1
|
Guo Z, Zhu Z, Lin X, Wang S, Wen Y, Wang L, Zhi L, Zhou J. Tumor microenvironment and immunotherapy for triple-negative breast cancer. Biomark Res 2024; 12:166. [PMID: 39741315 DOI: 10.1186/s40364-024-00714-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 12/20/2024] [Indexed: 01/02/2025] Open
Abstract
Triple-negative breast cancer (TNBC) is a subtype of breast cancer known for its high aggressiveness and poor prognosis. Conventional treatment of TNBC is challenging due to its heterogeneity and lack of clear targets. Recent advancements in immunotherapy have shown promise in treating TNBC, with immune checkpoint therapy playing a significant role in comprehensive treatment plans. The tumor microenvironment (TME), comprising immune cells, stromal cells, and various cytokines, plays a crucial role in TNBC progression and response to immunotherapy. The high presence of tumor-infiltrating lymphocytes and immune checkpoint proteins in TNBC indicates the potential of immunotherapeutic strategies. However, the complexity of the TME, while offering therapeutic targets, requires further exploration of its multiple roles in immunotherapy. In this review, we discuss the interaction mechanism between TME and TNBC immunotherapy based on the characteristics and composition of TME, and elaborate on and analyze the effect of TME on immunotherapy, the potential of TME as an immune target, and the ability of TME as a biomarker. Understanding these dynamics will offer new insights for enhancing therapeutic approaches and investigating stratification and prognostic markers for TNBC patients.
Collapse
Affiliation(s)
- Zijie Guo
- Department of Surgical Oncology, Affiliated Sir Run Shaw Hospital, Zhejiang University School of Medicine, No.3 East Qingchun Road, Hangzhou, 310016, Zhejiang, China
- Biomedical Research Center, Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, 310016, Zhejiang, China
| | - Ziyu Zhu
- Department of Surgical Oncology, Affiliated Sir Run Shaw Hospital, Zhejiang University School of Medicine, No.3 East Qingchun Road, Hangzhou, 310016, Zhejiang, China
- Biomedical Research Center, Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, 310016, Zhejiang, China
| | - Xixi Lin
- Department of Surgical Oncology, Affiliated Sir Run Shaw Hospital, Zhejiang University School of Medicine, No.3 East Qingchun Road, Hangzhou, 310016, Zhejiang, China
- Biomedical Research Center, Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, 310016, Zhejiang, China
| | - Shenkangle Wang
- Department of Surgical Oncology, Affiliated Sir Run Shaw Hospital, Zhejiang University School of Medicine, No.3 East Qingchun Road, Hangzhou, 310016, Zhejiang, China
- Biomedical Research Center, Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, 310016, Zhejiang, China
| | - Yihong Wen
- Biomedical Research Center, Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, 310016, Zhejiang, China
| | - Linbo Wang
- Department of Surgical Oncology, Affiliated Sir Run Shaw Hospital, Zhejiang University School of Medicine, No.3 East Qingchun Road, Hangzhou, 310016, Zhejiang, China.
- Biomedical Research Center, Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, 310016, Zhejiang, China.
| | - Lili Zhi
- Biomedical Research Center, Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, 310016, Zhejiang, China.
| | - Jichun Zhou
- Department of Surgical Oncology, Affiliated Sir Run Shaw Hospital, Zhejiang University School of Medicine, No.3 East Qingchun Road, Hangzhou, 310016, Zhejiang, China.
- Biomedical Research Center, Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, 310016, Zhejiang, China.
| |
Collapse
|
2
|
Wu B, Zhang B, Li B, Wu H, Jiang M. Cold and hot tumors: from molecular mechanisms to targeted therapy. Signal Transduct Target Ther 2024; 9:274. [PMID: 39420203 PMCID: PMC11491057 DOI: 10.1038/s41392-024-01979-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 08/20/2024] [Accepted: 09/12/2024] [Indexed: 10/19/2024] Open
Abstract
Immunotherapy has made significant strides in cancer treatment, particularly through immune checkpoint blockade (ICB), which has shown notable clinical benefits across various tumor types. Despite the transformative impact of ICB treatment in cancer therapy, only a minority of patients exhibit a positive response to it. In patients with solid tumors, those who respond well to ICB treatment typically demonstrate an active immune profile referred to as the "hot" (immune-inflamed) phenotype. On the other hand, non-responsive patients may exhibit a distinct "cold" (immune-desert) phenotype, differing from the features of "hot" tumors. Additionally, there is a more nuanced "excluded" immune phenotype, positioned between the "cold" and "hot" categories, known as the immune "excluded" type. Effective differentiation between "cold" and "hot" tumors, and understanding tumor intrinsic factors, immune characteristics, TME, and external factors are critical for predicting tumor response and treatment results. It is widely accepted that ICB therapy exerts a more profound effect on "hot" tumors, with limited efficacy against "cold" or "altered" tumors, necessitating combinations with other therapeutic modalities to enhance immune cell infiltration into tumor tissue and convert "cold" or "altered" tumors into "hot" ones. Therefore, aligning with the traits of "cold" and "hot" tumors, this review systematically delineates the respective immune characteristics, influencing factors, and extensively discusses varied treatment approaches and drug targets based on "cold" and "hot" tumors to assess clinical efficacy.
Collapse
Affiliation(s)
- Bo Wu
- Department of Neurology, The Fourth Affiliated Hospital, China Medical University, Shenyang, China
| | - Bo Zhang
- Department of Youth League Committee, The Fourth Affiliated Hospital, China Medical University, Shenyang, China
| | - Bowen Li
- Department of Pancreatic and Gastrointestinal Surgery, Ningbo No. 2 Hospital, Ningbo, China
| | - Haoqi Wu
- Department of Gynaecology and Obstetrics, The Second Hospital of Dalian Medical University, Dalian, China
| | - Meixi Jiang
- Department of Neurology, The Fourth Affiliated Hospital, China Medical University, Shenyang, China.
| |
Collapse
|
3
|
Yu H, Peng Z, Li X, Zhang Y. Establishment of a tumor-associated fibroblast associated gene score based on scRNA-seq to predict prognosis in patients with triple-negative breast cancer. PLoS One 2024; 19:e0311801. [PMID: 39418248 PMCID: PMC11486389 DOI: 10.1371/journal.pone.0311801] [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: 02/06/2024] [Accepted: 09/24/2024] [Indexed: 10/19/2024] Open
Abstract
The tumor microenvironment (TME) is emerging as a tool for the development of improved patient prognosis and the development of novel antitumor drugs. As the most important stromal cells in the tumor microenvironment, cancer-associated fibroblasts (CAFs) play an important role in the development of TNBC. The rise of single-cell sequencing technology has facilitated our study of the various cell types in TME. In this study, we interpreted the heterogeneity of TNBCs from the perspective of tumor-associated fibroblasts in the tumor microenvironment based on the TNBC single-cell sequencing dataset GSE118389, in the hope of providing help for individualised treatment. Combining the TCGA database and the GSE103091 dataset, four genes associated with CAFs in TNBC (CERCAM, KLF10, ECM1,HGF) were identified using the R package Seurat as well as correlation consensus clustering analysis. Meanwhile, qRT-PCR, WB and IHC experiments confirmed their expression in TNBC. Based on these genes, CAFs Score was established and validated to correlate with the prognosis of patients with TNBC, with patients in the high score group surviving significantly worse than those in the low score group (P<0.001). In addition, there were significant differences in immune cell infiltration and expression of immune checkpoints between the high and low scoring groups. Compared to Stage I & II, the CAFs Score was higher in Stage III & IV TNBC patients (P = 0.043) and higher in N1-3 TNBC patients than in N0 TNBC patients (P = 0.035). EMT scores were higher within the high CAFs Score group (P = 1.4e-11) and there was a positive correlation between Stemness Score and CAFs Score (R = 0.61, P = 3.6e-09). Drug sensitivity analysis combining the GSE128099 showed a higher sensitivity to Gemcitabine in the low CAFs Score group (P = 0.0048). We speculate that these four CAFs-related genes are likely to be involved in regulating gemcitabine resistance in TNBC patients.
