1
|
Li RF, Liu S, Gao Q, Fu M, Sun XY, Xiao M, Ge XY, Peng X. Inhibition of CDH11 Activates cGAS-STING by Stimulating Branched Chain Amino Acid Catabolism and Mitigates Lung Metastasis of Adenoid Cystic Carcinoma. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024:e2408751. [PMID: 39739317 DOI: 10.1002/advs.202408751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Revised: 10/10/2024] [Indexed: 01/02/2025]
Abstract
Salivary adenoid cystic carcinoma (SACC) is an intractable malignant tumor originates in the secretory glands and frequently metastasizes to the lungs. Hybrid epithelial-mesenchymal transition (EMT) cells within the tumors are correlated with augmented proliferative capacity and facilitation of lung metastasis. Single-cell RNA sequencing and spatial transcriptomic sequencing are employed to reveal the hybrid EMT subsets within the vascular fibroblast microenvironment. These hybrid EMT cells exhibit a pro-tumorigenic impact in vitro. Notably, cadherin 11 (CDH11), a specific marker for hybrid EMT cells, may exert its regulatory role in cellular function by interfering with branched-chain amino acids (BCAA) metabolism by inhibiting branched-chain ketoacid dehydrogenase to activate the mammalian target of the rapamycin pathway, thus making it a potential therapeutic target for SACC. Furthermore, celecoxib and its derivatives are specific CDH11 inhibitors that regulate BCAA metabolism, increase reactive oxygen species production, and subsequently activate the cyclic GMP-AMP synthase-stimulator of the interferongene pathway (cGAS-STING). They also inhibit lung metastasis in NOD-SCID mice in vivo. Overall, these findings suggest a promising treatment strategy that targets hybrid EMT cells to mitigate lung metastasis in SACC. Celecoxib may serve as a promising clinical intervention for the treatment of lung metastases in patients with SACC.
Collapse
Affiliation(s)
- Rui-Feng Li
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
- Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
- National Center for Stomatology, Beijing, 100081, P. R. China
- National Clinical Research Center for Oral Diseases, Beijing, 100081, P. R. China
- National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, BeiJing, 100081, P. R. China
| | - Shuo Liu
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
- Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
- National Center for Stomatology, Beijing, 100081, P. R. China
- National Clinical Research Center for Oral Diseases, Beijing, 100081, P. R. China
- National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, BeiJing, 100081, P. R. China
| | - Qian Gao
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
- Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
- National Center for Stomatology, Beijing, 100081, P. R. China
- National Clinical Research Center for Oral Diseases, Beijing, 100081, P. R. China
- National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, BeiJing, 100081, P. R. China
| | - Min Fu
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
- Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
- National Center for Stomatology, Beijing, 100081, P. R. China
- National Clinical Research Center for Oral Diseases, Beijing, 100081, P. R. China
- National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, BeiJing, 100081, P. R. China
| | - Xin-Yi Sun
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
- Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
- National Center for Stomatology, Beijing, 100081, P. R. China
- National Clinical Research Center for Oral Diseases, Beijing, 100081, P. R. China
- National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, BeiJing, 100081, P. R. China
| | - Mian Xiao
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
- Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
- National Center for Stomatology, Beijing, 100081, P. R. China
- National Clinical Research Center for Oral Diseases, Beijing, 100081, P. R. China
- National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, BeiJing, 100081, P. R. China
| | - Xi-Yuan Ge
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
- Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
- National Center for Stomatology, Beijing, 100081, P. R. China
- National Clinical Research Center for Oral Diseases, Beijing, 100081, P. R. China
- National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, BeiJing, 100081, P. R. China
| | - Xin Peng
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
- Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
- National Center for Stomatology, Beijing, 100081, P. R. China
- National Clinical Research Center for Oral Diseases, Beijing, 100081, P. R. China
- National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, BeiJing, 100081, P. R. China
| |
Collapse
|
2
|
Chen Y, Pan G, Yang Y, Wu H, Weng M, Wu Q, Gao Y, Li W. Tumor exosomal RNPEP promotes lung metastasis of liver cancer via inducing cancer-associated fibroblast activation. Cancer Sci 2024. [PMID: 39658708 DOI: 10.1111/cas.16417] [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: 05/20/2024] [Revised: 11/08/2024] [Accepted: 11/15/2024] [Indexed: 12/12/2024] Open
Abstract
Cancer-associated fibroblasts (CAFs) are essential players in the tumor microenvironment (TME) due to their roles in facilitating tumor progression and metastasis. It is worth noting that the high-metastatic hepatocellular carcinoma (HCC) cell-derived exosomes have exhibited the ability to transform normal fibroblasts into CAFs, which further fosters the lung metastasis of low-metastatic HCC cells. Yet, the mechanisms underlying this tumor exosome-induced metastatic niche formation are poorly explored. In this study, the secreted protein arginyl aminopeptidase (RNPEP) was highly expressed in the plasma of patients with HCC. In addition, high-metastatic HCC cells showed augmented RNPEP expression levels in their exosomes. These exosomes induced obvious CAF-like properties in the human fibroblast cell line MRC-5, as evidenced by the increased CAF marker expression, and enhanced migratory ability. More strikingly, the secretions from high-metastatic tumor exosome-educated MRC-5 cells increased tumor stemness and promoted epithelial-mesenchymal transition (EMT) in MHCC-97L cells, a low-metastatic HCC cell line. However, the knockdown of RNPEP in exosomes from high-metastatic HCC cells abated the changes described above. Animal studies in vivo highlighted the pro-tumor and pro-metastatic effects of exosomal RNPEP on MHCC-97L cells by inducing CAF activation. Furthermore, tumor-derived exosomal RNPEP induced the activation of NF-κB signaling in MRC-5 cells, a critical pathway associated with CAF activation. Collectively, these results provide novel insight into tumor-derived exosomal RNPEP for its crosstalk with CAFs during HCC lung metastasis.
Collapse
Affiliation(s)
- Yuankun Chen
- Department of Tropical and Liver Diseases, The Second Affiliated Hospital of Hainan Medical University, Haikou, China
- Department of Clinical Laboratory, The Second Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Gaofeng Pan
- Department of Infectious Diseases, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Department of Infectious Diseases, The First People's Hospital of Kunshan, Kunshan, China
| | - Yijun Yang
- Department of Tropical and Liver Diseases, The Second Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Haifeng Wu
- Department of Tropical and Liver Diseases, The Second Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Minhua Weng
- Department of Tropical and Liver Diseases, The Second Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Qiuping Wu
- Department of Tropical and Liver Diseases, The Second Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Yufeng Gao
- Department of Infectious Diseases, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Wenting Li
- Department of Infectious Diseases, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
3
|
Yang C, Shu J, Li Y, Zhao N, Liu X, Tian X, Sun Z, Tabish MS, Hong Y, Chen K, Sun M. Long non-coding RNAs are involved in the crosstalk between cancer-associated fibroblasts and tumor cells. Front Immunol 2024; 15:1469918. [PMID: 39717771 PMCID: PMC11663902 DOI: 10.3389/fimmu.2024.1469918] [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: 07/24/2024] [Accepted: 11/22/2024] [Indexed: 12/25/2024] Open
Abstract
The proliferation of tumors is not merely self-regulated by the cancer cells but is also intrinsically connected to the tumor microenvironment (TME). Within this complex TME, cancer-associated fibroblasts (CAFs) are pivotal in the modulation of tumor onset and progression. Rich signaling interactions exist between CAFs and tumor cells, which are crucial for tumor regulation. Long non-coding RNAs (LncRNAs) emerge from cellular transcription as a class of functionally diverse RNA molecules. Recent studies have revealed that LncRNAs are integral to the crosstalk between CAFs and tumor cells, with the capacity to modify cellular transcriptional activity and secretion profiles, thus facilitating CAFs activation, tumor proliferation, metastasis, drug resistance, and other related functionalities. This comprehensive review revisits the latest research on LncRNA-mediated interactions between CAFs and tumor cells, encapsulates the biological roles of LncRNAs, and delves into the molecular pathways from a broader perspective, aspiring to offer novel perspectives for a deeper comprehension of the etiology of tumors and the enhancement of therapeutic approaches.
Collapse
Affiliation(s)
- Chenbo Yang
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Tumor Pathology, Zhengzhou University, Zhengzhou, China
| | - Jiao Shu
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Tumor Pathology, Zhengzhou University, Zhengzhou, China
| | - Yiwei Li
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Tumor Pathology, Zhengzhou University, Zhengzhou, China
- Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Na Zhao
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Tumor Pathology, Zhengzhou University, Zhengzhou, China
| | - Xiaonan Liu
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Tumor Pathology, Zhengzhou University, Zhengzhou, China
| | - Xiangyu Tian
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Tumor Pathology, Zhengzhou University, Zhengzhou, China
| | - Zexin Sun
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Tumor Pathology, Zhengzhou University, Zhengzhou, China
- Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Muhammad Saud Tabish
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Tumor Pathology, Zhengzhou University, Zhengzhou, China
| | - Yichen Hong
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Tumor Pathology, Zhengzhou University, Zhengzhou, China
- Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Kuisheng Chen
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Tumor Pathology, Zhengzhou University, Zhengzhou, China
| | - Miaomiao Sun
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Tumor Pathology, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
4
|
Shen P, Ma Z, Xu X, Li W, Li Y. Dental pulp stem cells promote malignant transformation of oral epithelial cells through mitochondrial transfer. Med Mol Morphol 2024; 57:306-319. [PMID: 39122902 DOI: 10.1007/s00795-024-00403-1] [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: 05/05/2024] [Accepted: 08/01/2024] [Indexed: 08/12/2024]
Abstract
Oral epithelial dysplasia includes a range of clinical oral mucosal diseases with potentially malignant traits. Dental pulp stem cells (DPSCs) are potential candidates for cell-based therapies targeting various diseases. However, the effect of DPSCs on the progression of oral mucosal precancerous lesions remains unclear. Animal experiments were conducted to assess the effect of human DPSCs (hDPSCs). We measured the proliferation, motility and mitochondrial respiratory function of the human dysplastic oral keratinocyte (DOK) cells cocultured with hDPSCs. Mitochondrial transfer experiments were performed to determine the role mitochondria from hDPSCs in the malignant transformation of DOK cells. hDPSCs injection accelerated carcinogenesis in 4NQO-induced oral epithelial dysplasia in mice. Coculture with hDPSCs increased the proliferation, migration, invasion and mitochondrial respiratory function of DOK cells. Mitochondria from hDPSCs could be transferred to DOK cells, and activated mTOR signaling pathway in DOK cells. Our study demonstrates that hDPSCs activate the mTOR signaling pathway through mitochondrial transfer, promoting the malignant transformation of oral precancerous epithelial lesions.
Collapse
Affiliation(s)
- Peiqi Shen
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, Guangdong, 510055, People's Republic of China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, Guangdong, China
| | - Zeyi Ma
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, Guangdong, 510055, People's Republic of China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, Guangdong, China
| | - Xiaoqing Xu
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, Guangdong, 510055, People's Republic of China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, Guangdong, China
| | - Weiyu Li
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, Guangdong, 510055, People's Republic of China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, Guangdong, China
| | - Yaoyin Li
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, Guangdong, 510055, People's Republic of China.
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, Guangdong, China.
| |
Collapse
|
5
|
De Oliveira Macena Y, Cezar MEN, Lira CBF, De Oliveira LBDM, Almeida TN, Costa ADAV, De Araujo BMD, de Almeida Junior D, Dantas HM, De Mélo EC, de Araújo ST, de França Júnior RR, Dos Santos Lemos Gurgel MA, de Carvalho Fraga CA. The roles of periostin derived from cancer-associated fibroblasts in tumor progression and treatment response. Cancer Metastasis Rev 2024; 44:11. [PMID: 39614015 DOI: 10.1007/s10555-024-10233-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 11/21/2024] [Indexed: 12/01/2024]
Abstract
Periostin (POSTN), a matricellular protein predominantly secreted by cancer-associated fibroblasts (CAFs), has emerged as a key regulator of cancer progression and therapy response. This review provides an overview of recent findings regarding the diverse roles of periostin in cancer therapy and its potential as a therapeutic target. Studies have elucidated periostin's involvement in tumorigenesis, including tumor growth, metastasis, chemotherapy resistance, and modulation of the tumor microenvironment (TME). CAFs periostin + play a central role in shaping the TME by remodeling the extracellular matrix (ECM) and promoting immune evasion, thus promoting tumor cell survival and dissemination. Elevated periostin expression has been correlated with poor prognosis across multiple cancer types, suggesting its utility as a prognostic biomarker. Periostin has been implicated in mediating resistance to chemotherapy, with CAFs periostin + establishing a pro-tumorigenic niche that confers protection to cancer cells against cytotoxic therapies. Targeting periostin or its downstream effectors presents a promising strategy to overcome therapy resistance and enhance treatment efficacy. While significant progress has been made in understanding the biological functions of periostin in cancer, gaps persist in elucidating its precise mechanisms of action and clinical relevance. Future research should focus on deciphering the signaling pathways and molecular interactions underlying periostin-mediated effects in the TME. Prospective clinical studies are warranted to evaluate periostin as a predictive biomarker and therapeutic target in cancer patients.
Collapse
Affiliation(s)
- Yasmin De Oliveira Macena
- Universidade Federal de Alagoas, Campus Arapiraca, Centro de Ciências Médicas, Av. Manoel Severino Barbosa, Bom Sucesso, Arapiraca, AL, CEP 57309-005, Brazil
| | - Maria Eduarda Nunes Cezar
- Universidade Federal de Alagoas, Campus Arapiraca, Centro de Ciências Médicas, Av. Manoel Severino Barbosa, Bom Sucesso, Arapiraca, AL, CEP 57309-005, Brazil
| | - Cinthya Brunelly Ferreira Lira
- Universidade Federal de Alagoas, Campus Arapiraca, Centro de Ciências Médicas, Av. Manoel Severino Barbosa, Bom Sucesso, Arapiraca, AL, CEP 57309-005, Brazil
| | | | - Thais Noronha Almeida
- Universidade Federal de Alagoas, Campus Arapiraca, Centro de Ciências Médicas, Av. Manoel Severino Barbosa, Bom Sucesso, Arapiraca, AL, CEP 57309-005, Brazil
| | - Ana Dora Alecio Virtuoso Costa
- Universidade Federal de Alagoas, Campus Arapiraca, Centro de Ciências Médicas, Av. Manoel Severino Barbosa, Bom Sucesso, Arapiraca, AL, CEP 57309-005, Brazil
| | - Bernardo Mansur Dantas De Araujo
- Universidade Federal de Alagoas, Campus Arapiraca, Centro de Ciências Médicas, Av. Manoel Severino Barbosa, Bom Sucesso, Arapiraca, AL, CEP 57309-005, Brazil
| | - Durval de Almeida Junior
- Universidade Federal de Alagoas, Campus Arapiraca, Centro de Ciências Médicas, Av. Manoel Severino Barbosa, Bom Sucesso, Arapiraca, AL, CEP 57309-005, Brazil
| | - Henrique Macêdo Dantas
- Universidade Federal de Alagoas, Campus Arapiraca, Centro de Ciências Médicas, Av. Manoel Severino Barbosa, Bom Sucesso, Arapiraca, AL, CEP 57309-005, Brazil
| | - Emanuel Cézar De Mélo
- Universidade Federal de Alagoas, Campus Arapiraca, Centro de Ciências Médicas, Av. Manoel Severino Barbosa, Bom Sucesso, Arapiraca, AL, CEP 57309-005, Brazil
| | - Sandra Taveiros de Araújo
- Universidade Federal de Alagoas, Campus Arapiraca, Centro de Ciências Médicas, Av. Manoel Severino Barbosa, Bom Sucesso, Arapiraca, AL, CEP 57309-005, Brazil
| | - Raimundo Rodrigues de França Júnior
- Universidade Federal de Alagoas, Campus Arapiraca, Centro de Ciências Médicas, Av. Manoel Severino Barbosa, Bom Sucesso, Arapiraca, AL, CEP 57309-005, Brazil
| | - Maria Amélia Dos Santos Lemos Gurgel
- Universidade Federal de Alagoas, Campus Arapiraca, Centro de Ciências Médicas, Av. Manoel Severino Barbosa, Bom Sucesso, Arapiraca, AL, CEP 57309-005, Brazil
| | - Carlos Alberto de Carvalho Fraga
- Universidade Federal de Alagoas, Campus Arapiraca, Centro de Ciências Médicas, Av. Manoel Severino Barbosa, Bom Sucesso, Arapiraca, AL, CEP 57309-005, Brazil.
