stemness phenotype of TNBC cells. TN-BCSC-derived CHI3L1 increased CTLA4 expression in T cells through MAF, inhibiting CD8+ T cell cytotoxicity and inducing immunosuppression. Furthermore, the CTLA4+ T cells might secrete S100A4 to promote the stemness phenotype of TNBC cells. CONCLUSIONS TN-BCSC-derived CHI3L1 upregulates CTLA4 expression in T cells through MAF, suppressing the function of CD8+ T cells, which promotes the immune escape of TNBC.
Collapse
Affiliation(s)
- Shufeng Ji
- Special Medical Service Center, General Surgery, Zhujiang Hospital of Southern Medical University, No. 253, Middle Gongye Avenue, Haizhu District, Guangzhou, 510280, Guangdong, People's Republic of China
| | - Hao Yu
- Special Medical Service Center, General Surgery, Zhujiang Hospital of Southern Medical University, No. 253, Middle Gongye Avenue, Haizhu District, Guangzhou, 510280, Guangdong, People's Republic of China
| | - Dan Zhou
- Department of Breast Surgery, The First People's Hospital of Foshan, Foshan, 528000, People's Republic of China
| | - Xulong Fan
- Department of Breast Surgery, Maternity and Children's Healthcare Hospital of Foshan, Foshan, 528000, People's Republic of China
| | - Yan Duan
- Special Medical Service Center, General Surgery, Zhujiang Hospital of Southern Medical University, No. 253, Middle Gongye Avenue, Haizhu District, Guangzhou, 510280, Guangdong, People's Republic of China
| | - Yijiang Tan
- Special Medical Service Center, General Surgery, Zhujiang Hospital of Southern Medical University, No. 253, Middle Gongye Avenue, Haizhu District, Guangzhou, 510280, Guangdong, People's Republic of China
| | - Min Lang
- Special Medical Service Center, General Surgery, Zhujiang Hospital of Southern Medical University, No. 253, Middle Gongye Avenue, Haizhu District, Guangzhou, 510280, Guangdong, People's Republic of China
| | - Guoli Shao
- Special Medical Service Center, General Surgery, Zhujiang Hospital of Southern Medical University, No. 253, Middle Gongye Avenue, Haizhu District, Guangzhou, 510280, Guangdong, People's Republic of China.
| |
Collapse
|
10
|
Johansen AZ, Novitski SI, Hjaltelin JX, Theile S, Boisen MK, Brunak S, Madsen DH, Nielsen DL, Chen IM. Plasma YKL-40 is associated with prognosis in patients with metastatic pancreatic cancer receiving immune checkpoint inhibitors in combination with radiotherapy. Front Immunol 2023; 14:1228907. [PMID: 37744345 PMCID: PMC10513102 DOI: 10.3389/fimmu.2023.1228907] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 08/22/2023] [Indexed: 09/26/2023] Open
Abstract
Background YKL-40, also known as chitinase-3-like protein 1 (CHI3L1), is a secreted glycoprotein produced by various cell types including stromal, immune, and cancer cells. It contributes to cancer progression through tumor-promoting inflammation and has been shown to inhibit the cytotoxicity of T and NK lymphocytes. In vivo studies have demonstrated synergistic anti-cancer effects of blocking YKL-40 in combination with immune checkpoint inhibitors (ICIs). Biomarkers for the prediction of the response to ICIs are highly needed. We investigated the association between plasma YKL-40 and clinical benefit and survival in patients with metastatic pancreatic cancer (mPC) receiving ICIs and stereotactic body radiotherapy (SBRT). Methods Blood samples were collected from 84 patients with mPC who participated in the randomized phase II CheckPAC study, in which patients received nivolumab with or without ipilimumab combined with a single fraction of SBRT. Plasma YKL-40 was measured using a commercial ELISA kit. Results Elevated baseline plasma YKL-40 was an independent predictor of shorter overall survival (OS) (HR 2.19, 95% CI 1.21-3.95). A ≥ 40% decrease in plasma YKL-40 during treatment was associated with longer progression-free survival (p = 0.009) and OS (p = 0.0028). There was no correlation between plasma YKL-40 and the tumor burden marker CA19-9 at baseline or during treatment. Conclusion This study contributes new knowledge regarding YKL-40 as a predictor of clinical benefit from ICIs and radiotherapy. These exploratory results warrant further investigation of YKL-40 as a biomarker for patients treated with immunotherapies. Clinical trial registration Clinicaltrials.gov, identifier NCT02866383.