Collapse
Affiliation(s)
- Hao Yu
- Department of Breast Surgery, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
- Department of Breast Surgery, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Ziqi Peng
- Department of Breast Surgery, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Xing Li
- Department of Breast Surgery, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Yiqi Zhang
- Department of Breast Surgery, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| |
Collapse
|
4
|
Berdiaki A, Neagu M, Tzanakakis P, Spyridaki I, Pérez S, Nikitovic D. Extracellular Matrix Components and Mechanosensing Pathways in Health and Disease. Biomolecules 2024; 14:1186. [PMID: 39334952 PMCID: PMC11430160 DOI: 10.3390/biom14091186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 09/12/2024] [Accepted: 09/18/2024] [Indexed: 09/30/2024] Open
Abstract
Glycosaminoglycans (GAGs) and proteoglycans (PGs) are essential components of the extracellular matrix (ECM) with pivotal roles in cellular mechanosensing pathways. GAGs, such as heparan sulfate (HS) and chondroitin sulfate (CS), interact with various cell surface receptors, including integrins and receptor tyrosine kinases, to modulate cellular responses to mechanical stimuli. PGs, comprising a core protein with covalently attached GAG chains, serve as dynamic regulators of tissue mechanics and cell behavior, thereby playing a crucial role in maintaining tissue homeostasis. Dysregulation of GAG/PG-mediated mechanosensing pathways is implicated in numerous pathological conditions, including cancer and inflammation. Understanding the intricate mechanisms by which GAGs and PGs modulate cellular responses to mechanical forces holds promise for developing novel therapeutic strategies targeting mechanotransduction pathways in disease. This comprehensive overview underscores the importance of GAGs and PGs as key mediators of mechanosensing in maintaining tissue homeostasis and their potential as therapeutic targets for mitigating mechano-driven pathologies, focusing on cancer and inflammation.
Collapse
Affiliation(s)
- Aikaterini Berdiaki
- Department of Histology-Embryology, Medical School, University of Crete, 712 03 Heraklion, Greece; (A.B.); (P.T.); (I.S.)
| | - Monica Neagu
- Immunology Department, “Victor Babes” National Institute of Pathology, 050096 Bucharest, Romania;
| | - Petros Tzanakakis
- Department of Histology-Embryology, Medical School, University of Crete, 712 03 Heraklion, Greece; (A.B.); (P.T.); (I.S.)
| | - Ioanna Spyridaki
- Department of Histology-Embryology, Medical School, University of Crete, 712 03 Heraklion, Greece; (A.B.); (P.T.); (I.S.)
| | - Serge Pérez
- Centre de Recherche sur les Macromolécules Végétales (CERMAV), Centre National de la Recherche Scientifique (CNRS), University Grenoble Alpes, 38000 Grenoble, France;
| | - Dragana Nikitovic
- Department of Histology-Embryology, Medical School, University of Crete, 712 03 Heraklion, Greece; (A.B.); (P.T.); (I.S.)
| |
Collapse
|
5
|
Chen T, Wen Y, Song X, Zhang Z, Zhu J, Tian X, Zeng S, Li J. Rationally designed β-cyclodextrin-crosslinked polyacrylamide hydrogels for cell spheroid formation and 3D tumor model construction. Carbohydr Polym 2024; 339:122253. [PMID: 38823920 DOI: 10.1016/j.carbpol.2024.122253] [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: 05/08/2024] [Accepted: 05/09/2024] [Indexed: 06/03/2024]
Abstract
In vitro tumor models are essential for understanding tumor behavior and evaluating tumor biological properties. Hydrogels that can mimic the tumor extracellular matrix have become popular for creating 3D in vitro tumor models. However, designing biocompatible hydrogels with appropriate chemical and physical properties for constructing tumor models is still a challenge. In this study, we synthesized a series of β-cyclodextrin (β-CD)-crosslinked polyacrylamide hydrogels with different β-CD densities and mechanical properties and evaluated their potential for use in 3D in vitro tumor model construction, including cell capture and spheroid formation. By utilizing a combination of β-CD-methacrylate (CD-MA) and a small amount of N,N'-methylene bisacrylamide (BIS) as hydrogel crosslinkers and optimizing the CD-MA/BIS ratio, the hydrogels performed excellently for tumor cell 3D culture and spheroid formation. Notably, when we co-cultured L929 fibroblasts with HeLa tumor cells on the hydrogel surface, co-cultured spheroids were formed, showing that the hydrogel can mimic the complexity of the tumor extracellular matrix. This comprehensive investigation of the relationship between hydrogel mechanical properties and biocompatibility provides important insights for hydrogel-based in vitro tumor modeling and advances our understanding of the mechanisms underlying tumor growth and progression.
Collapse
Affiliation(s)
- Taili Chen
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan Province, China; Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, 15 Kent Ridge Crescent, Singapore 119276, Singapore
| | - Yuting Wen
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, 15 Kent Ridge Crescent, Singapore 119276, Singapore; National University of Singapore (Suzhou) Research Institute, Suzhou, Jiangsu 215000, China; National University of Singapore (Chongqing) Research Institute, Yubei District, Chongqing 401120, China.
| | - Xia Song
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, 15 Kent Ridge Crescent, Singapore 119276, Singapore
| | - Zhongxing Zhang
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, 15 Kent Ridge Crescent, Singapore 119276, Singapore
| | - Jingling Zhu
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, 15 Kent Ridge Crescent, Singapore 119276, Singapore; NUS Environmental Research Institute (NERI), National University of Singapore, 5A Engineering Drive 1, Singapore 117411, Singapore
| | - Xuehao Tian
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, 15 Kent Ridge Crescent, Singapore 119276, Singapore
| | - Shan Zeng
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan Province, China.
| | - Jun Li
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, 15 Kent Ridge Crescent, Singapore 119276, Singapore; National University of Singapore (Suzhou) Research Institute, Suzhou, Jiangsu 215000, China; National University of Singapore (Chongqing) Research Institute, Yubei District, Chongqing 401120, China; NUS Environmental Research Institute (NERI), National University of Singapore, 5A Engineering Drive 1, Singapore 117411, Singapore.
| |
Collapse
|
6
|
Miao S, Rodriguez BL, Gibbons DL. The Multifaceted Role of Neutrophils in NSCLC in the Era of Immune Checkpoint Inhibitors. Cancers (Basel) 2024; 16:2507. [PMID: 39061147 PMCID: PMC11274601 DOI: 10.3390/cancers16142507] [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: 06/05/2024] [Revised: 06/29/2024] [Accepted: 07/02/2024] [Indexed: 07/28/2024] Open
Abstract
Lung cancer is the most common cause of cancer-related death in both males and females in the U.S. and non-small-cell lung cancer (NSCLC) accounts for 85%. Although the use of first- or second-line immune checkpoint inhibitors (ICIs) exhibits remarkable clinical benefits, resistance to ICIs develops over time and dampens the efficacy of ICIs in patients. Tumor-associated neutrophils (TANs) have an important role in modulating the tumor microenvironment (TME) and tumor immune response. The major challenge in the field is to characterize the TANs in NSCLC TME and understand the link between TAN-related immunosuppression with ICI treatment response. In this review, we summarize the current studies of neutrophil interaction with malignant cells, T-cells, and other components in the TME. Ongoing clinical trials are aimed at utilizing reagents that have putative effects on tumor-associated neutrophils, in combination with ICI. Elevated neutrophil populations and neutrophil-associated factors could be potential therapeutic targets to enhance anti-PD1 treatment in NSCLC.
Collapse
Affiliation(s)
- Shucheng Miao
- Department of Thoracic Head & Neck Medical Oncology, MD Anderson Cancer Center, Houston, TX 77030, USA; (S.M.); (B.L.R.)
- The University of Texas MD Anderson Cancer Center, UTHealth at Houston Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| | - Bertha Leticia Rodriguez
- Department of Thoracic Head & Neck Medical Oncology, MD Anderson Cancer Center, Houston, TX 77030, USA; (S.M.); (B.L.R.)
| | - Don L. Gibbons
- Department of Thoracic Head & Neck Medical Oncology, MD Anderson Cancer Center, Houston, TX 77030, USA; (S.M.); (B.L.R.)
- The University of Texas MD Anderson Cancer Center, UTHealth at Houston Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| |
Collapse
|
7
|
Whately KM, Sengottuvel N, Edatt L, Srivastava S, Woods AT, Tsai YS, Porrello A, Zimmerman MP, Chack AC, Jefferys SR, Yacovone G, Kim DJ, Dudley AC, Amelio AL, Pecot CV. Spon1+ inflammatory monocytes promote collagen remodeling and lung cancer metastasis through lipoprotein receptor 8 signaling. JCI Insight 2024; 9:e168792. [PMID: 38716730 PMCID: PMC11141919 DOI: 10.1172/jci.insight.168792] [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: 01/12/2023] [Accepted: 03/21/2024] [Indexed: 05/12/2024] Open
Abstract
Lung cancer is the leading cause of cancer-related deaths in the world, and non-small cell lung cancer (NSCLC) is the most common subset. We previously found that infiltration of tumor inflammatory monocytes (TIMs) into lung squamous carcinoma (LUSC) tumors is associated with increased metastases and poor survival. To further understand how TIMs promote metastases, we compared RNA-Seq profiles of TIMs from several LUSC metastatic models with inflammatory monocytes (IMs) of non-tumor-bearing controls. We identified Spon1 as upregulated in TIMs and found that Spon1 expression in LUSC tumors corresponded with poor survival and enrichment of collagen extracellular matrix signatures. We observed SPON1+ TIMs mediate their effects directly through LRP8 on NSCLC cells, which resulted in TGF-β1 activation and robust production of fibrillar collagens. Using several orthogonal approaches, we demonstrated that SPON1+ TIMs were sufficient to promote NSCLC metastases. Additionally, we found that Spon1 loss in the host, or Lrp8 loss in cancer cells, resulted in a significant decrease of both high-density collagen matrices and metastases. Finally, we confirmed the relevance of the SPON1/LRP8/TGF-β1 axis with collagen production and survival in patients with NSCLC. Taken together, our study describes how SPON1+ TIMs promote collagen remodeling and NSCLC metastases through an LRP8/TGF-β1 signaling axis.
