1
|
Zhang Z, Wang R, Chen L. Drug Delivery System Targeting Cancer-Associated Fibroblast for Improving Immunotherapy. Int J Nanomedicine 2025; 20:483-503. [PMID: 39816375 PMCID: PMC11734509 DOI: 10.2147/ijn.s500591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Accepted: 12/16/2024] [Indexed: 01/18/2025] Open
Abstract
Cancer-associated fibroblasts (CAFs) are a heterogeneous population of non-malignant cells that play a crucial role in the tumor microenvironment, increasingly recognized as key contributors to cancer progression, metastasis, and treatment resistance. So, targeting CAFs has always been considered an important part of cancer immunotherapy. However, targeting CAFs to improve the efficacy of tumor therapy is currently a major challenge. Nanomaterials show their unique advantages in the whole process. At present, nanomaterials have achieved significant accomplishments in medical applications, particularly in the field of cancer-targeted therapy, showing enormous potential. It has been confirmed that nanomaterials can not only directly target CAFs, but also interact with the tumor microenvironment (TME) and immune cells to affect tumorigenesis. As for the cancer treatment, nanomaterials could enhance the therapeutic effect in many ways. Therefore, in this review, we first summarized the current understanding of the complex interactions between CAFs and TME, immune cells, and tumor cells. Next, we discussed common nanomaterials in modern medicine and their respective impacts on the TME, CAFs, and interactions with tumors. Finally, we focus on the application of nano drug delivery system targeting CAFs in cancer therapy.
Collapse
Affiliation(s)
- Zhongsong Zhang
- School of Clinical Medicine, Chengdu Medical College, Chengdu, 610550, People's Republic of China
| | - Rong Wang
- School of Clinical Medicine, Chengdu Medical College, Chengdu, 610550, People's Republic of China
| | - Long Chen
- School of Basic Medical Sciences, Chengdu Medical College, Chengdu, 610550, People's Republic of China
| |
Collapse
|
2
|
Guinn S, Perez B, Tandurella JA, Ramani M, Lee JW, Zabransky DJ, Kartalia E, Patel J, Zlomke H, Nicolson N, Shin S, Barrett B, Sun N, Hernandez A, Coyne E, Cannon C, Gross NE, Charmsaz S, Cho Y, Leatherman J, Lyman M, Mitchell J, Kagohara LT, Goggins MG, Lafaro KJ, He J, Shubert C, Burns W, Zheng L, Fertig EJ, Jaffee EM, Burkhart RA, Ho WJ, Zimmerman JW. Cancer associated fibroblasts drive epithelial to mesenchymal transition and classical to basal change in pancreatic ductal adenocarcinoma cells with loss of IL-8 expression. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.07.631784. [PMID: 39829906 PMCID: PMC11741337 DOI: 10.1101/2025.01.07.631784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) carries an extremely poor prognosis, in part resulting from cellular heterogeneity that supports overall tumorigenicity. Cancer associated fibroblasts (CAF) are key determinants of PDAC biology and response to systemic therapy. While CAF subtypes have been defined, the effects of patient-specific CAF heterogeneity and plasticity on tumor cell behavior remain unclear. Here, multi-omics was used to characterize the tumor microenvironment (TME) in tumors from patients undergoing curative-intent surgery for PDAC. In these same patients, matched tumor organoid and CAF lines were established to functionally validate the impact of CAFs on the tumor cells. CAFs were found to drive epithelial-mesenchymal transition (EMT) and a switch in tumor cell classificiaton from classical to basal subtype. Furthermore, we identified CAF-specific interleukin 8 (IL-8) as an important modulator of tumor cell subtype. Finally, we defined neighborhood relationships between tumor cell and T cell subsets. Statement of Significance This multidimensional analysis highlights the diverse role CAFs have in influencing other cell types in the TME, including epithelial-derived tumor and infiltrating immune cells. Our methods provide a platform for evaluating emerging therapeutic approaches and for studying mechanisms that dictate tumor behavior in a manner that reflects patient-specific heterogeneity.
Collapse
|
3
|
Lv K, He T. Cancer-associated fibroblasts: heterogeneity, tumorigenicity and therapeutic targets. MOLECULAR BIOMEDICINE 2024; 5:70. [PMID: 39680287 PMCID: PMC11649616 DOI: 10.1186/s43556-024-00233-8] [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: 07/24/2024] [Revised: 11/04/2024] [Accepted: 11/19/2024] [Indexed: 12/17/2024] Open
Abstract
Cancer, characterized by its immune evasion, active metabolism, and heightened proliferation, comprises both stroma and cells. Although the research has always focused on parenchymal cells, the non-parenchymal components must not be overlooked. Targeting cancer parenchymal cells has proven to be a formidable challenge, yielding limited success on a broad scale. The tumor microenvironment(TME), a critical niche for cancer cell survival, presents a novel way for cancer treatment. Cancer-associated fibroblast (CAF), as a main component of TME, is a dynamically evolving, dual-functioning stromal cell. Furthermore, their biological activities span the entire spectrum of tumor development, metastasis, drug resistance, and prognosis. A thorough understanding of CAFs functions and therapeutic advances holds significant clinical implications. In this review, we underscore the heterogeneity of CAFs by elaborating on their origins, types and function. Most importantly, by elucidating the direct or indirect crosstalk between CAFs and immune cells, the extracellular matrix, and cancer cells, we emphasize the tumorigenicity of CAFs in cancer. Finally, we highlight the challenges encountered in the exploration of CAFs and list targeted therapies for CAF, which have implications for clinical treatment.
Collapse
Affiliation(s)
- Keke Lv
- Department of Hepatopanreatobiliary Surgery, Changhai Hospital, 168 Changhai Road, Yangpu District, Shanghai, 200433, China
| | - Tianlin He
- Department of Hepatopanreatobiliary Surgery, Changhai Hospital, 168 Changhai Road, Yangpu District, Shanghai, 200433, China.
| |
Collapse
|
4
|
Cheng PSW, Zaccaria M, Biffi G. Functional heterogeneity of fibroblasts in primary tumors and metastases. Trends Cancer 2024:S2405-8033(24)00260-7. [PMID: 39674792 DOI: 10.1016/j.trecan.2024.11.005] [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: 08/29/2024] [Revised: 10/29/2024] [Accepted: 11/18/2024] [Indexed: 12/16/2024]
Abstract
Cancer-associated fibroblasts (CAFs) are abundant components of the tumor microenvironment (TME) of most solid malignancies and have emerged as key regulators of cancer progression and therapy response. Although recent technological advances have uncovered substantial CAF molecular heterogeneity at the single-cell level, defining functional roles for most described CAF populations remains challenging. With the aim of bridging CAF molecular and functional heterogeneity, this review focuses on recently identified functional interactions of CAF subtypes with malignant cells, immune cells, and other stromal cells in primary tumors and metastases. Dissecting the heterogeneous functional crosstalk of specific CAF populations with other components is starting to uncover candidate combinatorial strategies for therapeutically targeting the TME and cancer progression.
Collapse
Affiliation(s)
- Priscilla S W Cheng
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, CB2 0RE, UK
| | - Marta Zaccaria
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, CB2 0RE, UK
| | - Giulia Biffi
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, CB2 0RE, UK.
| |
Collapse
|
5
|
Yang C, Liu W, Powell CA, Wang Q. Heterogeneity and therapeutic implications of cancer-associated fibroblasts in lung cancer: Recent advances and future perspectives. CHINESE MEDICAL JOURNAL PULMONARY AND CRITICAL CARE MEDICINE 2024; 2:240-249. [PMID: 39834587 PMCID: PMC11742357 DOI: 10.1016/j.pccm.2024.08.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Indexed: 01/22/2025]
Abstract
Lung cancer is a leading cause of cancer-related mortality. The tumor microenvironment is a complex and heterogeneous cellular environment surrounding tumor cells, including cancer-associated fibroblasts (CAFs), blood vessels, immune cells, the extracellular matrix, and various cytokines secreted by cells. CAFs are highly heterogeneous and play crucial roles in lung cancer. This review highlights recent advances in the understanding of CAFs in lung cancer, focusing on their heterogeneity and functions in tumorigenesis, progression, angiogenesis, invasion, metastasis, therapy resistance, tumor immune suppression, and targeted therapy responses. Additionally, we explore the underlying mechanisms and the potential of CAFs as a target in the development of innovative therapies for lung cancer.
Collapse
Affiliation(s)
- Chunhui Yang
- Department of Clinical Laboratory, The Second Hospital, Dalian Medical University, Dalian, Liaoning 116023, China
| | - Wenwen Liu
- Translational Research Center for Lung Cancer, The Second Hospital, Dalian Medical University, Dalian, Liaoning 116023, China
| | - Charles A. Powell
- Division of Pulmonary, Critical Care, and Sleep Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Qi Wang
- Translational Research Center for Lung Cancer, The Second Hospital, Dalian Medical University, Dalian, Liaoning 116023, China
- Department of Respiratory Medicine, The Second Hospital, Dalian Medical University, Dalian, Liaoning 116023, China
| |
Collapse
|
6
|
Li J, Tang Y, Long F, Tian L, Tang A, Ding L, Chen J, Liu M. Integrating bulk RNA-seq and scRNA-seq analyses revealed the function and clinical value of thrombospondins in colon cancer. Comput Struct Biotechnol J 2024; 23:2251-2266. [PMID: 38827236 PMCID: PMC11140486 DOI: 10.1016/j.csbj.2024.05.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 05/10/2024] [Accepted: 05/13/2024] [Indexed: 06/04/2024] Open
Abstract
Background Acting as mediators in cell-matrix and cell-cell communication, matricellular proteins play a crucial role in cancer progression. Thrombospondins (TSPs), a type of matricellular glycoproteins, are key regulators in cancer biology with multifaceted roles. Although TSPs have been implicated in anti-tumor immunity and epithelial-mesenchymal transition (EMT) in several malignancies, their specific roles to colon cancer remain elusive. Addressing this knowledge gap is essential, as understanding the function of TSPs in colon cancer could identify new therapeutic targets and prognostic markers. Methods Analyzing 1981 samples from 10 high-throughput datasets, including six bulk RNA-seq, three scRNA-seq, and one spatial transcriptome dataset, our study investigated the prognostic relevance, risk stratification value, immune heterogeneity, and cellular origin of TSPs, as well as their influence on cancer-associated fibroblasts (CAFs). Utilizing survival analysis, unsupervised clustering, and functional enrichment, along with multiple correlation analyses of the tumor-microenvironment (TME) via Gene Set Variation Analysis (GSVA), spatial localization, Monocle2, and CellPhoneDB, we provided insights into the clinical and cellular implications of TSPs. Results First, we observed significant upregulation of THBS2 and COMP in colon cancer, both of which displayed significant prognostic value. Additionally, we detected a significant positive correlation between TSPs and immune cells, as well as marker genes of EMT. Second, based on TSPs expression, patients were divided into two clusters with distinct prognoses: the high TSPs expression group (TSPs-H) was characterized by pronounced immune and stromal cell infiltration, and notably elevated T-cell exhaustion scores. Subsequently, we found that THBS2 and COMP may be associated with the differentiation of CAFs into pan-iCAFs and pan-dCAFs, which are known for their heightened matrix remodeling activities. Moreover, THBS2 enhanced CAFs communication with vascular endothelial cells and monocyte-macrophages. CAFs expressing THBS2 (THBS2+ CAFs) demonstrated higher scores across multiple signaling pathways, including angiogenic, EMT, Hedgehog, Notch, Wnt, and TGF-β, when compared to THBS2- CAFs. These observations suggest that THBS2 may be associated with stronger pro-carcinogenic activity in CAFs. Conclusions This study revealed the crucial role of TSPs and the significant correlation between THBS2 and CAFs interactions in colon cancer progression, providing valuable insights for targeting TSPs to mitigate cancer progression.
Collapse
Affiliation(s)
- Jing Li
- Key Laboratory of Clinical Laboratory Diagnostics, College of Laboratory Medicine, Chongqing Medical University, Chongqing 400046, China
| | - Ying Tang
- Key Laboratory of Clinical Laboratory Diagnostics, College of Laboratory Medicine, Chongqing Medical University, Chongqing 400046, China
- Medical Laboratory, People's Hospital of Qingbaijiang District, Chengdu 61300, China
| | - Fei Long
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
- Center for Single-Cell Omics and Tumor Liquid Biopsy, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Luyao Tian
- Key Laboratory of Clinical Laboratory Diagnostics, College of Laboratory Medicine, Chongqing Medical University, Chongqing 400046, China
| | - Ao Tang
- Key Laboratory of Clinical Laboratory Diagnostics, College of Laboratory Medicine, Chongqing Medical University, Chongqing 400046, China
| | - LiHui Ding
- Key Laboratory of Clinical Laboratory Diagnostics, College of Laboratory Medicine, Chongqing Medical University, Chongqing 400046, China
| | - Juan Chen
- Key Laboratory of Clinical Laboratory Diagnostics, College of Laboratory Medicine, Chongqing Medical University, Chongqing 400046, China
| | - Mingwei Liu
- Key Laboratory of Clinical Laboratory Diagnostics, College of Laboratory Medicine, Chongqing Medical University, Chongqing 400046, China
| |
Collapse
|
7
|
Kumar S, Ali I, Abbas F, Shafiq F, Yadav AK, Ghate MD, Kumar D. In-silico identification and exploration of small molecule coumarin-1,2,3-triazole hybrids as potential EGFR inhibitors for targeting lung cancer. Mol Divers 2024; 28:4301-4324. [PMID: 38470555 DOI: 10.1007/s11030-024-10817-9] [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: 08/14/2023] [Accepted: 01/25/2024] [Indexed: 03/14/2024]
Abstract
Globally, lung cancer is a significant public health concern due to its role as the leading cause of cancer-related mortalities. The promising target of EGFR for lung cancer treatment has been identified, providing a potential avenue for more effective therapies. The purpose of the study was to design a library of 1843 coumarin-1,2,3-triazole hybrids and screen them based on a designed pharmacophore to identify potential inhibitors targeting EGFR in lung cancer with minimum or no side effects. Pharmacophore-based screening was carried out and 60 hits were obtained. To gain a better understanding of the binding interactions between the compounds and the targeted receptor, molecular docking was conducted on the 60 screened compounds. In-silico ADME and toxicity studies were also conducted to assess the drug-likeness and safety of the identified compounds. The results indicated that coumarin-1,2,3-triazole hybrids COUM-0849, COUM-0935, COUM-0414, COUM-1335, COUM-0276, and COUM-0484 exhibit dock score of - 10.2, - 10.2, - 10.1, - 10.1, - 10, - 10 while reference molecule - 7.9 kcal/mol for EGFR (PDB ID: 4HJO) respectively. The molecular docking and molecular dynamics simulations revealed that the identified compounds formed stable interactions with the active site of EGFR, indicating their potential as inhibitors. The in-silico ADME and toxicity studies showed that the compounds had favorable drug-likeness properties and low toxicity, further supporting their potential as therapeutic agents. Finally, we performed DFT studies on the best-selected ligands to gain further insights into their electronic properties. The findings of this study provide important insights into the potential of coumarin-1,2,3-triazole hybrids as promising EGFR inhibitors for the management of lung cancer.
Collapse
Affiliation(s)
- Sunil Kumar
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Sciences, Shoolini University, Solan, Himachal Pradesh, 173229, India
| | - Iqra Ali
- Department of Biosciences, COMSATS University Islamabad, Islamabad Campus, Islamabad, 45550, Pakistan
| | - Faheem Abbas
- Key Lab of Organic Optoelectronics and Molecular Engineering of Ministry of Education, Department of Chemistry, Tsinghua University, Beijing, 100084, People's Republic of China
| | - Faiza Shafiq
- Department of Chemistry, University of Agriculture, Faisalabad, 38040, Pakistan
| | - Ashok Kumar Yadav
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, 160014, India
| | - Manjunath D Ghate
- School of Pharmacy, National Forensic Sciences University, Gandhinagar, Gujarat, 382007, India
| | - Deepak Kumar
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Sciences, Shoolini University, Solan, Himachal Pradesh, 173229, India.
| |
Collapse
|
8
|
Papavassiliou KA, Sofianidi AA, Gogou VA, Papavassiliou AG. The Clinical Significance of Cancer-Associated Fibroblasts Classification in Non-Small Cell Lung Cancer. Cells 2024; 13:1909. [PMID: 39594657 PMCID: PMC11592919 DOI: 10.3390/cells13221909] [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: 11/13/2024] [Accepted: 11/18/2024] [Indexed: 11/28/2024] Open
Abstract
Malignant cells flourish within a specialized environment known as the tumor microenvironment (TME) [...].