- CRID - Centro de Pesquisa em Doenças Inflamatórias, Av. Bandeirantes, R. Paineiras, 3900 - Casa 03 - Vila Monte Alegre, Ribeirão Preto, SP, 14049-900, Brazil.
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Ciências Farmacêuticas (FCF), Universidade de São Paulo (USP), São Paulo, SP, Brazil.
| |
Collapse
|
6
|
Li X, Li N, Wang Y, Han Q, Sun B. Research Progress of Fibroblasts in Human Diseases. Biomolecules 2024; 14:1478. [PMID: 39595654 PMCID: PMC11591654 DOI: 10.3390/biom14111478] [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: 09/19/2024] [Revised: 11/16/2024] [Accepted: 11/18/2024] [Indexed: 11/28/2024] Open
Abstract
Fibroblasts, which originate from embryonic mesenchymal cells, are the predominant cell type seen in loose connective tissue. As the main components of the internal environment that cells depend on for survival, fibroblasts play an essential role in tissue development, wound healing, and the maintenance of tissue homeostasis. Furthermore, fibroblasts are also involved in several pathological processes, such as fibrosis, cancers, and some inflammatory diseases. In this review, we analyze the latest research progress on fibroblasts, summarize the biological characteristics and physiological functions of fibroblasts, and delve into the role of fibroblasts in disease pathogenesis and explore treatment approaches for fibroblast-related diseases.
Collapse
Affiliation(s)
| | | | | | | | - Boshi Sun
- Department of Colorectal Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China; (X.L.); (N.L.); (Y.W.); (Q.H.)
| |
Collapse
|
7
|
Chen H, Xu X, Li J, Xue Y, Li X, Zhang K, Jiang H, Liu X, Li M. Decoding tumor-fibrosis interplay: mechanisms, impact on progression, and innovative therapeutic strategies. Front Pharmacol 2024; 15:1491400. [PMID: 39534084 PMCID: PMC11555290 DOI: 10.3389/fphar.2024.1491400] [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: 09/04/2024] [Accepted: 10/09/2024] [Indexed: 11/16/2024] Open
Abstract
Malignant tumors are a category of diseases that possess invasive and metastatic capabilities, with global incidence and mortality rates remaining high. In recent years, the pivotal role of fibrosis in tumor progression, drug resistance, and immune evasion has increasingly been acknowledged. Fibrosis enhances the proliferation, migration, and invasion of tumor cells by modifying the composition and structure of the extracellular matrix, thereby offering protection for immune evasion by tumor cells. The activation of cancer-associated fibroblasts (CAFs) plays a significant role in this process, as they further exacerbate the malignant traits of tumors by secreting a variety of cytokines and growth factors. Anti-fibrotic tumor treatment strategies, including the use of anti-fibrotic drugs and inhibition of fibrosis-related signaling pathways such as Transforming Growth Factor-β (TGF-β), have demonstrated potential in delaying tumor progression and improving the effectiveness of chemotherapy, targeted therapy, and immunotherapy. In the future, by developing novel drugs that target the fibrotic microenvironment, new therapeutic options may be available for patients with various refractory tumors.
Collapse
Affiliation(s)
- Huiguang Chen
- Institute of Infection, Immunology, and Tumor Microenvironment, School of Medicine, Wuhan University of Science and Technology, Wuhan, China
| | - Xuexin Xu
- Institute of Infection, Immunology, and Tumor Microenvironment, School of Medicine, Wuhan University of Science and Technology, Wuhan, China
| | - Jingxian Li
- Institute of Infection, Immunology, and Tumor Microenvironment, School of Medicine, Wuhan University of Science and Technology, Wuhan, China
| | - Yu Xue
- Institute of Infection, Immunology, and Tumor Microenvironment, School of Medicine, Wuhan University of Science and Technology, Wuhan, China
| | - Xin Li
- Institute of Infection, Immunology, and Tumor Microenvironment, School of Medicine, Wuhan University of Science and Technology, Wuhan, China
| | - Kaiyu Zhang
- Institute of Infection, Immunology, and Tumor Microenvironment, School of Medicine, Wuhan University of Science and Technology, Wuhan, China
| | - Haihui Jiang
- Institute of Infection, Immunology, and Tumor Microenvironment, School of Medicine, Wuhan University of Science and Technology, Wuhan, China
| | - Xiaoliu Liu
- Institute of Infection, Immunology, and Tumor Microenvironment, School of Medicine, Wuhan University of Science and Technology, Wuhan, China
- Department of Anatomy, School of Medicine, Wuhan University of Science and Technology, Wuhan, China
| | - Mingzhe Li
- Department of Anatomy, School of Medicine, Wuhan University of Science and Technology, Wuhan, China
| |
Collapse
|
8
|
Wang H, Chen J, Chen X, Liu Y, Wang J, Meng Q, Wang H, He Y, Song Y, Li J, Ju Z, Xiao P, Qian J, Song Z. Cancer-Associated Fibroblasts Expressing Sulfatase 1 Facilitate VEGFA-Dependent Microenvironmental Remodeling to Support Colorectal Cancer. Cancer Res 2024; 84:3371-3387. [PMID: 39250301 DOI: 10.1158/0008-5472.can-23-3987] [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: 12/15/2023] [Revised: 04/18/2024] [Accepted: 07/16/2024] [Indexed: 09/11/2024]
Abstract
Tumor stroma plays a critical role in fostering tumor progression and metastasis. Cancer-associated fibroblasts (CAF) are a major component of the tumor stroma. Identifying the key molecular determinants for the protumor properties of CAFs could enable the development of more effective treatment strategies. In this study, through analyses of single-cell sequencing data, we identified a population of CAFs expressing high levels of sulfatase 1 (SULF1), which was associated with poor prognosis in patients with colorectal cancer. Colorectal cancer models using mice with conditional SULF1 knockout in fibroblasts revealed the tumor-supportive function of SULF1+ CAFs. Mechanistically, SULF1+ CAFs enhanced the release of VEGFA from heparan sulfate proteoglycan. The increased bioavailability of VEGFA initiated the deposition of extracellular matrix and enhanced angiogenesis. In addition, intestinal microbiota-produced butyrate suppressed SULF1 expression in CAFs through its histone deacetylase (HDAC) inhibitory activity. The insufficient butyrate production in patients with colorectal cancer increased the abundance of SULF1+ CAFs, thereby promoting tumor progression. Importantly, tumor growth inhibition by HDAC was dependent on SULF1 expression in CAFs, and patients with colorectal cancer with more SULF1+ CAFs were more responsive to treatment with the HDAC inhibitor chidamide. Collectively, these findings unveil the critical role of SULF1+ CAFs in colorectal cancer and provide a strategy to stratify patients with colorectal cancer for HDAC inhibitor treatment. Significance: SULF1+ cancer-associated fibroblasts play a tumor-promoting role in colorectal cancer by stimulating extracellular matrix deposition and angiogenesis and can serve as a biomarker for the therapeutic response to HDAC inhibitors in patients.
Collapse
Affiliation(s)
- Huijuan Wang
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Biological Treatment of Zhejiang Province, Hangzhou, China
- Key Laboratory of Integrated Traditional Chinese and Western Medicine Research on Anorectal Diseases of Zhejiang Province, Hangzhou, China
| | - Jiaxin Chen
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Biological Treatment of Zhejiang Province, Hangzhou, China
- Key Laboratory of Integrated Traditional Chinese and Western Medicine Research on Anorectal Diseases of Zhejiang Province, Hangzhou, China
| | - Xiaoyu Chen
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Biological Treatment of Zhejiang Province, Hangzhou, China
- Key Laboratory of Integrated Traditional Chinese and Western Medicine Research on Anorectal Diseases of Zhejiang Province, Hangzhou, China
| | - Yingqiang Liu
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Biological Treatment of Zhejiang Province, Hangzhou, China
- Key Laboratory of Integrated Traditional Chinese and Western Medicine Research on Anorectal Diseases of Zhejiang Province, Hangzhou, China
| | - Jiawei Wang
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Biological Treatment of Zhejiang Province, Hangzhou, China
- Key Laboratory of Integrated Traditional Chinese and Western Medicine Research on Anorectal Diseases of Zhejiang Province, Hangzhou, China
| | - Qing Meng
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Biological Treatment of Zhejiang Province, Hangzhou, China
- Key Laboratory of Integrated Traditional Chinese and Western Medicine Research on Anorectal Diseases of Zhejiang Province, Hangzhou, China
| | - Huogang Wang
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Biological Treatment of Zhejiang Province, Hangzhou, China
- Key Laboratory of Integrated Traditional Chinese and Western Medicine Research on Anorectal Diseases of Zhejiang Province, Hangzhou, China
| | - Ying He
- Huzhou Key Laboratory of Translational Medicine, Huzhou, China
| | - Yujia Song
- Hangzhou No. 14 High School, Hangzhou, China
| | - Jingyun Li
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Biological Treatment of Zhejiang Province, Hangzhou, China
- Key Laboratory of Integrated Traditional Chinese and Western Medicine Research on Anorectal Diseases of Zhejiang Province, Hangzhou, China
| | - Zhenyu Ju
- Key Laboratory of Regenerative Medicine of Ministry of Education, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Institute of Ageing and Regenerative Medicine, Jinan University, Guangzhou, China
| | - Peng Xiao
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Junbin Qian
- Zhejiang Key Laboratory of Precision Diagnosis and Therapy for Major Gynecological Diseases, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for Child Health, Hangzhou, China
| | - Zhangfa Song
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Biological Treatment of Zhejiang Province, Hangzhou, China
- Key Laboratory of Integrated Traditional Chinese and Western Medicine Research on Anorectal Diseases of Zhejiang Province, Hangzhou, China
| |
Collapse
|
9
|
Zhang T, Zheng B, Xia C, Wu P, Zheng B, Jiang L, Li J, Lv G, Zhou H, Huang W, Zou M. Hypoxic Upregulation of IER2 Increases Paracrine GMFG Signaling of Endoplasmic Reticulum Stress-CAF to Promote Chordoma Progression via Targeting ITGB1. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2405421. [PMID: 39207055 PMCID: PMC11515918 DOI: 10.1002/advs.202405421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 07/27/2024] [Indexed: 09/04/2024]
Abstract
Currently, the oncogenic mechanism of endoplasmic reticulum stress-CAF (ERS-CAF) subpopulation in chordoma remains unknown. Here, single-cell RNA sequencing, spatial transcriptomics, GeoMx Digital Spatial Profiler, data-independent acquisition proteomics, bulk RNA-seq, and multiplexed quantitative immunofluorescence are used to unveil the precise molecular mechanism of how ERS-CAF affected chordoma progression. Results show that hypoxic microenvironment reprograms CAFs into ERS-CAF subtype. Mechanistically, this occurrs via hypoxia-mediated transcriptional upregulation of IER2. Overexpression of IER2 in CAFs promotes chordoma progression, which can be impeded by IER2 knockdown or use of ERS inhibitors. IER2 also induces expression of ERS-CAF marker genes and results in production of a pro-tumorigenic paracrine GMFG signaling, which exert its biological function via directly binding to ITGB1 on tumor cells. ITGB1 inhibition attenuates tumor malignant progression, which can be partially reversed by exogenous GMFG intervention. Further analyses reveal a positive correlation between ITGB1high tumor cell counts and SPP1+ macrophage density, as well as the spatial proximity of these two cell types. Clinically, a significant correlation of high IER2/ITGB1 expression with tumor aggressive phenotype and poor patient survival is observed. Collectively, the findings suggest that ERS-CAF regulates SPP1+ macrophage to aggravate chordoma progression via the IER2/GMFG/ITGB1 axis, which may be targeted therapeutically in future.
Collapse
Affiliation(s)
- Tao‐Lan Zhang
- Department of PharmacyThe First Affiliated HospitalHengyang Medical SchoolUniversity of South ChinaHengyang421001China
| | - Bo‐Wen Zheng
- Department of PharmacyThe First Affiliated HospitalHengyang Medical SchoolUniversity of South ChinaHengyang421001China
- Musculoskeletal Tumor CenterPeking University People's HospitalPeking UniversityBeijing100044China
| | - Chao Xia
- Department of Spine SurgeryThe First Affiliated HospitalHengyang Medical SchoolUniversity of South ChinaHengyang421001China
| | - Peng‐Fei Wu
- Department of Genetics and EndocrinologyNational Children's Medical Center for South Central RegionGuangzhou Women and Children's Medical CenterGuangzhou Medical UniversityGuangzhouGuangdong510623China
| | - Bo‐Yv Zheng
- Department of Orthopedics SurgeryGeneral Hospital of the Central Theater CommandWuhan430061China
| | - Ling‐Xiang Jiang
- Department of Radiation OncologyMelvin and Bren Simon Comprehensive Cancer CenterIndiana University School of MedicineIndianapolisIN46202USA
| | - Jing Li
- Department of Spine SurgeryThe Second Xiangya HospitalCentral South UniversityChangsha410011China
| | - Guo‐Hua Lv
- Department of Spine SurgeryThe Second Xiangya HospitalCentral South UniversityChangsha410011China
| | - Hong Zhou
- Department of RadiologyThe First Affiliated HospitalHengyang Medical SchoolUniversity of South ChinaHengyang421001China
| | - Wei Huang
- The First Affiliated HospitalHealth Management CenterHengyang Medical SchoolUniversity of South ChinaHengyang421001China
| | - Ming‐Xiang Zou
- Department of Spine SurgeryThe First Affiliated HospitalHengyang Medical SchoolUniversity of South ChinaHengyang421001China
| |
Collapse
|
10
|
Li X, Chen T, Li X, Zhang H, Li Y, Zhang S, Luo S, Zheng T. Therapeutic targets of armored chimeric antigen receptor T cells navigating the tumor microenvironment. Exp Hematol Oncol 2024; 13:96. [PMID: 39350256 PMCID: PMC11440706 DOI: 10.1186/s40164-024-00564-w] [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: 05/07/2024] [Accepted: 09/17/2024] [Indexed: 10/04/2024] Open
Abstract
Chimeric antigen receptor (CAR) T cell therapy, which targets tumors with high specificity through the recognition of particular antigens, has emerged as one of the most rapidly advancing modalities in immunotherapy, demonstrating substantial success against hematological malignancies. However, previous generations of CAR-T cell therapy encountered numerous challenges in treating solid tumors, such as the lack of suitable targets, high immunosuppression, suboptimal persistence, and insufficient infiltration owing to the complexities of the tumor microenvironment, all of which limited their efficacy. In this review, we focus on the current therapeutic targets of fourth-generation CAR-T cells, also known as armored CAR-T cells, and explore the mechanisms by which these engineered cells navigate the tumor microenvironment by targeting its various components. Enhancing CAR-T cells with these therapeutic targets holds promise for improving their effectiveness against solid tumors, thus achieving substantial clinical value and advancing the field of CAR-T cell therapy. Additionally, we discuss potential strategies to overcome existing challenges and highlight novel targets that could further enhance the efficacy of CAR-T cell therapy in treating solid tumors.
Collapse
Affiliation(s)
- Xianjun Li
- Harbin Medical University Cancer Hospital, Harbin, 150081, China
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, 150081, China
- Heilongjiang Province Key Laboratory of Molecular Oncology, Harbin,150081, China
| | - Tianjun Chen
- Harbin Medical University Cancer Hospital, Harbin, 150081, China
- Heilongjiang Province Key Laboratory of Molecular Oncology, Harbin,150081, China
| | - Xuehan Li
- Harbin Medical University Cancer Hospital, Harbin, 150081, China
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, 150081, China
- Heilongjiang Province Key Laboratory of Molecular Oncology, Harbin,150081, China
| | - Hanyu Zhang
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Yingjing Li
- Harbin Medical University Cancer Hospital, Harbin, 150081, China
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, 150081, China
- Heilongjiang Province Key Laboratory of Molecular Oncology, Harbin,150081, China
| | - Shuyuan Zhang
- Harbin Medical University Cancer Hospital, Harbin, 150081, China
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, 150081, China
- Heilongjiang Province Key Laboratory of Molecular Oncology, Harbin,150081, China
| | - Shengnan Luo
- Harbin Medical University Cancer Hospital, Harbin, 150081, China
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, 150081, China
- Heilongjiang Province Key Laboratory of Molecular Oncology, Harbin,150081, China
| | - Tongsen Zheng
- Harbin Medical University Cancer Hospital, Harbin, 150081, China.