Collapse
Affiliation(s)
| | - Nisitha Sengottuvel
- UNC Lineberger Comprehensive Cancer Center and
- Department of Genetics and Molecular Biology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Lincy Edatt
- UNC Lineberger Comprehensive Cancer Center and
| | - Sonal Srivastava
- Department of Tumor Microenvironment and Metastasis, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - Allison T. Woods
- UNC Lineberger Comprehensive Cancer Center and
- Department of Cell Biology and Physiology and
| | - Yihsuan S. Tsai
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | | | - Matthew P. Zimmerman
- UNC Lineberger Comprehensive Cancer Center and
- Department of Cell Biology and Physiology and
| | - Aaron C. Chack
- UNC Lineberger Comprehensive Cancer Center and
- Department of Genetics and Molecular Biology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | | | | | - Dae Joong Kim
- Department of Microbiology, Immunology, and Cancer Biology and
| | - Andrew C. Dudley
- Department of Microbiology, Immunology, and Cancer Biology and
- UVA Comprehensive Cancer Center, The University of Virginia, Charlottesville, Virginia, USA
| | - Antonio L. Amelio
- Department of Tumor Microenvironment and Metastasis, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
- Department of Head and Neck-Endocrine Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - Chad V. Pecot
- UNC Lineberger Comprehensive Cancer Center and
- Division of Oncology and
- RNA Discovery Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
8
|
Guo S, Chen P, Yang Y, Wei W, Pan Y, Zeng F, Fan L, Wang W. Tumor-to-stroma cd8 + t cells ratio combined with cancer-associated fibroblasts: an innovative approach to predicting lymph node metastases of cervical cancer. J Cancer Res Clin Oncol 2024; 150:93. [PMID: 38369672 PMCID: PMC10874907 DOI: 10.1007/s00432-023-05578-1] [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: 10/08/2023] [Accepted: 12/02/2023] [Indexed: 02/20/2024]
Abstract
PURPOSE Precise identification of lymph node metastases is vital for the management of cervical cancer. However, the existing diagnostic methods for lymph node metastases have certain drawbacks. In this study, we aim to explore the expression of cancer-associated fibroblasts (CAFs) and tumor-to-stroma CD8+ T cells ratio (CD8+ T cells T:S ratio) and its association with lymph node metastases of cervical cancer. METHODS Hundred and ten cervical cancer tissues and 39 biopsy tissues from patients were investigated immunocytochemically for the expression of CAFs and CD8+ T cells. The statistical correlation analysis was carried out using the SPSS system. RESULTS A strong and statistically significant negative correlation (r= - 0.690; P < 0.001) was observed between CAF density and CD8+ T cells T:S ratio. Not only were CAFs density and CD8+ T cells T:S ratio correlated with lymph node metastases respectively (P < 0.001), but the combination of them also significantly correlated with lymph node metastases (P < 0.001). Then, we constructed the combined diagnosis model (Logit (P) = - 4.446 + 0.300 × CAFs + 0.752 × CD8+ T cells T:S Ratio) of cervical cancer lymph node metastases. ROC curves analysis showed that the ROC curves areas for CAFs, CD8+ T cells T:S ratio, and a combination of both are 0.879, 0.747, and 0.951. Then, the prediction model was verified by biopsy specimens and consistent results were obtained. CONCLUSIONS The combination of CAF density and CD8+ T cells T:S ratio has a significant predictive value for lymph node metastases in patients with cervical cancer.
Collapse
Affiliation(s)
- Shuangshuang Guo
- Department of Obstetrics and Gynecology, Guangzhou Medical University, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong, China
| | - Peiyu Chen
- Department of Obstetrics and Gynecology, Guangzhou Medical University, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong, China
| | - Yang Yang
- The Six Affiliated Hospital, Guangzhou Medical University, Qingyuan, 511518, Guangdong, China
| | - Wenfei Wei
- Department of Gynecology, Zhuhai People's Hospital (Zhuhai Hospital Affiliated With Jinan University), Zhuhai, Guangdong, China
| | - YuHua Pan
- Department of Obstetrics and Gynecology, Guangzhou Medical University, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong, China
| | - Fanke Zeng
- Department of Obstetrics and Gynecology, Guangzhou Medical University, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong, China
| | - Liangsheng Fan
- Department of Obstetrics and Gynecology, Guangzhou Medical University, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong, China.
| | - Wei Wang
- Department of Obstetrics and Gynecology, Guangzhou Medical University, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong, China.
| |
Collapse
|
9
|
Shen Y, Zou Y, Bie B, Lv Y. Hierarchically Released Liquid Metal Nanoparticles for Mild Photothermal Therapy/Chemotherapy of Breast Cancer Bone Metastases via Remodeling Tumor Stromal Microenvironment. Adv Healthc Mater 2023; 12:e2301080. [PMID: 37436138 DOI: 10.1002/adhm.202301080] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 07/08/2023] [Accepted: 07/10/2023] [Indexed: 07/13/2023]
Abstract
Currently, the treatment strategy for bone metastasis is mainly to inhibit the growth of tumor cells and the activity of osteoclasts, while ignoring the influence of the tumor stromal microenvironment (TSM) on the progression of bone metastasis. Herein, a dual-target liquid metal (LM)-based drug delivery system (DDS) with favorable photothermal performance is designed to spatially program the delivery of multiple therapeutic agents to enhance the treatment of bone metastasis through TSM remodeling. Briefly, mesoporous silicon-coated LM is integrated into zeolitic imidazolate framework-8 (ZIF-8) with both bone-seeking and tumor-targeting capacities. Curcumin (Cur), a tumor microenvironment modulator, is encapsulated into ZIF-8, and doxorubicin (DOX) is enclosed inside mesoporous silicon. Specific accumulation of the LM-based DDS in bone metastases first relieves the tumor stroma by releasing Cur in response to the acidic tumor microenvironment and then releases DOX deep into the tumor under near-infrared light irradiation. The combined strategy of the LM-based DDS and mild photothermal therapy has been shown to effectively restrain cross-talk between osteoclasts and tumor cells by inhibiting the secretion of transforming growth factor-β, degrading extracellular matrix components, and increasing infiltration of CD4+ and CD8+ T cells, which provides a promising strategy for the treatment of bone metastases.
Collapse
Affiliation(s)
- Yaping Shen
- Mechanobiology and Regenerative Medicine Laboratory, Bioengineering College, Chongqing University, 174 Shazheng Street, Shapingba District, Chongqing, 400044, China
| | - Yang Zou
- State Key Laboratory of New Textile Materials and Advanced Processing Technologies, School of Environmental Engineering, Wuhan Textile University, No. 1 Sunshine Avenue, Jiangxia District, Wuhan, Hubei Province, 430200, China
| | - Binglin Bie
- State Key Laboratory of New Textile Materials and Advanced Processing Technologies, Wuhan Textile University, No. 1 Sunshine Avenue, Jiangxia District, Wuhan, Hubei Province, 430200, China
| | - Yonggang Lv
- State Key Laboratory of New Textile Materials and Advanced Processing Technologies, Wuhan Textile University, No. 1 Sunshine Avenue, Jiangxia District, Wuhan, Hubei Province, 430200, China
| |
Collapse
|
10
|
Liu Y, Hu Y, Xue J, Li J, Yi J, Bu J, Zhang Z, Qiu P, Gu X. Advances in immunotherapy for triple-negative breast cancer. Mol Cancer 2023; 22:145. [PMID: 37660039 PMCID: PMC10474743 DOI: 10.1186/s12943-023-01850-7] [Citation(s) in RCA: 80] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Accepted: 08/26/2023] [Indexed: 09/04/2023] Open
Abstract
BACKGROUND Immunotherapy has recently emerged as a treatment strategy which stimulates the human immune system to kill tumor cells. Tumor immunotherapy is based on immune editing, which enhances the antigenicity of tumor cells and increases the tumoricidal effect of immune cells. It also suppresses immunosuppressive molecules, activates or restores immune system function, enhances anti-tumor immune responses, and inhibits the growth f tumor cell. This offers the possibility of reducing mortality in triple-negative breast cancer (TNBC). MAIN BODY Immunotherapy approaches for TNBC have been diversified in recent years, with breakthroughs in the treatment of this entity. Research on immune checkpoint inhibitors (ICIs) has made it possible to identify different molecular subtypes and formulate individualized immunotherapy schedules. This review highlights the unique tumor microenvironment of TNBC and integrates and analyzes the advances in ICI therapy. It also discusses strategies for the combination of ICIs with chemotherapy, radiation therapy, targeted therapy, and emerging treatment methods such as nanotechnology, ribonucleic acid vaccines, and gene therapy. Currently, numerous ongoing or completed clinical trials are exploring the utilization of immunotherapy in conjunction with existing treatment modalities for TNBC. The objective of these investigations is to assess the effectiveness of various combined immunotherapy approaches and determine the most effective treatment regimens for patients with TNBC. CONCLUSION This review provides insights into the approaches used to overcome drug resistance in immunotherapy, and explores the directions of immunotherapy development in the treatment of TNBC.