Collapse
Affiliation(s)
- Kostas A. Papavassiliou
- First University Department of Respiratory Medicine, ‘Sotiria’ Chest Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (K.A.P.); (V.A.G.)
| | - Amalia A. Sofianidi
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Vassiliki A. Gogou
- First University Department of Respiratory Medicine, ‘Sotiria’ Chest Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (K.A.P.); (V.A.G.)
| | - Athanasios G. Papavassiliou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| |
Collapse
|
9
|
Chen X, Zhou Z, Yazgan Z, Xie L, Rossi F, Liu Y, Zhang B, Polanco PM, Zeh HJ, Kim AC, Huang H. Single-cell resolution spatial analysis of antigen-presenting cancer-associated fibroblast niches. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.15.623232. [PMID: 39605724 PMCID: PMC11601292 DOI: 10.1101/2024.11.15.623232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Recent studies have identified a unique subtype of cancer-associated fibroblasts (CAFs) termed antigen-presenting CAFs (apCAFs), which remain the least understood CAF subtype. To gain a comprehensive understanding of the origin and function apCAFs, we construct a fibroblast molecular atlas across 14 types of solid tumors. Our integration study unexpectedly reveals two distinct apCAF lineages present in most cancer types: one associated with mesothelial-like cells and the other with fibrocytes. Using a high-resolution single-cell spatial imaging platform, we characterize the spatial niches of these apCAF lineages. We find that mesothelial-like apCAFs are located near cancer cells, while fibrocyte-like apCAFs are associated with tertiary lymphoid structures. Additionally, we discover that both apCAF lineages can up-regulate the secreted protein SPP1, which facilitates primary tumor formation and peritoneal metastasis. Taken together, this study offers an unprecedented resolution in analyzing apCAF lineages and their spatial niches.
Collapse
|
10
|
Qiu X, Li S, Fan T, Zhang Y, Wang B, Zhang B, Zhang M, Zhang L. Advances and prospects in tumor infiltrating lymphocyte therapy. Discov Oncol 2024; 15:630. [PMID: 39514075 PMCID: PMC11549075 DOI: 10.1007/s12672-024-01410-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 10/01/2024] [Indexed: 11/16/2024] Open
Abstract
Tumor-infiltrating lymphocyte (TIL) therapy in adoptive T-cell therapy (ACT) has already caused durable regression in a variety of cancer types due to T-cell persistence, clinical activity, and duration of objective response and safety. TILs are composed of polyclonal effector T-cells specific to heterogenetic tumor antigens, reasonably providing a promising means for tumor therapy. In addition, their expansion in vitro can release them from the suppressive tumor microenvironment. Even though significant advances have been made in the procedure of TIL therapy, from TIL isolation, modification, expansion, and infusion back to the patient to target the tumor, strategy optimization is always ongoing to overcome drawbacks such as a complex process, options for the lineage differentiation status of TILs, and sufficient trafficking of TILs to the tumor. In this review, we summarize the current advances of TIL therapy, raise problem-based optimization strategies, and provide future perspectives on next-generation TIL therapy as a potential avenue for enhancing cell-based immunotherapy.
Collapse
Affiliation(s)
- Xu Qiu
- The Department of Immunology, School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
| | - Shengjun Li
- Clinical Laboratory, Qingdao Women and Children's Hospital, Qingdao, Shandong, China
| | - Tianyu Fan
- The Department of Immunology, School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
- Taian City Central Hospital, Taian, Shandong, China
| | - Yue Zhang
- The Department of Immunology, School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
| | - Bin Wang
- The Department of Pathogenic Biology, School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
| | - Bei Zhang
- The Department of Immunology, School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
| | - Mingzhe Zhang
- The Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Li Zhang
- The Department of Immunology, School of Basic Medicine, Qingdao University, Qingdao, Shandong, China.
| |
Collapse
|
11
|
Park JG, Roh PR, Kang MW, Cho SW, Hwangbo S, Jung HD, Kim HU, Kim JH, Yoo JS, Han JW, Jang JW, Choi JY, Yoon SK, You YK, Choi HJ, Ryu JY, Sung PS. Intrahepatic IgA complex induces polarization of cancer-associated fibroblasts to matrix phenotypes in the tumor microenvironment of HCC. Hepatology 2024; 80:1074-1086. [PMID: 38466639 DOI: 10.1097/hep.0000000000000772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 01/13/2024] [Indexed: 03/13/2024]
Abstract
BACKGROUND AND AIMS Cancer-associated fibroblasts (CAFs) play key roles in the tumor microenvironment. IgA contributes to inflammation and dismantling antitumor immunity in the human liver. In this study, we aimed to elucidate the effects of the IgA complex on CAFs in Pil Soo Sung the tumor microenvironment of HCC. APPROACH AND RESULTS CAF dynamics in HCC tumor microenvironment were analyzed through single-cell RNA sequencing of HCC samples. CAFs isolated from 50 HCC samples were treated with mock or serum-derived IgA dimers in vitro. Progression-free survival of patients with advanced HCC treated with atezolizumab and bevacizumab was significantly longer in those with low serum IgA levels ( p <0.05). Single-cell analysis showed that subcluster proportions in the CAF-fibroblast activation protein-α matrix were significantly increased in patients with high serum IgA levels. Flow cytometry revealed a significant increase in the mean fluorescence intensity of fibroblast activation protein in the CD68 + cells from patients with high serum IgA levels ( p <0.001). We confirmed CD71 (IgA receptor) expression in CAFs, and IgA-treated CAFs exhibited higher programmed death-ligand 1 expression levels than those in mock-treated CAFs ( p <0.05). Coculture with CAFs attenuated the cytotoxic function of activated CD8 + T cells. Interestingly, activated CD8 + T cells cocultured with IgA-treated CAFs exhibited increased programmed death-1 expression levels than those cocultured with mock-treated CAFs ( p <0.05). CONCLUSIONS Intrahepatic IgA induced polarization of HCC-CAFs into more malignant matrix phenotypes and attenuates cytotoxic T-cell function. Our study highlighted their potential roles in tumor progression and immune suppression.
Collapse
Affiliation(s)
- Jong Geun Park
- The Catholic University Liver Research Center, College of Medicine, Department of Biomedicine & Health Sciences, The Catholic University of Korea, Seoul, Republic of Korea
| | - Pu Reun Roh
- The Catholic University Liver Research Center, College of Medicine, Department of Biomedicine & Health Sciences, The Catholic University of Korea, Seoul, Republic of Korea
| | - Min Woo Kang
- The Catholic University Liver Research Center, College of Medicine, Department of Biomedicine & Health Sciences, The Catholic University of Korea, Seoul, Republic of Korea
| | - Sung Woo Cho
- The Catholic University Liver Research Center, College of Medicine, Department of Biomedicine & Health Sciences, The Catholic University of Korea, Seoul, Republic of Korea
| | - Suhyun Hwangbo
- Department of Genomic Medicine, Seoul National University Hospital, Daehak-ro, Jongno-gu, Seoul, Republic of Korea
| | - Hae Deok Jung
- Department of Chemical and Biomolecular Engineering (BK21 four), Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Hyun Uk Kim
- Department of Chemical and Biomolecular Engineering (BK21 four), Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Ji Hoon Kim
- The Catholic University Liver Research Center, College of Medicine, Department of Biomedicine & Health Sciences, The Catholic University of Korea, Seoul, Republic of Korea
- Department of Internal Medicine, Uijeongbu St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jae-Sung Yoo
- The Catholic University Liver Research Center, College of Medicine, Department of Biomedicine & Health Sciences, The Catholic University of Korea, Seoul, Republic of Korea
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, College of Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Republic of Korea
| | - Ji Won Han
- The Catholic University Liver Research Center, College of Medicine, Department of Biomedicine & Health Sciences, The Catholic University of Korea, Seoul, Republic of Korea
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, College of Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jeong Won Jang
- The Catholic University Liver Research Center, College of Medicine, Department of Biomedicine & Health Sciences, The Catholic University of Korea, Seoul, Republic of Korea
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, College of Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jong Young Choi
- The Catholic University Liver Research Center, College of Medicine, Department of Biomedicine & Health Sciences, The Catholic University of Korea, Seoul, Republic of Korea
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, College of Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Republic of Korea
| | - Seung Kew Yoon
- The Catholic University Liver Research Center, College of Medicine, Department of Biomedicine & Health Sciences, The Catholic University of Korea, Seoul, Republic of Korea
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, College of Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Republic of Korea
| | - Young Kyoung You
- Department of Surgery, The Catholic University of Korea, Seoul, Republic of Korea
| | - Ho Joong Choi
- Department of Surgery, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jae Yong Ryu
- Department of Biotechnology, Duksung Women's University, Seoul, Korea
| | - Pil Soo Sung
- The Catholic University Liver Research Center, College of Medicine, Department of Biomedicine & Health Sciences, The Catholic University of Korea, Seoul, Republic of Korea
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, College of Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Republic of Korea
| |
Collapse
|
12
|
Yu S, Wang S, Wang X, Xu X. The axis of tumor-associated macrophages, extracellular matrix proteins, and cancer-associated fibroblasts in oncogenesis. Cancer Cell Int 2024; 24:335. [PMID: 39375726 PMCID: PMC11459962 DOI: 10.1186/s12935-024-03518-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 09/29/2024] [Indexed: 10/09/2024] Open
Abstract
The extracellular matrix (ECM) is a complex, dynamic network of multiple macromolecules that serve as a crucial structural and physical scaffold for neighboring cells. In the tumor microenvironment (TME), ECM proteins play a significant role in mediating cellular communication between cancer-associated fibroblasts (CAFs) and tumor-associated macrophages (TAMs). Revealing the ECM modification of the TME necessitates the intricate signaling cascades that transpire among diverse cell populations and ECM proteins. The advent of single-cell sequencing has enabled the identification and refinement of specific cellular subpopulations, which has substantially enhanced our comprehension of the intricate milieu and given us a high-resolution perspective on the diversity of ECM proteins. However, it is essential to integrate single-cell data and establish a coherent framework. In this regard, we present a comprehensive review of the relationships among ECM, TAMs, and CAFs. This encompasses insights into the ECM proteins released by TAMs and CAFs, signaling integration in the TAM-ECM-CAF axis, and the potential applications and limitations of targeted therapies for CAFs. This review serves as a reliable resource for focused therapeutic strategies while highlighting the crucial role of ECM proteins as intermediates in the TME.
Collapse
Affiliation(s)
- Shuhong Yu
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Siyu Wang
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Xuanyu Wang
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Ximing Xu
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|
13
|
Si Y, Zhao Z, Meng X, Zhao K. RNA-seq and bulk RNA-seq data analysis of cancer-related fibroblasts (CAF) in LUAD to construct a CAF-based risk signature. Sci Rep 2024; 14:23243. [PMID: 39369095 PMCID: PMC11455853 DOI: 10.1038/s41598-024-74336-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 09/25/2024] [Indexed: 10/07/2024] Open
Abstract
Angiogenesis, metastasis, and resistance to therapy are all facilitated by cancer-associated fibroblasts (CAFs). A CAF-based risk signature can be used to predict patients' prognoses for Lung adenocarcinoma (LUAD) based on CAF characteristics. The Gene Expression Omnibus (GEO) database was used to gather signal-cell RNA sequencing (scRNA-seq) data for this investigation. The GEO and TCGA databases were used to gather bulk RNA-seq and microarray data for LUAD. The scRNA-seq data were analyzed using the Seurat R program based on the CAF markers. Our goal was to use differential expression analysis to discover differentially expressed genes (DEGs) across normal and tumor samples in the TCGA dataset. Pearson correlation analysis was utilized to discover prognostic genes related with CAF, followed by univariate Cox regression analysis. Using Lasso regression, a risk signature based on CAF-related prognostic genes was created. A nomogram model was created based on the clinical and pathological aspects. 5 CAF clusters were identified in LUAD, 4 of which were associated with prognosis. From 2811 DEGs, 1002 genes were found to be significantly correlated with CAF clusters, which led to the creation of a risk signature with 8 genes. These 8 genes were primarily connected with 41 pathways, such as antigen paocessing and presentation, apoptosis, and cell cycle. Meanwhile, the risk signature was significantly associated with stromal and immune scores, as well as some immune cells. Multivariate analysis revealed that risk signature was an independent prognostic factor for LUAD, and its value in predicting immunotherapeutic outcomes was confirmed. A novel nomogram integrating the stage and CAF-based risk signature was constructed, which exhibited favorable predictability and reliability in the prognosis prediction of LUAD. CAF-based risk signatures can be effective in predicting the prognosis of LUAD, and they may provide new strategies for cancer treatments by interpreting the response of LUAD to immunotherapy.
Collapse
Affiliation(s)
- Youjiao Si
- Department of Radiology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Zhonghua Zhao
- Department of Oncology, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Xiangjiao Meng
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Kaikai Zhao
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, China.
| |
Collapse
|
14
|
Furugaki K, Fujimura T, Sakaguchi N, Watanabe Y, Uchibori K, Miyauchi E, Hayashi H, Katayama R, Yoshiura S. Combined blockade of GPX4 and activated EGFR/HER3 bypass pathways inhibits the development of ALK-inhibitor-induced tolerant persister cells in ALK-fusion-positive lung cancer. Mol Oncol 2024. [PMID: 39369284 DOI: 10.1002/1878-0261.13746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 07/24/2024] [Accepted: 09/24/2024] [Indexed: 10/07/2024] Open
Abstract
Cancers can develop resistance to treatment with ALK tyrosine kinase inhibitors (ALK-TKIs) via emergence of a subpopulation of drug-tolerant persister (DTP) cells that can survive initial drug treatment long enough to acquire genetic aberrations. DTP cells are thus a potential therapeutic target. We generated alectinib-induced DTP cells from a patient-derived ALK+ non-small-cell lung cancer (NSCLC) cell line and screened 3114 agents in the anticancer compounds library (TargetMol). We identified phospholipid hydroperoxide glutathione peroxidase GPX4 as being involved in promoting the survival of DTP cells. GPX4 was found to be upregulated in DTP cells and to promote cell survival by suppressing reactive oxygen species (ROS) accumulation; GPX4 inhibitors blocked this upregulation and facilitated ROS-mediated cell death. Activated bypass signals involving epidermal growth factor receptor (EGFR)/receptor tyrosine-protein kinase erbB-3 (HER3) were also identified in DTP cells, and co-treatment with EGFR-TKI plus ALK-TKI enhanced ROS levels. Triple combination with an ALK-TKI plus a bypass pathway inhibitor plus a GPX4 inhibitor suppressed cell growth and induced intracellular ROS accumulation more greatly than did treatment with each agent alone. The combined inhibition of ALK plus inhibition of activated bypass signals plus inhibition of GPX4 may be a potent therapeutic strategy for patients with ALK+ NSCLC to prevent the development of resistance to ALK-TKIs and lead to tumor eradication.
Collapse
Affiliation(s)
- Koh Furugaki
- Product Research Department, Chugai Pharmaceutical Co., Ltd, Yokohama, Japan
| | - Takaaki Fujimura
- Product Research Department, Chugai Pharmaceutical Co., Ltd, Yokohama, Japan
| | - Narumi Sakaguchi
- Biological Technology Department, Chugai Pharmaceutical Co., Ltd, Yokohama, Japan
| | - Yasutaka Watanabe
- Division of Thoracic Oncology, Saitama Cancer Center, Saitama, Japan
| | - Ken Uchibori
- Department of Thoracic Medical Oncology, The Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Eisaku Miyauchi
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Hidetoshi Hayashi
- Department of Medical Oncology, Kindai University Faculty of Medicine, Sayama, Japan
| | - Ryohei Katayama
- Division of Experimental Chemotherapy, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Shigeki Yoshiura
- Product Research Department, Chugai Pharmaceutical Co., Ltd, Yokohama, Japan
| |
Collapse
|
15
|
Li Y, Xie T, Wang S, Yang L, Hao X, Wang Y, Hu X, Wang L, Li J, Ying J, Xing P. Mechanism exploration and model construction for small cell transformation in EGFR-mutant lung adenocarcinomas. Signal Transduct Target Ther 2024; 9:261. [PMID: 39353908 PMCID: PMC11445518 DOI: 10.1038/s41392-024-01981-3] [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: 05/15/2024] [Revised: 09/03/2024] [Accepted: 09/14/2024] [Indexed: 10/03/2024] Open
Abstract
Small-cell lung cancer (SCLC) transformation accounts for 3-14% of resistance in EGFR-TKI relapsed lung adenocarcinomas (LUADs), with unknown molecular mechanisms and optimal treatment strategies. We performed transcriptomic analyses (including bulk and spatial transcriptomics) and multiplex immunofluorescence on pre-treated samples from LUADs without transformation after EGFR-TKI treatment (LUAD-NT), primary SCLCs (SCLC-P) and LUADs with transformation after EGFR-TKI treatment (before transformation: LUAD-BT; after transformation: SCLC-AT). Our study found that LUAD-BT exhibited potential transcriptomic characteristics for transformation compared with LUAD-NT. We identified several pathways that shifted during transformation, and the transformation might be promoted by epigenetic alterations (such as HDAC10, HDAC1, DNMT3A) within the tumor cells instead of within the tumor microenvironment. For druggable pathways, transformed-SCLC were proved to be less dependent on EGF signaling but more relied on FGF signaling, while VEGF-VEGFR pathway remained active, indicating potential treatments after transformation. We also found transformed-SCLC showed an immuno-exhausted status which was associated with the duration of EGFR-TKI before transformation. Besides, SCLC-AT exhibited distinct molecular subtypes from SCLC-P. Moreover, we constructed an ideal 4-marker model based on transcriptomic and IHC data to predict SCLC transformation, which obtained a sensitivity of 100% and 87.5%, a specificity of 95.7% and 100% in the training and test cohorts, respectively. We provided insights into the molecular mechanisms of SCLC transformation and the differences between SCLC-AT and SCLC-P, which might shed light on prevention strategies and subsequent therapeutic strategies for SCLC transformation in the future.