Collapse
Affiliation(s)
- Astrid Z. Johansen
- National Center for Cancer Immune Therapy, Department of Oncology, Copenhagen University Hospital – Herlev and Gentofte, Herlev, Denmark
| | - Sif I. Novitski
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jessica X. Hjaltelin
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Susann Theile
- Department of Oncology, Copenhagen University Hospital – Herlev and Gentofte, Herlev, Denmark
| | - Mogens K. Boisen
- Department of Oncology, Copenhagen University Hospital – Herlev and Gentofte, Herlev, Denmark
| | - Søren Brunak
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Daniel H. Madsen
- National Center for Cancer Immune Therapy, Department of Oncology, Copenhagen University Hospital – Herlev and Gentofte, Herlev, Denmark
- Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Dorte L. Nielsen
- Department of Oncology, Copenhagen University Hospital – Herlev and Gentofte, Herlev, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Inna M. Chen
- Department of Oncology, Copenhagen University Hospital – Herlev and Gentofte, Herlev, Denmark
| |
Collapse
|
11
|
Dumontet C, Reichert JM, Senter PD, Lambert JM, Beck A. Antibody-drug conjugates come of age in oncology. Nat Rev Drug Discov 2023; 22:641-661. [PMID: 37308581 DOI: 10.1038/s41573-023-00709-2] [Citation(s) in RCA: 197] [Impact Index Per Article: 98.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/20/2023] [Indexed: 06/14/2023]
Abstract
Antibody-drug conjugates (ADCs) combine the specificity of monoclonal antibodies with the potency of highly cytotoxic agents, potentially reducing the severity of side effects by preferentially targeting their payload to the tumour site. ADCs are being increasingly used in combination with other agents, including as first-line cancer therapies. As the technology to produce these complex therapeutics has matured, many more ADCs have been approved or are in late-phase clinical trials. The diversification of antigenic targets as well as bioactive payloads is rapidly broadening the scope of tumour indications for ADCs. Moreover, novel vector protein formats as well as warheads targeting the tumour microenvironment are expected to improve the intratumour distribution or activation of ADCs, and consequently their anticancer activity for difficult-to-treat tumour types. However, toxicity remains a key issue in the development of these agents, and better understanding and management of ADC-related toxicities will be essential for further optimization. This Review provides a broad overview of the recent advances and challenges in ADC development for cancer treatment.
Collapse
Affiliation(s)
- Charles Dumontet
- CRCL INSERM 1052/CNRS 5286, University of Lyon, Hospices Civils de Lyon, Lyon, France.
| | | | | | | | - Alain Beck
- Institut de Recherche Pierre Fabre, CIPF, Saint-Julien-en-Genevois, France
| |
Collapse
|
12
|
Lipinski B, Arras P, Pekar L, Klewinghaus D, Boje AS, Krah S, Zimmermann J, Klausz K, Peipp M, Siegmund V, Evers A, Zielonka S. NKp46-specific single domain antibodies enable facile engineering of various potent NK cell engager formats. Protein Sci 2023; 32:e4593. [PMID: 36775946 PMCID: PMC9951198 DOI: 10.1002/pro.4593] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 02/07/2023] [Accepted: 02/09/2023] [Indexed: 02/14/2023]
Abstract
Herein, we describe the generation of potent NK cell engagers (NKCEs) based on single domain antibodies (sdAbs) specific for NKp46 harboring the humanized Fab version of Cetuximab for tumor targeting. After immunization of camelids, a plethora of different VHH domains were retrieved by yeast surface display. Upon reformatting into Fc effector-silenced NKCEs targeting NKp46 and EGFR in a strictly monovalent fashion, the resulting bispecific antibodies elicited potent NK cell-mediated killing of EGFR-overexpressing tumor cells with potencies (EC50 killing) in the picomolar range. This was further augmented via co-engagement of Fcγ receptor IIIa (FcγRIIIa). Importantly, NKp46-specific sdAbs enabled the construction of various NKCE formats with different geometries and valencies which displayed favorable biophysical and biochemical properties without further optimization. By this means, killing capacities were further improved significantly. Hence, NKp46-specific sdAbs are versatile building blocks for the construction of different NKCE formats.