Collapse
Affiliation(s)
- Yang Liu
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning Province, China
| | - Yueting Hu
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning Province, China
| | - Jinqi Xue
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning Province, China
| | - Jingying Li
- Department of Health Management, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning Province, China
| | - Jiang Yi
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning Province, China
| | - Jiawen Bu
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning Province, China
| | - Zhenyong Zhang
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning Province, China.
| | - Peng Qiu
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning Province, China.
| | - Xi Gu
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning Province, China.
| |
Collapse
|
11
|
Zhao Z, Mak TK, Shi Y, Li K, Huo M, Zhang C. Integrative analysis of cancer-associated fibroblast signature in gastric cancer. Heliyon 2023; 9:e19217. [PMID: 37809716 PMCID: PMC10558323 DOI: 10.1016/j.heliyon.2023.e19217] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 08/11/2023] [Accepted: 08/16/2023] [Indexed: 10/10/2023] Open
Abstract
Background CAFs regulate the signaling of GC cells by promoting their migration, invasion, and proliferation and the function of immune cells as well as their location and migration in the TME by remodeling the extracellular matrix (ECM). This study explored the understanding of the heterogeneity of CAFs in TME and laid the groundwork for GC biomarker and precision treatment development. Methods The scRNA-seq and bulk RNA-seq datasets were obtained from GEO and TCGA. The prognostic significance of various CAFs subtypes was investigated using ssGSEA combined with Kaplan-Meier analysis. POSTN expression in GC tissues and CAFs was detected using immunohistochemistry, immunofluorescence, and Western blotting. Differential expression analysis identified the differentially expressed genes (DEGs) between normal and tumor samples in TCGA-STAD. Pearson correlation analysis identified DEGs associated with adverse prognosis CAF subtype, and univariate Cox regression analysis determined prognostic genes associated with CAFs. LASSO regression analysis and Multivariate Cox regression were used to build a prognosis model for CAFs. Results We identified five CAFs subtypes in GC, with the CAF_0 subtype associated with poor prognosis. The abundance of CAF_0 correlated with T stage, clinical stage, histological type, and immune cell infiltration levels. Periostin (POSTN) exhibited increased expression in both GC tissues and CAFs and was linked to poor prognosis in GC patients. Through LASSO and multivariate Cox regression analysis, three genes (CXCR4, MATN3, and KIF24) were selected to create the CAFs-score. We developed a nomogram to facilitate the clinical application of the CAFs-score. Notably, the CAFs signature showed significant correlations with immune cells, stromal components, and immunological scores, suggesting its pivotal role in the tumor microenvironment (TME). Furthermore, CAFs-score demonstrated prognostic value in assessing immunotherapy outcomes, highlighting its potential as a valuable biomarker to guide therapeutic decisions. Conclusion CAF_0 subtype in TME is the cause of poor prognosis in GC patients. Furthermore, CAFs-score constructed from the CAF_0 subtype can be used to determine the clinical prognosis, immune infiltration, clinicopathological characteristics, and assessment of personalized treatment of GC patients.
Collapse
Affiliation(s)
- Zidan Zhao
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Tsz Kin Mak
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Yuntao Shi
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Kuan Li
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Mingyu Huo
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Changhua Zhang
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China
| |
Collapse
|
12
|
Park J, Kim JS, Yang G, Lee H, Shim G, Lee J, Oh YK. Lysyl oxidase-responsive anchoring nanoparticles for modulation of the tumor immune microenvironment. J Control Release 2023; 360:376-391. [PMID: 37406820 DOI: 10.1016/j.jconrel.2023.06.041] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 05/30/2023] [Accepted: 06/30/2023] [Indexed: 07/07/2023]
Abstract
In the tumor microenvironment, lysyl oxidase (LOX) is known to play a key role in stabilizing the tumor extracellular matrix. Here, we designed LOX-responsive nanoparticles to interact with the collagen matrix of the tumor microenvironment. Collagen-coated and imiquimod-loaded polydopamine nanoparticles (CPN/IQ) could form crosslinked structures with the collagen matrix via LOX. In vitro, anchoring of CPN/IQ nanoparticles was observed with LOX-secreting CT26 cells, but this was blocked by a LOX inhibitor. In CT26 tumor-bearing mice, co-administration of nanoparticles plus the LOX inhibitor did not significantly alter the antitumor efficacy among nanoparticles. In the absence of the LOX inhibitor, however, a single administration of CPN/IQ could provide sustained responsiveness to near-infrared irradiation and ablation of primary tumors. In the primary tumor microenvironment, CPN/IQ lowered the Treg cell population but increased the cytotoxic CD3+CD8+ T cell population. In splenic dendritic cells, CPN/IQ treatment significantly increased the CD11c+CD86+ and CD11c+CD80+ cell populations. In a CT26 distant tumor-rechallenge model, CPN/IQ treatment increased the cytotoxic CD3+CD8+ T cell population and provided 100% survival of mice until 64 days. This study indicates the feasibility of tumor immune microenvironment modulation using LOX-responsive size-transforming nanoparticles. Although we tested the concept in a CT26 cell-derived tumor model, the concept of LOX-responsive collagen matrix- anchoring nanoparticles may be broadly applied to other tumor tissues with LOX-rich tumor microenvironments.
Collapse
Affiliation(s)
- Jinwon Park
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Republic of Korea
| | - Jung Suk Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Republic of Korea
| | - Geon Yang
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Republic of Korea
| | - Hobin Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Republic of Korea
| | - Gayong Shim
- School of Systems Biomedical Science, Soongsil University, Seoul 06978, Republic of Korea.
| | - Jaiwoo Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Republic of Korea.
| | - Yu-Kyoung Oh
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Republic of Korea.
| |
Collapse
|
13
|
Prieto-Fernández L, Montoro-Jiménez I, de Luxan-Delgado B, Otero-Rosales M, Rodrigo JP, Calvo F, García-Pedrero JM, Álvarez-Teijeiro S. Dissecting the functions of cancer-associated fibroblasts to therapeutically target head and neck cancer microenvironment. Biomed Pharmacother 2023; 161:114502. [PMID: 37002578 DOI: 10.1016/j.biopha.2023.114502] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 02/22/2023] [Accepted: 03/07/2023] [Indexed: 03/15/2023] Open
Abstract
Head and neck cancers (HNC) are a diverse group of aggressive malignancies with high morbidity and mortality, leading to almost half-million deaths annually worldwide. A better understanding of the molecular processes governing tumor formation and progression is crucial to improve current diagnostic and prognostic tools as well as to develop more personalized treatment strategies. Tumors are highly complex and heterogeneous structures in which growth and dissemination is not only governed by the cancer cells intrinsic mechanisms, but also by the surrounding tumor microenvironment (TME). Cancer-associated fibroblasts (CAFs) emerge as predominant TME components and key players in the generation of permissive conditions that ultimately impact in tumor progression and metastatic dissemination. Although CAFs were initially considered a consequence of tumor development, it is now well established that they actively contribute to numerous cancer hallmarks i.e., tumor cell growth, migration and invasion, cancer cell stemness, angiogenesis, metabolic reprograming, inflammation, and immune system modulation. In this scenario, therapeutic strategies targeting CAF functions could potentially have a major impact in cancer therapeutics, providing avenues for new treatment options or for improving efficacy in established approaches. This review is focused on thoroughly dissecting existing evidences supporting the contribution of CAFs in HNC biology with an emphasis on current knowledge of the key molecules and pathways involved in CAF-tumor crosstalk, and their potential as novel biomarkers and/or therapeutic targets to effectively interfere the tumor-stroma crosstalk for HNC patients benefit. involved in CAF-tumor crosstalk, and their potential as novel biomarkers and/or therapeutic targets to effec- tively interfere the tumor-stroma crosstalk for HNC patients benefit.