Collapse
Affiliation(s)
- Yan Li
- State Key Laboratory of Molecular Oncology, Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Tongji Xie
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
- Department of Pulmonary and Critical Care Medicine, Beijing Hospital, National Centre of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Shouzheng Wang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
- Department of Medical Oncology, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, 101149, China
| | - Lin Yang
- State Key Laboratory of Molecular Oncology, Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Xuezhi Hao
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yan Wang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Xingsheng Hu
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Lin Wang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Junling Li
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Jianming Ying
- State Key Laboratory of Molecular Oncology, Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Puyuan Xing
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
16
|
Cheng X, Cao Y, Liu X, Li Y, Li Q, Gao D, Yu Q. Single-cell and spatial omics unravel the spatiotemporal biology of tumour border invasion and haematogenous metastasis. Clin Transl Med 2024; 14:e70036. [PMID: 39350478 PMCID: PMC11442492 DOI: 10.1002/ctm2.70036] [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: 05/05/2024] [Revised: 08/14/2024] [Accepted: 09/16/2024] [Indexed: 10/04/2024] Open
Abstract
Solid tumours exhibit a well-defined architecture, comprising a differentiated core and a dynamic border that interfaces with the surrounding tissue. This border, characterised by distinct cellular morphology and molecular composition, serves as a critical determinant of the tumour's invasive behaviour. Notably, the invasive border of the primary tumour represents the principal site for intravasation of metastatic cells. These cells, known as circulating tumour cells (CTCs), function as 'seeds' for distant dissemination and display remarkable heterogeneity. Advancements in spatial sequencing technology are progressively unveiling the spatial biological features of tumours. However, systematic investigations specifically targeting the characteristics of the tumour border remain scarce. In this comprehensive review, we illuminate key biological insights along the tumour body-border-haematogenous metastasis axis over the past five years. We delineate the distinctive landscape of tumour invasion boundaries and delve into the intricate heterogeneity and phenotype of CTCs, which orchestrate haematogenous metastasis. These insights have the potential to explain the basis of tumour invasion and distant metastasis, offering new perspectives for the development of more complex and precise clinical interventions and treatments.
Collapse
Affiliation(s)
- Xifu Cheng
- Department of Gastroenterology and Hepatologythe Second Affiliated HospitalJiangxi Medical CollegeNanchang UniversityNanchangChina
- Department of Pathogen Biology and ImmunologySchool of Basic Medical SciencesJiangxi Medical CollegeNanchang UniversityNanchangChina
| | - Yuke Cao
- Department of Gastroenterology and Hepatologythe Second Affiliated HospitalJiangxi Medical CollegeNanchang UniversityNanchangChina
| | - Xiangyi Liu
- Queen Mary SchoolJiangxi Medical CollegeNanchang UniversityNanchangChina
| | - Yuanheng Li
- Queen Mary SchoolJiangxi Medical CollegeNanchang UniversityNanchangChina
| | - Qing Li
- Department of Oncologythe Second Affiliated HospitalJiangxi Medical CollegeNanchang UniversityNanchangChina
| | - Dian Gao
- Department of Gastroenterology and Hepatologythe Second Affiliated HospitalJiangxi Medical CollegeNanchang UniversityNanchangChina
- Department of Pathogen Biology and ImmunologySchool of Basic Medical SciencesJiangxi Medical CollegeNanchang UniversityNanchangChina
| | - Qiongfang Yu
- Department of Gastroenterology and Hepatologythe Second Affiliated HospitalJiangxi Medical CollegeNanchang UniversityNanchangChina
| |
Collapse
|
17
|
Zhang S, Yang Y, Zhang L, Liu Y, Guo Z, Wu J, Zhou W, Hong Z, Zhang W. Identification and Validation of a Prognostic Signature Based on Fibroblast Immune-related Genes to Predict the Prognosis and Therapeutic Response of renal clear cell carcinoma by Integrated Analysis of Single-Cell and Bulk RNA Sequencing Data. J Cancer 2024; 15:5942-5955. [PMID: 39440053 PMCID: PMC11493018 DOI: 10.7150/jca.100194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 08/24/2024] [Indexed: 10/25/2024] Open
Abstract
Background: The importance of fibroblasts in cancer progression is becoming more acknowledged, particularly the significance of their immune-related genes. However, the precise roles these genes play in fibroblasts throughout tumor development remains unclear. Exploring how these genes function in advancing kidney renal clear cell carcinoma (KIRC) could provide answers to these uncertainties. Material and method: The Cancer Genome Atlas (TCGA) database served as the source of data for KIRC patients. We distinguished fibroblast immune-related genes (FIGs), which are used to construct risk score. Further analysis conducted including enrichment analysis, assessment of tumor mutation burden (TMB), evaluation of tumor microenvironment (TME), analysis of immune cell infiltration, and drug sensitivity prediction. Result: The risk score using 6 FIGs effectively predicts the outcomes for KIRC patients. Nomogram which is based on the risk score and clinical data, demonstrated superior predictive performance compared to the version without the risk score. Enrichment analysis identified that coagulation pathway predominates in high-risk group, the protein secretion pathway is prevalent in low-risk patients' cohort. The adverse prognosis in high-risk patient cohort could be linked to an elevated TMB. TME analysis showed that high-risk group's tumor tissues contain more immune and stromal cells. Furthermore, the amount of regulatory T cells increases with the risk score. Low-risk group response better to immunotherapy. Finally, RT-qPCR confirmed the differential expression of FIGs in KIRC patients. Conclusion: This risk score and nomogram are valuable tools assessing KIRC patients' prognosis. Poorer prognosis in high-risk categories may have relationship with activation of coagulation pathway and a higher TMB.
Collapse
Affiliation(s)
- Shuwen Zhang
- Department of Thoracic Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China, 330006
- Queen Mary School, Jiangxi medical college, Nanchang University, Nanchang, China, 330006
| | - Yuqian Yang
- Queen Mary School, Jiangxi medical college, Nanchang University, Nanchang, China, 330006
- Department of Urology Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China, 330006
| | - Liyi Zhang
- Queen Mary School, Jiangxi medical college, Nanchang University, Nanchang, China, 330006
- Department of Urology Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China, 330006
| | - Yijiang Liu
- Department of Urology Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China, 330006
- Jiangxi medical college, Nanchang University, Nanchang, China, 330006
| | - Zihun Guo
- Department of Thoracic Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China, 330006
| | - Jiajun Wu
- Department of Thoracic Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China, 330006
| | - Weijun Zhou
- Department of Thoracic Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China, 330006
| | - Zhengdong Hong
- Department of Urology Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China, 330006
| | - Wenxiong Zhang
- Department of Thoracic Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China, 330006
| |
Collapse
|
18
|
Vella N, Fenech AG, Petroni Magri V. 3D cell culture models in research: applications to lung cancer pharmacology. Front Pharmacol 2024; 15:1438067. [PMID: 39376603 PMCID: PMC11456561 DOI: 10.3389/fphar.2024.1438067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 08/26/2024] [Indexed: 10/09/2024] Open
Abstract
Lung cancer remains one of the leading causes of cancer-related mortality worldwide, necessitating innovative research methodologies to improve treatment outcomes and develop novel strategies. The advent of three-dimensional (3D) cell cultures has marked a significant advancement in lung cancer research, offering a more physiologically relevant model compared to traditional two-dimensional (2D) cultures. This review elucidates the various types of 3D cell culture models currently used in lung cancer pharmacology, including spheroids, organoids and engineered tissue models, having pivotal roles in enhancing our understanding of lung cancer biology, facilitating drug development, and advancing precision medicine. 3D cell culture systems mimic the complex spatial architecture and microenvironment of lung tumours, providing critical insights into the cellular and molecular mechanisms of tumour progression, metastasis and drug responses. Spheroids, derived from commercialized cell lines, effectively model the tumour microenvironment (TME), including the formation of hypoxic and nutrient gradients, crucial for evaluating the penetration and efficacy of anti-cancer therapeutics. Organoids and tumouroids, derived from primary tissues, recapitulate the heterogeneity of lung cancers and are instrumental in personalized medicine approaches, supporting the simulation of in vivo pharmacological responses in a patient-specific context. Moreover, these models have been co-cultured with various cell types and biomimicry extracellular matrix (ECM) components to further recapitulate the heterotypic cell-cell and cell-ECM interactions present within the lung TME. 3D cultures have been significantly contributing to the identification of novel therapeutic targets and the understanding of resistance mechanisms against conventional therapies. Therefore, this review summarizes the latest findings in drug research involving lung cancer 3D models, together with the common laboratory-based assays used to study drug effects. Additionally, the integration of 3D cell cultures into lung cancer drug development workflows and precision medicine is discussed. This integration is pivotal in accelerating the translation of laboratory findings into clinical applications, thereby advancing the landscape of lung cancer treatment. By closely mirroring human lung tumours, these models not only enhance our understanding of the disease but also pave the way for the development of more effective and personalized therapeutic strategies.
Collapse
Affiliation(s)
| | - Anthony G. Fenech
- Department of Clinical Pharmacology and Therapeutics, Faculty of Medicine and Surgery, University of Malta, Msida, Malta
| | | |
Collapse
|
19
|
Guo T, Xu J. Cancer-associated fibroblasts: a versatile mediator in tumor progression, metastasis, and targeted therapy. Cancer Metastasis Rev 2024; 43:1095-1116. [PMID: 38602594 PMCID: PMC11300527 DOI: 10.1007/s10555-024-10186-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Accepted: 03/31/2024] [Indexed: 04/12/2024]
Abstract
Tumor microenvironment (TME) has been demonstrated to play a significant role in tumor initiation, progression, and metastasis. Cancer-associated fibroblasts (CAFs) are the major component of TME and exhibit heterogeneous properties in their communication with tumor cells. This heterogeneity of CAFs can be attributed to various origins, including quiescent fibroblasts, mesenchymal stem cells (MSCs), adipocytes, pericytes, endothelial cells, and mesothelial cells. Moreover, single-cell RNA sequencing has identified diverse phenotypes of CAFs, with myofibroblastic CAFs (myCAFs) and inflammatory CAFs (iCAFs) being the most acknowledged, alongside newly discovered subtypes like antigen-presenting CAFs (apCAFs). Due to these heterogeneities, CAFs exert multiple functions in tumorigenesis, cancer stemness, angiogenesis, immunosuppression, metabolism, and metastasis. As a result, targeted therapies aimed at the TME, particularly focusing on CAFs, are rapidly developing, fueling the promising future of advanced tumor-targeted therapy.
Collapse
Affiliation(s)
- Tianchen Guo
- Women's Reproductive Health Laboratory of Zhejiang Province, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, Zhejiang, China
| | - Junfen Xu
- Department of Gynecologic Oncology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, Zhejiang, China.
| |
Collapse
|
20
|
Stephan A, Suhrmann JH, Skowron MA, Che Y, Poschmann G, Petzsch P, Kresbach C, Wruck W, Pongratanakul P, Adjaye J, Stühler K, Köhrer K, Schüller U, Nettersheim D. Molecular and epigenetic ex vivo profiling of testis cancer-associated fibroblasts and their interaction with germ cell tumor cells and macrophages. Matrix Biol 2024; 132:10-23. [PMID: 38851302 DOI: 10.1016/j.matbio.2024.06.001] [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: 02/14/2024] [Revised: 05/10/2024] [Accepted: 06/03/2024] [Indexed: 06/10/2024]
Abstract
Germ cell tumors (GCT) are the most common solid tumors in young men of age 15 - 40. In previous studies, we profiled the interaction of GCT cells with cells of the tumor microenvironment (TM), which showed that especially the 3D interaction of fibroblasts (FB) or macrophages with GCT cells influenced the growth behavior and cisplatin response as well as the transcriptome and secretome of the tumor cells, suggesting that the crosstalk of these cells with GCT cells is crucial for tumor progression and therapy outcome. In this study, we shed light on the mechanisms of activation of cancer-associated fibroblasts (CAF) in the GCT setting and their effects on GCT cells lines and the monocyte cell line THP-1. Ex vivo cultures of GCT-derived CAF were established and characterized molecularly and epigenetically by performing DNA methylation arrays, RNA sequencing, and mass spectrometry-based secretome analysis. We demonstrated that the activation state of CAF is influenced by their former prevailing tumor environment in which they have resided. Hereby, we postulate that seminoma (SE) and embryonal carcinoma (EC) activate CAF, while teratoma (TER) play only a minor role in CAF formation. In turn, CAF influence proliferation and the expression of cisplatin sensitivity-related factors in GCT cells lines as well as polarization of in vitro-induced macrophages by the identified effector molecules IGFBP1, LGALS3BP, LYVE1, and PTX3. Our data suggests that the vital interaction of CAF with GCT cells and with macrophages has a huge influence on shaping the extracellular matrix as well as on recruitment of immune cells to the TM. In conclusion, therapeutically interfering with CAF and / or macrophages in addition to the standard therapy might slow-down progression of GCT and re-shaping of the TM to a tumor-promoting environment. Significance: The interaction of CAF with GCT and macrophages considerably influences the microenvironment. Thus, therapeutically interfering with CAF might slow-down progression of GCT and re-shaping of the microenvironment to a tumor-promoting environment.
Collapse
Affiliation(s)
- Alexa Stephan
- Department of Urology, Urological Research Laboratory, Translational UroOncology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Jan-Henrik Suhrmann
- Department of Urology, Urological Research Laboratory, Translational UroOncology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Margaretha A Skowron
- Department of Urology, Urological Research Laboratory, Translational UroOncology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Yue Che
- Department of Urology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Gereon Poschmann
- Molecular Proteomics Laboratory (MPL), Biological and Medical Research Centre (BMFZ), Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Patrick Petzsch
- Genomics and Transcriptomics Laboratory, Biological and Medical Research Centre (BMFZ), Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Catena Kresbach
- Institute of Neuropathology, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Wasco Wruck
- Institute for Stem cell Research and Regenerative Medicine, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Pailin Pongratanakul
- Department of Urology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - James Adjaye
- Institute for Stem cell Research and Regenerative Medicine, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Kai Stühler
- Molecular Proteomics Laboratory (MPL), Biological and Medical Research Centre (BMFZ), Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Karl Köhrer
- Genomics and Transcriptomics Laboratory, Biological and Medical Research Centre (BMFZ), Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Ulrich Schüller
- Institute of Neuropathology, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Daniel Nettersheim
- Department of Urology, Urological Research Laboratory, Translational UroOncology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany; Center for Integrated Oncology Aachen, Bonn, Cologne, Düsseldorf (CIO ABCD), Germany.
| |
Collapse
|
21
|
Sun X, Wu LF, Altschuler SJ, Hata AN. Targeting therapy-persistent residual disease. NATURE CANCER 2024; 5:1298-1304. [PMID: 39289594 DOI: 10.1038/s43018-024-00819-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 07/26/2024] [Indexed: 09/19/2024]
Abstract
Disease relapse driven by acquired drug resistance limits the effectiveness of most systemic anti-cancer agents. Targeting persistent cancer cells in residual disease before relapse has emerged as a potential strategy for enhancing the efficacy and the durability of current therapies. However, barriers remain to implementing persister-directed approaches in the clinic. This Perspective discusses current preclinical and clinical complexities and outlines key steps toward the development of clinical strategies that target therapy-persistent residual disease.