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, 150081, China.
- Department of Phase 1 Trials Center, Harbin Medical University Cancer Hospital, Harbin, 150081, China.
- Heilongjiang Province Key Laboratory of Molecular Oncology, Harbin,150081, China.
| |
Collapse
|
11
|
Zhang F, Ma Y, Li D, Wei J, Chen K, Zhang E, Liu G, Chu X, Liu X, Liu W, Tian X, Yang Y. Cancer associated fibroblasts and metabolic reprogramming: unraveling the intricate crosstalk in tumor evolution. J Hematol Oncol 2024; 17:80. [PMID: 39223656 PMCID: PMC11367794 DOI: 10.1186/s13045-024-01600-2] [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: 06/25/2024] [Accepted: 08/20/2024] [Indexed: 09/04/2024] Open
Abstract
Metabolic reprogramming provides tumors with an energy source and biofuel to support their survival in the malignant microenvironment. Extensive research into the intrinsic oncogenic mechanisms of the tumor microenvironment (TME) has established that cancer-associated fibroblast (CAFs) and metabolic reprogramming regulates tumor progression through numerous biological activities, including tumor immunosuppression, chronic inflammation, and ecological niche remodeling. Specifically, immunosuppressive TME formation is promoted and mediators released via CAFs and multiple immune cells that collectively support chronic inflammation, thereby inducing pre-metastatic ecological niche formation, and ultimately driving a vicious cycle of tumor proliferation and metastasis. This review comprehensively explores the process of CAFs and metabolic regulation of the dynamic evolution of tumor-adapted TME, with particular focus on the mechanisms by which CAFs promote the formation of an immunosuppressive microenvironment and support metastasis. Existing findings confirm that multiple components of the TME act cooperatively to accelerate the progression of tumor events. The potential applications and challenges of targeted therapies based on CAFs in the clinical setting are further discussed in the context of advancing research related to CAFs.
Collapse
Affiliation(s)
- Fusheng Zhang
- Department of Hepatobiliary and Pancreatic Surgery, Peking University First Hospital, Beijing, 100034, China
| | - Yongsu Ma
- Department of Hepatobiliary and Pancreatic Surgery, Peking University First Hospital, Beijing, 100034, China
| | - Dongqi Li
- Department of Hepatobiliary and Pancreatic Surgery, Peking University First Hospital, Beijing, 100034, China
| | - Jianlei Wei
- Key laboratory of Microecology-immune Regulatory Network and Related Diseases School of Basic Medicine, Jiamusi University, Jiamusi, Heilongjiang Province, 154007, China
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Key Laboratory of Carcinogenesis and Translational Research, Peking University Health Science Center, Beijing, 100191, China
| | - Kai Chen
- Department of Hepatobiliary and Pancreatic Surgery, Peking University First Hospital, Beijing, 100034, China
| | - Enkui Zhang
- Department of Hepatobiliary and Pancreatic Surgery, Peking University First Hospital, Beijing, 100034, China
| | - Guangnian Liu
- Department of Hepatobiliary and Pancreatic Surgery, Peking University First Hospital, Beijing, 100034, China
| | - Xiangyu Chu
- Department of Hepatobiliary and Pancreatic Surgery, Peking University First Hospital, Beijing, 100034, China
| | - Xinxin Liu
- Department of Hepatobiliary and Pancreatic Surgery, Peking University First Hospital, Beijing, 100034, China
| | - Weikang Liu
- Department of Hepatobiliary and Pancreatic Surgery, Peking University First Hospital, Beijing, 100034, China
| | - Xiaodong Tian
- Department of Hepatobiliary and Pancreatic Surgery, Peking University First Hospital, Beijing, 100034, China.
| | - Yinmo Yang
- Department of Hepatobiliary and Pancreatic Surgery, Peking University First Hospital, Beijing, 100034, China.
| |
Collapse
|
12
|
Chen J, Sun JJ, Ma YW, Zhu MQ, Hu J, Lu QJ, Cai ZG. Cancer-associated fibroblasts derived exosomal LINC01833 promotes the occurrence of non-small cell lung cancer through miR-335-5p -VAPA axis. J Biochem Mol Toxicol 2024; 38:e23769. [PMID: 39152098 DOI: 10.1002/jbt.23769] [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: 01/22/2024] [Revised: 03/25/2024] [Accepted: 07/05/2024] [Indexed: 08/19/2024]
Abstract
Cancer-associated fibroblasts (CAFs) are an important component of the tumor microenvironment (TME) and can induce functional polarization of tumor macrophages. This study aimed to explore the effect of CAFs-derived exosome LINC01833 on the malignant biological behavior of non-small cell lung cancer (NSCLC) cells and its mechanism. Tumor tissues (n = 3) and adjacent noncancerous tissues (n = 3) were collected from patients with NSCLC, and fibroblasts (CAF, NF) were isolated from the two tissues. Expression of LINC01833/miR-335-5p/VAPA in NSCLC clinical tissues and cell lines was detected by RT-qPCR. Exosomes of CAFs and NFs were isolated by ultracentrifugation. Cell proliferation, migration, invasion, and M2 macrophage polarization were detected by MTT, transwell, wound-healing assay, and flow cytometry assay, while western blot was used to verify the expression of M2 macrophage polarization-related proteins. Tumor volume weight and M2 macrophage polarization were detected by tumor xenografts in nude mice. LINC01833 was highly expressed in NSCLC tumor tissues and cells. Knockdown of LINC01833 exosomes could significantly inhibit proliferation, migration, invasion of NSCLC cells, and M2 macrophage polarization of THP-1 cells, while simultaneous knockdown of miR-335-5p on the above basis could reverse the effect of knockdown of LINC01833. In vivo experiments also indicated that knockdown of LINC01833 exosomes suppressed tumor growth and M2 macrophage polarization. CAF-derived LINC01833 exosomes can promote the proliferation, migration and invasion of NSCLC cells and M2 macrophage polarization by inhibiting miR-335-5p and regulating VAPA activity.
Collapse
Affiliation(s)
- Jie Chen
- Naval Medical Center of PLA, Thoracic and Cardiac Surgery, Naval Medical University, Shanghai, China
| | - Jian-Jun Sun
- Naval Medical Center of PLA, Thoracic and Cardiac Surgery, Naval Medical University, Shanghai, China
| | - Ya-Wen Ma
- Department of Cardiology, Naval Medical Center of PLA, Naval Medical University, Shanghai, China
| | - Meng-Qin Zhu
- Naval Medical Center of PLA, Thoracic and Cardiac Surgery, Naval Medical University, Shanghai, China
| | - Jing Hu
- Naval Medical Center of PLA, Thoracic and Cardiac Surgery, Naval Medical University, Shanghai, China
| | - Qi-Jue Lu
- Naval Medical Center of PLA, Thoracic and Cardiac Surgery, Naval Medical University, Shanghai, China
| | - Zhi-Gang Cai
- Naval Medical Center of PLA, Thoracic and Cardiac Surgery, Naval Medical University, Shanghai, China
| |
Collapse
|
13
|
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
|
14
|
Hu Z, Guo X, Li Z, Meng Z, Huang S. The neoantigens derived from transposable elements - A hidden treasure for cancer immunotherapy. Biochim Biophys Acta Rev Cancer 2024; 1879:189126. [PMID: 38849060 DOI: 10.1016/j.bbcan.2024.189126] [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: 01/16/2024] [Revised: 05/26/2024] [Accepted: 06/02/2024] [Indexed: 06/09/2024]
Abstract
Neoantigen-based therapy is a promising approach that selectively activates the immune system of the host to recognize and eradicate cancer cells. Preliminary clinical trials have validated the feasibility, safety, and immunogenicity of personalized neoantigen-directed vaccines, enhancing their effectiveness and broad applicability in immunotherapy. While many ongoing oncological trials concentrate on neoantigens derived from mutations, these targets do not consistently provoke an immune response in all patients harboring the mutations. Additionally, tumors like ovarian cancer, which have a low tumor mutational burden (TMB), may be less amenable to mutation-based neoantigen therapies. Recent advancements in next-generation sequencing and bioinformatics have uncovered a rich source of neoantigens from non-canonical RNAs associated with transposable elements (TEs). Considering the substantial presence of TEs in the human genome and the proven immunogenicity of TE-derived neoantigens in various tumor types, this review investigates the latest findings on TE-derived neoantigens, examining their clinical implications, challenges, and unique advantages in enhancing tumor immunotherapy.
Collapse
Affiliation(s)
- Zhixiang Hu
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, and Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xinyi Guo
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, and Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ziteng Li
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, and Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhiqiang Meng
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, and Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
| | - Shenglin Huang
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, and Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
15
|
Xie Y, Ma J, Tang W, Zhang Y, Zhang C, Chen Y. Efficacy and Safety Evaluation of 177Lu-FAP-2286 in the Treatment of Advanced Lung Cancer. Clin Nucl Med 2024; 49:830-837. [PMID: 39102810 DOI: 10.1097/rlu.0000000000005297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/07/2024]
Abstract
PURPOSE The aim of this study was to evaluate the efficacy and safety of peptide-targeted radionuclide therapy (PTRT) with 177Lu-FAP-2286 in advanced lung cancer. PATIENTS AND METHODS This single-center prospective study included 9 patients diagnosed with advanced lung cancer. These patients met the inclusion criteria and received PTRT with 177Lu-FAP-2286. Short-term efficacy was assessed using RECIST 1.1 and PERCIST 1.0 criteria. Long-term efficacy was evaluated through overall survival, progression-free survival (PFS), overall response rate, EORTC QLQ-C30 v3.0, Eastern Cooperative Oncology Group, and Karnofsky Performance Status. Toxicity response was assessed using CTCAE v5.0. RESULTS The results based on RECIST 1.1 and PERCIST 1.0 criteria were comparable, with 44% of patients showing a partial metabolic response, 33.3% with stable metabolic disease, and 22.22% with progressive metabolic disease. The highest metabolic response after treatment reached 66.89%, and the overall response rate could reach 77.78%. In the long-term efficacy assessment, the median overall survival and PFS were 10 months and 6 months, respectively. The 2 patients with the lowest PFS (3 months) started PTRT relatively late. EORTC QLQ-C30 v3.0, Eastern Cooperative Oncology Group, and Karnofsky Performance Status scores showed that the overall health status, symptom response, and quality of life of patients improved after 177Lu-FAP-2286 treatment. The most noticeable improvements in clinical symptoms were dyspnea and cancer-related pain. No grade III/IV toxicity events were observed during follow-up period, and fibrinogen decreased significantly after treatment. CONCLUSIONS 177Lu-FAP-2286 has the potential to be a viable PTRT option for patients with advanced lung cancer.
Collapse
|
16
|
Wang ZB, Zhang X, Fang C, Liu XT, Liao QJ, Wu N, Wang J. Immunotherapy and the ovarian cancer microenvironment: Exploring potential strategies for enhanced treatment efficacy. Immunology 2024; 173:14-32. [PMID: 38618976 DOI: 10.1111/imm.13793] [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: 09/15/2023] [Accepted: 04/05/2024] [Indexed: 04/16/2024] Open
Abstract
Despite progress in cancer immunotherapy, ovarian cancer (OC) prognosis continues to be disappointing. Recent studies have shed light on how not just tumour cells, but also the complex tumour microenvironment, contribute to this unfavourable outcome of OC immunotherapy. The complexities of the immune microenvironment categorize OC as a 'cold tumour'. Nonetheless, understanding the precise mechanisms through which the microenvironment influences the effectiveness of OC immunotherapy remains an ongoing scientific endeavour. This review primarily aims to dissect the inherent characteristics and behaviours of diverse cells within the immune microenvironment, along with an exploration into its reprogramming and metabolic changes. It is expected that these insights will elucidate the operational dynamics of the immune microenvironment in OC and lay a theoretical groundwork for improving the efficacy of immunotherapy in OC management.
Collapse
Affiliation(s)
- Zhi-Bin Wang
- Hunan Gynecological Tumor Clinical Research Center; Hunan Key Laboratory of Cancer Metabolism; Hunan Cancer Hospital, and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
- Public Service Platform of Tumor Organoids Technology, Changsha, China
| | - Xiu Zhang
- Hunan Gynecological Tumor Clinical Research Center; Hunan Key Laboratory of Cancer Metabolism; Hunan Cancer Hospital, and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
- Public Service Platform of Tumor Organoids Technology, Changsha, China
| | - Chao Fang
- Hunan Gynecological Tumor Clinical Research Center; Hunan Key Laboratory of Cancer Metabolism; Hunan Cancer Hospital, and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
- Hunan Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, Changsha Medical University, Changsha, China
| | - Xiao-Ting Liu
- The Second People's Hospital of Hunan Province, Changsha, China
| | - Qian-Jin Liao
- Hunan Gynecological Tumor Clinical Research Center; Hunan Key Laboratory of Cancer Metabolism; Hunan Cancer Hospital, and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
- Public Service Platform of Tumor Organoids Technology, Changsha, China
| | - Nayiyuan Wu
- Hunan Gynecological Tumor Clinical Research Center; Hunan Key Laboratory of Cancer Metabolism; Hunan Cancer Hospital, and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
- Public Service Platform of Tumor Organoids Technology, Changsha, China
| | - Jing Wang
- Hunan Gynecological Tumor Clinical Research Center; Hunan Key Laboratory of Cancer Metabolism; Hunan Cancer Hospital, and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
- Public Service Platform of Tumor Organoids Technology, Changsha, China
| |
Collapse
|
17
|
Zhou Z, Mai Y, Zhang G, Wang Y, Sun P, Jing Z, Li Z, Xu Y, Han B, Liu J. Emerging role of immunogenic cell death in cancer immunotherapy: Advancing next-generation CAR-T cell immunotherapy by combination. Cancer Lett 2024; 598:217079. [PMID: 38936505 DOI: 10.1016/j.canlet.2024.217079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 06/11/2024] [Accepted: 06/18/2024] [Indexed: 06/29/2024]
Abstract
Immunogenic cell death (ICD) is a stress-driven form of regulated cell death (RCD) in which dying tumor cells' specific signaling pathways are activated to release damage-associated molecular patterns (DAMPs), leading to the robust anti-tumor immune response as well as a reversal of the tumor immune microenvironment from "cold" to "hot". Chimeric antigen receptor (CAR)-T cell therapy, as a landmark in anti-tumor immunotherapy, plays a formidable role in hematologic malignancies but falls short in solid tumors. The Gordian knot of CAR-T cells for solid tumors includes but is not limited to, tumor antigen heterogeneity or absence, physical and immune barriers of tumors. The combination of ICD induction therapy and CAR-T cell immunotherapy is expected to promote the intensive use of CAR-T cell in solid tumors. In this review, we summarize the characteristics of ICD, stress-responsive mechanism, and the synergistic effect of various ICD-based therapies with CAR-T cells to effectively improve anti-tumor capacity.
Collapse
Affiliation(s)
- Zhaokai Zhou
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Yumiao Mai
- Department of Pediatrics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Ge Zhang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Henan Province Key Laboratory of Cardiac Injury and Repair, Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, Henan, 450052, China
| | - Yingjie Wang
- Department of Pediatrics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Pan Sun
- Department of Pediatrics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Zhaohe Jing
- Department of Pediatrics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Zhengrui Li
- Department of Oral and Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yudi Xu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Bo Han
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Jian Liu
- Department of Pediatrics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China.
| |
Collapse
|
18
|
Mu Y, Zhang Z, Zhou H, Ma L, Wang DA. Applications of nanotechnology in remodeling the tumour microenvironment for glioblastoma treatment. Biomater Sci 2024; 12:4045-4064. [PMID: 38993162 DOI: 10.1039/d4bm00665h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/13/2024]
Abstract
With the increasing research and deepening understanding of the glioblastoma (GBM) tumour microenvironment (TME), novel and more effective therapeutic strategies have been proposed. The GBM TME involves intricate interactions between tumour and non-tumour cells, promoting tumour progression. Key therapeutic goals for GBM treatment include improving the immunosuppressive microenvironment, enhancing the cytotoxicity of immune cells against tumours, and inhibiting tumour growth and proliferation. Consequently, remodeling the GBM TME using nanotechnology has emerged as a promising approach. Nanoparticle-based drug delivery enables targeted delivery, thereby improving treatment specificity, facilitating combination therapies, and optimizing drug metabolism. This review provides an overview of the GBM TME and discusses the methods of remodeling the GBM TME using nanotechnology. Specifically, it explores the application of nanotechnology in ameliorating immune cell immunosuppression, inducing immunogenic cell death, stimulating, and recruiting immune cells, regulating tumour metabolism, and modulating the crosstalk between tumours and other cells.