Collapse
Affiliation(s)
- Llara Prieto-Fernández
- Department of Otolaryngology, Hospital Universitario Central de Asturias and Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), University of Oviedo, Oviedo, Spain; Instituto Universitario de Oncología del Principado de Asturias (IUOPA), University of Oviedo, Oviedo, Spain; CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
| | - Irene Montoro-Jiménez
- Department of Otolaryngology, Hospital Universitario Central de Asturias and Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), University of Oviedo, Oviedo, Spain; Instituto Universitario de Oncología del Principado de Asturias (IUOPA), University of Oviedo, Oviedo, Spain; CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
| | - Beatriz de Luxan-Delgado
- Department of Otolaryngology, Hospital Universitario Central de Asturias and Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), University of Oviedo, Oviedo, Spain; Instituto Universitario de Oncología del Principado de Asturias (IUOPA), University of Oviedo, Oviedo, Spain
| | - María Otero-Rosales
- Department of Otolaryngology, Hospital Universitario Central de Asturias and Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), University of Oviedo, Oviedo, Spain; Instituto Universitario de Oncología del Principado de Asturias (IUOPA), University of Oviedo, Oviedo, Spain
| | - Juan P Rodrigo
- Department of Otolaryngology, Hospital Universitario Central de Asturias and Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), University of Oviedo, Oviedo, Spain; Instituto Universitario de Oncología del Principado de Asturias (IUOPA), University of Oviedo, Oviedo, Spain; CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
| | - Fernando Calvo
- Instituto de Biomedicina y Biotecnología de Cantabria (Consejo Superior de Investigaciones Científicas, Universidad de Cantabria), Santander, Spain
| | - Juana M García-Pedrero
- Department of Otolaryngology, Hospital Universitario Central de Asturias and Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), University of Oviedo, Oviedo, Spain; Instituto Universitario de Oncología del Principado de Asturias (IUOPA), University of Oviedo, Oviedo, Spain; CIBERONC, Instituto de Salud Carlos III, Madrid, Spain.
| | - Saúl Álvarez-Teijeiro
- Department of Otolaryngology, Hospital Universitario Central de Asturias and Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), University of Oviedo, Oviedo, Spain; Instituto Universitario de Oncología del Principado de Asturias (IUOPA), University of Oviedo, Oviedo, Spain; CIBERONC, Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
14
|
Lin D, Shen Y, Liang T. Oncolytic virotherapy: basic principles, recent advances and future directions. Signal Transduct Target Ther 2023; 8:156. [PMID: 37041165 PMCID: PMC10090134 DOI: 10.1038/s41392-023-01407-6] [Citation(s) in RCA: 121] [Impact Index Per Article: 60.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 03/05/2023] [Accepted: 03/14/2023] [Indexed: 04/13/2023] Open
Abstract
Oncolytic viruses (OVs) have attracted growing awareness in the twenty-first century, as they are generally considered to have direct oncolysis and cancer immune effects. With the progress in genetic engineering technology, OVs have been adopted as versatile platforms for developing novel antitumor strategies, used alone or in combination with other therapies. Recent studies have yielded eye-catching results that delineate the promising clinical outcomes that OVs would bring about in the future. In this review, we summarized the basic principles of OVs in terms of their classifications, as well as the recent advances in OV-modification strategies based on their characteristics, biofunctions, and cancer hallmarks. Candidate OVs are expected to be designed as "qualified soldiers" first by improving target fidelity and safety, and then equipped with "cold weapons" for a proper cytocidal effect, "hot weapons" capable of activating cancer immunotherapy, or "auxiliary weapons" by harnessing tactics such as anti-angiogenesis, reversed metabolic reprogramming and decomposing extracellular matrix around tumors. Combinations with other cancer therapeutic agents have also been elaborated to show encouraging antitumor effects. Robust results from clinical trials using OV as a treatment congruously suggested its significance in future application directions and challenges in developing OVs as novel weapons for tactical decisions in cancer treatment.
Collapse
Affiliation(s)
- Danni Lin
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou, Zhejiang, China
- The Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Yinan Shen
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou, Zhejiang, China
- The Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Tingbo Liang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
- Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou, Zhejiang, China.
- The Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, Zhejiang, China.
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, China.
| |
Collapse
|
15
|
Feng D, Xiong Q, Wei Q, Yang L. Cellular landscape of tumour microenvironment in prostate cancer. Immunology 2023; 168:199-202. [PMID: 35146758 DOI: 10.1111/imm.13456] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 02/03/2022] [Indexed: 02/05/2023] Open
Abstract
Prostate cancer is still a public health priority in men and the impact of this disease will be more pronounced with the ageing of the world's population. Clinical heterogeneity of prostate cancer is reflected in spatial and clonal genomic diversity. Accumulating evidence demonstrates that the malignant behaviour of cancer is not only attributed to cancer cells but also fundamentally affected by stromal activity and controlled by various mechanisms of the tumour microenvironment. Data on prostate cancer in this study was derived from seven GEO datasets and the TCGA database. We analyzed the tumour microenvironment of prostate cancer in terms of clinical process, T stage and Gleason score using EPIC and xCell algorithms. We also analyzed the common immune checkpoints. In this study, we confirmed remarkable tumour tissue remodelling in the development of prostate cancer and further demonstrated the importance of cancer-related fibroblasts in the biochemical recurrence and metastasis for patients with prostate cancer undergoing radical radiotherapy or prostatectomy. In addition, we found that NRP1, CD200, TNFSF18 and CD80 might be the potential targets for prostate cancer.
Collapse
Affiliation(s)
- Dechao Feng
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Qiao Xiong
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Qiang Wei
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Lu Yang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
16
|
Xu M, Zhang T, Xia R, Wei Y, Wei X. Targeting the tumor stroma for cancer therapy. Mol Cancer 2022; 21:208. [PMID: 36324128 PMCID: PMC9628074 DOI: 10.1186/s12943-022-01670-1] [Citation(s) in RCA: 128] [Impact Index Per Article: 42.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 10/13/2022] [Accepted: 10/13/2022] [Indexed: 11/06/2022] Open
Abstract
Tumors are comprised of both cancer cells and surrounding stromal components. As an essential part of the tumor microenvironment, the tumor stroma is highly dynamic, heterogeneous and commonly tumor-type specific, and it mainly includes noncellular compositions such as the extracellular matrix and the unique cancer-associated vascular system as well as a wide variety of cellular components including activated cancer-associated fibroblasts, mesenchymal stromal cells, pericytes. All these elements operate with each other in a coordinated fashion and collectively promote cancer initiation, progression, metastasis and therapeutic resistance. Over the past few decades, numerous studies have been conducted to study the interaction and crosstalk between stromal components and neoplastic cells. Meanwhile, we have also witnessed an exponential increase in the investigation and recognition of the critical roles of tumor stroma in solid tumors. A series of clinical trials targeting the tumor stroma have been launched continually. In this review, we introduce and discuss current advances in the understanding of various stromal elements and their roles in cancers. We also elaborate on potential novel approaches for tumor-stroma-based therapeutic targeting, with the aim to promote the leap from bench to bedside.
Collapse
Affiliation(s)
- Maosen Xu
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, West China Hospital, National Clinical Research Center for Geriatrics, Sichuan University, No. 17, Block 3, Southern Renmin Road, 610041, Chengdu, Sichuan, PR China
| | - Tao Zhang
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, West China Hospital, National Clinical Research Center for Geriatrics, Sichuan University, No. 17, Block 3, Southern Renmin Road, 610041, Chengdu, Sichuan, PR China
| | - Ruolan Xia
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, West China Hospital, National Clinical Research Center for Geriatrics, Sichuan University, No. 17, Block 3, Southern Renmin Road, 610041, Chengdu, Sichuan, PR China
| | - Yuquan Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, West China Hospital, National Clinical Research Center for Geriatrics, Sichuan University, No. 17, Block 3, Southern Renmin Road, 610041, Chengdu, Sichuan, PR China
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, West China Hospital, National Clinical Research Center for Geriatrics, Sichuan University, No. 17, Block 3, Southern Renmin Road, 610041, Chengdu, Sichuan, PR China.