Collapse
Affiliation(s)
- Xiaoxiao Sun
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
| | - Lani F Wu
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA.
| | - Steven J Altschuler
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA.
| | - Aaron N Hata
- Massachusetts General Hospital Cancer Center, Boston, MA, USA.
- Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
22
|
Muralidharan H, Hansen T, Steinle A, Schumacher D, Stickeler E, Maurer J. Breast Cancer Stem Cells Upregulate IRF6 in Stromal Fibroblasts to Induce Stromagenesis. Cells 2024; 13:1466. [PMID: 39273037 PMCID: PMC11393902 DOI: 10.3390/cells13171466] [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: 06/29/2024] [Revised: 08/11/2024] [Accepted: 08/25/2024] [Indexed: 09/15/2024] Open
Abstract
The microenvironment of a cancer stem cell (CSC) niche is often found in coexistence with cancer-associated fibroblasts (CAFs). Here, we show the first in-depth analysis of the interaction between primary triple-negative breast cancer stem cells (BCSCs) with fibroblasts. Using 2D co-culture models with specific seeding ratios, we identified stromal fibroblast aggregation at the BCSC cluster periphery, and, on closer observation, the aggregated fibroblasts was found to encircle BCSC clusters in nematic organization. In addition, collagen type I and fibronectin accumulation were also found at the BCSC-stromal periphery. MACE-Seq analysis of BCSC-encapsulating fibroblasts displayed the transformation of stromal fibroblasts to CAFs and the upregulation of fibrosis regulating genes of which the Interferon Regulatory Factor 6 (IRF6) gene was identified. Loss of function experiments with the IRF6 gene decreased fibroblast encapsulation around BCSC clusters in 2D co-cultures. In BCSC xenografts, fibroblast IRF6 expression led to an increase in the stromal area and fibroblast density in tumors, in addition to a reduction in necrotic growth. Based on our findings, we propose that fibroblast IRF6 function is an important factor in the development of the stromal microenvironment and in sustaining the BCSC tumor niche.
Collapse
Affiliation(s)
- Harshini Muralidharan
- Department of Obstetrics and Gynecology, University Hospital Aachen (UKA), 52074 Aachen, Germany
| | - Thomas Hansen
- Department of Obstetrics and Gynecology, University Hospital Aachen (UKA), 52074 Aachen, Germany
| | - Anja Steinle
- Department of Obstetrics and Gynecology, University Hospital Aachen (UKA), 52074 Aachen, Germany
| | - David Schumacher
- Department of Anesthesiology, University Hospital, RWTH Aachen University, 52074 Aachen, Germany
- Department of Nephrology and Clinical Immunology, RWTH Aachen University, 52074 Aachen, Germany
| | - Elmar Stickeler
- Department of Obstetrics and Gynecology, University Hospital Aachen (UKA), 52074 Aachen, Germany
- Center for Integrated Oncology (CIO), Aachen, Bonn, Cologne, Düsseldorf (ABCD), Venusberg-Campus 1, 53127 Bonn, Germany
| | - Jochen Maurer
- Department of Obstetrics and Gynecology, University Hospital Aachen (UKA), 52074 Aachen, Germany
- Center for Integrated Oncology (CIO), Aachen, Bonn, Cologne, Düsseldorf (ABCD), Venusberg-Campus 1, 53127 Bonn, Germany
| |
Collapse
|
23
|
Hu Q, Remsing Rix LL, Desai B, Miroshnychenko D, Li X, Welsh EA, Fang B, Wright GM, Chaudhary N, Kroeger JL, Doebele RC, Koomen JM, Haura EB, Marusyk A, Rix U. Cancer-associated fibroblasts confer ALK inhibitor resistance in EML4-ALK -driven lung cancer via concurrent integrin and MET signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.27.609975. [PMID: 39253447 PMCID: PMC11383036 DOI: 10.1101/2024.08.27.609975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/11/2024]
Abstract
Cancer-associated fibroblasts (CAFs) are associated with tumor progression and modulate drug sensitivity of cancer cells. However, the underlying mechanisms are often incompletely understood and crosstalk between tumor cells and CAFs involves soluble secreted as well as adhesion proteins. Interrogating a panel of non-small cell lung cancer (NSCLC) cell lines driven by EML4-ALK fusions, we observed substantial CAF-mediated drug resistance to clinical ALK tyrosine kinase inhibitors (TKIs). Array-based cytokine profiling of fibroblast-derived conditioned- media identified HGF-MET signaling as a major contributor to CAF-mediated paracrine resistance that can be overcome by MET TKIs. However, 'Cell Type specific labeling using Amino acid Precursors' (CTAP)-based expression and phosphoproteomics in direct coculture also highlighted a critical role for the fibronectin-integrin pathway. Flow cytometry analysis confirmed activation of integrin β1 (ITGB1) in lung cancer cells by CAF coculture. Treatment with pharmacological inhibitors, cancer cell-specific silencing or CRISPR-Cas9-mediated knockout of ITGB1 overcame adhesion protein-mediated resistance. Concurrent targeting of MET and integrin signaling effectively abrogated CAF-mediated resistance of EML4-ALK -driven NSCLC cells to ALK TKIs in vitro . Consistently, combination of the ALK TKI alectinib with the MET TKI capmatinib and/or the integrin inhibitor cilengitide was significantly more efficacious than single agent treatment in suppressing tumor growth using an in vivo EML4-ALK -dependent allograft mouse model of NSCLC. In summary, these findings emphasize the complexity of resistance-associated crosstalk between CAFs and cancer cells, which can involve multiple concurrent signaling pathways, and illustrate how comprehensive elucidation of paracrine and juxtacrine resistance mechanisms can inform on more effective therapeutic approaches.
Collapse
|
24
|
Chen M, Chen F, Gao Z, Li X, Hu L, Yang S, Zhao S, Song Z. CAFs and T cells interplay: The emergence of a new arena in cancer combat. Biomed Pharmacother 2024; 177:117045. [PMID: 38955088 DOI: 10.1016/j.biopha.2024.117045] [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: 04/25/2024] [Revised: 06/11/2024] [Accepted: 06/25/2024] [Indexed: 07/04/2024] Open
Abstract
The interaction between the immune system and the tumor matrix has a huge impact on the progression and treatment of cancer. This paper summarizes and discusses the crosstalk between T cells and cancer-associated fibroblasts (CAFs). CAFs can also produce inhibitors that counteract the function of T cells and promote tumor immune escape, while T cells can also engage in complex two-way interactions with CAFs through direct cell contact, the exchange of soluble factors such as cytokines, and the remodeling of the extracellular matrix. Precise targeted intervention can effectively reverse tumor-promoting crosstalk between T cells and CAFs, improve anti-tumor immune response, and provide a new perspective for cancer treatment. Therefore, it is important to deeply understand the mechanism of crosstalk between T cells and CAFs. This review aims to outline the underlying mechanisms of these interactions and discuss potential therapeutic strategies that may become fundamental tools in the treatment of cancer, especially hard-to-cure cancers.
Collapse
Affiliation(s)
- Minjie Chen
- Department of Surgery, the Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Fei Chen
- Department of Surgery, the Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Zhaofeng Gao
- Department of Surgery, the Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Xiaoping Li
- Department of Surgery, the Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Lingyu Hu
- Department of Surgery, the Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Shuying Yang
- Department of intensive medicine, The Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China.
| | - Siqi Zhao
- Department of Surgery, the Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China.
| | - Zhengwei Song
- Department of Surgery, the Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China.
| |
Collapse
|
25
|
Huang Y, Wang X, Wen C, Wang J, Zhou H, Wu L. Cancer-associated fibroblast-derived colony-stimulating factor 2 confers acquired osimertinib resistance in lung adenocarcinoma via promoting ribosome biosynthesis. MedComm (Beijing) 2024; 5:e653. [PMID: 39036343 PMCID: PMC11260172 DOI: 10.1002/mco2.653] [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: 11/22/2023] [Revised: 06/02/2024] [Accepted: 06/04/2024] [Indexed: 07/23/2024] Open
Abstract
Acquired resistance is a major obstacle to the therapeutic efficacy of osimertinib in lung adenocarcinoma (LUAD), but the underlying mechanisms are still not fully understood. Cancer-associated fibroblasts (CAFs) are the most abundant stromal cell type in LUAD tumor-microenvironment (TME) and have emerged as a key player in chemoresistance. However, the function of CAFs in osimertinib resistance is still unclear. Here, we showed that CAFs derived from osimertinib-resistant LUAD tissues (CAFOR) produced much more colony-stimulating factor 2 (CSF2) than those isolated from osimertinib-sensitive tissues. CAFOR-derived CSF2 activated the Janus kinase 2 (JAK2)/Signal transducer and activator of transcription 3 (STAT3) signaling pathway and upregulated lnc-CSRNP3 in LUAD cells. Lnc-CSRNP3 then promoted the expression of nearby gene CSRNP3 by recruiting chromodomain helicase DNA binding protein 9 (CHD9) and inhibited the phosphatase activity of the serine/threonine protein phosphatase 1 catalytic subunit α (PP1α), thereby induced osimertinib resistance by enhancing ribosome biogenesis. Collectively, our study reveals a critical role for CAFs in the development of osimertinib resistance and identifies the CSF2 pathway as an attractive target for monitoring osimertinib efficacy and overcoming osimertinib resistance in LUAD.
Collapse
Affiliation(s)
- Yutang Huang
- Institute of Life SciencesChongqing Medical UniversityChongqingChina
| | - Xiaoqing Wang
- Institute of Life SciencesChongqing Medical UniversityChongqingChina
| | - Chunjie Wen
- Institute of Life SciencesChongqing Medical UniversityChongqingChina
| | - Jingchan Wang
- School of StomatologyChongqing Medical UniversityChongqingChina
| | - Honghao Zhou
- Institute of Life SciencesChongqing Medical UniversityChongqingChina
- Pharmacogenetics Research InstituteInstitute of Clinical PharmacologyCentral South UniversityChangshaChina
| | - Lanxiang Wu
- Institute of Life SciencesChongqing Medical UniversityChongqingChina
| |
Collapse
|
26
|
Chen S, Hu T, Zhao J, Zhu Q, Wang J, Huang Z, Xiang C, Zhao R, Zhu C, Lu S, Han Y. Novel molecular subtypes of METex14 non-small cell lung cancer with distinct biological and clinical significance. NPJ Precis Oncol 2024; 8:159. [PMID: 39060379 PMCID: PMC11282101 DOI: 10.1038/s41698-024-00642-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 07/09/2024] [Indexed: 07/28/2024] Open
Abstract
Not all MET exon 14 skipping (METex14) NSCLC patients benefited from MET inhibitors. We hypothesized an inter-tumoral heterogeneity in METex14 NSCLC. Investigations at genomic and transcriptomic level were conducted in METex14 NSCLC samples from stage I-III and recurrent/metastatic patients as discovery and validation cohort. Four molecular subtypes were discovered. MET-Driven subtype, with the worst prognosis, displayed MET overexpression, enrichment of MET-related pathways, and higher infiltration of fibroblast and regulatory T cells. Immune-Activated subtype having the most idea long-term survival, had higher tertiary lymphoid structures, spatial co-option of PD-L1+ cancer cells, and GZMK+ CD8+ T cell. FGFR- and Bypass-Activated subtypes displayed FGFR2 overexpression and enrichments of multiple oncogenic pathways respectively. In the validation cohort, patients with MET-Driven subtype had better response to MET inhibitors than those with MET overexpression. Thus, molecular subtypes of METex14 NSCLC with distinct biological and clinical significance may indicate more precise therapeutic strategies for METex14 NSCLC patients.
Collapse
Affiliation(s)
- Shengnan Chen
- Department of Pathology, Shanghai Chest Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Tao Hu
- Department of Medicine, Amoy Diagnostics Co., Ltd., Xiamen, China
| | - Jikai Zhao
- Department of Pathology, Shanghai Chest Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Qian Zhu
- Department of Pathology, Shanghai Chest Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Jin Wang
- Department of Medicine, Amoy Diagnostics Co., Ltd., Xiamen, China
| | - Zhan Huang
- Department of Medicine, Amoy Diagnostics Co., Ltd., Xiamen, China
| | - Chan Xiang
- Department of Pathology, Shanghai Chest Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Ruiying Zhao
- Department of Pathology, Shanghai Chest Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Changbin Zhu
- Department of Medicine, Amoy Diagnostics Co., Ltd., Xiamen, China.
| | - Shun Lu
- Department of Oncology, Shanghai Chest Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China.
| | - Yuchen Han
- Department of Pathology, Shanghai Chest Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China.
| |
Collapse
|
27
|
Sajjadi SF, Salehi N, Sadeghi M. Comprehensive integrated single-cell RNA sequencing analysis of brain metastasis and glioma microenvironment: Contrasting heterogeneity landscapes. PLoS One 2024; 19:e0306220. [PMID: 39058687 PMCID: PMC11280140 DOI: 10.1371/journal.pone.0306220] [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: 01/31/2024] [Accepted: 06/12/2024] [Indexed: 07/28/2024] Open
Abstract
Understanding the specific type of brain malignancy, source of brain metastasis, and underlying transformation mechanisms can help provide better treatment and less harm to patients. The tumor microenvironment plays a fundamental role in cancer progression and affects both primary and metastatic cancers. The use of single-cell RNA sequencing to gain insights into the heterogeneity profiles in the microenvironment of brain malignancies is useful for guiding treatment decisions. To comprehensively investigate the heterogeneity in gliomas and brain metastasis originating from different sources (lung and breast), we integrated data from three groups of single-cell RNA-sequencing datasets obtained from GEO. We gathered and processed single-cell RNA sequencing data from 90,168 cells obtained from 17 patients. We then employed the R package Seurat for dataset integration. Next, we clustered the data within the UMAP space and acquired differentially expressed genes for cell categorization. Our results underscore the significance of macrophages as abundant and pivotal constituents of gliomas. In contrast, lung-to-brain metastases exhibit elevated numbers of AT2, cytotoxic CD4+ T, and exhausted CD8+ T cells. Conversely, breast-to-brain metastases are characterized by an abundance of epithelial and myCAF cells. Our study not only illuminates the variation in the TME between brain metastasis with different origins but also opens the door to utilizing established markers for these cell types to differentiate primary brain metastatic cancers.
Collapse
Affiliation(s)
- Seyedeh Fatemeh Sajjadi
- School of Biological Science, Institute for Research in Fundamental Sciences (IPM), Tehran, Iran
| | - Najmeh Salehi
- School of Biological Science, Institute for Research in Fundamental Sciences (IPM), Tehran, Iran
| | - Mehdi Sadeghi
- School of Biological Science, Institute for Research in Fundamental Sciences (IPM), Tehran, Iran
- National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran
| |
Collapse
|
28
|
Li K, Wang R, Liu GW, Peng ZY, Wang JC, Xiao GD, Tang SC, Du N, Zhang J, Zhang J, Ren H, Sun X, Yang YP, Liu DP. Refining the optimal CAF cluster marker for predicting TME-dependent survival expectancy and treatment benefits in NSCLC patients. Sci Rep 2024; 14:16766. [PMID: 39034310 PMCID: PMC11271481 DOI: 10.1038/s41598-024-55375-0] [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: 07/11/2023] [Accepted: 02/22/2024] [Indexed: 07/23/2024] Open
Abstract
The tumor microenvironment (TME) plays a pivotal role in the onset, progression, and treatment response of cancer. Among the various components of the TME, cancer-associated fibroblasts (CAFs) are key regulators of both immune and non-immune cellular functions. Leveraging single-cell RNA sequencing (scRNA) data, we have uncovered previously hidden and promising roles within this specific CAF subgroup, paving the way for its clinical application. However, several critical questions persist, primarily stemming from the heterogeneous nature of CAFs and the use of different fibroblast markers in various sample analyses, causing confusion and hindrance in their clinical implementation. In this groundbreaking study, we have systematically screened multiple databases to identify the most robust marker for distinguishing CAFs in lung cancer, with a particular focus on their potential use in early diagnosis, staging, and treatment response evaluation. Our investigation revealed that COL1A1, COL1A2, FAP, and PDGFRA are effective markers for characterizing CAF subgroups in most lung adenocarcinoma datasets. Through comprehensive analysis of treatment responses, we determined that COL1A1 stands out as the most effective indicator among all CAF markers. COL1A1 not only deciphers the TME signatures related to CAFs but also demonstrates a highly sensitive and specific correlation with treatment responses and multiple survival outcomes. For the first time, we have unveiled the distinct roles played by clusters of CAF markers in differentiating various TME groups. Our findings confirm the sensitive and unique contributions of CAFs to the responses of multiple lung cancer therapies. These insights significantly enhance our understanding of TME functions and drive the translational application of extensive scRNA sequence results. COL1A1 emerges as the most sensitive and specific marker for defining CAF subgroups in scRNA analysis. The CAF ratios represented by COL1A1 can potentially serve as a reliable predictor of treatment responses in clinical practice, thus providing valuable insights into the influential roles of TME components. This research marks a crucial step forward in revolutionizing our approach to cancer diagnosis and treatment.