Collapse
Affiliation(s)
- Yulei Mu
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR 999077, China.
- Karolinska Institutet Ming Wai Lau Centre for Reparative Medicine, HKSTP, Sha Tin, Hong Kong SAR
| | - Zhen Zhang
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR 999077, China.
| | - Huiqun Zhou
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR 999077, China.
- Karolinska Institutet Ming Wai Lau Centre for Reparative Medicine, HKSTP, Sha Tin, Hong Kong SAR
| | - Liang Ma
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR 999077, China.
| | - Dong-An Wang
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR 999077, China.
- Karolinska Institutet Ming Wai Lau Centre for Reparative Medicine, HKSTP, Sha Tin, Hong Kong SAR
- Centre for Neuromusculoskeletal Restorative Medicine, InnoHK, HKSTP, Sha Tin, Hong Kong SAR 999077, China
| |
Collapse
|
19
|
Li Y, Shen X, Ding H, Zhang Y, Pan D, Su L, Wu Y, Fang Z, Zhou J, Gong Q, Luo K. Dendritic nanomedicine enhances chemo-immunotherapy by disturbing metabolism of cancer-associated fibroblasts for deep penetration and activating function of immune cells. Acta Pharm Sin B 2024; 14:3680-3696. [PMID: 39220877 PMCID: PMC11365400 DOI: 10.1016/j.apsb.2024.03.010] [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: 01/25/2024] [Revised: 02/21/2024] [Accepted: 02/28/2024] [Indexed: 09/04/2024] Open
Abstract
Inefficient drug penetration hurdled by the stroma in the tumor tissue leads to a diminished therapeutic effect for drugs and a reduced infiltration level of immune cells. Herein, we constructed a PEGylated dendritic epirubicin (Epi) prodrug (Epi-P4D) to regulate the metabolism of cancer-associated fibroblasts (CAFs), thus enhancing Epi penetration into both multicellular tumor spheroids (MTSs) and tumor tissues in mouse colon cancer (CT26), mouse breast cancer (4T1) and human breast cancer (MDA-MB-231) models. Enhanced cytotoxicity against CT26 MTSs and remarkable antitumor efficacy of Epi-P4D were ascribed to reduced fibronectin, α-SMA, and collagen secretion. Besides, thinning of the tumor tissue stroma and efficient eradication of tumor cells promoted the immunogenic cell death effect for dendritic cell (DC) maturation and subsequent immune activation, including elevating the CD4+ T cell population, reducing CD4+ and CD8+ T cell hyperactivation and exhaustion, and amplifying the natural killer (NK) cell proportion and effectively activating them. As a result, this dendritic nanomedicine thinned the stroma of tumor tissues to enhance drug penetration and facilitate immune cell infiltration for elevated antitumor efficacy.
Collapse
Affiliation(s)
- Yunkun Li
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xiaoding Shen
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Haitao Ding
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yuxin Zhang
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Dayi Pan
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Liping Su
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yahui Wu
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zaixiang Fang
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jie Zhou
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Qiyong Gong
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
- Functional and Molecular Imaging Key Laboratory of Sichuan Province, and Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu 610041, China
- Department of Radiology, West China Xiamen Hospital of Sichuan University, Xiamen 361021, China
| | - Kui Luo
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
- Functional and Molecular Imaging Key Laboratory of Sichuan Province, and Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu 610041, China
| |
Collapse
|
20
|
Jia N, Wang Q, Li W, Chen D, Hu H. Membrane Fusion Liposomes Deliver Antifibrotic and Chemotherapeutic Drugs Sequentially to Enhance Tumor Treatment Efficacy by Reshaping Tumor Microenvironment. Adv Healthc Mater 2024; 13:e2400219. [PMID: 38657266 DOI: 10.1002/adhm.202400219] [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: 01/19/2024] [Revised: 04/14/2024] [Indexed: 04/26/2024]
Abstract
The intricate tumor microenvironment in triple-negative breast cancer (TNBC) hampers chemotherapy and immunotherapy efficacy due to dense extracellular matrix (ECM) by tumor-associated fibroblasts (TAFs). Nanoparticle-based therapies, especially "all-in-one" nanoparticles, have shown great potential in combined drug delivery strategies to reshape the tumor microenvironment and enhance therapeutic efficiency. However, these "all-in-one" nanoparticles suffer from limitations in targeting different target cells, uncontrollable dosing ratio, and disregarding the impact of delivery schedules. This study prepared cell membrane fusion liposomes (TAFsomes and CCMsomes) to load FDA-approved antifibrotic drug pirfenidone (PFD/TAFsomes) and antitumor drug doxorubicin (DOX/CCMsomes). These liposomes can specifically target TAFs cells and tumor cells, and combined administration can effectively inhibit TAFs activity, reshape the tumor microenvironment (TME), and significantly enhance the tumor chemotherapy efficacy. Combined drug delivery defeats "all-in-one" liposomes (DOX/PFD/Liposomes, DOX/PFD/TAFsomes, and DOX/PFD/CCMsomes) by flexibly adjusting the drug delivery ratio. Moreover, an asynchronous delivery strategy that optimizes the administration schedule not only further improves the therapeutic effect, but also amplifies the effectiveness of α-PD-L1 immunotherapy by modulating the tumor immune microenvironment. This delivery strategy provides a personalized treatment approach with clinical translation potential, providing new ideas for enhancing the therapeutic effect against solid tumors such as TNBC.
Collapse
Affiliation(s)
- Nan Jia
- School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province, 110016, China
| | - Qi Wang
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, AL, 36849, USA
| | - Wenpan Li
- School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province, 110016, China
| | - Dawei Chen
- School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province, 110016, China
| | - Haiyang Hu
- School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province, 110016, China
| |
Collapse
|
21
|
Gong XT, Zhuang J, Chong KC, Xu Q, Ling X, Cao L, Wu M, Yang J, Liu B. Far-Red Aggregation-Induced Emission Hydrogel-Reinforced Tissue Clearing for 3D Vasculature Imaging of Whole Lung and Whole Tumor. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2402853. [PMID: 39003614 DOI: 10.1002/adma.202402853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Revised: 06/18/2024] [Indexed: 07/15/2024]
Abstract
Understanding the vascular formation and distribution in metastatic lung tumors is a significant challenge due to autofluorescence, antibody/dye diffusion in dense tumor, and fluorophore stability when exposed to solvent-based clearing agents. Here, an approach is presented that redefines 3D vasculature imaging within metastatic tumor, peritumoral lung tissue, and normal lung. Specifically, a far-red aggregation-induced emission nanoparticle with surface amino groups (termed as TSCN nanoparticle, TSCNNP) is designed for in situ formation of hydrogel (TSCNNP@Gel) inside vasculatures to provide structural support and enhance the fluorescence in solvent-based tissue clearing method. Using this TSCNNP@Gel-reinforced tissue clearing imaging approach, the critical challenges are successfully overcome and comprehensive visualization of the whole pulmonary vasculature up to 2 µm resolution is enabled, including its detailed examination in metastatic tumors. Importantly, features of tumor-associated vasculature in 3D panoramic views are unveiled, providing the potential to determine tumor stages, predict tumor progression, and facilitate the histopathological diagnosis of various tumor types.
Collapse
Affiliation(s)
- Xiao-Ting Gong
- Joint School of National University of Singapore and Tianjin University, International Campus of Tianjin University, Binhai New City, Fuzhou, 350207, China
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore, 117585, Singapore
| | - Jiahao Zhuang
- Joint School of National University of Singapore and Tianjin University, International Campus of Tianjin University, Binhai New City, Fuzhou, 350207, China
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore, 117585, Singapore
| | - Kok Chan Chong
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore, 117585, Singapore
| | - Qun Xu
- School of Life Sciences, Peking University, Beijing, 100871, China
| | - Xia Ling
- Joint School of National University of Singapore and Tianjin University, International Campus of Tianjin University, Binhai New City, Fuzhou, 350207, China
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore, 117585, Singapore
| | - Lei Cao
- Joint School of National University of Singapore and Tianjin University, International Campus of Tianjin University, Binhai New City, Fuzhou, 350207, China
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore, 117585, Singapore
| | - Min Wu
- Joint School of National University of Singapore and Tianjin University, International Campus of Tianjin University, Binhai New City, Fuzhou, 350207, China
| | - Jing Yang
- School of Life Sciences, Peking University, Beijing, 100871, China
| | - Bin Liu
- Joint School of National University of Singapore and Tianjin University, International Campus of Tianjin University, Binhai New City, Fuzhou, 350207, China
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore, 117585, Singapore
| |
Collapse
|
22
|
Yin J, Zhu W, Feng S, Yan P, Qin S. The role of cancer-associated fibroblasts in the invasion and metastasis of colorectal cancer. Front Cell Dev Biol 2024; 12:1375543. [PMID: 39139454 PMCID: PMC11319178 DOI: 10.3389/fcell.2024.1375543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 07/11/2024] [Indexed: 08/15/2024] Open
Abstract
Colorectal cancer (CRC) is the third most common cancer and has ranked the third leading cause in cancerassociated death globally. Metastasis is the leading cause of death in colorectal cancer patients. The role of tumor microenvironment (TME) in colorectal cancer metastasis has received increasing attention. As the most abundant cell type in the TME of solid tumors, cancer-associated fibroblasts (CAFs) have been demonstrated to have multiple functions in advancing tumor growth and metastasis. They can remodel the extracellular matrix (ECM) architecture, promote epithelial-mesenchymal transition (EMT), and interact with cancer cells or other stromal cells by secreting growth factors, cytokines, chemokines, and exosomes, facilitating tumor cell invasion into TME and contributing to distant metastasis. This article aims to analyze the sources and heterogeneity of CAFs in CRC, as well as their role in invasion and metastasis, in order to provide new insights into the metastasis mechanism of CRC and its clinical applications.
Collapse
Affiliation(s)
- Jinjin Yin
- Department of Pharmacy, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Wenting Zhu
- Department of Pharmacy, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Senling Feng
- Department of Pharmacy, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Pengke Yan
- Department of Pharmacy, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Shumin Qin
- Department of Gastroenterology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangzhou, China
| |
Collapse
|
23
|
Lyu P, Gu X, Wang F, Sun H, Zhou Q, Yang S, Yuan W. Advances in targeting cancer-associated fibroblasts through single-cell spatial transcriptomic sequencing. Biomark Res 2024; 12:73. [PMID: 39075612 PMCID: PMC11287900 DOI: 10.1186/s40364-024-00622-9] [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: 03/01/2024] [Accepted: 07/18/2024] [Indexed: 07/31/2024] Open
Abstract
Cancer-associated fibroblasts (CAFs) are the major components of the tumor microenvironment and are related to tumor proliferation, metastasis, relapse, and drug resistance. With the development of sequencing technologies, single-cell RNA sequencing has become a popular method for identifying CAFs in the tumor microenvironment. Whereas the drawbacks of CAFs, such as the lack of a spatial landscape, still exist, recent research has utilized spatial transcriptomics combined with single-cell RNA sequencing to address this issue. These multiomics analyses can resolve the single-cell resolution problem in spatial transcriptomics. In this review, we summarized the recent literature regarding the targeting of CAFs to address drug resistance, angiogenesis, metabolic reprogramming and metastasis in tumor tissue.
Collapse
Affiliation(s)
- Pin Lyu
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Erqi District, Zhengzhou, 450000, Henan, China
| | - Xiaoming Gu
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Erqi District, Zhengzhou, 450000, Henan, China
| | - Fuqi Wang
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Erqi District, Zhengzhou, 450000, Henan, China
| | - Haifeng Sun
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Erqi District, Zhengzhou, 450000, Henan, China
| | - Quanbo Zhou
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Erqi District, Zhengzhou, 450000, Henan, China
| | - Shuaixi Yang
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Erqi District, Zhengzhou, 450000, Henan, China
| | - Weitang Yuan
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Erqi District, Zhengzhou, 450000, Henan, China.
| |
Collapse
|
24
|
Meng X, Liu Z, Deng L, Yang Y, Zhu Y, Sun X, Hao Y, He Y, Fu J. Hydrogen Therapy Reverses Cancer-Associated Fibroblasts Phenotypes and Remodels Stromal Microenvironment to Stimulate Systematic Anti-Tumor Immunity. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2401269. [PMID: 38757665 PMCID: PMC11267370 DOI: 10.1002/advs.202401269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Revised: 04/02/2024] [Indexed: 05/18/2024]
Abstract
Tumor microenvironment (TME) plays an important role in the tumor progression. Among TME components, cancer-associated fibroblasts (CAFs) show multiple tumor-promoting effects and can induce tumor immune evasion and drug-resistance. Regulating CAFs can be a potential strategy to augment systemic anti-tumor immunity. Here, the study observes that hydrogen treatment can alleviate intracellular reactive oxygen species of CAFs and reshape CAFs' tumor-promoting and immune-suppressive phenotypes. Accordingly, a controllable and TME-responsive hydrogen therapy based on a CaCO3 nanoparticles-coated magnesium system (Mg-CaCO3) is developed. The hydrogen therapy by Mg-CaCO3 can not only directly kill tumor cells, but also inhibit pro-tumor and immune suppressive factors in CAFs, and thus augment immune activities of CD4+ T cells. As implanted in situ, Mg-CaCO3 can significantly suppress tumor growth, turn the "cold" primary tumor into "hot", and stimulate systematic anti-tumor immunity, which is confirmed by the bilateral tumor transplantation models of "cold tumor" (4T1 cells) and "hot tumor" (MC38 cells). This hydrogen therapy system reverses immune suppressive phenotypes of CAFs, thus providing a systematic anti-tumor immune stimulating strategy by remodeling tumor stromal microenvironment.
Collapse
Affiliation(s)
- Xiaoyan Meng
- Department of Oral Maxillofacial & Head and Neck OncologyShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200011P. R. China
- College of StomatologyNational Center for StomatologyNational Clinical Research Center for Oral DiseasesShanghai Key Laboratory of StomatologyShanghai Jiao Tong UniversityShanghai200011P. R. China
| | - Zhonglong Liu
- Department of Oral Maxillofacial & Head and Neck OncologyShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200011P. R. China
- College of StomatologyNational Center for StomatologyNational Clinical Research Center for Oral DiseasesShanghai Key Laboratory of StomatologyShanghai Jiao Tong UniversityShanghai200011P. R. China
| | - Liang Deng
- Shanghai Key Laboratory of Orthopaedic ImplantDepartment of Orthopaedic SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200011P. R. China
- Clinical and Translational Research Center for 3D Printing TechnologyShanghai Engineering Research Center of Innovative Orthopaedic Instruments and Personalized MedicineShanghai200011P. R. China
| | - Yangzi Yang
- Department of Orthopedic SurgerySpine CenterChangzheng HospitalNavy Medical UniversityNo. 415 Fengyang RoadShanghai200003P. R. China
| | - Yingchun Zhu
- Key Laboratory of Inorganic Coating MaterialsShanghai Institute of CeramicsChinese Academy of SciencesShanghai200050P. R. China
| | - Xiaoying Sun
- College of SciencesShanghai UniversityShanghai200444P. R. China
| | - Yongqiang Hao
- Shanghai Key Laboratory of Orthopaedic ImplantDepartment of Orthopaedic SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200011P. R. China
- Clinical and Translational Research Center for 3D Printing TechnologyShanghai Engineering Research Center of Innovative Orthopaedic Instruments and Personalized MedicineShanghai200011P. R. China
| | - Yue He
- Department of Oral Maxillofacial & Head and Neck OncologyShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200011P. R. China
- College of StomatologyNational Center for StomatologyNational Clinical Research Center for Oral DiseasesShanghai Key Laboratory of StomatologyShanghai Jiao Tong UniversityShanghai200011P. R. China
| | - Jingke Fu
- Shanghai Key Laboratory of Orthopaedic ImplantDepartment of Orthopaedic SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200011P. R. China
- Clinical and Translational Research Center for 3D Printing TechnologyShanghai Engineering Research Center of Innovative Orthopaedic Instruments and Personalized MedicineShanghai200011P. R. China
| |
Collapse
|
25
|
Xu W, Ye J, Cao Z, Zhao Y, Zhu Y, Li L. Glucocorticoids in lung cancer: Navigating the balance between immunosuppression and therapeutic efficacy. Heliyon 2024; 10:e32357. [PMID: 39022002 PMCID: PMC11252876 DOI: 10.1016/j.heliyon.2024.e32357] [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: 03/22/2024] [Revised: 06/03/2024] [Accepted: 06/03/2024] [Indexed: 07/20/2024] Open
Abstract
Glucocorticoids (GCs), a class of hormones secreted by the adrenal glands, are released into the bloodstream to maintain homeostasis and modulate responses to various stressors. These hormones function by binding to the widely expressed GC receptor (GR), thereby regulating a wide range of pathophysiological processes, especially in metabolism and immunity. The role of GCs in the tumor immune microenvironment (TIME) of lung cancer (LC) has been a focal point of research. As immunosuppressive agents, GCs exert a crucial impact on the occurrence, progression, and treatment of LC. In the TIME of LC, GCs act as a constantly swinging pendulum, simultaneously offering tumor-suppressive properties while diminishing the efficacy of immune-based therapies. The present study reviews the role and mechanisms of GCs in the TIME of LC.