Collapse
Affiliation(s)
- Britta Lipinski
- Protein Engineering and Antibody TechnologiesMerck Healthcare KGaADarmstadtGermany
- Institute for Organic Chemistry and BiochemistryTechnical University of DarmstadtDarmstadtGermany
| | - Paul Arras
- Protein Engineering and Antibody TechnologiesMerck Healthcare KGaADarmstadtGermany
| | - Lukas Pekar
- Protein Engineering and Antibody TechnologiesMerck Healthcare KGaADarmstadtGermany
| | - Daniel Klewinghaus
- Protein Engineering and Antibody TechnologiesMerck Healthcare KGaADarmstadtGermany
| | - Ammelie Svea Boje
- Division of Antibody‐Based Immunotherapy, Department of Internal Medicine IIUniversity Hospital Schleswig‐Holstein and Christian‐Albrechts‐University KielKielGermany
| | - Simon Krah
- Protein Engineering and Antibody TechnologiesMerck Healthcare KGaADarmstadtGermany
| | - Jasmin Zimmermann
- Protein Engineering and Antibody TechnologiesMerck Healthcare KGaADarmstadtGermany
- Institute for Organic Chemistry and BiochemistryTechnical University of DarmstadtDarmstadtGermany
| | - Katja Klausz
- Division of Antibody‐Based Immunotherapy, Department of Internal Medicine IIUniversity Hospital Schleswig‐Holstein and Christian‐Albrechts‐University KielKielGermany
| | - Matthias Peipp
- Division of Antibody‐Based Immunotherapy, Department of Internal Medicine IIUniversity Hospital Schleswig‐Holstein and Christian‐Albrechts‐University KielKielGermany
| | | | - Andreas Evers
- Computational Chemistry and BiologyMerck Healthcare KGaADarmstadtGermany
| | - Stefan Zielonka
- Protein Engineering and Antibody TechnologiesMerck Healthcare KGaADarmstadtGermany
- Institute for Organic Chemistry and BiochemistryTechnical University of DarmstadtDarmstadtGermany
| |
Collapse
|
13
|
Chan AML, Cheah JM, Lokanathan Y, Ng MH, Law JX. Natural Killer Cell-Derived Extracellular Vesicles as a Promising Immunotherapeutic Strategy for Cancer: A Systematic Review. Int J Mol Sci 2023; 24:ijms24044026. [PMID: 36835438 PMCID: PMC9964266 DOI: 10.3390/ijms24044026] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 01/30/2023] [Accepted: 02/03/2023] [Indexed: 02/19/2023] Open
Abstract
Cancer is the second leading contributor to global deaths caused by non-communicable diseases. The cancer cells are known to interact with the surrounding non-cancerous cells, including the immune cells and stromal cells, within the tumor microenvironment (TME) to modulate the tumor progression, metastasis and resistance. Currently, chemotherapy and radiotherapy are the standard treatments for cancers. However, these treatments cause a significant number of side effects, as they damage both the cancer cells and the actively dividing normal cells indiscriminately. Hence, a new generation of immunotherapy using natural killer (NK) cells, cytotoxic CD8+ T-lymphocytes or macrophages was developed to achieve tumor-specific targeting and circumvent the adverse effects. However, the progression of cell-based immunotherapy is hindered by the combined action of TME and TD-EVs, which render the cancer cells less immunogenic. Recently, there has been an increase in interest in using immune cell derivatives to treat cancers. One of the highly potential immune cell derivatives is the NK cell-derived EVs (NK-EVs). As an acellular product, NK-EVs are resistant to the influence of TME and TD-EVs, and can be designed for "off-the-shelf" use. In this systematic review, we examine the safety and efficacy of NK-EVs to treat various cancers in vitro and in vivo.