Collapse
Affiliation(s)
- Kai Li
- Department of Otorhinolaryngology‑Head and Neck Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Rui Wang
- Department of Thoracic Surgery and Oncology, Cancer Centre, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, Shaanxi, China
| | - Guo-Wei Liu
- Department of Thoracic Surgery, Qinghai Provincial People's Hospital, Gonghe Road No. 2, Chengdong District, Xining, 810007, Qinghai, China
| | - Zi-Yang Peng
- School of Future Technology, National Local Joint Engineering Research Center for Precision Surgery and Regenerative Medicine, Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Ji-Chang Wang
- Department of Vascular Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Guo-Dong Xiao
- Oncology Department, The First Affiliated Hospital of Zhengzhou University, Zheng Zhou, 450052, Henan, China
| | - Shou-Ching Tang
- Section of Hematology Oncology, Department of Internal Medicine, LSUHSC Cancer Center, School of Medicine, 1700 Tulane Avenue, New Orleans, LA, 70112, USA
| | - Ning Du
- Department of Thoracic Surgery and Oncology, Cancer Centre, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, Shaanxi, China
| | - Jia Zhang
- Department of Thoracic Surgery and Oncology, Cancer Centre, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, Shaanxi, China
| | - Jing Zhang
- Department of Thoracic Surgery and Oncology, Cancer Centre, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, Shaanxi, China
| | - Hong Ren
- Department of Thoracic Surgery and Oncology, Cancer Centre, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, Shaanxi, China
| | - Xin Sun
- Department of Thoracic Surgery and Oncology, Cancer Centre, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, Shaanxi, China
- Department of Pathology, Anatomy and Cell Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - Yi-Ping Yang
- Department of Radiotherapy, Shaanxi Provincial Tumor Hospital, 309 Yanta W Rd, Yanta District, Xi'an, 710063, Shaanxi, China.
| | - Da-Peng Liu
- Department of Thoracic Surgery and Oncology, Cancer Centre, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, Shaanxi, China.
| |
Collapse
|
29
|
Zhang C, Sun YX, Yi DC, Jiang BY, Yan LX, Liu ZD, Peng LS, Zhang WJ, Sun H, Chen ZY, Wang DH, Peng D, Chen SA, Li SQ, Zhang Z, Tan XY, Yang J, Zhao ZY, Zhang WT, Su J, Li YS, Liao RQ, Dong S, Xu CR, Zhou Q, Yang XN, Wu YL, Zhang ZM, Zhong WZ. Neoadjuvant sintilimab plus chemotherapy in EGFR-mutant NSCLC: Phase 2 trial interim results (NEOTIDE/CTONG2104). Cell Rep Med 2024; 5:101615. [PMID: 38897205 PMCID: PMC11293361 DOI: 10.1016/j.xcrm.2024.101615] [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: 11/02/2023] [Revised: 01/31/2024] [Accepted: 05/23/2024] [Indexed: 06/21/2024]
Abstract
The clinical efficacy of neoadjuvant immunotherapy plus chemotherapy remains elusive in localized epidermal growth factor receptor (EGFR)-mutant non-small cell lung cancer (NSCLC). Here, we report interim results of a Simon's two-stage design, phase 2 trial using neoadjuvant sintilimab with carboplatin and nab-paclitaxel in resectable EGFR-mutant NSCLC. All 18 patients undergo radical surgery, with one patient experiencing surgery delay. Fourteen patients exhibit confirmed radiological response, with 44% achieving major pathological response (MPR) and no pathological complete response (pCR). Similar genomic alterations are observed before and after treatment without influencing the efficacy of subsequent EGFR-tyrosine kinase inhibitors (TKIs) in vitro. Infiltration and T cell receptor (TCR) clonal expansion of CCR8+ regulatory T (Treg)hi/CXCL13+ exhausted T (Tex)lo cells define a subtype of EGFR-mutant NSCLC highly resistant to immunotherapy, with the phenotype potentially serving as a promising signature to predict immunotherapy efficacy. Informed circulating tumor DNA (ctDNA) detection in EGFR-mutant NSCLC could help identify patients nonresponsive to neoadjuvant immunochemotherapy. These findings provide supportive data for the utilization of neoadjuvant immunochemotherapy and insight into immune resistance in EGFR-mutant NSCLC.
Collapse
Affiliation(s)
- Chao Zhang
- Department of Pulmonary Surgery, Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China; Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China; School of Medicine, South China University of Technology, Guangzhou, China
| | - Yu-Xuan Sun
- School of Life Sciences, Peking University, Beijing, China
| | - Ding-Cheng Yi
- School of Life Sciences, Peking University, Beijing, China
| | - Ben-Yuan Jiang
- Department of Pulmonary Surgery, Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China; Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Li-Xu Yan
- Department of Pathology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Ze-Dao Liu
- School of Life Sciences, Peking University, Beijing, China
| | - Li-Shan Peng
- Department of Pulmonary Surgery, Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China; Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Wen-Jie Zhang
- School of Life Sciences, Peking University, Beijing, China
| | - Hao Sun
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Zhi-Yong Chen
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China; Department of Radiation Therapy, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | | | - Di Peng
- Burning Rock Biotech, Guangzhou, China
| | | | - Si-Qi Li
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Ze Zhang
- Institute of Biomedical Research, Yunnan University, Kunming, China
| | - Xiao-Yue Tan
- PET Center, Department of Nuclear Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Jie Yang
- Department of Pulmonary Surgery, Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China; Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Zhang-Yi Zhao
- School of Life Sciences, Peking University, Beijing, China
| | - Wan-Ting Zhang
- School of Life Sciences, Peking University, Beijing, China
| | - Jian Su
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Yang-Si Li
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Ri-Qiang Liao
- Department of Pulmonary Surgery, Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China; Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Song Dong
- Department of Pulmonary Surgery, Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China; Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Chong-Rui Xu
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Qing Zhou
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Xue-Ning Yang
- Department of Pulmonary Surgery, Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China; Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Yi-Long Wu
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Ze-Min Zhang
- School of Life Sciences, Peking University, Beijing, China; BIOPIC, Beijing Advanced Innovation Center for Genomics, Peking University, Beijing, China
| | - Wen-Zhao Zhong
- Department of Pulmonary Surgery, Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China; Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China.
| |
Collapse
|
30
|
Zhu C, Chen Z, Wang S, Cao J, Cheng Y, Zheng M. Single-cell analyzing of tumor microenvironment and cell adhesion between early and late-stage lung cancer. Mol Immunol 2024; 171:1-11. [PMID: 38696904 DOI: 10.1016/j.molimm.2024.04.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 04/14/2024] [Accepted: 04/24/2024] [Indexed: 05/04/2024]
Abstract
Lung adenocarcinoma (LUAD) is a highly heterogeneous disease that threaten human life with serious incidence and high mortality. High heterogeneity of tumor microenvironment (TME) was reported in multiple studies. However, the factor of controlling the tumor migration progression between eary and late-stage LUAD is still not fully understood. In this study, we conducted a comprehensive analysis of single-cell RNA sequencing (scRNA-seq) data of LUAD obtained from the GEO database. The identification of cell clusters revealed significant expansion of epithelial cells in late-stage patients. Interpretation of the cell-cell communication results between early-stage and late-stage patient samples indicated that early tumor cells may interact with epithelial cells through the TGF-β pathway to promote tumor progression. The cell cycle analysis demonstrated a significant increase in the number of cells in the G2 and M phases in late-stage lung cancer. Further analysis using Non-negative Matrix Factorization (NMF) revealed early-stage cell-specific gene features involved in cell adhesion-related biological processes. Among these, the Tensin (TNS) gene family, particularly TNS1, showed high expression in epithelial cells and fibroblasts of early-stage samples, specifically associated with cell adhesion. Survival analysis using TCGA database for LUAD demonstrated that patients with high expression of TNS1 exhibited significantly higher overall survival rates compared to those with low expression. Immunofluorescence experiments have demonstrated co-expression of TNS1 with fibroblast and tumor cell markers (α-SMA and EPCAM). Immunohistochemistry experiments further validated the significantly higher expression levels of TNS1 in early-stage LUAD tissues compared to late-stage lung cancer tissues (P<0.05). Pathway experiments have shown that early-stage tumor patients with high expression of TNS1 exhibit stronger phosphorylation levels of Akt and mTOR, indicating a more potent activation of the Akt/mTOR signaling pathway. In conclusion, the results of this study demonstrate that TNS1 is an adhesive molecule in the immune microenvironment of early-stage tumor cells, and it may serve as a novel prognostic marker for lug cancer.
Collapse
Affiliation(s)
- Chaonan Zhu
- Department of Thoracic Surgery, North China University of Science and Technology Affiliated Hospital, Tangshan, Hebei 063000, China.
| | - Zhiquan Chen
- Department of Thoracic Surgery, North China University of Science and Technology Affiliated Hospital, Tangshan, Hebei 063000, China
| | - Shuai Wang
- Department of Thoracic Surgery, North China University of Science and Technology Affiliated Hospital, Tangshan, Hebei 063000, China
| | - Junmei Cao
- Department of Thoracic Surgery, North China University of Science and Technology Affiliated Hospital, Tangshan, Hebei 063000, China
| | - Yuan Cheng
- Department of Thoracic Surgery, North China University of Science and Technology Affiliated Hospital, Tangshan, Hebei 063000, China
| | - Maogen Zheng
- Department of Thoracic Surgery, North China University of Science and Technology Affiliated Hospital, Tangshan, Hebei 063000, China
| |
Collapse
|
31
|
Sentek H, Braun A, Budeus B, Klein D. Non-small cell lung cancer cells and concomitant cancer therapy induce a resistance-promoting phenotype of tumor-associated mesenchymal stem cells. Front Oncol 2024; 14:1406268. [PMID: 39011489 PMCID: PMC11246879 DOI: 10.3389/fonc.2024.1406268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Accepted: 05/30/2024] [Indexed: 07/17/2024] Open
Abstract
Introduction The tumor microenvironment gained attraction over the last decades as stromal cells significantly impact on tumor development, progression and metastasis, and immune evasion as well as on cancer therapy resistance. We previously reported that lung-resident mesenchymal stem cells (MSCs) were mobilized and activated in non-small cell lung cancer (NSCLC) progression and could even mediate radiation resistance in co-cultured NSCLC cells. Methods We investigated how MSCs were affected by NSCLC cells in combination with cancer (radiation) therapy in indirect co-cultures using tumor-conditioned medium and Transwells or direct three-dimensional NSCLC-MSC spheroid co-cultures in order to unravel the resistance-mediating action of tumor-associated MSCs. Results Although no obvious phenotypic and functional alterations in MSCs following NSCLC co-culture could be observed, MSC senescence was induced following co-applied radiotherapy (RT). Global gene expression profiling, in combination with gene set enrichment analysis upon treatment, was used to confirm the senescent phenotype of irradiated MSC and to reveal relevant senescence-associated secretory phenotype (SASP) factors that could meditate NSCLC RT resistance. We identified senescent tumor-associated MSC-derived serine proteinase inhibitor (serpin) E1/PAI1 as potential SASP factor mediating NSCLC progression and RT resistance. Discussion Specified intra-tumor-stroma interactions and cell type-specific pro-tumorigenic functions could not only improve lung cancer classification but could even be used for a more precise profiling of individual patients, finally paving an additional way for the discovery of potential drug targets for NSCLC patients.
Collapse
Affiliation(s)
| | | | | | - Diana Klein
- Institute of Cell Biology (Cancer Research), University of Duisburg-Essen, University Hospital, Essen, Germany
| |
Collapse
|
32
|
Miti T, Desai B, Miroshnychenko D, Basanta D, Marusyk A. Dissecting the Spatially Restricted Effects of Microenvironment-Mediated Resistance on Targeted Therapy Responses. Cancers (Basel) 2024; 16:2405. [PMID: 39001467 PMCID: PMC11240540 DOI: 10.3390/cancers16132405] [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: 05/09/2024] [Revised: 06/16/2024] [Accepted: 06/26/2024] [Indexed: 07/16/2024] Open
Abstract
The response of tumors to anti-cancer therapies is defined not only by cell-intrinsic therapy sensitivities but also by local interactions with the tumor microenvironment. Fibroblasts that make tumor stroma have been shown to produce paracrine factors that can strongly reduce the sensitivity of tumor cells to many types of targeted therapies. Moreover, a high stroma/tumor ratio is generally associated with poor survival and reduced therapy responses. However, in contrast to advanced knowledge of the molecular mechanisms responsible for stroma-mediated resistance, its effect on the ability of tumors to escape therapeutic eradication remains poorly understood. To a large extent, this gap of knowledge reflects the challenge of accounting for the spatial aspects of microenvironmental resistance, especially over longer time frames. To address this problem, we integrated spatial inferences of proliferation-death dynamics from an experimental animal model of targeted therapy responses with spatial mathematical modeling. With this approach, we dissected the impact of tumor/stroma distribution, magnitude and distance of stromal effects. While all of the tested parameters affected the ability of tumor cells to resist elimination, spatial patterns of stroma distribution within tumor tissue had a particularly strong impact.
Collapse
Affiliation(s)
- Tatiana Miti
- Department of Integrative Mathematical Oncology, H. Lee Moffitt Cancer Centre and Research Institute, Tampa, FL 33612, USA;
| | - Bina Desai
- Department of Tumor Microenvironment and Metastasis, H. Lee Moffitt Cancer Centre and Research Institute, Tampa, FL 33612, USA (D.M.)
- Cancer Biology Ph.D. Program, University of South Florida, Tampa, FL 33620, USA
| | - Daria Miroshnychenko
- Department of Tumor Microenvironment and Metastasis, H. Lee Moffitt Cancer Centre and Research Institute, Tampa, FL 33612, USA (D.M.)
| | - David Basanta
- Department of Integrative Mathematical Oncology, H. Lee Moffitt Cancer Centre and Research Institute, Tampa, FL 33612, USA;
| | - Andriy Marusyk
- Department of Tumor Microenvironment and Metastasis, H. Lee Moffitt Cancer Centre and Research Institute, Tampa, FL 33612, USA (D.M.)
| |
Collapse
|
33
|
Xiao Y, Wang Z, Gu M, Wei P, Wang X, Li W. Cancer-associated fibroblasts: heterogeneity and their role in the tumor immune response. Clin Exp Med 2024; 24:126. [PMID: 38864912 PMCID: PMC11169017 DOI: 10.1007/s10238-024-01375-3] [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: 02/11/2024] [Accepted: 05/13/2024] [Indexed: 06/13/2024]
Abstract
In recent decades, many reports have been published on the composition and function of the tumor microenvironment (TME), among which cancer-associated fibroblasts (CAFs) have received much attention. CAFs have different degrees of heterogeneity in terms of their origin, phenotype, and function and can be divided into different subpopulations. These subgroups may play different roles in the occurrence and development of tumors. In addition, CAFs are closely associated with tumor immunity and have been found to regulate immune cell activity and to suppress the tumor immune response. In this review, we systematize the heterogeneity and characteristics of CAFs, discuss how specific CAF subgroups contribute to cancer progression by inducing an immunosuppressive microenvironment, and finally, we examine the future clinical applications of CAF subgroups.