Collapse
Affiliation(s)
| | | | - Zhendong Cao
- Department of Respiration, The Second Affiliated Hospital of Nanjing University of Traditional Chinese Medicine (Jiangsu Second Hospital of Traditional Chinese Medicine), Nanjing, Jiangsu, 210017, China
| | - Yupei Zhao
- Department of Respiration, The Second Affiliated Hospital of Nanjing University of Traditional Chinese Medicine (Jiangsu Second Hospital of Traditional Chinese Medicine), Nanjing, Jiangsu, 210017, China
| | - Yimin Zhu
- Department of Respiration, The Second Affiliated Hospital of Nanjing University of Traditional Chinese Medicine (Jiangsu Second Hospital of Traditional Chinese Medicine), Nanjing, Jiangsu, 210017, China
| | - Lei Li
- Department of Respiration, The Second Affiliated Hospital of Nanjing University of Traditional Chinese Medicine (Jiangsu Second Hospital of Traditional Chinese Medicine), Nanjing, Jiangsu, 210017, China
| |
Collapse
|
26
|
Shi YX, Dai PH, Chen T, Yan JH. Comprehensive analysis and experimental verification reveal the molecular characteristics of EGLN3 in pan-cancer and its relationship with the proliferation and apoptosis of lung cancer. Heliyon 2024; 10:e33206. [PMID: 39021988 PMCID: PMC11253545 DOI: 10.1016/j.heliyon.2024.e33206] [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: 03/28/2024] [Revised: 06/17/2024] [Accepted: 06/17/2024] [Indexed: 07/20/2024] Open
Abstract
Background Egl-9 family hypoxia-inducible factor 3 (EGLN3) is involved in the regulation of tumor microenvironment and tumor progression. However, its biological function and clinical significance in various cancers remain unclear. Methods RNA-seq, immunofluorescence, and single-cell sequencing were used to investigate the expression landscape of EGLN3 in pan-cancer. The TISCH2 and CancerSEA databases were used for single-cell function analysis of EGLN3 in tumors. TIMER2.0 database was used to explain the relationship between EGLN3 expression and immune cell infiltration. In addition, the LinkedOmics database was used to perform KEGG enrichment analysis of EGLN3 in pan-cancer. siRNA was used to silence gene expression. CCK8, transwell migration assay, flow cytometry analysis, RT-PCR, and western blotting were used to explore biological function of EGLN3. Results The results showed that EGLN3 was highly expressed in a variety of tumors, and was mainly localized to the cytosol. EGLN3 expression is associated with immunoinfiltration of a variety of immune cells, including macrophages in the tumor immune microenvironment and tumor-associated fibroblasts. Functional experiments revealed that EGLN3 knockdown could inhibit cell proliferation, migration, and promote cell apoptosis. In addition, we found that Bax expression was up-regulated and Bcl-2 expression was down-regulated in the si-EGLN3 group. Taken together, as a potential oncogene, EGLN3 is involved in the regulation of tumor malignant process, especially tumor cell apoptosis. Conclusion We comprehensively investigated the expression pattern, single-cell function, immune infiltration level and regulated signaling pathway of EGLN3 in pan-cancer. We found that EGLN3 is an important hypoxia and immune-related gene that may serve as a potential target for tumor immunotherapy.
Collapse
Affiliation(s)
- Yuan-Xiang Shi
- Institute of Clinical Medicine, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Peng-Hui Dai
- Department of Pathology, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Tao Chen
- School of Medicine, Hunan Normal University, Changsha, China
| | - Jian-Hua Yan
- Department of Cardiac Thoracic Surgery, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan, China
| |
Collapse
|
27
|
Yang N, Huang Y, Wang X, Wang D, Yao D, Ren G. Fibronectin-Targeting Dual-Modal MR/NIRF Imaging Contrast Agents for Diagnosis of Gastric Cancer and Peritoneal Metastasis. Bioconjug Chem 2024; 35:843-854. [PMID: 38775802 DOI: 10.1021/acs.bioconjchem.4c00208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
The prevalence and fatality rates of gastric cancer (GC) remain elevated, with advanced stages presenting a grim prognosis. Noninvasive diagnosis of GC cancer often proves challenging until the disease has progressed to an advanced stage or metastasized. Initially, the level of fibronectin (FN) in cancer-associated fibroblasts (CAFs) of GC was at least 3.7 times higher than that in normal fibroblasts. Herein, two FN-targeting magnetic resonance/near-infrared fluorescence (MR/NIRF) imaging contrast agents were developed to detect GC and peritoneal metastasis noninvasively. The probes CREKA-Cy7-(Gd-DOTA) and CREKA-Cy7-(Gd-DOTA)3 demonstrated significant FN-targeting capability (with dissociation constants of 1.0 and 2.1 mM) and effective MR imaging performance (with proton relaxivity values of 9.66 and 27.44 mM-1 s-1 at 9.4 T, 37 °C). In vivo imaging revealed a high signal-to-noise ratio and successful visualization of GC metastasis using NIRF imaging as well as successful tumor detection in MR imaging. Therefore, this study highlights the potential of FN-targeting probes for GC diagnosis and aids in the advancement of new diagnostic strategies for the clinical detection of GC.
Collapse
Affiliation(s)
- Ningxin Yang
- Department of Radiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Yuelin Huang
- Shanghai University of Sport, Shanghai 200438, China
| | - Xiaoyu Wang
- Department of Radiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Dengbin Wang
- Department of Radiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Defan Yao
- Department of Radiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
- Shanghai University of Sport, Shanghai 200438, China
| | - Gang Ren
- Department of Radiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| |
Collapse
|
28
|
Li Q, Peng G, Liu H, Wang L, Lu R, Li L. Molecular mechanisms of secretory autophagy and its potential role in diseases. Life Sci 2024; 347:122653. [PMID: 38663839 DOI: 10.1016/j.lfs.2024.122653] [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: 03/28/2024] [Accepted: 04/17/2024] [Indexed: 04/29/2024]
Abstract
Autophagy is a cellular degradation system that recycles or degrades damaged organelles, viral particles, and aggregated proteins through the lysosomal pathway. Autophagy plays an indispensable role in cellular homeostasis and communication processes. An interesting aspect is that autophagy also mediates the secretion of cellular contents, a process known as secretory autophagy. Secretory autophagy differs from macroautophagy, which sequesters recruited proteins, organelles, or viral particles into autophagosomes and degrades these sequesters in lysosomes, while the secretory autophagy pathway participates in the extracellular export of cellular contents sequestered by autophagosomes through autophagy and endosomal modulators. Recent evidence reveals that secretory autophagy is pivotal in the occurrence and progression of diseases. In this review, we summarize the molecular mechanisms of secretory autophagy. Furthermore, we review the impact of secretory autophagy on diseases, including cancer, viral infectious diseases, neurodegenerative diseases, and cardiovascular diseases. Considering the pleiotropic actions of secretory autophagy on diseases, studying the mechanism of secretory autophagy may help to understand the relevant pathophysiological processes.
Collapse
Affiliation(s)
- Qin Li
- Institute of Pharmacy and Pharmacology, University of South China, Hengyang 421001, Hunan, China
| | - Guolong Peng
- Institute of Pharmacy and Pharmacology, University of South China, Hengyang 421001, Hunan, China
| | - Huimei Liu
- Institute of Pharmacy and Pharmacology, University of South China, Hengyang 421001, Hunan, China
| | - Liwen Wang
- Institute of Pharmacy and Pharmacology, University of South China, Hengyang 421001, Hunan, China
| | - Ruirui Lu
- Institute of Pharmacy and Pharmacology, University of South China, Hengyang 421001, Hunan, China.
| | - Lanfang Li
- Institute of Pharmacy and Pharmacology, University of South China, Hengyang 421001, Hunan, China.
| |
Collapse
|
29
|
Benmelech S, Le T, McKay M, Nam J, Subramaniam K, Tellez D, Vlasak G, Mak M. Biophysical and biochemical aspects of immune cell-tumor microenvironment interactions. APL Bioeng 2024; 8:021502. [PMID: 38572312 PMCID: PMC10990568 DOI: 10.1063/5.0195244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Accepted: 03/19/2024] [Indexed: 04/05/2024] Open
Abstract
The tumor microenvironment (TME), composed of and influenced by a heterogeneous set of cancer cells and an extracellular matrix, plays a crucial role in cancer progression. The biophysical aspects of the TME (namely, its architecture and mechanics) regulate interactions and spatial distributions of cancer cells and immune cells. In this review, we discuss the factors of the TME-notably, the extracellular matrix, as well as tumor and stromal cells-that contribute to a pro-tumor, immunosuppressive response. We then discuss the ways in which cells of the innate and adaptive immune systems respond to tumors from both biochemical and biophysical perspectives, with increased focus on CD8+ and CD4+ T cells. Building upon this information, we turn to immune-based antitumor interventions-specifically, recent biophysical breakthroughs aimed at improving CAR-T cell therapy.
Collapse
Affiliation(s)
- Shoham Benmelech
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut 06511, USA
| | - Thien Le
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut 06511, USA
| | - Maggie McKay
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut 06511, USA
| | - Jungmin Nam
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut 06511, USA
| | - Krupakar Subramaniam
- Department of Molecular, Cellular, and Developmental Biology, Yale University, New Haven, Connecticut 06511, USA
| | - Daniela Tellez
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut 06511, USA
| | - Grace Vlasak
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut 06511, USA
| | - Michael Mak
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut 06511, USA
| |
Collapse
|
30
|
Zhang Y, Fang Z, Pan D, Li Y, Zhou J, Chen H, Li Z, Zhu M, Li C, Qin L, Ren X, Gong Q, Luo K. Dendritic Polymer-Based Nanomedicines Remodel the Tumor Stroma: Improve Drug Penetration and Enhance Antitumor Immune Response. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2401304. [PMID: 38469918 DOI: 10.1002/adma.202401304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 03/09/2024] [Indexed: 03/13/2024]
Abstract
The dense extracellular matrix (ECM) in solid tumors, contributed by cancer-associated fibroblasts (CAFs), hinders penetration of drugs and diminishes their therapeutic outcomes. A sequential treatment strategy of remodeling the ECM via a CAF modifier (dasatinib, DAS) is proposed to promote penetration of an immunogenic cell death (ICD) inducer (epirubicin, Epi) via apoptotic vesicles, ultimately enhancing the treatment efficacy against breast cancer. Dendritic poly(oligo(ethylene glycol) methyl ether methacrylate) (POEGMA)-based nanomedicines (poly[OEGMA-Dendron(G2)-Gly-Phe-Leu-Gly-DAS] (P-DAS) and poly[OEGMA-Dendron(G2)-hydrazone-Epi] (P-Epi)) are developed for sequential delivery of DAS and Epi, respectively. P-DAS reprograms CAFs to reduce collagen by downregulating collagen anabolism and energy metabolism, thereby reducing the ECM deposition. The regulated ECM can enhance tumor penetration of P-Epi to strengthen its ICD effect, leading to an amplified antitumor immune response. In breast cancer-bearing mice, this approach alleviates the ECM barrier, resulting in reduced tumor burden and increased cytotoxic T lymphocyte infiltration, and more encouragingly, synergizes effectively with anti-programmed cell death 1 (PD-1) therapy, significantly inhibiting tumor growth and preventing lung metastasis. Furthermore, systemic toxicity is barely detectable after sequential treatment with P-DAS and P-Epi. This approach opens a new avenue for treating desmoplastic tumors by metabolically targeting CAFs to overcome the ECM barrier.
Collapse
Affiliation(s)
- Yuxin Zhang
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zaixiang Fang
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Dayi Pan
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yunkun Li
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jie Zhou
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Hongying Chen
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zhiqian Li
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Mengli Zhu
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Cong Li
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Liwen Qin
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xiangyi Ren
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Qiyong Gong
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
- Functional and Molecular Imaging Key Laboratory of Sichuan Province, and Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, 610041, China
- Department of Radiology, West China Xiamen Hospital of Sichuan University, Xiamen, 361021, China
| | - Kui Luo
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
- Functional and Molecular Imaging Key Laboratory of Sichuan Province, and Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, 610041, China
| |
Collapse
|
31
|
Tang H, You T, Ge H, Gao J, Wang Y, Bai C, Sun Z, Han Q, Zhao RC. Anlotinib may enhance the efficacy of anti-PD1 therapy by inhibiting the AKT pathway and promoting the apoptosis of CAFs in lung adenocarcinoma. Int Immunopharmacol 2024; 133:112053. [PMID: 38615380 DOI: 10.1016/j.intimp.2024.112053] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Revised: 03/30/2024] [Accepted: 04/06/2024] [Indexed: 04/16/2024]
Abstract
Although PD-1 inhibitors have revolutionized the treatment paradigm of non-small cell lung cancer (NSCLC), their efficacy in treating NSCLC has remained unsatisfactory. Targeting cancer-associated fibroblasts (CAFs) is a potential approach for improving the immunotherapy response. Multitarget antiangiogenic tyrosine kinase receptor inhibitors (TKIs) can enhance the efficacy of PD-1 inhibitors in NSCLC patients. However, the effects and mechanisms of antiangiogenic TKIs on CAFs have not been elucidated. In this study, we first compared anlotinib with other antiangiogenic TKIs and confirmed the superior efficacy of anlotinib. Furthermore, we established NSCLC-associated CAF models and found that anlotinib impaired CAF viability and migration capacity and contributed to CAF apoptosis and cell cycle arrest in the G2/M phase. Moreover, anlotinib treatment attenuated the capacity of CAFs to recruit lung cancer cells and macrophages. Experiments in animal models suggested that anlotinib could enhance the efficacy of anti-PD1 therapy in NSCLC and affect CAF proliferation and apoptosis. Anlotinib increased the abundance of tumor-infiltrating CD8 + T cells, and PD-1 inhibitor-induced cytotoxicity to tumor cells was achieved through the transformation of the tumor microenvironment (TME) caused by anlotinib, which may partly explain the synergistic antitumor effect of anlotinib and PD-1 inhibitors. Mechanistically, anlotinib affects CAF apoptosis and cell viability at least in part by inhibiting the AKT pathway. In conclusion, our study suggested that anlotinib could regulate the TME, inhibit the AKT pathway and promote CAF apoptosis, providing new insights into the antitumor effect of anlotinib and improving the efficacy of immunotherapy.