| |
Collapse
|
17
|
Berndt A, Gaßler N, Franz M. Invasion-Associated Reorganization of Laminin 332 in Oral Squamous Cell Carcinomas: The Role of the Laminin γ2 Chain in Tumor Biology, Diagnosis, and Therapy. Cancers (Basel) 2022; 14:cancers14194903. [PMID: 36230826 PMCID: PMC9564360 DOI: 10.3390/cancers14194903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 09/30/2022] [Accepted: 10/03/2022] [Indexed: 11/23/2022] Open
Abstract
Simple Summary The destructive growth of carcinomas is associated with crossing the border between the epithelial and the connective tissue parts of an organ. One component of this borderline, the basement membrane, is the heterotrimeric laminin 332, which mediates the adhesion of basal epithelial cells. This protein, in particular its gamma 2 chain, is fundamentally reorganized during tumor cell invasion. Specific deposition patterns of laminin 332 are also present in oral squamous cell carcinomas and have been shown to be of high diagnostic and predictive value. Furthermore, laminin 332 restructuring is associated with important tumor biological processes, e.g., stromal activation, the development of a motile phenotype, and tumor spreading. In this review, current knowledge in the field is summarized and the recommendation to consider laminin 332 as a promising grading and monitoring parameter and as a potential therapeutic target is discussed. Abstract Invasion of the connective tissue by carcinoma cells is accompanied by disintegration and reorganization of the hemidesmosomes, which connect the basement membrane to the basal epithelial cells. In terms of mediating the basement membrane, i.e., basal cell interactions, the heterotrimeric laminin 332 is the most important bridging molecule. Due to this distinct function, laminin 332, especially its gamma 2 chain, came into the focus of cancer research. Specific de novo synthesis and deposition patterns of laminin 332 are evident upon development and progression of oral squamous cell carcinomas (OSCCs). Loss from the basement membrane, cytoplasmic accumulation, and extracellular deposition are associated with crucial processes such as stromal activation and immune response, epithelial to mesenchymal transition, and tumor cell budding. In networks with components of the tumor microenvironment, altered expression of laminin 332 chains, proteolytic processing, and interaction with integrin receptors seem to promote cancer cell migration. Indeed, reorganization patterns are shown to have a high diagnostic and prognostic value. Here, we summarize the current knowledge on laminin 332 reorganization in OSCCs with special focus on its gamma 2 chain and provide, based on the current literature, evidence on its promising role as a grading and monitoring parameter and as a potential therapeutic target.
Collapse
Affiliation(s)
- Alexander Berndt
- Section Pathology, Institute of Legal Medicine, University Hospital Jena, 07747 Jena, Germany
- Correspondence: ; Tel.: +49-3641-939-70-61
| | - Nikolaus Gaßler
- Section Pathology, Institute of Legal Medicine, University Hospital Jena, 07747 Jena, Germany
| | - Marcus Franz
- Department of Internal Medicine I, University Hospital Jena, 07747 Jena, Germany
| |
Collapse
|
18
|
Vasudevan J, Jiang K, Fernandez J, Lim CT. Extracellular matrix mechanobiology in cancer cell migration. Acta Biomater 2022; 163:351-364. [PMID: 36243367 DOI: 10.1016/j.actbio.2022.10.016] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 09/11/2022] [Accepted: 10/06/2022] [Indexed: 11/01/2022]
Abstract
The extracellular matrix (ECM) is pivotal in modulating tumor progression. Besides chemically stimulating tumor cells, it also offers physical support that orchestrates the sequence of events in the metastatic cascade upon dynamically modulating cell mechanosensation. Understanding this translation between matrix biophysical cues and intracellular signaling has led to rapid growth in the interdisciplinary field of cancer mechanobiology in the last decade. Substantial efforts have been made to develop novel in vitro tumor mimicking platforms to visualize and quantify the mechanical forces within the tissue that dictate tumor cell invasion and metastatic growth. This review highlights recent findings on tumor matrix biophysical cues such as fibrillar arrangement, crosslinking density, confinement, rigidity, topography, and non-linear mechanics and their implications on tumor cell behavior. We also emphasize how perturbations in these cues alter cellular mechanisms of mechanotransduction, consequently enhancing malignancy. Finally, we elucidate engineering techniques to individually emulate the mechanical properties of tumors that could help serve as toolkits for developing and testing ECM-targeted therapeutics on novel bioengineered tumor platforms. STATEMENT OF SIGNIFICANCE: Disrupted ECM mechanics is a driving force for transitioning incipient cells to life-threatening malignant variants. Understanding these ECM changes can be crucial as they may aid in developing several efficacious drugs that not only focus on inducing cytotoxic effects but also target specific matrix mechanical cues that support and enhance tumor invasiveness. Designing and implementing an optimal tumor mimic can allow us to predictively map biophysical cue-modulated cell behaviors and facilitate the design of improved lab-grown tumor models with accurately controlled structural features. This review focuses on the abnormal changes within the ECM during tumorigenesis and its implications on tumor cell-matrix mechanoreciprocity. Additionally, it accentuates engineering approaches to produce ECM features of varying levels of complexity which is critical for improving the efficiency of current engineered tumor tissue models.
Collapse
|
19
|
Parayath NN, Gandham SK, Amiji MM. Tumor-targeted miRNA nanomedicine for overcoming challenges in immunity and therapeutic resistance. Nanomedicine (Lond) 2022; 17:1355-1373. [PMID: 36255330 PMCID: PMC9706370 DOI: 10.2217/nnm-2022-0130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
miRNA are critical messengers in the tumor microenvironment (TME) that influence various processes leading to immune suppression, tumor progression, metastasis and resistance. Strategies to modulate miRNAs in the TME have important implications in overcoming these challenges. However, miR delivery to specific cells in the TME has been challenging. This review discusses nanomedicine strategies to achieve cell-specific delivery of miRNAs. The key goal of delivery is to activate the tumor immune landscape as well as to prevent chemotherapy resistance. Specifically, the use of hyaluronic acid-based nanoparticle miRNA delivery to the TME is discussed. The discussion is focused on miRNA-125b for reprogramming tumor-associated macrophages to overcome immunosuppression and miRNA-let-7b to overcome resistance to anticancer chemotherapeutics because both these miRNAs have been extensively evaluated for delivery with hyaluronic acid-based delivery systems.
Collapse
Affiliation(s)
- Neha N Parayath
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, Northeastern University, Boston, MA 02115, USA
| | - Srujan K Gandham
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, Northeastern University, Boston, MA 02115, USA
| | - Mansoor M Amiji
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, Northeastern University, Boston, MA 02115, USA,Department of Chemical Engineering, College of Engineering, Northeastern University, Boston, MA 02115, USA,Author for correspondence: Tel.: +1 617 373 3137;
| |
Collapse
|
20
|
Liang ZQ, He RQ, Luo JY, Huang ZG, Li J, Zhong LY, Chen JH, Huang SN, Shi L, Wei KL, Zeng JH, Zeng JJ, Chen G. Downregulated Dual-Specificity Protein Phosphatase 1 in Ovarian Carcinoma: A Comprehensive Study With Multiple Methods. Pathol Oncol Res 2022; 28:1610404. [PMID: 35911442 PMCID: PMC9336223 DOI: 10.3389/pore.2022.1610404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Accepted: 06/23/2022] [Indexed: 11/13/2022]
Abstract
Introduction: We aimed to explore the abnormal expression of dual-specificity protein phosphatase 1 (DUSP1) and its latent molecular mechanisms in ovarian carcinoma (OVCA). Materials and Methods: Two clinical cohorts collected from two different hospitals were used to evaluate the expression of DUSP1 protein in OVCA tissues. RNA-sequencing and microarray datasets were utilised to verify DUSP1 expression at mRNA levels in both OVCA tissues and in the peripheral blood of OVCA patients. Furthermore, an integrated calculation was performed to pool the standard mean difference (SMD) from each cohort in order to comprehensively assess the expression of DUSP1 in OVCA. Furthermore, we examined the relationship among DUSP1, tumour microenvironment (TME), and chemotherapy resistance in OVCA. Moreover, we used pathway enrichment analysis to explore the underlying mechanisms of DUSP1 in OVCA. Results: A pooled SMD of −1.19 (95% CI [−2.00, −0.38], p = 0.004) with 1,240 samples revealed that DUSP1 was downregulated in OVCA at both mRNA and protein levels. The area under the receiver operating characteristic curve of 0.9235 indicated the downregulated DUSP1 in peripheral blood may have a non-invasive diagnostic value in OVCA. Through six algorithms, we identified that DUSP1 may related to tumour-infiltrating T cells and cancer associated fibroblasts (CAFs) in OVCA. Pathway enrichment demonstrated that DUSP1 might participate in the mitogen-activated protein kinase (MAPK) signalling pathway. Furthermore, DUSP1 may have relations with chemotherapy resistance, and a favourable combining affinity was observed in the paclitaxel-DUSP1 docking model. Conclusion: DUSP1 was downregulated in OVCA, and this decreasing trend may affect the infiltration of CAFs. Finally, DUSP1 may have a targeting relation with paclitaxel and participate in MAPK signaling pathways.