Collapse
Affiliation(s)
- Yuxuan Xiao
- Cancer Research Center, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Ziyu Wang
- Cancer Research Center, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Meng Gu
- Cancer Research Center, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Panjian Wei
- Cancer Research Center, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Xiaojue Wang
- Cancer Research Center, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Weiying Li
- Cancer Research Center, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China.
| |
Collapse
|
34
|
Cheon I, Lee S, Oh S, Ahn YH. miR-200-mediated inactivation of cancer-associated fibroblasts via targeting of NRP2-VEGFR signaling attenuates lung cancer invasion and metastasis. MOLECULAR THERAPY. NUCLEIC ACIDS 2024; 35:102194. [PMID: 38766528 PMCID: PMC11101731 DOI: 10.1016/j.omtn.2024.102194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 04/18/2024] [Indexed: 05/22/2024]
Abstract
Cancer-associated fibroblasts (CAFs) play a substantial role in promoting cancer cell motility, drug resistance, angiogenesis, and metastasis; therefore, extensive research has been conducted to determine their mode of activation. We aimed to identify whether miRNA-200 (miR-200), a widely recognized suppressor of epithelial-mesenchymal transition, prevents CAFs from promoting cancer progression. Overexpression of miR-200 prevented CAFs from promoting lung cancer cell migration, invasion, tumorigenicity, and metastasis. Additionally, miR-200 suppressed the ability of CAFs to recruit and polarize macrophages toward the M2 phenotype, as well as the migration and tube formation of vascular endothelial cells. NRP2, a co-receptor of vascular endothelial growth factor receptor (VEGFR), was confirmed to be a target of miR-200, which mediates the functional activity of miR-200 in CAFs. NRP2-VEGFR signaling facilitates the secretion of VEGF-D and pleiotrophin from CAFs, leading to the activation of cancer cell migration and invasion. These findings suggest that miR-200 remodels CAFs to impede cancer progression and metastasis and that miR-200 and NRP2 are potential therapeutic targets in the treatment of lung cancer.
Collapse
Affiliation(s)
- Inyoung Cheon
- Department of Molecular Medicine and Inflammation-Cancer Microenvironment Research Center, College of Medicine, Ewha Womans University, Seoul 07804, Korea
| | - Sieun Lee
- Department of Molecular Medicine and Inflammation-Cancer Microenvironment Research Center, College of Medicine, Ewha Womans University, Seoul 07804, Korea
| | - Seonyeong Oh
- Department of Molecular Medicine and Inflammation-Cancer Microenvironment Research Center, College of Medicine, Ewha Womans University, Seoul 07804, Korea
| | - Young-Ho Ahn
- Department of Molecular Medicine and Inflammation-Cancer Microenvironment Research Center, College of Medicine, Ewha Womans University, Seoul 07804, Korea
| |
Collapse
|
35
|
Enfield KS, Colliver E, Lee C, Magness A, Moore DA, Sivakumar M, Grigoriadis K, Pich O, Karasaki T, Hobson PS, Levi D, Veeriah S, Puttick C, Nye EL, Green M, Dijkstra KK, Shimato M, Akarca AU, Marafioti T, Salgado R, Hackshaw A, Jamal-Hanjani M, van Maldegem F, McGranahan N, Glass B, Pulaski H, Walk E, Reading JL, Quezada SA, Hiley CT, Downward J, Sahai E, Swanton C, Angelova M. Spatial Architecture of Myeloid and T Cells Orchestrates Immune Evasion and Clinical Outcome in Lung Cancer. Cancer Discov 2024; 14:1018-1047. [PMID: 38581685 PMCID: PMC11145179 DOI: 10.1158/2159-8290.cd-23-1380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 02/27/2024] [Accepted: 03/22/2024] [Indexed: 04/08/2024]
Abstract
Understanding the role of the tumor microenvironment (TME) in lung cancer is critical to improving patient outcomes. We identified four histology-independent archetype TMEs in treatment-naïve early-stage lung cancer using imaging mass cytometry in the TRACERx study (n = 81 patients/198 samples/2.3 million cells). In immune-hot adenocarcinomas, spatial niches of T cells and macrophages increased with clonal neoantigen burden, whereas such an increase was observed for niches of plasma and B cells in immune-excluded squamous cell carcinomas (LUSC). Immune-low TMEs were associated with fibroblast barriers to immune infiltration. The fourth archetype, characterized by sparse lymphocytes and high tumor-associated neutrophil (TAN) infiltration, had tumor cells spatially separated from vasculature and exhibited low spatial intratumor heterogeneity. TAN-high LUSC had frequent PIK3CA mutations. TAN-high tumors harbored recently expanded and metastasis-seeding subclones and had a shorter disease-free survival independent of stage. These findings delineate genomic, immune, and physical barriers to immune surveillance and implicate neutrophil-rich TMEs in metastasis. SIGNIFICANCE This study provides novel insights into the spatial organization of the lung cancer TME in the context of tumor immunogenicity, tumor heterogeneity, and cancer evolution. Pairing the tumor evolutionary history with the spatially resolved TME suggests mechanistic hypotheses for tumor progression and metastasis with implications for patient outcome and treatment. This article is featured in Selected Articles from This Issue, p. 897.
Collapse
Affiliation(s)
- Katey S.S. Enfield
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Emma Colliver
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Claudia Lee
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Alastair Magness
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, United Kingdom
| | - David A. Moore
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, United Kingdom
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, United Kingdom
- Department of Cellular Pathology, University College London Hospitals, London, United Kingdom
| | - Monica Sivakumar
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, United Kingdom
| | - Kristiana Grigoriadis
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, United Kingdom
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, United Kingdom
- Cancer Genome Evolution Research Group, Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, United Kingdom
| | - Oriol Pich
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Takahiro Karasaki
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, United Kingdom
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, United Kingdom
- Cancer Metastasis Laboratory, University College London Cancer Institute, London, United Kingdom
| | - Philip S. Hobson
- Flow Cytometry, The Francis Crick Institute, London, United Kingdom
| | - Dina Levi
- Flow Cytometry, The Francis Crick Institute, London, United Kingdom
| | - Selvaraju Veeriah
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, United Kingdom
| | - Clare Puttick
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, United Kingdom
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, United Kingdom
- Cancer Genome Evolution Research Group, Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, United Kingdom
| | - Emma L. Nye
- Experimental Histopathology, The Francis Crick Institute, London, United Kingdom
| | - Mary Green
- Experimental Histopathology, The Francis Crick Institute, London, United Kingdom
| | - Krijn K. Dijkstra
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Masako Shimato
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Ayse U. Akarca
- Department of Cellular Pathology, University College London Hospitals, London, United Kingdom
| | - Teresa Marafioti
- Department of Cellular Pathology, University College London Hospitals, London, United Kingdom
| | - Roberto Salgado
- Department of Pathology, ZAS Hospitals, Antwerp, Belgium
- Division of Research, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Allan Hackshaw
- Cancer Research UK and University College London Cancer Trials Centre, London, United Kingdom
| | | | - Mariam Jamal-Hanjani
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, United Kingdom
- Cancer Metastasis Laboratory, University College London Cancer Institute, London, United Kingdom
- Department of Oncology, University College London Hospitals, London, United Kingdom
| | - Febe van Maldegem
- Oncogene Biology Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Nicholas McGranahan
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, United Kingdom
- Cancer Genome Evolution Research Group, Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, United Kingdom
| | | | | | | | - James L. Reading
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, United Kingdom
- Pre-cancer Immunology Laboratory, University College London Cancer Institute, London, United Kingdom
- Immune Regulation and Tumour Immunotherapy Group, Cancer Immunology Unit, Research Department of Haematology, University College London Cancer Institute, London, United Kingdom
| | - Sergio A. Quezada
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, United Kingdom
- Immune Regulation and Tumour Immunotherapy Group, Cancer Immunology Unit, Research Department of Haematology, University College London Cancer Institute, London, United Kingdom
| | - Crispin T. Hiley
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, United Kingdom
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, United Kingdom
| | - Julian Downward
- Oncogene Biology Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Erik Sahai
- Tumour Cell Biology Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Charles Swanton
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, United Kingdom
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, United Kingdom
- Department of Oncology, University College London Hospitals, London, United Kingdom
| | - Mihaela Angelova
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, United Kingdom
| |
Collapse
|
36
|
Hou Y, Yang Z, Xiang B, Liu J, Geng L, Xu D, Zhan M, Xu Y, Zhang B. Metformin is a potential therapeutic for COVID-19/LUAD by regulating glucose metabolism. Sci Rep 2024; 14:12406. [PMID: 38811809 PMCID: PMC11137110 DOI: 10.1038/s41598-024-63081-0] [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: 03/13/2024] [Accepted: 05/24/2024] [Indexed: 05/31/2024] Open
Abstract
Lung adenocarcinoma (LUAD) is the most common and aggressive subtype of lung cancer, and coronavirus disease 2019 (COVID-19) has become a serious public health threat worldwide. Patients with LUAD and COVID-19 have a poor prognosis. Therefore, finding medications that can be used to treat COVID-19/LUAD patients is essential. Bioinformatics analysis was used to identify 20 possible metformin target genes for the treatment of COVID-19/LUAD. PTEN and mTOR may serve as hub target genes of metformin. Metformin may be able to cure COVID-19/LUAD comorbidity through energy metabolism, oxidoreductase NADH activity, FoxO signalling pathway, AMPK signalling system, and mTOR signalling pathway, among other pathways, according to the results of bioinformatic research. Metformin has ability to inhibit the proliferation of A549 cells, according to the results of colony formation and proliferation assays. In A549 cells, metformin increased glucose uptake and lactate generation, while decreasing ATP synthesis and the NAD+/NADH ratio. In summary, PTEN and mTOR may be potential targets of metformin for the treatment of COVID-19/LUAD. The mechanism by which metformin inhibits lung adenocarcinoma cell proliferation may be related to glucose metabolism regulated by PI3K/AKT signalling and mTOR signalling pathways. Our study provides a new theoretical basis for the treatment of COVID-19/LUAD.
Collapse
Affiliation(s)
- Yongwang Hou
- Clinical Laboratory, The First Affiliated Hospital of Hebei North University, Zhangjiakou, 075000, Hebei, China.
| | - Zhicong Yang
- Central Laboratory, The First Affiliated Hospital of Hebei North University, Zhangjiakou, 075000, Hebei, China
| | - Baoli Xiang
- Respiratory Department, The First Affiliated Hospital of Hebei North University, Zhangjiakou, 075000, Hebei, China
| | - Jiangmin Liu
- Clinical Laboratory, The First Affiliated Hospital of Hebei North University, Zhangjiakou, 075000, Hebei, China
| | - Lina Geng
- Clinical Laboratory, The First Affiliated Hospital of Hebei North University, Zhangjiakou, 075000, Hebei, China
| | - Dandan Xu
- Central Laboratory, The First Affiliated Hospital of Hebei North University, Zhangjiakou, 075000, Hebei, China
| | - Minghua Zhan
- Clinical Laboratory, The First Affiliated Hospital of Hebei North University, Zhangjiakou, 075000, Hebei, China
| | - Yuhuan Xu
- Clinical Laboratory, The First Affiliated Hospital of Hebei North University, Zhangjiakou, 075000, Hebei, China
| | - Bin Zhang
- Clinical Laboratory, The First Affiliated Hospital of Hebei North University, Zhangjiakou, 075000, Hebei, China.
| |
Collapse
|
37
|
Dhungel N, Dragoi AM. Exploring the multifaceted role of direct interaction between cancer cells and fibroblasts in cancer progression. Front Mol Biosci 2024; 11:1379971. [PMID: 38863965 PMCID: PMC11165130 DOI: 10.3389/fmolb.2024.1379971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 04/24/2024] [Indexed: 06/13/2024] Open
Abstract
The interaction between the tumor microenvironment (TME) and the cancer cells is a complex and mutually beneficial system that leads to rapid cancer cells proliferation, metastasis, and resistance to therapy. It is now recognized that cancer cells are not isolated, and tumor progression is governed among others, by many components of the TME. The reciprocal cross-talk between cancer cells and their microenvironment can be indirect through the secretion of extracellular matrix (ECM) proteins and paracrine signaling through exosomes, cytokines, and growth factors, or direct by cell-to-cell contact mediated by cell surface receptors and adhesion molecules. Among TME components, cancer-associated fibroblasts (CAFs) are of unique interest. As one of the most abundant components of the TME, CAFs play key roles in the reorganization of the extracellular matrix, facilitating metastasis and chemotherapy evasion. Both direct and indirect roles have been described for CAFs in modulating tumor progression. In this review, we focus on recent advances in understanding the role of direct contact between cancer cells and cancer-associated fibroblasts (CAFs) in driving tumor development and metastasis. We also summarize recent findings on the role of direct contact between cancer cells and CAFs in chemotherapy resistance.
Collapse
Affiliation(s)
- Nilu Dhungel
- Department of Molecular and Cellular Physiology, LSUHSC, Shreveport, LA, United States
| | - Ana-Maria Dragoi
- Department of Molecular and Cellular Physiology, LSUHSC, Shreveport, LA, United States
- Feist-Weiller Cancer Center, INLET Core, LSUHSC, Shreveport, LA, United States
| |
Collapse
|
38
|
Jiang Z, Zhang Z, You X, Ye L, Lin W, Liu L, Cao Y, Pan J. Crosstalk among T cells, epithelial cells, and fibroblasts identifies a prognostic signature in oral squamous cell carcinoma. Oral Dis 2024; 30:1956-1969. [PMID: 37491735 DOI: 10.1111/odi.14688] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 06/19/2023] [Accepted: 07/10/2023] [Indexed: 07/27/2023]
Abstract
OBJECTIVES This study aimed to analyze the crosstalk network among T cells, epithelial cells, and fibroblasts in the tumor microenvironment of oral squamous cell carcinoma (OSCC) and to determine their prognostic values. MATERIALS AND METHODS Single-cell subpopulation identification and communication analysis identified crosstalk markers. The least absolute shrinkage and selection operator Cox analysis identified key prognostic features by integrating the bulk transcriptome and clinical parameters. Functional analysis and immune infiltration were explored to determine possible mechanisms. RESULTS Interactions between epithelial cells and fibroblasts primarily involve MIF, MK, PTN, IGF, EGF, and PERIOSTIN, whereas T cells interact with epithelial cells and fibroblasts through MIF, CXCL, PAR, IFN, and EGF signals. We constructed a novel prognostic feature comprising 13 crosstalk genes: HBEGF, FGF7, GRN, ITGB5, CXCR6, ERBB2, AREG, F2RL2, NAMPT, KLK12, HMGB2, TUBA1B, and KLRD1. Patients were stratified based on the RiskScore. Functional analysis revealed that the high-risk group was enriched in immunosuppressive pathways (p < 0.001). Immune checkpoints including PD-1, PD-L1, and CTLA4 were more highly expressed in the high-risk group (p < 0.05). CONCLUSIONS The crosstalk network among T cells, epithelial cells, and fibroblasts is complex and may have implications for prognosis and clinical treatments of OSCC patients.
Collapse
Affiliation(s)
- Zhishen Jiang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Zijian Zhang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xiaotong You
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Li Ye
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Weimin Lin
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Liu Liu
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Conservative Dentistry and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yubin Cao
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jian Pan
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
39
|
Mathieson L, Koppensteiner L, Dorward DA, O'Connor RA, Akram AR. Cancer-associated fibroblasts expressing fibroblast activation protein and podoplanin in non-small cell lung cancer predict poor clinical outcome. Br J Cancer 2024; 130:1758-1769. [PMID: 38582812 PMCID: PMC11130154 DOI: 10.1038/s41416-024-02671-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 03/21/2024] [Accepted: 03/22/2024] [Indexed: 04/08/2024] Open
Abstract
BACKGROUND Cancer-associated fibroblasts (CAFs) are a dominant cell type in the stroma of non-small cell lung cancer (NSCLC). Fibroblast heterogeneity reflects subpopulations of CAFs, which can influence prognosis and treatment efficacy. We describe the subtypes of CAFs in NSCLC. METHODS Primary human NSCLC resections were assessed by flow cytometry and multiplex immunofluorescence for markers of fibroblast activation which allowed identification of CAF subsets. Survival data were analysed for our NSCLC cohort consisting of 163 patients to understand prognostic significance of CAF subsets. RESULTS We identified five CAF populations, termed CAF S1-S5. CAF-S5 represents a previously undescribed population, and express FAP and PDPN but lack the myofibroblast marker αSMA, whereas CAF-S1 populations express all three. CAF-S5 are spatially further from tumour regions then CAF-S1 and scRNA data demonstrate an inflammatory phenotype. The presence of CAF-S1 or CAF-S5 is correlated to worse survival outcome in NSCLC, despite curative resection, highlighting the prognostic importance of CAF subtypes in NSCLC. TCGA data suggest the predominance of CAF-S5 has a poor prognosis across several cancer types. CONCLUSION This study describes the fibroblast heterogeneity in NSCLC and the prognostic importance of the novel CAF-S5 subset where its presence correlates to worse survival outcome.