Collapse
Affiliation(s)
- Hui Tang
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Tingting You
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Hui Ge
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Jingxi Gao
- Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Peking Union Medical College Hospital, Center of Excellence in Tissue Engineering Chinese Academy of Medical Sciences, Beijing Key Laboratory, Beijing, China
| | - Yingyi Wang
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| | - Chunmei Bai
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| | - Zhao Sun
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| | - Qin Han
- Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Peking Union Medical College Hospital, Center of Excellence in Tissue Engineering Chinese Academy of Medical Sciences, Beijing Key Laboratory, Beijing, China.
| | - Robert Chunhua Zhao
- Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Peking Union Medical College Hospital, Center of Excellence in Tissue Engineering Chinese Academy of Medical Sciences, Beijing Key Laboratory, Beijing, China; School of Life Sciences, Shanghai University, Shanghai, China.
| |
Collapse
|
32
|
Chen X, Xia D, Zeng X, Meng L, Wang Y, Li H, Zhang J, Zhao Z, Zhuang R, Fang J, Zhang X, Guo Z. Rational Design and Pharmacomodulation of 18F-Labeled Biotin/FAPI-Conjugated Heterodimers. J Med Chem 2024; 67:8361-8371. [PMID: 38726551 DOI: 10.1021/acs.jmedchem.4c00544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/24/2024]
Abstract
Due to the complex heterogeneity in different cancer types, the heterodimeric strategy has been intensively practiced to improve the effectiveness of tumor diagnostics. In this study, we developed a series of novel 18F-labeled biotin/FAPI-conjugated heterobivalent radioligands ([18F]AlF-NSFB, [18F]AlF-NSFBP2, and [18F]AlF-NSFBP4), synergistically targeting both fibroblast activation protein (FAP) and biotin receptor (BR), to enhance specific tumor uptake and retention. The in vitro and in vivo biological properties of these dual-targeting tracers were evaluated, with a particular focus on positron emission tomography imaging in A549 and HT1080-FAP tumor-bearing mice. Notably, in comparison to the corresponding FAP-targeted monomer [18F]AlF-NSF, biotin/FAPI-conjugated heterodimers exhibited a high uptake in tumor and prolong retention. In conclusion, as a proof-of-concept study, the findings validated the superiority of biotin/FAPI-conjugated heterodimers and the positive influence of biotin and linker on pharmacokinetics of radioligands. Within them, the bispecific [18F]AlF-NSFBP4 holds significant promise as a candidate for further clinical translational studies.
Collapse
Affiliation(s)
- Xuedong Chen
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, 4221-116 Xiang'An South Rd, Xiamen 361102, China
| | - Dongsheng Xia
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, 4221-116 Xiang'An South Rd, Xiamen 361102, China
| | - Xueyuan Zeng
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, 4221-116 Xiang'An South Rd, Xiamen 361102, China
| | - Lingxin Meng
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, 4221-116 Xiang'An South Rd, Xiamen 361102, China
| | - Yanjie Wang
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, 4221-116 Xiang'An South Rd, Xiamen 361102, China
| | - Huifeng Li
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, 4221-116 Xiang'An South Rd, Xiamen 361102, China
| | - Jingru Zhang
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, 4221-116 Xiang'An South Rd, Xiamen 361102, China
| | - Zuoquan Zhao
- Theranostics and Translational Research Center, Institute of Clinical Medicine, Department of Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 1 Shuaifuyuan, Dongcheng District, Beijing 100730, China
| | - Rongqiang Zhuang
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, 4221-116 Xiang'An South Rd, Xiamen 361102, China
| | - Jianyang Fang
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, 4221-116 Xiang'An South Rd, Xiamen 361102, China
| | - Xianzhong Zhang
- Theranostics and Translational Research Center, Institute of Clinical Medicine, Department of Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 1 Shuaifuyuan, Dongcheng District, Beijing 100730, China
| | - Zhide Guo
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, 4221-116 Xiang'An South Rd, Xiamen 361102, China
| |
Collapse
|
33
|
Mori JO, Elhussin I, Brennen WN, Graham MK, Lotan TL, Yates CC, De Marzo AM, Denmeade SR, Yegnasubramanian S, Nelson WG, Denis GV, Platz EA, Meeker AK, Heaphy CM. Prognostic and therapeutic potential of senescent stromal fibroblasts in prostate cancer. Nat Rev Urol 2024; 21:258-273. [PMID: 37907729 PMCID: PMC11058122 DOI: 10.1038/s41585-023-00827-x] [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] [Accepted: 10/03/2023] [Indexed: 11/02/2023]
Abstract
The stromal component of the tumour microenvironment in primary and metastatic prostate cancer can influence and promote disease progression. Within the prostatic stroma, fibroblasts are one of the most prevalent cell types associated with precancerous and cancerous lesions; they have a vital role in the structural composition, organization and integrity of the extracellular matrix. Fibroblasts within the tumour microenvironment can undergo cellular senescence, which is a stable arrest of cell growth and a phenomenon that is emerging as a recognized hallmark of cancer. Supporting the idea that cellular senescence has a pro-tumorigenic role, a subset of senescent cells exhibits a senescence-associated secretory phenotype (SASP), which, along with increased inflammation, can promote prostate cancer cell growth and survival. These cellular characteristics make targeting senescent cells and/or modulating SASP attractive as a potential preventive or therapeutic option for prostate cancer.
Collapse
Affiliation(s)
- Joakin O Mori
- Department of Medicine, Boston University Chobanian & Avedisian School of Medicine and Boston Medical Center, Boston, MA, USA
| | - Isra Elhussin
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Urology and the James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - W Nathaniel Brennen
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Urology and the James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Mindy K Graham
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Tamara L Lotan
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Urology and the James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Clayton C Yates
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Urology and the James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Angelo M De Marzo
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Urology and the James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Samuel R Denmeade
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Urology and the James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Srinivasan Yegnasubramanian
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Urology and the James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - William G Nelson
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Urology and the James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Gerald V Denis
- Department of Medicine, Boston University Chobanian & Avedisian School of Medicine and Boston Medical Center, Boston, MA, USA
- Department of Pharmacology and Experimental Therapeutics, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Elizabeth A Platz
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Urology and the James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Alan K Meeker
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Urology and the James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Christopher M Heaphy
- Department of Medicine, Boston University Chobanian & Avedisian School of Medicine and Boston Medical Center, Boston, MA, USA.
- Department of Pathology and Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA.
| |
Collapse
|
34
|
Sun J, Du R, Li X, Liu C, Wang D, He X, Li G, Zhang K, Wang S, Hao Q, Zhang Y, Li M, Gao Y, Zhang C. CD63 + cancer-associated fibroblasts confer CDK4/6 inhibitor resistance to breast cancer cells by exosomal miR-20. Cancer Lett 2024; 588:216747. [PMID: 38403110 DOI: 10.1016/j.canlet.2024.216747] [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: 11/14/2023] [Revised: 02/12/2024] [Accepted: 02/19/2024] [Indexed: 02/27/2024]
Abstract
Cyclin-dependent kinase 4 (CDK4) and CDK6 inhibitors (CDK4/6i) have rapidly received Food and Drug Administration (FDA) approval as a new type of therapy for patients with advanced hormone receptor-positive breast cancer. However, with the widespread application of CDK4/6i, drug resistance has become a new challenge for clinical practice and has greatly limited the treatment effect. Here, the whole microenvironment landscape of ER+ breast cancer tumors was revealed through single-cell RNA sequencing, and a specific subset of cancer-associated fibroblasts (CD63+ CAFs) was identified as highly enriched in CDK4/6i resistant tumor tissues. Then, we found that CD63+ CAFs can distinctly promote resistance to CDK4/6i in breast cancer cells and tumor xenografts. In addition, it was discovered that miR-20 is markedly enriched in the CD63+ CAFs-derived exosomes, which are used to communicate with ER+ breast cancer cells, leading to CDK4/6i resistance. Furthermore, exosomal miR-20 could directly target the RB1 mRNA 3'UTR and negatively regulate RB1 expression to decrease CDK4/6i sensitivity in breast cancer cells. Most importantly, we designed and synthesized cRGD-miR-20 sponge nanoparticles and found that they can enhance the therapeutic effect of CDK4/6i in breast cancer. In summary, our findings reveal that CD63+ CAFs can promote CDK4/6i resistance via exosomal miR-20, which induces the downregulation of RB1 in breast cancer cells, and suggest that CD63+ CAFs may be a novel therapeutic target to enhance CDK4/6i sensitivity.
Collapse
Affiliation(s)
- Jiahui Sun
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, 710032, Xi'an, PR China
| | - Ruoxin Du
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, 710032, Xi'an, PR China
| | - Xiaoju Li
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, 710032, Xi'an, PR China; Bioinformatics Center of AMMS, Beijing, 100850, PR China
| | - Chenlin Liu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, 710032, Xi'an, PR China
| | - Donghui Wang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, 710032, Xi'an, PR China
| | - Xiangmei He
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, 710032, Xi'an, PR China
| | - Guodong Li
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, 710032, Xi'an, PR China
| | - Kuo Zhang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, 710032, Xi'an, PR China
| | - Shuning Wang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, 710032, Xi'an, PR China
| | - Qiang Hao
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, 710032, Xi'an, PR China
| | - Yingqi Zhang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, 710032, Xi'an, PR China
| | - Meng Li
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, 710032, Xi'an, PR China.
| | - Yuan Gao
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, 710032, Xi'an, PR China.
| | - Cun Zhang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, 710032, Xi'an, PR China.
| |
Collapse
|
35
|
Chen C, Liu J, Lin X, Xiang A, Ye Q, Guo J, Rui T, Xu J, Hu S. Crosstalk between cancer-associated fibroblasts and regulated cell death in tumors: insights into apoptosis, autophagy, ferroptosis, and pyroptosis. Cell Death Discov 2024; 10:189. [PMID: 38649701 PMCID: PMC11035635 DOI: 10.1038/s41420-024-01958-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 04/10/2024] [Accepted: 04/11/2024] [Indexed: 04/25/2024] Open
Abstract
Cancer-associated fibroblasts (CAFs), the main stromal component of the tumor microenvironment (TME), play multifaceted roles in cancer progression through paracrine signaling, exosome transfer, and cell interactions. Attractively, recent evidence indicates that CAFs can modulate various forms of regulated cell death (RCD) in adjacent tumor cells, thus involving cancer proliferation, therapy resistance, and immune exclusion. Here, we present a brief introduction to CAFs and basic knowledge of RCD, including apoptosis, autophagy, ferroptosis, and pyroptosis. In addition, we further summarize the different types of RCD in tumors that are mediated by CAFs, as well as the effects of these modes of RCD on CAFs. This review will deepen our understanding of the interactions between CAFs and RCD and might offer novel therapeutic avenues for future cancer treatments.
Collapse
Affiliation(s)
- Cong Chen
- Department of Breast Surgery, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, China
| | - Jian Liu
- Department of Breast Surgery, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, China
| | - Xia Lin
- Department of Breast Surgery, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, China
| | - Aizhai Xiang
- Department of Breast Surgery, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, China
| | - Qianwei Ye
- Department of Breast Surgery, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, China
| | - Jufeng Guo
- Department of Breast Surgery, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, China
| | - Tao Rui
- Department of Breast Surgery, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, China
| | - Jian Xu
- Department of Central Laboratory, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, China
| | - Shufang Hu
- Department of Breast Surgery, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, China.
| |
Collapse
|
36
|
Xue X, Wang X, Pang M, Yu L, Qian J, Li X, Tian M, Lu C, Xiao C, Liu Y. An exosomal strategy for targeting cancer-associated fibroblasts mediated tumors desmoplastic microenvironments. J Nanobiotechnology 2024; 22:196. [PMID: 38644492 PMCID: PMC11032607 DOI: 10.1186/s12951-024-02452-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 04/01/2024] [Indexed: 04/23/2024] Open
Abstract
Tumors desmoplastic microenvironments are characterized by abundant stromal cells and extracellular matrix (ECM) deposition. Cancer-associated fibroblasts (CAFs), as the most abundant of all stromal cells, play significant role in mediating microenvironments, which not only remodel ECM to establish unique pathological barriers to hinder drug delivery in desmoplastic tumors, but also talk with immune cells and cancer cells to promote immunosuppression and cancer stem cells-mediated drug resistance. Thus, CAFs mediated desmoplastic microenvironments will be emerging as promising strategy to treat desmoplastic tumors. However, due to the complexity of microenvironments and the heterogeneity of CAFs in such tumors, an effective deliver system should be fully considered when designing the strategy of targeting CAFs mediated microenvironments. Engineered exosomes own powerful intercellular communication, cargoes delivery, penetration and targeted property of desired sites, which endow them with powerful theranostic potential in desmoplastic tumors. Here, we illustrate the significance of CAFs in tumors desmoplastic microenvironments and the theranostic potential of engineered exosomes targeting CAFs mediated desmoplastic microenvironments in next generation personalized nano-drugs development.
Collapse
Affiliation(s)
- Xiaoxia Xue
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Xiangpeng Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Mingshi Pang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Liuchunyang Yu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Jinxiu Qian
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Xiaoyu Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Meng Tian
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Cheng Lu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| | - Cheng Xiao
- Institute of Clinical Medicine, China-Japan Friendship Hospital, Beijing, 100029, China.
| | - Yuanyan Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China.
| |
Collapse
|
37
|
Gou Z, Li J, Liu J, Yang N. The hidden messengers: cancer associated fibroblasts-derived exosomal miRNAs as key regulators of cancer malignancy. Front Cell Dev Biol 2024; 12:1378302. [PMID: 38694824 PMCID: PMC11061421 DOI: 10.3389/fcell.2024.1378302] [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: 01/29/2024] [Accepted: 04/08/2024] [Indexed: 05/04/2024] Open
Abstract
Cancer-associated fibroblasts (CAFs), a class of stromal cells in the tumor microenvironment (TME), play a key role in controlling cancer cell invasion and metastasis, immune evasion, angiogenesis, and resistance to chemotherapy. CAFs mediate their activities by secreting soluble chemicals, releasing exosomes, and altering the extracellular matrix (ECM). Exosomes contain various biomolecules, such as nucleic acids, lipids, and proteins. microRNA (miRNA), a 22-26 nucleotide non-coding RNA, can regulate the cellular transcription processes. Studies have shown that miRNA-loaded exosomes secreted by CAFs engage in various regulatory communication networks with other TME constituents. This study focused on the roles of CAF-derived exosomal miRNAs in generating cancer malignant characteristics, including immune modulation, tumor growth, migration and invasion, epithelial-mesenchymal transition (EMT), and treatment resistance. This study thoroughly examines miRNA's dual regulatory roles in promoting and suppressing cancer. Thus, changes in the CAF-derived exosomal miRNAs can be used as biomarkers for the diagnosis and prognosis of patients, and their specificity can be used to develop newer therapies. This review also discusses the pressing problems that require immediate attention, aiming to inspire researchers to explore more novel avenues in this field.
Collapse
Affiliation(s)
- Zixuan Gou
- Bethune First Clinical School of Medicine, The First Hospital of Jilin University, Changchun, China
| | - Jiannan Li
- Department of General Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Jianming Liu
- Department of Otolaryngology Head and Neck Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Na Yang
- Department of Clinical Pharmacy, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
38
|
Ten A, Kumeiko V, Farniev V, Gao H, Shevtsov M. Tumor Microenvironment Modulation by Cancer-Derived Extracellular Vesicles. Cells 2024; 13:682. [PMID: 38667297 PMCID: PMC11049026 DOI: 10.3390/cells13080682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Revised: 04/06/2024] [Accepted: 04/11/2024] [Indexed: 04/28/2024] Open
Abstract
The tumor microenvironment (TME) plays an important role in the process of tumorigenesis, regulating the growth, metabolism, proliferation, and invasion of cancer cells, as well as contributing to tumor resistance to the conventional chemoradiotherapies. Several types of cells with relatively stable phenotypes have been identified within the TME, including cancer-associated fibroblasts (CAFs), tumor-associated macrophages (TAMs), neutrophils, and natural killer (NK) cells, which have been shown to modulate cancer cell proliferation, metastasis, and interaction with the immune system, thus promoting tumor heterogeneity. Growing evidence suggests that tumor-cell-derived extracellular vesicles (EVs), via the transfer of various molecules (e.g., RNA, proteins, peptides, and lipids), play a pivotal role in the transformation of normal cells in the TME into their tumor-associated protumorigenic counterparts. This review article focuses on the functions of EVs in the modulation of the TME with a view to how exosomes contribute to the transformation of normal cells, as well as their importance for cancer diagnosis and therapy.