Collapse
Affiliation(s)
- Zi-Qian Liang
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Rong-Quan He
- Department of Medical Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Jia-Yuan Luo
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Zhi-Guang Huang
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Jie Li
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Lu-Yang Zhong
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Jun-Hong Chen
- Department of Pathology, Maternal and Child Health Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Su-Ning Huang
- Department of Radiotherapy, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Lin Shi
- Department of Pathology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Kang-Lai Wei
- Department of Pathology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Jiang-Hui Zeng
- Department of Clinical Laboratory, The Third Affiliated Hospital of Guangxi Medical University/Nanning Second People’s Hospital, Nanning, China
| | - Jing-Jing Zeng
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Gang Chen
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- *Correspondence: Gang Chen,
| |
Collapse
|
21
|
Liu J, Lei B, Yu X, Li Y, Deng Y, Yang G, Li Z, Liu T, Ye L. Combining Immune-Related Genes For Delineating the Extracellular Matrix and Predicting Hormone Therapy and Neoadjuvant Chemotherapy Benefits In Breast Cancer. Front Immunol 2022; 13:888339. [PMID: 35911730 PMCID: PMC9331652 DOI: 10.3389/fimmu.2022.888339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 06/20/2022] [Indexed: 11/23/2022] Open
Abstract
Breast cancer (BC) is the most prevalent cancer in women worldwide. A systematic approach to BC treatment, comprising adjuvant and neoadjuvant chemotherapy (NAC), as well as hormone therapy, forms the foundation of the disease’s therapeutic strategy. The extracellular matrix (ECM) is a dynamic network that exerts a robust biological effect on the tumor microenvironment (TME), and it is highly regulated by several immunological components, such as chemokines and cytokines. It has been established that the ECM promotes the development of an immunosuppressive TME. Therefore, while analyzing the ECM of BC, immune-related genes must be considered. In this study, we used bioinformatic approaches to identify the most valuable ECM-related immune genes. We used weighted gene co-expression network analysis to identify the immune-related genes that potentially regulate the ECM and then combined them with the original ECM-related gene set for further analysis. Least absolute shrinkage and selection operator (LASSO) regression and SurvivalRandomForest were used to narrow our ECM-related gene list and establish an ECM index (ECMI) to better delineate the ECM signature. We stratified BC patients into ECMI high and low groups and evaluated their clinical, biological, and genomic characteristics. We found that the ECMI is highly correlated with long-term BC survival. In terms of the biological process, this index is positively associated with the cell cycle, DNA damage repair, and homologous recombination but negatively with processes involved in angiogenesis and epithelial–mesenchymal transition. Furthermore, the tumor mutational burden, copy number variation, and DNA methylation levels were found to be related to the ECMI. In the Metabric cohort, we demonstrated that hormone therapy is more effective in patients with a low ECMI. Additionally, differentially expressed genes from the ECM-related gene list were extracted from patients with a pathologic complete response (pCR) to NAC and with residual disease (RD) to construct a neural network model for predicting the chance of achieving pCR individually. Finally, we performed qRT-PCR to validate our findings and demonstrate the important role of the gene OGN in predicting the pCR rate. In conclusion, delineation of the ECM signature with immune-related genes is anticipated to aid in the prediction of the prognosis of patients with BC and the benefits of hormone therapy and NAC in BC patients.
Collapse
Affiliation(s)
- Jianyu Liu
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Bo Lei
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Xin Yu
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yingpu Li
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yuhan Deng
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Guang Yang
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Zhigao Li
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China
- *Correspondence: Tong Liu, ; Zhigao Li, ; Leiguang Ye,
| | - Tong Liu
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China
- *Correspondence: Tong Liu, ; Zhigao Li, ; Leiguang Ye,
| | - Leiguang Ye
- Department of Oncology, Harbin Medical University, Harbin, China
- *Correspondence: Tong Liu, ; Zhigao Li, ; Leiguang Ye,
| |
Collapse
|
22
|
Chen H, Tu W, Lu Y, Zhang Y, Xu Y, Chen X, Zhu M, Liu Y. Low-dose X-ray irradiation combined with FAK inhibitors improves the immune microenvironment and confers sensitivity to radiotherapy in pancreatic cancer. Biomed Pharmacother 2022; 151:113114. [PMID: 35594704 DOI: 10.1016/j.biopha.2022.113114] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 05/06/2022] [Accepted: 05/10/2022] [Indexed: 11/30/2022] Open
Abstract
Radiation therapy offers limited clinical benefits for patients with pancreatic cancer, partly as a result of the predominantly immunosuppressive microenvironment characteristic of this specific type of cancer. A large number of abnormal blood vessels and high-density fibrous matrices in pancreatic cancer will lead to hypoxia within tumor tissue and hinder immune cell infiltration. We used low-dose X-ray irradiation, also known as low-dose radiation therapy (LDRT), to normalize the blood vessels in pancreatic cancer, while simultaneously administering an inhibitor of focal adhesion kinase (FAK) to reduce pancreatic cancer fibrosis. We found that this treatment successfully reduced pancreatic cancer hypoxia, increased immune cell infiltration, and increased sensitivity to radiation therapy for pancreatic cancer.
Collapse
Affiliation(s)
- Huanliang Chen
- Department of Radiation Oncology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201620, China
| | - Wenzhi Tu
- Department of Radiation Oncology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201620, China
| | - Yue Lu
- Department of Radiotherapy, Huangpu Branch of the Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200002, China
| | - Yingzi Zhang
- Department of Radiation Oncology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201620, China
| | - Yiqing Xu
- Department of Radiation Oncology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201620, China
| | - Xuming Chen
- Department of Radiation Oncology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201620, China
| | - Meiling Zhu
- Department of Oncology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China.
| | - Yong Liu
- Department of Radiation Oncology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201620, China.
| |
Collapse
|
23
|
Shah K, Mallik SB, Gupta P, Iyer A. Targeting Tumour-Associated Fibroblasts in Cancers. Front Oncol 2022; 12:908156. [PMID: 35814453 PMCID: PMC9258494 DOI: 10.3389/fonc.2022.908156] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 05/23/2022] [Indexed: 11/13/2022] Open
Abstract
Tumours develop within complex tissue environments consisting of aberrant oncogenic cancer cells, diverse innate and adaptive immune cells, along with structural stromal cells, extracellular matrix and vascular networks, and many other cellular and non-cellular soluble constituents. Understanding the heterogeneity and the complex interplay between these cells remains a key barrier in treating tumours and cancers. The immune status of the pre-tumour and tumour milieu can dictate if the tumour microenvironment (TME) supports either a pro-malignancy or an anti-malignancy phenotype. Identification of the factors and cell types that regulate the dysfunction of the TME is crucial in order to understand and modulate the immune status of tumours. Among these cell types, tumour-associated fibroblasts are emerging as a major component of the TME that is often correlated with poor prognosis and therapy resistance, including immunotherapies. Thus, a deeper understanding of the complex roles of tumour-associated fibroblasts in regulating tumour immunity and cancer therapy could provide new insight into targeting the TME in various human cancers. In this review, we summarize recent studies investigating the role of immune and key stromal cells in regulating the immune status of the TME and discuss the therapeutic potential of targeting stromal cells, especially tumour-associated fibroblasts, within the TME as an adjuvant therapy to sensitize immunosuppressive tumours and prevent cancer progression, chemo-resistance and metastasis.
Collapse
Affiliation(s)
- Kairav Shah
- Alembic Discovery & Innovation, Alembic Pharmaceuticals, Hyderabad, India
| | | | - Praveer Gupta
- Alembic Discovery & Innovation, Alembic Pharmaceuticals, Hyderabad, India
| | - Abishek Iyer
- Alembic Discovery & Innovation, Alembic Pharmaceuticals, Hyderabad, India
| |
Collapse
|
24
|
Zhang P, Qin C, Liu N, Zhou X, Chu X, Lv F, Gu Y, Yin L, Liu J, Zhou J, Huo M. The programmed site-specific delivery of LY3200882 and PD-L1 siRNA boosts immunotherapy for triple-negative breast cancer by remodeling tumor microenvironment. Biomaterials 2022; 284:121518. [DOI: 10.1016/j.biomaterials.2022.121518] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 04/02/2022] [Accepted: 04/08/2022] [Indexed: 12/17/2022]
|
25
|
Willumsen N, Jensen C, Green G, Nissen NI, Neely J, Nelson DM, Pedersen RS, Frederiksen P, Chen IM, Boisen MK, Johansen AZ, Madsen DH, Svane IM, Lipton A, Leitzel K, Ali SM, Erler JT, Hurkmans DP, Mathijssen RHJ, Aerts J, Eslam M, George J, Christiansen C, Bissel MJ, Karsdal MA. Fibrotic activity quantified in serum by measurements of type III collagen pro-peptides can be used for prognosis across different solid tumor types. Cell Mol Life Sci 2022; 79:204. [PMID: 35332383 PMCID: PMC8948122 DOI: 10.1007/s00018-022-04226-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 02/15/2022] [Accepted: 02/28/2022] [Indexed: 12/12/2022]
Abstract
Due to activation of fibroblast into cancer-associated fibroblasts, there is often an increased deposition of extracellular matrix and fibrillar collagens, e.g. type III collagen, in the tumor microenvironment (TME) that leads to tumor fibrosis (desmoplasia). Tumor fibrosis is closely associated with treatment response and poor prognosis for patients with solid tumors. To assure that the best possible treatment option is provided for patients, there is medical need for identifying patients with high (or low) fibrotic activity in the TME. Measuring unique collagen fragments such as the pro-peptides released into the bloodstream during fibrillar collagen deposition in the TME can provide a non-invasive measure of the fibrotic activity. Based on data from 8 previously published cohorts, this review provides insight into the prognostic value of quantifying tumor fibrosis by measuring the pro-peptide of type III collagen in serum of a total of 1692 patients with different solid tumor types and discusses the importance of tumor fibrosis for understanding prognosis and for potentially guiding future drug development efforts that aim at overcoming the poor outcome associated with a fibrotic TME.