Collapse
Affiliation(s)
- Alvin Man Lung Chan
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
- Ming Medical Sdn Bhd, D3-3 (2nd Floor), Block D3 Dana 1 Commercial Centre, Jalan PJU 1a/22, Petaling Jaya 47101, Malaysia
| | - Jin Min Cheah
- Ming Medical Sdn Bhd, D3-3 (2nd Floor), Block D3 Dana 1 Commercial Centre, Jalan PJU 1a/22, Petaling Jaya 47101, Malaysia
| | - Yogeswaran Lokanathan
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| | - Min Hwei Ng
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| | - Jia Xian Law
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
- Correspondence: ; Tel.: +60-391-457677
| |
Collapse
|
14
|
Li F, Liu S. Focusing on NK cells and ADCC: A promising immunotherapy approach in targeted therapy for HER2-positive breast cancer. Front Immunol 2022; 13:1083462. [PMID: 36601109 PMCID: PMC9806173 DOI: 10.3389/fimmu.2022.1083462] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Accepted: 12/05/2022] [Indexed: 12/23/2022] Open
Abstract
Human epidermal growth factor receptor 2 (HER2)-positive breast cancer has a high metastatic potential. Monoclonal antibodies (mAbs) that target HER2, such as trastuzumab and pertuzumab, are the cornerstone of adjuvant therapy for HER2-positive breast cancer. A growing body of preclinical and clinical evidence points to the importance of innate immunity mediated by antibody-dependent cellular cytotoxicity (ADCC) in the clinical effect of mAbs on the resulting anti-tumor response. In this review, we provide an overview of the role of natural killer (NK) cells and ADCC in targeted therapy of HER2-positive breast cancer, including the biological functions of NK cells and the role of NK cells and ADCC in anti-HER2 targeted drugs. We then discuss regulatory mechanisms and recent strategies to leverage our knowledge of NK cells and ADCC as an immunotherapy approach for HER2-positive breast cancer.
Collapse
|
15
|
Zhan M, Guo Y, Shen M, Shi X. Nanomaterial‐Boosted Tumor Immunotherapy Through Natural Killer Cells. ADVANCED NANOBIOMED RESEARCH 2022; 2. [DOI: 10.1002/anbr.202200096] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025] Open
Abstract
Natural killer (NK)‐cell immunotherapy as an alternative to T‐cell immunotherapy has been widely used in clinical cell immunotherapy of various tumors. Despite the surprising findings, the widespread applications of NK cells are still limited by the insufficient expansion and short lifespan of adoptive NK cells in vivo, the poor penetration of NK cells in solid tumors, as well as the immunosuppressive tumor microenvironment that may cause the inactivation of NK cells. Fortunately, the emergence of nanomaterials provides many opportunities to address these vexing problems, thus overcoming the barriers faced by NK cells and promoting the tumor inhibitory efficacy of NK cells. Herein, the recent advances in the rational design of nanomaterials for boosting the NK cell‐based immunotherapy, mainly through enhancing NK cell engagement with tumors, boosting NK cell activation or expansion, as well as redirecting NK cells to tumor cells, are reviewed. Lastly, the design and preparation of next‐generation nanomaterials that aim to further boost the NK cell‐based immunotherapy are briefly discussed.