Collapse
Affiliation(s)
- Artem Ten
- School of Medicine and Life Sciences, Far Eastern Federal University, 690922 Vladivostok, Russia; (A.T.); (V.K.); (V.F.)
| | - Vadim Kumeiko
- School of Medicine and Life Sciences, Far Eastern Federal University, 690922 Vladivostok, Russia; (A.T.); (V.K.); (V.F.)
| | - Vladislav Farniev
- School of Medicine and Life Sciences, Far Eastern Federal University, 690922 Vladivostok, Russia; (A.T.); (V.K.); (V.F.)
| | - Huile Gao
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610064, China;
| | - Maxim Shevtsov
- School of Medicine and Life Sciences, Far Eastern Federal University, 690922 Vladivostok, Russia; (A.T.); (V.K.); (V.F.)
- Laboratory of Biomedical Nanotechnologies, Institute of Cytology of the Russian Academy of Sciences, Tikhoretsky Ave., 4, 194064 St. Petersburg, Russia
- Personalized Medicine Centre, Almazov National Medical Research Centre, Akkuratova Str., 2, 197341 St. Petersburg, Russia
- Department of Radiation Oncology, Technishe Universität München (TUM), Klinikum Rechts der Isar, Ismaninger Str., 22, 81675 Munich, Germany
| |
Collapse
|
39
|
Huang Q, Ge Y, He Y, Wu J, Tong Y, Shang H, Liu X, Ba X, Xia D, Peng E, Chen Z, Tang K. The Application of Nanoparticles Targeting Cancer-Associated Fibroblasts. Int J Nanomedicine 2024; 19:3333-3365. [PMID: 38617796 PMCID: PMC11012801 DOI: 10.2147/ijn.s447350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Accepted: 03/23/2024] [Indexed: 04/16/2024] Open
Abstract
Cancer-associated fibroblasts (CAF) are the most abundant stromal cells in the tumor microenvironment (TME), especially in solid tumors. It has been confirmed that it can not only interact with tumor cells to promote cancer progression and metastasis, but also affect the infiltration and function of immune cells to induce chemotherapy and immunotherapy resistance. So, targeting CAF has been considered an important method in cancer treatment. The rapid development of nanotechnology provides a good perspective to improve the efficiency of targeting CAF. At present, more and more researches have focused on the application of nanoparticles (NPs) in targeting CAF. These studies explored the effects of different types of NPs on CAF and the multifunctional nanomedicines that can eliminate CAF are able to enhance the EPR effect which facilitate the anti-tumor effect of themselves. There also exist amounts of studies focusing on using NPs to inhibit the activation and function of CAF to improve the therapeutic efficacy. The application of NPs targeting CAF needs to be based on an understanding of CAF biology. Therefore, in this review, we first summarized the latest progress of CAF biology, then discussed the types of CAF-targeting NPs and the main strategies in the current. The aim is to elucidate the application of NPs in targeting CAF and provide new insights for engineering nanomedicine to enhance immune response in cancer treatment.
Collapse
Affiliation(s)
- Qiu Huang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, People’s Republic of China
| | - Yue Ge
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, People’s Republic of China
| | - Yu He
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, People’s Republic of China
| | - Jian Wu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, People’s Republic of China
| | - Yonghua Tong
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, People’s Republic of China
| | - Haojie Shang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, People’s Republic of China
| | - Xiao Liu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, People’s Republic of China
| | - Xiaozhuo Ba
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, People’s Republic of China
| | - Ding Xia
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, People’s Republic of China
| | - Ejun Peng
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, People’s Republic of China
| | - Zhiqiang Chen
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, People’s Republic of China
| | - Kun Tang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, People’s Republic of China
| |
Collapse
|
40
|
Fang B, Lu Y, Li X, Wei Y, Ye D, Wei G, Zhu Y. Targeting the tumor microenvironment, a new therapeutic approach for prostate cancer. Prostate Cancer Prostatic Dis 2024:10.1038/s41391-024-00825-z. [PMID: 38565910 DOI: 10.1038/s41391-024-00825-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 03/17/2024] [Accepted: 03/21/2024] [Indexed: 04/04/2024]
Abstract
BACKGROUND A growing number of studies have shown that in addition to adaptive immune cells such as CD8 + T cells and CD4 + T cells, various other cellular components within prostate cancer (PCa) tumor microenvironment (TME), mainly tumor-associated macrophages (TAMs), cancer-associated fibroblasts (CAFs) and myeloid-derived suppressor cells (MDSCs), have been increasingly recognized as important modulators of tumor progression and promising therapeutic targets. OBJECTIVE In this review, we aim to delineate the mechanisms by which TAMs, CAFs and MDSCs interact with PCa cells in the TME, summarize the therapeutic advancements targeting these cells and discuss potential new therapeutic avenues. METHODS We searched PubMed for relevant studies published through December 10 2023 on TAMs, CAFs and MDSCs in PCa. RESULTS TAMs, CAFs and MDSCs play a critical role in the tumorigenesis, progression, and metastasis of PCa. Moreover, they substantially mediate therapeutic resistance against conventional treatments including anti-androgen therapy, chemotherapy, and immunotherapy. Therapeutic interventions targeting these cellular components have demonstrated promising effects in preclinical models and several clinical trials for PCa, when administrated alone, or combined with other anti-cancer therapies. However, the lack of reliable biomarkers for patient selection and incomplete understanding of the mechanisms underlying the interactions between these cellular components and PCa cells hinder their clinical translation and utility. CONCLUSION New therapeutic strategies targeting TAMs, CAFs, and MDSCs in PCa hold promising prospects. Future research endeavors should focus on a more comprehensive exploration of the specific mechanisms by which these cells contribute to PCa, aiming to identify additional drug targets and conduct more clinical trials to validate the safety and efficacy of these treatment strategies.
Collapse
Affiliation(s)
- Bangwei Fang
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Genitourinary Cancer Institute, Shanghai, 200032, China
| | - Ying Lu
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences, Shanghai Medical College of Fudan University, Shanghai, China
| | - Xiaomeng Li
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Genitourinary Cancer Institute, Shanghai, 200032, China
| | - Yu Wei
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Genitourinary Cancer Institute, Shanghai, 200032, China
| | - Dingwei Ye
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Genitourinary Cancer Institute, Shanghai, 200032, China
| | - Gonghong Wei
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences, Shanghai Medical College of Fudan University, Shanghai, China
| | - Yao Zhu
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
- Shanghai Genitourinary Cancer Institute, Shanghai, 200032, China.
| |
Collapse
|
41
|
Qian ZY, Pan YQ, Li XX, Chen YX, Wu HX, Liu ZX, Kosar M, Bartek J, Wang ZX, Xu RH. Modulator of TMB-associated immune infiltration (MOTIF) predicts immunotherapy response and guides combination therapy. Sci Bull (Beijing) 2024; 69:803-822. [PMID: 38320897 DOI: 10.1016/j.scib.2024.01.025] [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: 08/01/2023] [Revised: 11/04/2023] [Accepted: 12/07/2023] [Indexed: 02/08/2024]
Abstract
Patients with high tumor mutational burden (TMB) levels do not consistently respond to immune checkpoint inhibitors (ICIs), possibly because a high TMB level does not necessarily result in adequate infiltration of CD8+ T cells. Using bulk ribonucleic acid sequencing (RNA-seq) data from 9311 tumor samples across 30 cancer types, we developed a novel tool called the modulator of TMB-associated immune infiltration (MOTIF), which comprises genes that can determine the extent of CD8+ T cell infiltration prompted by a certain TMB level. We confirmed that MOTIF can accurately reflect the integrity and defects of the cancer-immunity cycle. By analyzing 84 human single-cell RNA-seq datasets from 32 types of solid tumors, we revealed that MOTIF can provide insights into the diverse roles of various cell types in the modulation of CD8+ T cell infiltration. Using pretreatment RNA-seq data from 13 ICI-treated cohorts, we validated the use of MOTIF in predicting CD8+ T cell infiltration and ICI efficacy. Among the components of MOTIF, we identified EMC3 as a negative regulator of CD8+ T cell infiltration, which was validated via in vivo studies. Additionally, MOTIF provided guidance for the potential combinations of programmed death 1 blockade with certain immunostimulatory drugs to facilitate CD8+ T cell infiltration and improve ICI efficacy.
Collapse
Affiliation(s)
- Zheng-Yu Qian
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou 510060, China
| | - Yi-Qian Pan
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou 510060, China
| | - Xue-Xin Li
- Science for Life Laboratory, Division of Genome Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm S-171 21, Sweden; Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang 110032, China
| | - Yan-Xing Chen
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou 510060, China
| | - Hao-Xiang Wu
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou 510060, China
| | - Ze-Xian Liu
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou 510060, China; Bioinformatics Platform, Sun Yat-sen University Cancer Center, Guangzhou 510060, China; Laboratory of Artificial Intelligence and Data Science, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Martin Kosar
- Science for Life Laboratory, Division of Genome Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm S-171 21, Sweden; Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Haining 314400, China; Edinburgh Medical School, Biomedical Sciences, College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh EH1 1LT, UK
| | - Jiri Bartek
- Science for Life Laboratory, Division of Genome Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm S-171 21, Sweden; Danish Cancer Society Research Center, Copenhagen DK-2100, Denmark.
| | - Zi-Xian Wang
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou 510060, China; Laboratory of Artificial Intelligence and Data Science, Sun Yat-sen University Cancer Center, Guangzhou 510060, China.
| | - Rui-Hua Xu
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou 510060, China; Laboratory of Artificial Intelligence and Data Science, Sun Yat-sen University Cancer Center, Guangzhou 510060, China.
| |
Collapse
|
42
|
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
|
43
|
Li Y, Zhang C, Jiang A, Lin A, Liu Z, Cheng X, Wang W, Cheng Q, Zhang J, Wei T, Luo P. Potential anti-tumor effects of regulatory T cells in the tumor microenvironment: a review. J Transl Med 2024; 22:293. [PMID: 38509593 PMCID: PMC10953261 DOI: 10.1186/s12967-024-05104-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Accepted: 03/15/2024] [Indexed: 03/22/2024] Open
Abstract
Regulatory T cells (Tregs) expressing the transcription factor FoxP3 are essential for maintaining immunological balance and are a significant component of the immunosuppressive tumor microenvironment (TME). Single-cell RNA sequencing (ScRNA-seq) technology has shown that Tregs exhibit significant plasticity and functional diversity in various tumors within the TME. This results in Tregs playing a dual role in the TME, which is not always centered around supporting tumor progression as typically believed. Abundant data confirms the anti-tumor activities of Tregs and their correlation with enhanced patient prognosis in specific types of malignancies. In this review, we summarize the potential anti-tumor actions of Tregs, including suppressing tumor-promoting inflammatory responses and boosting anti-tumor immunity. In addition, this study outlines the spatial and temporal variations in Tregs function to emphasize that their predictive significance in malignancies may change. It is essential to comprehend the functional diversity and potential anti-tumor effects of Tregs to improve tumor therapy strategies.
Collapse
Affiliation(s)
- Yu Li
- The Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Cangang Zhang
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Aimin Jiang
- Department of Urology, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Anqi Lin
- The Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Zaoqu Liu
- Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing, China
- Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Department of Pathophysiology, Peking Union Medical College, Beijing, 100730, China
| | - Xiangshu Cheng
- College of Bioinformatics Science and Technology, Harbin Medical University, 157 Baojian Road. Nangang District, Harbin, Heilongiiang, China
| | - Wanting Wang
- Institute of Molecular and Translational Medicine, and Department of Biochemistry and Molecular Biology, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, Shaanxi, China
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Jian Zhang
- The Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China.
| | - Ting Wei
- The Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China.
| | - Peng Luo
- The Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
44
|
Sofias AM, Guo B, Xu J, Lammers T. Image-guided drug delivery: Biomedical and imaging advances. Adv Drug Deliv Rev 2024; 206:115187. [PMID: 38272184 DOI: 10.1016/j.addr.2024.115187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2024]
Affiliation(s)
- Alexandros Marios Sofias
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging (ExMI), RWTH Aachen University Hospital, Aachen, Germany.
| | - Bing Guo
- School of Science, Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application, Harbin Institute of Technology, Shenzhen 518055, China.
| | - Jian Xu
- Institute of Low-Dimensional Materials Genome Initiative, College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen 518060, China.
| | - Twan Lammers
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging (ExMI), RWTH Aachen University Hospital, Aachen, Germany.
| |
Collapse
|
45
|
Cao L, Ouyang H. Intercellular crosstalk between cancer cells and cancer-associated fibroblasts via exosomes in gastrointestinal tumors. Front Oncol 2024; 14:1374742. [PMID: 38463229 PMCID: PMC10920350 DOI: 10.3389/fonc.2024.1374742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 02/08/2024] [Indexed: 03/12/2024] Open
Abstract
Gastrointestinal (GI) tumors are a significant global health threat, with high rates of morbidity and mortality. Exosomes contain various biologically active molecules like nucleic acids, proteins, and lipids and can serve as messengers for intercellular communication. They play critical roles in the exchange of information between tumor cells and the tumor microenvironment (TME). The TME consists of mesenchymal cells and components of the extracellular matrix (ECM), with fibroblasts being the most abundant cell type in the tumor mesenchyme. Cancer-associated fibroblasts (CAFs) are derived from normal fibroblasts and mesenchymal stem cells that are activated in the TME. CAFs can secrete exosomes to modulate cell proliferation, invasion, migration, drug resistance, and other biological processes in tumors. Additionally, tumor cells can manipulate the function and behavior of fibroblasts through direct cell-cell interactions. This review provides a summary of the intercellular crosstalk between GI tumor cells and CAFs through exosomes, along with potential underlying mechanisms.
Collapse
Affiliation(s)
- Longyang Cao
- Department of Gastroenterology, The First Peoples' Hospital of Hangzhou Linan District, Hangzhou, China
| | - Hong Ouyang
- Department of Gastroenterology, The First Peoples' Hospital of Hangzhou Linan District, Hangzhou, China
| |
Collapse
|
46
|
Cheng N, Wang B, Xu J, Xue L, Ying J. Tumor stroma ratio, tumor stroma maturity, tumor-infiltrating immune cells in relation to prognosis, and neoadjuvant therapy response in esophagogastric junction adenocarcinoma. Virchows Arch 2024:10.1007/s00428-024-03755-2. [PMID: 38383941 DOI: 10.1007/s00428-024-03755-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 01/14/2024] [Accepted: 01/27/2024] [Indexed: 02/23/2024]
Abstract
Accurate predictions on prognosis and neoadjuvant therapy response are crucial for esophagogastric junction adenocarcinoma (EGJA) patients. Therefore, we aimed to investigate the predictive abilities of several indicators, including tumor stroma ratio (TSR), tumor stroma maturity (TSM), and the density and spatial distribution of tumor-infiltrating immune cells (TIICs), such as T cells, B cells, and tumor-associated macrophages (TAMs). Resection and biopsy specimens of a total of 695 patients were included, obtained from the National Cancer Center (NCC) and The Cancer Genome Atlas (TCGA) cohorts. TSR and TSM were evaluated based on histological assessment. TIICs were quantified by QuPath following immunohistochemical (IHC) staining in resection specimens, while the Klintrup-Mäkinen (KM) grade was employed for evaluating TIIC in biopsy specimens. Patients with high stromal levels or immature stroma had relatively worse prognoses. Furthermore, high CD8+T cell count in the tumor periphery, as well as low CD68+ TAM count either in the tumor center or in the tumor periphery, was an independent favorable prognostic factor. Significantly, the combination model incorporating TSM and CD163+TAMs emerged as an independent prognostic factor in both two independent cohorts (HR 3.644, 95% CI 1.341-9.900, p = 0.011 and HR 1.891, 95% CI 1.195-2.99, p = 0.006, respectively). Additionally, high stromal levels in preoperative biopsies correlated with poor neoadjuvant therapy response (p < 0.05). In conclusion, our findings suggest that TSR, TSM, CD8+T cell, CD68+TAMs, and CD163+TAMs predict the prognosis to some extent in patients with EGJA. Notably, the combined model incorporating TSM and CD163+TAM can contribute significantly to prognostic stratification. Additionally, high stromal levels evaluated in preoperative biopsy specimens correlated with poor neoadjuvant therapy response.