Collapse
Affiliation(s)
- Layla Mathieson
- Centre for Inflammation Research, Institute of Regeneration and Repair, University of Edinburgh, 5 Little France Dr, Edinburgh BioQuarter, Edinburgh, EH16 4UU, UK
- Translational Healthcare Technologies Group, Centre for Inflammation Research, Institute of Regeneration and Repair, University of Edinburgh, 5 Little France Dr, Edinburgh BioQuarter, Edinburgh, EH16 4UU, UK
| | - Lilian Koppensteiner
- Centre for Inflammation Research, Institute of Regeneration and Repair, University of Edinburgh, 5 Little France Dr, Edinburgh BioQuarter, Edinburgh, EH16 4UU, UK
- Translational Healthcare Technologies Group, Centre for Inflammation Research, Institute of Regeneration and Repair, University of Edinburgh, 5 Little France Dr, Edinburgh BioQuarter, Edinburgh, EH16 4UU, UK
| | - David A Dorward
- Department of Pathology, Royal Infirmary of Edinburgh, Edinburgh, UK
| | - Richard A O'Connor
- Centre for Inflammation Research, Institute of Regeneration and Repair, University of Edinburgh, 5 Little France Dr, Edinburgh BioQuarter, Edinburgh, EH16 4UU, UK
- Translational Healthcare Technologies Group, Centre for Inflammation Research, Institute of Regeneration and Repair, University of Edinburgh, 5 Little France Dr, Edinburgh BioQuarter, Edinburgh, EH16 4UU, UK
| | - Ahsan R Akram
- Centre for Inflammation Research, Institute of Regeneration and Repair, University of Edinburgh, 5 Little France Dr, Edinburgh BioQuarter, Edinburgh, EH16 4UU, UK.
- Translational Healthcare Technologies Group, Centre for Inflammation Research, Institute of Regeneration and Repair, University of Edinburgh, 5 Little France Dr, Edinburgh BioQuarter, Edinburgh, EH16 4UU, UK.
- Cancer Research UK Scotland Centre, Institute of Genetics & Cancer, The University of Edinburgh, Crewe Road South, Edinburgh, EH4 2XR, UK.
| |
Collapse
|
40
|
Desai B, Miti T, Prabhakaran S, Miroshnychenko D, Henry M, Marusyk V, Gatenbee C, Bui M, Scott J, Altrock PM, Haura E, Anderson ARA, Basanta D, Marusyk A. Peristromal niches protect lung cancers from targeted therapies through a combined effect of multiple molecular mediators. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.24.590626. [PMID: 38712093 PMCID: PMC11071426 DOI: 10.1101/2024.04.24.590626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Targeted therapies directed against oncogenic signaling addictions, such as inhibitors of ALK in ALK+ NSCLC often induce strong and durable clinical responses. However, they are not curative in metastatic cancers, as some tumor cells persist through therapy, eventually developing resistance. Therapy sensitivity can reflect not only cell-intrinsic mechanisms but also inputs from stromal microenvironment. Yet, the contribution of tumor stroma to therapeutic responses in vivo remains poorly defined. To address this gap of knowledge, we assessed the contribution of stroma-mediated resistance to therapeutic responses to the frontline ALK inhibitor alectinib in xenograft models of ALK+ NSCLC. We found that stroma-proximal tumor cells are partially protected against cytostatic effects of alectinib. This effect is observed not only in remission, but also during relapse, indicating the strong contribution of stroma-mediated resistance to both persistence and resistance. This therapy-protective effect of the stromal niche reflects a combined action of multiple mechanisms, including growth factors and extracellular matrix components. Consequently, despite improving alectinib responses, suppression of any individual resistance mechanism was insufficient to fully overcome the protective effect of stroma. Focusing on shared collateral sensitivity of persisters offered a superior therapeutic benefit, especially when using an antibody-drug conjugate with bystander effect to limit therapeutic escape. These findings indicate that stroma-mediated resistance might be the major contributor to both residual and progressing disease and highlight the limitation of focusing on suppressing a single resistance mechanism at a time.
Collapse
|
41
|
Li X, Li W, Wang J, Wang Q, Liang M, Chen S, Ba W, Fang J. Establishment of a novel microfluidic co-culture system for simultaneous analysis of multiple indicators of gefitinib sensitivity in colorectal cancer cells. Mikrochim Acta 2024; 191:279. [PMID: 38647729 DOI: 10.1007/s00604-024-06362-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 04/08/2024] [Indexed: 04/25/2024]
Abstract
The therapeutic effect of gefitinib on colorectal cancer (CRC) is unclear, but it has been reported that stromal cells in the tumor microenvironment may have an impact on drug sensitivity. Herein, we established a microfluidic co-culture system and explored the sensitivity of CRC cells co-cultured with cancer-associated fibroblasts (CAFs) to gefitinib. The system consisted of a multichannel chip and a Petri dish. The chambers in the chip and dish were designed to continuously supply nutrients for long-term cell survival and create chemokine gradients for driving cell invasion without any external equipment. Using this system, the proliferation and invasiveness of cells were simultaneously evaluated by quantifying the area of cells and the migration distance of cells. In addition, the system combined with live cell workstation could evaluate the dynamic drug response of co-cultured cells and track individual cell trajectories in real-time. When CRC cells were co-cultured with CAFs, CAFs promoted CRC cell proliferation and invasion and reduced the sensitivity of cells to gefitinib through the exosomes secreted by CAFs. Furthermore, the cells that migrated out of the chip were collected, and EMT-related markers were determined by immunofluorescent and western blot assays. The results demonstrated that CAFs affected the response of CRC cells to gefitinib by inducing EMT, providing new ideas for further research on the resistance mechanism of gefitinib. This suggests that targeting CAFs or exosomes might be a new approach to enhance CRC sensitivity to gefitinib, and our system could be a novel platform for investigating the crosstalk between tumor cells and CAFs and understanding multiple biological changes of the tumor cells in the tumor microenvironment.
Collapse
Affiliation(s)
- Xin Li
- Department of Cell Biology, Key Laboratory of Cell Biology, Key Laboratory of Medical Cell Biology, Ministry of Education, Ministry of Public Health, China Medical University, Shenyang, 110122, PR China
| | - Wanming Li
- Department of Cell Biology, Key Laboratory of Cell Biology, Key Laboratory of Medical Cell Biology, Ministry of Education, Ministry of Public Health, China Medical University, Shenyang, 110122, PR China
| | - Jie Wang
- Department of Cell Biology, Key Laboratory of Cell Biology, Key Laboratory of Medical Cell Biology, Ministry of Education, Ministry of Public Health, China Medical University, Shenyang, 110122, PR China
| | - Qun Wang
- Department of Cell Biology, Key Laboratory of Cell Biology, Key Laboratory of Medical Cell Biology, Ministry of Education, Ministry of Public Health, China Medical University, Shenyang, 110122, PR China
| | - Menghu Liang
- Department of Cell Biology, Key Laboratory of Cell Biology, Key Laboratory of Medical Cell Biology, Ministry of Education, Ministry of Public Health, China Medical University, Shenyang, 110122, PR China
| | - Shuo Chen
- Department of Cell Biology, Key Laboratory of Cell Biology, Key Laboratory of Medical Cell Biology, Ministry of Education, Ministry of Public Health, China Medical University, Shenyang, 110122, PR China
| | - Wei Ba
- Department of Cell Biology, Key Laboratory of Cell Biology, Key Laboratory of Medical Cell Biology, Ministry of Education, Ministry of Public Health, China Medical University, Shenyang, 110122, PR China
| | - Jin Fang
- Department of Cell Biology, Key Laboratory of Cell Biology, Key Laboratory of Medical Cell Biology, Ministry of Education, Ministry of Public Health, China Medical University, Shenyang, 110122, PR China.
| |
Collapse
|
42
|
Liu R, Zhao Y, Su S, Kwabil A, Njoku PC, Yu H, Li X. Unveiling cancer dormancy: Intrinsic mechanisms and extrinsic forces. Cancer Lett 2024; 591:216899. [PMID: 38649107 DOI: 10.1016/j.canlet.2024.216899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 04/06/2024] [Accepted: 04/12/2024] [Indexed: 04/25/2024]
Abstract
Tumor cells disseminate in various distant organs at early stages of cancer progression. These disseminated tumor cells (DTCs) can stay dormant/quiescent without causing patient symptoms for years or decades. These dormant tumor cells survive despite curative treatments by entering growth arrest, escaping immune surveillance, and/or developing drug resistance. However, these dormant cells can reactivate to proliferate, causing metastatic progression and/or relapse, posing a threat to patients' survival. It's unclear how cancer cells maintain dormancy and what triggers their reactivation. What are better approaches to prevent metastatic progression and relapse through harnessing cancer dormancy? To answer these remaining questions, we reviewed the studies of tumor dormancy and reactivation in various types of cancer using different model systems, including the brief history of dormancy studies, the intrinsic characteristics of dormant cells, and the external cues at the cellular and molecular levels. Furthermore, we discussed future directions in the field and the strategies for manipulating dormancy to prevent metastatic progression and recurrence.
Collapse
Affiliation(s)
- Ruihua Liu
- School of Life Sciences, Inner Mongolia University, Hohhot, Inner Mongolia Autonomous Region, 010070, China; Department of Cell and Cancer Biology, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH, 43614, USA
| | - Yawei Zhao
- Department of Cell and Cancer Biology, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH, 43614, USA
| | - Shang Su
- Department of Cell and Cancer Biology, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH, 43614, USA
| | - Augustine Kwabil
- Department of Cell and Cancer Biology, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH, 43614, USA
| | - Prisca Chinonso Njoku
- Department of Cell and Cancer Biology, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH, 43614, USA
| | - Haiquan Yu
- School of Life Sciences, Inner Mongolia University, Hohhot, Inner Mongolia Autonomous Region, 010070, China.
| | - Xiaohong Li
- Department of Cell and Cancer Biology, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH, 43614, USA.
| |
Collapse
|
43
|
Zhang Y, Hu Q, Pei Y, Luo H, Wang Z, Xu X, Zhang Q, Dai J, Wang Q, Fan Z, Fang Y, Ye M, Li B, Chen M, Xue Q, Zheng Q, Zhang S, Huang M, Zhang T, Gu J, Xiong Z. A patient-specific lung cancer assembloid model with heterogeneous tumor microenvironments. Nat Commun 2024; 15:3382. [PMID: 38643164 PMCID: PMC11032376 DOI: 10.1038/s41467-024-47737-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 04/08/2024] [Indexed: 04/22/2024] Open
Abstract
Cancer models play critical roles in basic cancer research and precision medicine. However, current in vitro cancer models are limited by their inability to mimic the three-dimensional architecture and heterogeneous tumor microenvironments (TME) of in vivo tumors. Here, we develop an innovative patient-specific lung cancer assembloid (LCA) model by using droplet microfluidic technology based on a microinjection strategy. This method enables precise manipulation of clinical microsamples and rapid generation of LCAs with good intra-batch consistency in size and cell composition by evenly encapsulating patient tumor-derived TME cells and lung cancer organoids inside microgels. LCAs recapitulate the inter- and intratumoral heterogeneity, TME cellular diversity, and genomic and transcriptomic landscape of their parental tumors. LCA model could reconstruct the functional heterogeneity of cancer-associated fibroblasts and reflect the influence of TME on drug responses compared to cancer organoids. Notably, LCAs accurately replicate the clinical outcomes of patients, suggesting the potential of the LCA model to predict personalized treatments. Collectively, our studies provide a valuable method for precisely fabricating cancer assembloids and a promising LCA model for cancer research and personalized medicine.
Collapse
Affiliation(s)
- Yanmei Zhang
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, China
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing, 100084, China
- Biomanufacturing and Engineering Living Systems Innovation International Talents Base (111 Base), Beijing, 100084, China
- Institute of New Materials and Advanced Manufacturing, Beijing Academy of Science and Technology, Beijing, 100089, China
| | - Qifan Hu
- MOE Key Laboratory of Bioinformatics, BNRIST Bioinformatics Division, Department of Automation, Tsinghua University, Beijing, 100084, China
| | - Yuquan Pei
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Thoracic Surgery II, Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Hao Luo
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, China
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing, 100084, China
- Biomanufacturing and Engineering Living Systems Innovation International Talents Base (111 Base), Beijing, 100084, China
| | - Zixuan Wang
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, China
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing, 100084, China
- Biomanufacturing and Engineering Living Systems Innovation International Talents Base (111 Base), Beijing, 100084, China
| | - Xinxin Xu
- Medical School of Chinese PLA, Beijing, 100853, China
| | - Qing Zhang
- Institute of New Materials and Advanced Manufacturing, Beijing Academy of Science and Technology, Beijing, 100089, China
| | - Jianli Dai
- Institute of New Materials and Advanced Manufacturing, Beijing Academy of Science and Technology, Beijing, 100089, China
| | - Qianqian Wang
- Institute of New Materials and Advanced Manufacturing, Beijing Academy of Science and Technology, Beijing, 100089, China
| | - Zilian Fan
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, China
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing, 100084, China
- Biomanufacturing and Engineering Living Systems Innovation International Talents Base (111 Base), Beijing, 100084, China
| | - Yongcong Fang
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, China
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing, 100084, China
- Biomanufacturing and Engineering Living Systems Innovation International Talents Base (111 Base), Beijing, 100084, China
| | - Min Ye
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, China
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing, 100084, China
- Biomanufacturing and Engineering Living Systems Innovation International Talents Base (111 Base), Beijing, 100084, China
| | - Binhan Li
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, China
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing, 100084, China
- Biomanufacturing and Engineering Living Systems Innovation International Talents Base (111 Base), Beijing, 100084, China
| | - Mailin Chen
- Department of Radiology, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Qi Xue
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Qingfeng Zheng
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Shulin Zhang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Miao Huang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Thoracic Surgery II, Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Ting Zhang
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, China
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing, 100084, China
- Biomanufacturing and Engineering Living Systems Innovation International Talents Base (111 Base), Beijing, 100084, China
| | - Jin Gu
- MOE Key Laboratory of Bioinformatics, BNRIST Bioinformatics Division, Department of Automation, Tsinghua University, Beijing, 100084, China.
| | - Zhuo Xiong
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, China.
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing, 100084, China.
- Biomanufacturing and Engineering Living Systems Innovation International Talents Base (111 Base), Beijing, 100084, China.
| |
Collapse
|
44
|
Petroni G, Pillozzi S, Antonuzzo L. Exploiting Tertiary Lymphoid Structures to Stimulate Antitumor Immunity and Improve Immunotherapy Efficacy. Cancer Res 2024; 84:1199-1209. [PMID: 38381540 PMCID: PMC11016894 DOI: 10.1158/0008-5472.can-23-3325] [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: 10/23/2023] [Revised: 01/04/2024] [Accepted: 02/19/2024] [Indexed: 02/23/2024]
Abstract
Tumor-associated tertiary lymphoid structures (TLS) have been associated with favorable clinical outcomes and response to immune checkpoint inhibitors in many cancer types, including non-small cell lung cancer. Although the detailed cellular and molecular mechanisms underlying these clinical associations have not been fully elucidated, growing preclinical and clinical studies are helping to elucidate the mechanisms at the basis of TLS formation, composition, and regulation of immune responses. However, a major challenge remains how to exploit TLS to enhance naïve and treatment-mediated antitumor immune responses. Here, we discuss the current understanding of tumor-associated TLS, preclinical models that can be used to study them, and potential therapeutic interventions to boost TLS formation, with a particular focus on lung cancer research.
Collapse
Affiliation(s)
- Giulia Petroni
- Department of Experimental and Clinical Medicine, University of Florence, Firenze, Italy
| | - Serena Pillozzi
- Department of Experimental and Clinical Biomedical Sciences 'Mario Serio', University of Florence, Firenze, Italy
| | - Lorenzo Antonuzzo
- Department of Experimental and Clinical Medicine, University of Florence, Firenze, Italy
- Clinical Oncology Unit, Careggi University Hospital, Firenze, Italy
| |
Collapse
|
45
|
Papavassiliou KA, Adamopoulos C, Papavassiliou AG. Many faces, many places: delving deeper into CAF heterogeneity in NSCLC. Trends Cancer 2024; 10:277-279. [PMID: 38395732 DOI: 10.1016/j.trecan.2024.02.004] [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: 02/11/2024] [Accepted: 02/14/2024] [Indexed: 02/25/2024]
Abstract
In a recent study published in Cancer Cell, Cords et al. employed multiplexed imaging mass cytometry to analyze cancer-associated fibroblast (CAF) heterogeneity in 1070 NSCLC patients. This work defined good and poor prognostic CAF phenotypes, the latter associated with metastasis and chemoresistance, as well as revealed that CAF spatial location correlates with immune cell infiltration and clinical outcome.