Collapse
Affiliation(s)
- Mengsi Zhan
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine College of Biological Science and Medical Engineering Donghua University Shanghai 201620 P.R. China
| | - Yunqi Guo
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine College of Biological Science and Medical Engineering Donghua University Shanghai 201620 P.R. China
| | - Mingwu Shen
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine College of Biological Science and Medical Engineering Donghua University Shanghai 201620 P.R. China
| | - Xiangyang Shi
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine College of Biological Science and Medical Engineering Donghua University Shanghai 201620 P.R. China
| |
Collapse
|
16
|
Resistance to Trastuzumab. Cancers (Basel) 2022; 14:cancers14205115. [PMID: 36291900 PMCID: PMC9600208 DOI: 10.3390/cancers14205115] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Revised: 10/12/2022] [Accepted: 10/17/2022] [Indexed: 11/17/2022] Open
Abstract
Simple Summary Trastuzumab is a humanized antibody that has significantly improved the management and treatment outcomes of patients with cancers that overexpress HER2. Many research groups, both in academia and industry, have contributed towards understanding the various mechanisms engaged by trastuzumab to mediate its anti-tumor effects. Nevertheless, data from several clinical studies have indicated that a significant proportion of patients exhibit primary or acquired resistance to trastuzumab therapy. In this article, we discuss underlying mechanisms that contribute towards to resistance. Furthermore, we discuss the potential strategies to overcome some of the mechanisms of resistance to enhance the therapeutic efficacy of trastuzumab and other therapies based on it. Abstract One of the most impactful biologics for the treatment of breast cancer is the humanized monoclonal antibody, trastuzumab, which specifically recognizes the HER2/neu (HER2) protein encoded by the ERBB2 gene. Useful for both advanced and early breast cancers, trastuzumab has multiple mechanisms of action. Classical mechanisms attributed to trastuzumab action include cell cycle arrest, induction of apoptosis, and antibody-dependent cell-mediated cytotoxicity (ADCC). Recent studies have identified the role of the adaptive immune system in the clinical actions of trastuzumab. Despite the multiple mechanisms of action, many patients demonstrate resistance, primary or adaptive. Newly identified molecular and cellular mechanisms of trastuzumab resistance include induction of immune suppression, vascular mimicry, generation of breast cancer stem cells, deregulation of long non-coding RNAs, and metabolic escape. These newly identified mechanisms of resistance are discussed in detail in this review, particularly considering how they may lead to the development of well-rationalized, patient-tailored combinations that improve patient survival.
Collapse
|
17
|
Stock S, Benmebarek MR, Kluever AK, Darowski D, Jost C, Stubenrauch KG, Benz J, Freimoser-Grundschober A, Moessner E, Umana P, Subklewe M, Endres S, Klein C, Kobold S. Chimeric antigen receptor T cells engineered to recognize the P329G-mutated Fc part of effector-silenced tumor antigen-targeting human IgG1 antibodies enable modular targeting of solid tumors. J Immunother Cancer 2022; 10:jitc-2022-005054. [PMID: 35902133 PMCID: PMC9341194 DOI: 10.1136/jitc-2022-005054] [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] [Accepted: 06/26/2022] [Indexed: 11/03/2022] Open
Abstract
BACKGROUND Chimeric antigen receptor (CAR) T cell therapy has proven its clinical utility in hematological malignancies. Optimization is still required for its application in solid tumors. Here, the lack of cancer-specific structures along with tumor heterogeneity represent a critical barrier to safety and efficacy. Modular CAR T cells indirectly binding the tumor antigen through CAR-adaptor molecules have the potential to reduce adverse events and to overcome antigen heterogeneity. We hypothesized that a platform utilizing unique traits of clinical grade antibodies for selective CAR targeting would come with significant advantages. Thus, we developed a P329G-directed CAR targeting the P329G mutation in the Fc part of tumor-targeting human antibodies containing P329G L234A/L235A (LALA) mutations for Fc silencing. METHODS A single chain variable fragment-based second generation P329G-targeting CAR was retrovirally transduced into primary human T cells. These CAR T cells were combined with IgG1 antibodies carrying P329G LALA mutations in their Fc part targeting epidermal growth factor receptor (EGFR), mesothelin (MSLN) or HER2/neu. Mesothelioma, pancreatic and breast cancer cell lines expressing the respective antigens were used as target cell lines. Efficacy was evaluated in vitro and in vivo in xenograft mouse models. RESULTS Unlike CD16-CAR T cells, which bind human IgG in a non-selective manner, P329G-targeting CAR T cells revealed specific effector functions only when combined with antibodies carrying P329G LALA mutations in their Fc part. P329G-targeting CAR T cells cannot be activated by an excess of human IgG. P329G-directed CAR T cells combined with a MSLN-targeting P329G-mutated antibody mediated pronounced in vitro and in vivo antitumor efficacy in mesothelioma and pancreatic cancer models. Combined with a HER2-targeting antibody, P329G-targeting CAR T cells showed substantial in vitro activation, proliferation, cytokine production and cytotoxicity against HER2-expressing breast cancer cell lines and induced complete tumor eradication in a breast cancer xenograft mouse model. The ability of the platform to target multiple antigens sequentially was shown in vitro and in vivo. CONCLUSIONS P329G-targeting CAR T cells combined with antigen-binding human IgG1 antibodies containing the P329G Fc mutation mediate pronounced in vitro and in vivo effector functions in different solid tumor models, warranting further clinical translation of this concept.