Collapse
Affiliation(s)
- Na Cheng
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 17 Panjiayuan, Chaoyang District, Beijing, 100021, China
| | - Bingzhi Wang
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 17 Panjiayuan, Chaoyang District, Beijing, 100021, China
| | - Jiaqi Xu
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 17 Panjiayuan, Chaoyang District, Beijing, 100021, China
| | - Liyan Xue
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 17 Panjiayuan, Chaoyang District, Beijing, 100021, China.
| | - Jianming Ying
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 17 Panjiayuan, Chaoyang District, Beijing, 100021, China.
| |
Collapse
|
47
|
Sharma NK, Bahot A, Sekar G, Bansode M, Khunteta K, Sonar PV, Hebale A, Salokhe V, Sinha BK. Understanding Cancer's Defense against Topoisomerase-Active Drugs: A Comprehensive Review. Cancers (Basel) 2024; 16:680. [PMID: 38398072 PMCID: PMC10886629 DOI: 10.3390/cancers16040680] [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: 01/25/2024] [Accepted: 02/02/2024] [Indexed: 02/25/2024] Open
Abstract
In recent years, the emergence of cancer drug resistance has been one of the crucial tumor hallmarks that are supported by the level of genetic heterogeneity and complexities at cellular levels. Oxidative stress, immune evasion, metabolic reprogramming, overexpression of ABC transporters, and stemness are among the several key contributing molecular and cellular response mechanisms. Topo-active drugs, e.g., doxorubicin and topotecan, are clinically active and are utilized extensively against a wide variety of human tumors and often result in the development of resistance and failure to therapy. Thus, there is an urgent need for an incremental and comprehensive understanding of mechanisms of cancer drug resistance specifically in the context of topo-active drugs. This review delves into the intricate mechanistic aspects of these intracellular and extracellular topo-active drug resistance mechanisms and explores the use of potential combinatorial approaches by utilizing various topo-active drugs and inhibitors of pathways involved in drug resistance. We believe that this review will help guide basic scientists, pre-clinicians, clinicians, and policymakers toward holistic and interdisciplinary strategies that transcend resistance, renewing optimism in the ongoing battle against cancer.
Collapse
Affiliation(s)
- Nilesh Kumar Sharma
- Cancer and Translational Research Centre Dr. D.Y. Patil Biotechnology & Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune 411033, Maharashtra, India; (N.K.S.); (A.B.); (G.S.); (M.B.); (K.K.); (P.V.S.); (A.H.); (V.S.)
| | - Anjali Bahot
- Cancer and Translational Research Centre Dr. D.Y. Patil Biotechnology & Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune 411033, Maharashtra, India; (N.K.S.); (A.B.); (G.S.); (M.B.); (K.K.); (P.V.S.); (A.H.); (V.S.)
| | - Gopinath Sekar
- Cancer and Translational Research Centre Dr. D.Y. Patil Biotechnology & Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune 411033, Maharashtra, India; (N.K.S.); (A.B.); (G.S.); (M.B.); (K.K.); (P.V.S.); (A.H.); (V.S.)
| | - Mahima Bansode
- Cancer and Translational Research Centre Dr. D.Y. Patil Biotechnology & Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune 411033, Maharashtra, India; (N.K.S.); (A.B.); (G.S.); (M.B.); (K.K.); (P.V.S.); (A.H.); (V.S.)
| | - Kratika Khunteta
- Cancer and Translational Research Centre Dr. D.Y. Patil Biotechnology & Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune 411033, Maharashtra, India; (N.K.S.); (A.B.); (G.S.); (M.B.); (K.K.); (P.V.S.); (A.H.); (V.S.)
| | - Priyanka Vijay Sonar
- Cancer and Translational Research Centre Dr. D.Y. Patil Biotechnology & Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune 411033, Maharashtra, India; (N.K.S.); (A.B.); (G.S.); (M.B.); (K.K.); (P.V.S.); (A.H.); (V.S.)
| | - Ameya Hebale
- Cancer and Translational Research Centre Dr. D.Y. Patil Biotechnology & Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune 411033, Maharashtra, India; (N.K.S.); (A.B.); (G.S.); (M.B.); (K.K.); (P.V.S.); (A.H.); (V.S.)
| | - Vaishnavi Salokhe
- Cancer and Translational Research Centre Dr. D.Y. Patil Biotechnology & Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune 411033, Maharashtra, India; (N.K.S.); (A.B.); (G.S.); (M.B.); (K.K.); (P.V.S.); (A.H.); (V.S.)
| | - Birandra Kumar Sinha
- Mechanistic Toxicology Branch, Division of Translational Toxicology, National Institute of Environmental Health Sciences, Research Triangle Park, Durham, NC 27709, USA
| |
Collapse
|
48
|
Wei J, Ge X, Qian Y, Jiang K, Chen X, Lu W, Yang H, Fu D, Fang Y, Zhou X, Xiao Q, Tang Y, Ding K. Development and verification of a combined immune- and cancer-associated fibroblast related prognostic signature for colon adenocarcinoma. Front Immunol 2024; 15:1291938. [PMID: 38312843 PMCID: PMC10834644 DOI: 10.3389/fimmu.2024.1291938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Accepted: 01/04/2024] [Indexed: 02/06/2024] Open
Abstract
Introduction To better understand the role of immune escape and cancer-associated fibroblasts (CAFs) in colon adenocarcinoma (COAD), an integrative analysis of the tumor microenvironment was performed using a set of 12 immune- and CAF-related genes (ICRGs). Methods Univariate and least absolute shrinkage and selection operator (LASSO) Cox regression analyses were used to establish a prognostic signature based on the expression of these 12 genes (S1PR5, AEN, IL20RB, FGF9, OSBPL1A, HSF4, PCAT6, FABP4, KIF15, ZNF792, CD1B and GLP2R). This signature was validated in both internal and external cohorts and was found to have a higher C-index than previous COAD signatures, confirming its robustness and reliability. To make use of this signature in clinical settings, a nomogram incorporating ICRG signatures and key clinical parameters, such as age and T stage, was developed. Finally, the role of S1PR5 in the immune response of COAD was validated through in vitro cytotoxicity experiments. Results The developed nomogram exhibited slightly improved predictive accuracy compared to the ICRG signature alone, as indicated by the areas under the receiver operating characteristic curves (AUC, nomogram:0.838; ICRGs:0.807). The study also evaluated the relationships between risk scores (RS) based on the expression of the ICRGs and other key immunotherapy variables, including immune checkpoint expression, immunophenoscore (IPS), and microsatellite instability (MSI). Integration of these variables led to more precise prediction of treatment efficacy, enabling personalized immunotherapy for COAD patients. Knocking down S1PR5 can enhance the efficacy of PD-1 monoclonal antibody, promoting the cytotoxicity of T cells against HCT116 cells ((p<0.05). Discussion These findings indicate that the ICRG signature may be a valuable tool for predicting prognostic risk, evaluating the efficacy of immunotherapy, and tailoring personalized treatment options for patients with COAD.
Collapse
Affiliation(s)
- Jingsun Wei
- Department of Colorectal Surgery and Oncology (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Department of Colorectal Surgery and Oncology, Zhejiang Provincial Clinical Research Center for Cancer, Hangzhou, Zhejiang, China
- Department of Colorectal Surgery and Oncology, Cancer Center of Zhejiang University, Hangzhou, Zhejiang, China
| | - Xiaoxu Ge
- Department of Colorectal Surgery and Oncology (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Department of Colorectal Surgery and Oncology, Zhejiang Provincial Clinical Research Center for Cancer, Hangzhou, Zhejiang, China
- Department of Colorectal Surgery and Oncology, Cancer Center of Zhejiang University, Hangzhou, Zhejiang, China
| | - Yucheng Qian
- Department of Colorectal Surgery and Oncology (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Department of Colorectal Surgery and Oncology, Zhejiang Provincial Clinical Research Center for Cancer, Hangzhou, Zhejiang, China
- Department of Colorectal Surgery and Oncology, Cancer Center of Zhejiang University, Hangzhou, Zhejiang, China
| | - Kai Jiang
- Department of Colorectal Surgery and Oncology (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Department of Colorectal Surgery and Oncology, Zhejiang Provincial Clinical Research Center for Cancer, Hangzhou, Zhejiang, China
- Department of Colorectal Surgery and Oncology, Cancer Center of Zhejiang University, Hangzhou, Zhejiang, China
| | - Xin Chen
- Department of Colorectal Surgery and Oncology (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Department of Colorectal Surgery and Oncology, Zhejiang Provincial Clinical Research Center for Cancer, Hangzhou, Zhejiang, China
- Department of Colorectal Surgery and Oncology, Cancer Center of Zhejiang University, Hangzhou, Zhejiang, China
| | - Wei Lu
- Department of Colorectal Surgery and Oncology (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Department of Colorectal Surgery and Oncology, Zhejiang Provincial Clinical Research Center for Cancer, Hangzhou, Zhejiang, China
- Department of Colorectal Surgery and Oncology, Cancer Center of Zhejiang University, Hangzhou, Zhejiang, China
| | - Hang Yang
- Department of Colorectal Surgery and Oncology (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Department of Colorectal Surgery and Oncology, Zhejiang Provincial Clinical Research Center for Cancer, Hangzhou, Zhejiang, China
- Department of Colorectal Surgery and Oncology, Cancer Center of Zhejiang University, Hangzhou, Zhejiang, China
| | - Dongliang Fu
- Department of Colorectal Surgery and Oncology (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Department of Colorectal Surgery and Oncology, Zhejiang Provincial Clinical Research Center for Cancer, Hangzhou, Zhejiang, China
- Department of Colorectal Surgery and Oncology, Cancer Center of Zhejiang University, Hangzhou, Zhejiang, China
| | - Yimin Fang
- Department of Colorectal Surgery and Oncology (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Department of Colorectal Surgery and Oncology, Zhejiang Provincial Clinical Research Center for Cancer, Hangzhou, Zhejiang, China
- Department of Colorectal Surgery and Oncology, Cancer Center of Zhejiang University, Hangzhou, Zhejiang, China
| | - Xinyi Zhou
- Department of Colorectal Surgery and Oncology (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Department of Colorectal Surgery and Oncology, Zhejiang Provincial Clinical Research Center for Cancer, Hangzhou, Zhejiang, China
- Department of Colorectal Surgery and Oncology, Cancer Center of Zhejiang University, Hangzhou, Zhejiang, China
| | - Qian Xiao
- Department of Colorectal Surgery and Oncology (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Department of Colorectal Surgery and Oncology, Zhejiang Provincial Clinical Research Center for Cancer, Hangzhou, Zhejiang, China
- Department of Colorectal Surgery and Oncology, Cancer Center of Zhejiang University, Hangzhou, Zhejiang, China
| | - Yang Tang
- Department of Colorectal Surgery and Oncology (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Department of Colorectal Surgery and Oncology, Zhejiang Provincial Clinical Research Center for Cancer, Hangzhou, Zhejiang, China
- Department of Colorectal Surgery and Oncology, Cancer Center of Zhejiang University, Hangzhou, Zhejiang, China
| | - Kefeng Ding
- Department of Colorectal Surgery and Oncology (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Department of Colorectal Surgery and Oncology, Zhejiang Provincial Clinical Research Center for Cancer, Hangzhou, Zhejiang, China
- Department of Colorectal Surgery and Oncology, Cancer Center of Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
49
|
Cen X, Li M, Yao A, Zheng Y, Lai W. Immune infiltration and clinical significance analyses of the cancer-associated fibroblast-related signature in skin cutaneous melanoma. J Gene Med 2024; 26:e3614. [PMID: 37847069 DOI: 10.1002/jgm.3614] [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: 03/17/2023] [Revised: 07/14/2023] [Accepted: 09/26/2023] [Indexed: 10/18/2023] Open
Abstract
BACKGROUND Skin cutaneous melanoma (SKCM) is one of the most aggressive cancers with high mortality rates. Cancer-associated fibroblasts (CAFs) play essential roles in tumor growth, metastasis and the establishment of a pro-tumor microenvironment. This study aimed to establish a CAF-related signature for providing a new perspective for indicating prognosis and guiding therapeutic regimens of SKCM patients. METHODS In this study, the CAF-related genes were screened out based on melanoma-associated fibroblast markers identified from single-cell transcriptome analysis in the Gene Expression Omnibus (GEO) database and a CAF-related module identified from weighted gene co-expression analysis using The Cancer Genome Atlas (TCGA) dataset. We extracted these gene expression data of SKCM samples from TCGA and constructed a prognostic CAF-related signature. The prediction abilities of the signature for survival prognosis, tumor immune landscape and responses to chemo-/immunotherapies were evaluated in the TCGA-SKCM cohort. RESULTS We suggested that CAFs were significantly involved in the clinical outcomes of SKCM. A 10-gene CAF-related model was constructed, and the high-CAF risk group exhibited immunosuppressive features and worse prognosis. Patients with high CAF score were more likely to not respond to immune checkpoint inhibitors but were more sensitive to some chemotherapeutic agents, suggesting a potential approach of chemotherapy/anti-CAF combination treatment to improve the SKCM patient response rate of current immunotherapies. CONCLUSIONS The CAF-related risk score could serve as a robust prognostic indicator and personal assessment of this score could uncover the degree of immunosuppression and provide treatment strategies to improve outcomes in clinical decision-making in SKCM patients.
Collapse
Affiliation(s)
- Xintao Cen
- Department of Dermatology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Mengna Li
- Department of Dermatology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Amin Yao
- Department of Dermatology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yue Zheng
- Department of Dermatology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Wei Lai
- Department of Dermatology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| |
Collapse
|
50
|
Oleynikova NA, Sycheva IN, Mikhailov IA, Zhestkov IA, Varenzov MG, Malkov PG. [Patterns of CAF expression in tumors of the genitourinary system]. Arkh Patol 2024; 86:28-35. [PMID: 39686894 DOI: 10.17116/patol20248606128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2024]
Abstract
Cancer-associated fibroblasts (CAF), as the most important component of the tumor microenvironment, have been the subject of targeted study in recent years. There is increasing evidence of CAF heterogeneity and their different roles in tumor progression. However, there is no single marker identifying CAF, and there is also no understanding of the similarity of CAF markers in various tumors of the genitourinary system. OBJECTIVE To evaluate the response of CAF markers - FAP and PDGFRα+β in bladder cancer and prostate cancer. MATERIAL AND METHODS An immunohistochemical study with antibodies to FAP and PDGFRα+β was conducted on material from 34 patients with prostate cancer and 44 patients with bladder cancer in order to identify statistical relationships with stage, morphological characteristics and survival, as well as to understand the applicability of the same markers for cancer of different localizations. RESULTS An increased FAP reaction was shown in prostate cancer with metastases (p=0.0239), a significantly higher number of recurrences in the group with a high PDGFRa+β (3+) reaction (p=0.0018); the presence of FAP reaction only in invasive bladder cancers (p=0.0094) and bladder cancers with high grade dysplasia (p=0.0000234), the absence of a relationship between the level of PDGFRa+β and clinical and morphological characteristics in bladder cancer. CONCLUSION Our results confirm the heterogeneity of CAF in both bladder and prostate tumors and indicate that FAP is one of CAF subpopulation marker. Considering that FAP is expressed in a population of CAFs that lead to resistance to treatment with immune checkpoint inhibitors in melanoma, the FAP marker may also be diagnostically significant for tumors of the genitourinary system, especially given the fact that bladder tumors are treated with the BCG vaccine.
Collapse
Affiliation(s)
| | - I N Sycheva
- Lomonosov Moscow State University, Moscow, Russia
| | - I A Mikhailov
- Lomonosov Moscow State University, Moscow, Russia
- Center of Expertise and Quality Control of Healthcare, Moscow, Russia
| | - I A Zhestkov
- Lomonosov Moscow State University, Moscow, Russia
| | - M G Varenzov
- Lomonosov Moscow State University, Moscow, Russia
| | - P G Malkov
- Lomonosov Moscow State University, Moscow, Russia
- Russian Medical Academy of Continuous Professional Education, Moscow, Russia
| |
Collapse
|