Collapse
Affiliation(s)
- Kostas A Papavassiliou
- First University Department of Respiratory Medicine, 'Sotiria' Hospital, Medical School, National and Kapodistrian University of Athens, Athens 11527, Greece
| | - Christos Adamopoulos
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Athens 11527, Greece; Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Athanasios G Papavassiliou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Athens 11527, Greece.
| |
Collapse
|
46
|
Croizer H, Mhaidly R, Kieffer Y, Gentric G, Djerroudi L, Leclere R, Pelon F, Robley C, Bohec M, Meng A, Meseure D, Romano E, Baulande S, Peltier A, Vincent-Salomon A, Mechta-Grigoriou F. Deciphering the spatial landscape and plasticity of immunosuppressive fibroblasts in breast cancer. Nat Commun 2024; 15:2806. [PMID: 38561380 PMCID: PMC10984943 DOI: 10.1038/s41467-024-47068-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 03/19/2024] [Indexed: 04/04/2024] Open
Abstract
Although heterogeneity of FAP+ Cancer-Associated Fibroblasts (CAF) has been described in breast cancer, their plasticity and spatial distribution remain poorly understood. Here, we analyze trajectory inference, deconvolute spatial transcriptomics at single-cell level and perform functional assays to generate a high-resolution integrated map of breast cancer (BC), with a focus on inflammatory and myofibroblastic (iCAF/myCAF) FAP+ CAF clusters. We identify 10 spatially-organized FAP+ CAF-related cellular niches, called EcoCellTypes, which are differentially localized within tumors. Consistent with their spatial organization, cancer cells drive the transition of detoxification-associated iCAF (Detox-iCAF) towards immunosuppressive extracellular matrix (ECM)-producing myCAF (ECM-myCAF) via a DPP4- and YAP-dependent mechanism. In turn, ECM-myCAF polarize TREM2+ macrophages, regulatory NK and T cells to induce immunosuppressive EcoCellTypes, while Detox-iCAF are associated with FOLR2+ macrophages in an immuno-protective EcoCellType. FAP+ CAF subpopulations accumulate differently according to the invasive BC status and predict invasive recurrence of ductal carcinoma in situ (DCIS), which could help in identifying low-risk DCIS patients eligible for therapeutic de-escalation.
Collapse
Affiliation(s)
- Hugo Croizer
- Institut Curie, Stress and Cancer Laboratory, Equipe Labélisée par la Ligue Nationale Contre le Cancer, PSL Research University, 26, Rue d'Ulm, F-75248, Paris, France
- Inserm, U830, 26, Rue d'Ulm, F-75005, Paris, France
| | - Rana Mhaidly
- Institut Curie, Stress and Cancer Laboratory, Equipe Labélisée par la Ligue Nationale Contre le Cancer, PSL Research University, 26, Rue d'Ulm, F-75248, Paris, France
- Inserm, U830, 26, Rue d'Ulm, F-75005, Paris, France
| | - Yann Kieffer
- Institut Curie, Stress and Cancer Laboratory, Equipe Labélisée par la Ligue Nationale Contre le Cancer, PSL Research University, 26, Rue d'Ulm, F-75248, Paris, France
- Inserm, U830, 26, Rue d'Ulm, F-75005, Paris, France
| | - Geraldine Gentric
- Institut Curie, Stress and Cancer Laboratory, Equipe Labélisée par la Ligue Nationale Contre le Cancer, PSL Research University, 26, Rue d'Ulm, F-75248, Paris, France
- Inserm, U830, 26, Rue d'Ulm, F-75005, Paris, France
| | - Lounes Djerroudi
- Institut Curie, Stress and Cancer Laboratory, Equipe Labélisée par la Ligue Nationale Contre le Cancer, PSL Research University, 26, Rue d'Ulm, F-75248, Paris, France
- Inserm, U830, 26, Rue d'Ulm, F-75005, Paris, France
- Department of Diagnostic and Theragnostic Medicine, Institut Curie Hospital Group, 26, Rue d'Ulm, F-75248, Paris, France
| | - Renaud Leclere
- Department of Diagnostic and Theragnostic Medicine, Institut Curie Hospital Group, 26, Rue d'Ulm, F-75248, Paris, France
| | - Floriane Pelon
- Institut Curie, Stress and Cancer Laboratory, Equipe Labélisée par la Ligue Nationale Contre le Cancer, PSL Research University, 26, Rue d'Ulm, F-75248, Paris, France
- Inserm, U830, 26, Rue d'Ulm, F-75005, Paris, France
| | - Catherine Robley
- Institut Curie, Stress and Cancer Laboratory, Equipe Labélisée par la Ligue Nationale Contre le Cancer, PSL Research University, 26, Rue d'Ulm, F-75248, Paris, France
- Inserm, U830, 26, Rue d'Ulm, F-75005, Paris, France
| | - Mylene Bohec
- Institut Curie, PSL Research University, ICGex Next-Generation Sequencing Platform, 75005, Paris, France
- Institut Curie, PSL Research University, Single Cell Initiative, 75005, Paris, France
| | - Arnaud Meng
- Institut Curie, Stress and Cancer Laboratory, Equipe Labélisée par la Ligue Nationale Contre le Cancer, PSL Research University, 26, Rue d'Ulm, F-75248, Paris, France
- Inserm, U830, 26, Rue d'Ulm, F-75005, Paris, France
| | - Didier Meseure
- Department of Diagnostic and Theragnostic Medicine, Institut Curie Hospital Group, 26, Rue d'Ulm, F-75248, Paris, France
| | - Emanuela Romano
- Department of Medical Oncology, Center for Cancer Immunotherapy, Institut Curie, 26, Rue d'Ulm, F-75248, Paris, France
| | - Sylvain Baulande
- Institut Curie, PSL Research University, ICGex Next-Generation Sequencing Platform, 75005, Paris, France
- Institut Curie, PSL Research University, Single Cell Initiative, 75005, Paris, France
| | - Agathe Peltier
- Institut Curie, Stress and Cancer Laboratory, Equipe Labélisée par la Ligue Nationale Contre le Cancer, PSL Research University, 26, Rue d'Ulm, F-75248, Paris, France
- Inserm, U830, 26, Rue d'Ulm, F-75005, Paris, France
| | - Anne Vincent-Salomon
- Department of Diagnostic and Theragnostic Medicine, Institut Curie Hospital Group, 26, Rue d'Ulm, F-75248, Paris, France
| | - Fatima Mechta-Grigoriou
- Institut Curie, Stress and Cancer Laboratory, Equipe Labélisée par la Ligue Nationale Contre le Cancer, PSL Research University, 26, Rue d'Ulm, F-75248, Paris, France.
- Inserm, U830, 26, Rue d'Ulm, F-75005, Paris, France.
| |
Collapse
|
47
|
Imodoye SO, Adedokun KA, Bello IO. From complexity to clarity: unravelling tumor heterogeneity through the lens of tumor microenvironment for innovative cancer therapy. Histochem Cell Biol 2024; 161:299-323. [PMID: 38189822 DOI: 10.1007/s00418-023-02258-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/06/2023] [Indexed: 01/09/2024]
Abstract
Despite the tremendous clinical successes recorded in the landscape of cancer therapy, tumor heterogeneity remains a formidable challenge to successful cancer treatment. In recent years, the emergence of high-throughput technologies has advanced our understanding of the variables influencing tumor heterogeneity beyond intrinsic tumor characteristics. Emerging knowledge shows that drivers of tumor heterogeneity are not only intrinsic to cancer cells but can also emanate from their microenvironment, which significantly favors tumor progression and impairs therapeutic response. Although much has been explored to understand the fundamentals of the influence of innate tumor factors on cancer diversity, the roles of the tumor microenvironment (TME) are often undervalued. It is therefore imperative that a clear understanding of the interactions between the TME and other tumor intrinsic factors underlying the plastic molecular behaviors of cancers be identified to develop patient-specific treatment strategies. This review highlights the roles of the TME as an emerging factor in tumor heterogeneity. More particularly, we discuss the role of the TME in the context of tumor heterogeneity and explore the cutting-edge diagnostic and therapeutic approaches that could be used to resolve this recurring clinical conundrum. We conclude by speculating on exciting research questions that can advance our understanding of tumor heterogeneity with the goal of developing customized therapeutic solutions.
Collapse
Affiliation(s)
- Sikiru O Imodoye
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA.
| | - Kamoru A Adedokun
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Ibrahim O Bello
- Department of Oral Medicine and Diagnostic Sciences, College of Dentistry, King Saud University, Riyadh, Saudi Arabia.
- Department of Pathology, University of Helsinki, Haartmaninkatu 3, 00014, Helsinki, Finland.
| |
Collapse
|
48
|
Zhao R, Ding Y, Han R, Ding R, Liu J, Zhu C, Ding D, Bao M. Prognostic correlation between specialized capillary endothelial cells and lung adenocarcinoma. Heliyon 2024; 10:e28236. [PMID: 38533005 PMCID: PMC10963648 DOI: 10.1016/j.heliyon.2024.e28236] [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: 09/30/2023] [Revised: 03/12/2024] [Accepted: 03/14/2024] [Indexed: 03/28/2024] Open
Abstract
Background In-depth analysis of the functional changes occurring in endothelial cells (ECs) involved in capillary formation can help to elucidate the mechanism of tumour vascular growth. Methods Appropriate datasets were retrieved from the GEO database to obtain single-cell data on LUAD samples and adjacent normal tissue samples. ECs were selected by an automatic annotation program in R and further subdivided based on reported EC marker genes. Functional changes in different types of capillary ECs were then visualized, and the concrete expression was classified by genetic data in the TCGA. Finally, a prognostic model was constructed to predict immunoinfiltration status, survival and drug therapy effects. Results The LUAD data contained in the GSE183219 dataset were suitable for our analysis. After dimensionality reduction analysis and cell annotation, EC general capillary and EC aerocyte subsets as capillary specialized phenotypes showed a series of functional changes in tumour samples, with a total of 108 genes found to undergo functional changes. Use of CellPhoneDB revealed a close interaction of activity between ECs. After integration of TCGA, GSE68465 and GSE11969 datasets, the genes obtained were analysed by cluster analysis and risk model construction, identifying 8 genes. Drug sensitivity, immune cell and molecular differences can be accurately predicted. Conclusions EC general capillary and EC aerocyte subsets are recognized capillary ECs in the tumour microenvironment, and the functional changes between them are relevant to the prognosis and treatment of LUAD patients and have the potential to be used in target therapy.
Collapse
Affiliation(s)
- Rongchang Zhao
- Department of Oncology, Taixing People's Hospital, Taixing, China
| | - Yan Ding
- Department of Oncology, Taixing People's Hospital, Taixing, China
| | - Rongbo Han
- Department of Oncology, The Fourth Affiliated Hospital Of Nanjing Medical University, Nanjing, China
| | - Rongjie Ding
- Department of Oncology, Taixing People's Hospital, Taixing, China
| | - Jun Liu
- Department of Oncology, Taixing People's Hospital, Taixing, China
| | - Chunrong Zhu
- Department of Oncology, First Affiliated Hospital of Soochow University, Suzhou, China
| | - Dan Ding
- Department of Oncology, Taixing People's Hospital, Taixing, China
| | - Minhui Bao
- Department of Oncology, Taixing People's Hospital, Taixing, China
| |
Collapse
|
49
|
Luan Q, Pulido I, Isagirre A, Carretero J, Zhou J, Shimamura T, Papautsky I. Deciphering fibroblast-induced drug resistance in non-small cell lung carcinoma through patient-derived organoids in agarose microwells. LAB ON A CHIP 2024; 24:2025-2038. [PMID: 38410967 PMCID: PMC11209828 DOI: 10.1039/d3lc01044a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/28/2024]
Abstract
Patient-derived organoids (PDOs) serve as invaluable 3D tumor models, retaining the histological complexity and genetic heterogeneity found in primary tumors. However, the limitation of small sample volumes and the lack of tailored platforms have hindered the research using PDOs. Within the tumor microenvironment, cancer-associated fibroblasts play a pivotal role in influencing drug sensitivity. In this study, we introduce an agarose microwell platform designed for PDO-based tumor and tumor microenvironment models, enabling rapid drug screening and resistance studies with small sample volumes. These microwells, constructed using 3D printing molds, feature a U-shaped bottom and 200 μm diameter. We successfully generated co-culture spheroids of non-small cell lung carcinoma (NSCLC) cells, including NCI-H358 or A549, and NSCLC PDOs F231 or F671 with fibroblast cell line, WI-38. Our results demonstrate the production of uniformly-sized spheroids (coefficient of variation <30%), high viability (>80% after 1 week), and fibroblast-induced drug resistance. The PDOs maintained their viability (>81% after 2 weeks) and continued to proliferate. Notably, when exposed to adagrasib, a KRASG12C inhibitor, we observed reduced cytotoxicity in KRASG12C-mutant spheroids when co-cultured with fibroblasts or their supernatant. The fibroblast supernatant sustained proliferative signals in tumor models. Taking into account the physical features, viability, and drug resistance acquired through supernatants from the fibroblasts, our platform emerges as a suitable platform for in vitro tumor modeling and the evaluation of drug efficacy using patient-derived tissues.
Collapse
Affiliation(s)
- Qiyue Luan
- Department of Biomedical Engineering, University of Illinois Chicago, Chicago, 851 S. Morgan Street, 218 SEO, Chicago, IL 60607, USA.
| | - Ines Pulido
- Department of Surgery, University of Illinois Chicago, Chicago, IL 60612, USA
- University of Illinois Cancer Center, Chicago, IL 60612, USA
| | - Angelique Isagirre
- Department of Biomedical Engineering, University of Illinois Chicago, Chicago, 851 S. Morgan Street, 218 SEO, Chicago, IL 60607, USA.
| | - Julian Carretero
- Departament de Fisiologia, Facultat de Farmacia, Universitat de Valencia, Burjassot, 46010, Spain
| | - Jian Zhou
- Department of Biomedical Engineering, University of Illinois Chicago, Chicago, 851 S. Morgan Street, 218 SEO, Chicago, IL 60607, USA.
- University of Illinois Cancer Center, Chicago, IL 60612, USA
| | - Takeshi Shimamura
- Department of Surgery, University of Illinois Chicago, Chicago, IL 60612, USA
- University of Illinois Cancer Center, Chicago, IL 60612, USA
| | - Ian Papautsky
- Department of Biomedical Engineering, University of Illinois Chicago, Chicago, 851 S. Morgan Street, 218 SEO, Chicago, IL 60607, USA.
- University of Illinois Cancer Center, Chicago, IL 60612, USA
| |
Collapse
|
50
|
Zhou D, Zheng L. Recent advances in cancer-associated fibroblast: Biomarkers, signaling pathways, and therapeutic opportunities. Chin Med J (Engl) 2024; 137:638-650. [PMID: 38420743 PMCID: PMC10950138 DOI: 10.1097/cm9.0000000000003031] [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: 11/27/2023] [Indexed: 03/02/2024] Open
Abstract
ABSTRACT Anti-cancer therapies usually focus on tumor cells, but non-tumor stromal components in the tumor microenvironment also play vital roles in tumor initiation and progression, which may be the prognostic factors and potential therapeutic targets. Cancer-associated fibroblasts (CAFs) are the essential component in the tumor environment, exhibiting high heterogeneity in their cell origin and phenotype with diverse functions that influence tumor angiogenesis, immune systems, and metabolism. Single-cell RNA sequencing and genetically engineered mouse models have increased our understanding of CAF diversity, and many subtypes have been defined. However, the precise functions of these subtypes need to be studied and validated. Studies of signaling pathways and epigenetic changes in CAFs facilitate understanding of the phenotypes of CAFs and the crosstalk between tumor cells and CAFs to provide potential therapeutic targets. Some clinical trials, including phase III trials targeting CAFs, have been performed recently. However, few of these trials have generated promising results, which indicates that the complexity of CAFs in the tumor microenvironment remains largely unknown, and in-depth investigations of CAFs should be performed. This review summarizes the research on CAFs, focusing on the heterogeneity of their phenotypes and functions, specific signaling pathways, and the therapeutic strategies involving CAFs. Additionally, we briefly discuss the current technologies commonly used in CAF studies and describe the challenges and future perspectives of CAF research.
Collapse
Affiliation(s)
- Donger Zhou
- Department of Hepatobiliary-Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Lei Zheng
- Sidney Kimmel Comprehensive Cancer Center, The Skip Viragh Pancreatic Cancer Center for Clinical Research and Care, and The Bloomberg-Kimmel Institute for Immunotherapy at Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| |
Collapse
|