Collapse
Affiliation(s)
- Sophia Stock
- Department of Medicine IV, Division of Clinical Pharmacology, University Hospital, Ludwig Maximilian University (LMU), Munich, Germany .,Department of Medicine III, University Hospital, Ludwig Maximilian University (LMU), Munich, Germany.,German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - Mohamed-Reda Benmebarek
- Department of Medicine IV, Division of Clinical Pharmacology, University Hospital, Ludwig Maximilian University (LMU), Munich, Germany.,National Cancer Institute (NCI), Bethesda, Maryland, USA
| | - Anna-Kristina Kluever
- Department of Medicine IV, Division of Clinical Pharmacology, University Hospital, Ludwig Maximilian University (LMU), Munich, Germany
| | - Diana Darowski
- Roche Innovation Center Zurich, Roche Pharma Research & Early Development, Schlieren, Switzerland.,Innovent Biologics (Suzhou) Co., Ltd, Suzhou, Jiangsu, China
| | - Christian Jost
- Roche Innovation Center Zurich, Roche Pharma Research & Early Development, Schlieren, Switzerland.,Athebio AG, Schlieren, Switzerland
| | | | - Joerg Benz
- Roche Innovation Center Basel, Basel, Switzerland
| | | | - Ekkehard Moessner
- Roche Innovation Center Zurich, Roche Pharma Research & Early Development, Schlieren, Switzerland
| | - Pablo Umana
- Roche Innovation Center Zurich, Roche Pharma Research & Early Development, Schlieren, Switzerland
| | - Marion Subklewe
- Department of Medicine III, University Hospital, Ludwig Maximilian University (LMU), Munich, Germany.,German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - Stefan Endres
- Department of Medicine IV, Division of Clinical Pharmacology, University Hospital, Ludwig Maximilian University (LMU), Munich, Germany.,German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany.,Einheit für Klinische Pharmakologie (EKLiP), Helmholtz Zentrum München, German Research Center for Environmental Health (HMGU), Neuherberg, Germany
| | - Christian Klein
- Roche Innovation Center Zurich, Roche Pharma Research & Early Development, Schlieren, Switzerland
| | - Sebastian Kobold
- Department of Medicine IV, Division of Clinical Pharmacology, University Hospital, Ludwig Maximilian University (LMU), Munich, Germany .,German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany.,Einheit für Klinische Pharmakologie (EKLiP), Helmholtz Zentrum München, German Research Center for Environmental Health (HMGU), Neuherberg, Germany
| |
Collapse
|
18
|
Wotschel N, Montero PO, Wels WS, Tonn T, Eitler J. Live-cell imaging for analysis of the NK cell immunological synapse. Methods Cell Biol 2022. [PMID: 37516519 DOI: 10.1016/bs.mcb.2022.09.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The immunological synapse (IS) between NK cells and cancer cells is instrumental for the initiation of tumor-specific cytotoxicity. Improper function of processes at the IS can lead to NK cell unresponsiveness, contributing to tumor immune escape. Critical steps at the IS include target cell recognition, conjugation of NK cell and cancer cell, cytotoxic granule convergence to the microtubule-organizing center (MTOC), granule polarization to the IS, and degranulation. Here, we describe confocal live-cell imaging methods for the analysis of these processes at the immunological synapse, with a focus on mechanisms of cancer cell resistance facilitating escape from NK cell cytotoxicity.
Collapse
|