Collapse
Affiliation(s)
| | - Christina Jensen
- Nordic Bioscience, Herlev Hovedgade 205-207, 2730, Herlev, Denmark
| | | | - Neel I Nissen
- Nordic Bioscience, Herlev Hovedgade 205-207, 2730, Herlev, Denmark
| | | | | | | | | | - Inna M Chen
- Department of Oncology, Herlev and Gentofte Hospital, Copenhagen University Hospital, Herlev, Denmark
| | - Mogens K Boisen
- Department of Oncology, Herlev and Gentofte Hospital, Copenhagen University Hospital, Herlev, Denmark
| | - Astrid Z Johansen
- Department of Oncology, Herlev and Gentofte Hospital, Copenhagen University Hospital, Herlev, Denmark
| | - Daniel H Madsen
- Center for Cancer Immune Therapy, Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
| | - Inge Marie Svane
- Center for Cancer Immune Therapy, Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
| | - Allan Lipton
- Penn State Hershey Medical Center, Hershey, PA, USA
| | - Kim Leitzel
- Penn State Hershey Medical Center, Hershey, PA, USA
| | | | - Janine T Erler
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark
| | - Daan P Hurkmans
- Department of Pathology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Ron H J Mathijssen
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Joachim Aerts
- Department of Pulmonology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Mohammed Eslam
- Storr Liver Centre, Westmead Institute for Medical Research, Westmead Hospital and University of Sydney, Sydney, NSW, Australia
| | - Jacob George
- Storr Liver Centre, Westmead Institute for Medical Research, Westmead Hospital and University of Sydney, Sydney, NSW, Australia
| | | | - Mina J Bissel
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA, 94720, USA
| | - Morten A Karsdal
- Nordic Bioscience, Herlev Hovedgade 205-207, 2730, Herlev, Denmark
| |
Collapse
|
26
|
Gong F, Yang Y, Wen L, Wang C, Li J, Dai J. An Overview of the Role of Mechanical Stretching in the Progression of Lung Cancer. Front Cell Dev Biol 2022; 9:781828. [PMID: 35004682 PMCID: PMC8740071 DOI: 10.3389/fcell.2021.781828] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 12/09/2021] [Indexed: 12/19/2022] Open
Abstract
Cells and tissues in the human body are subjected to mechanical forces of varying degrees, such as tension or pressure. During tumorigenesis, physical factors, especially mechanical factors, are involved in tumor development. As lung tissue is influenced by movements associated with breathing, it is constantly subjected to cyclical stretching and retraction; therefore, lung cancer cells and lung cancer-associated fibroblasts (CAFs) are constantly exposed to mechanical load. Thus, to better explore the mechanisms involved in lung cancer progression, it is necessary to consider factors involved in cell mechanics, which may provide a more comprehensive analysis of tumorigenesis. The purpose of this review is: 1) to provide an overview of the anatomy and tissue characteristics of the lung and the presence of mechanical stimulation; 2) to summarize the role of mechanical stretching in the progression of lung cancer; and 3) to describe the relationship between mechanical stretching and the lung cancer microenvironment, especially CAFs.
Collapse
Affiliation(s)
- Fengying Gong
- Department of Traditional Chinese Medicine, Nanfang Hospital of Southern Medical University, Guangzhou, China
| | - Yuchao Yang
- Guangdong Provincial Key Laboratory of Medical Biomechanics and Guangdong Engineering Research Center for Translation of Medical 3D Printing Application and National Key Discipline of Human Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Liangtao Wen
- Shiyue City Community Health Service Center, Shenzhen Integrated Traditional Chinese and Western Medicine Hospital, Shenzhen, China
| | - Congrong Wang
- Department of Laboratory Medicine, Nanfang Hospital of Southern Medical University, Guangzhou, China
| | - Jingjun Li
- Department of Traditional Chinese Medicine, Nanfang Hospital of Southern Medical University, Guangzhou, China
| | - Jingxing Dai
- Guangdong Provincial Key Laboratory of Medical Biomechanics and Guangdong Engineering Research Center for Translation of Medical 3D Printing Application and National Key Discipline of Human Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| |
Collapse
|
27
|
Ibrahim AM, Hamid MA, Althiab RA, Shariff AHM, Zulkifli RM. In vitro fibroblasts viability and migration stimulation of Acalypha indica: an insight on wound healing activity. FUTURE JOURNAL OF PHARMACEUTICAL SCIENCES 2021. [DOI: 10.1186/s43094-021-00333-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
The current study investigates the antioxidant activity of Acalypha indica aerial parts and root ethanolic extracts and explore whether these extracts will stimulate fibroblasts viability and ability to migrate.
Results
Aerial parts extract exhibited higher DPPH scavenging activity compared to root extract with IC50 of 62 µg/mL and 206 µg/mL, respectively. Both aerial parts and root extracts showed low cytotoxicity towards fibroblasts with 753 µg/mL LD50 for aerial parts and undetected LD50 for root extract. Additionally, aerial parts extract significantly induces fibroblasts proliferation up to 134%. Wound closure investigation showed a significant closure percentage for aerial parts compared to untreated control with 75% at 1 µg/mL and high closure percentage with 70% at 0.1 µg/mL for root extract compared to only 59% closure percentage for untreated control after 48 h of the study.
Conclusions
This study provided evidence for A. indica to have great wound healing potential. The finding builds the scientific background in future to utilise the high antioxidant activity of A. indica and its ability to stimulate fibroblasts migration and proliferation for further applications.
Collapse
|
28
|
Diehl V, Huber LS, Trebicka J, Wygrecka M, Iozzo RV, Schaefer L. The Role of Decorin and Biglycan Signaling in Tumorigenesis. Front Oncol 2021; 11:801801. [PMID: 34917515 PMCID: PMC8668865 DOI: 10.3389/fonc.2021.801801] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 11/09/2021] [Indexed: 12/12/2022] Open
Abstract
The complex and adaptive nature of malignant neoplasm constitute a major challenge for the development of effective anti-oncogenic therapies. Emerging evidence has uncovered the pivotal functions exerted by the small leucine-rich proteoglycans, decorin and biglycan, in affecting tumor growth and progression. In their soluble forms, decorin and biglycan act as powerful signaling molecules. By receptor-mediated signal transduction, both proteoglycans modulate key processes vital for tumor initiation and progression, such as autophagy, inflammation, cell-cycle, apoptosis, and angiogenesis. Despite of their structural homology, these two proteoglycans interact with distinct cell surface receptors and thus modulate distinct signaling pathways that ultimately affect cancer development. In this review, we summarize growing evidence for the complex roles of decorin and biglycan signaling in tumor biology and address potential novel therapeutic implications.
Collapse
Affiliation(s)
- Valentina Diehl
- Institute of Pharmacology and Toxicology, Goethe University, Frankfurt, Germany
| | - Lisa Sophie Huber
- Institute of Pharmacology and Toxicology, Goethe University, Frankfurt, Germany
| | - Jonel Trebicka
- Department of Internal Medicine I, Goethe University, Frankfurt, Germany
| | - Malgorzata Wygrecka
- Center for Infection and Genomics of the Lung, Member of the German Center for Lung Research, University of Giessen and Marburg Lung Center, Giessen, Germany
| | - Renato V. Iozzo
- Department of Pathology, Anatomy and Cell Biology and the Translational Cellular Oncology Program, Sidney Kimmel Cancer Center, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA, United States
| | - Liliana Schaefer
- Institute of Pharmacology and Toxicology, Goethe University, Frankfurt, Germany
| |
Collapse
|
29
|
Chen P, Lin C, Yang S, Chang Y. Oral submucous fibrosis stimulates invasion and epithelial-mesenchymal transition in oral squamous cell carcinoma by activating MMP-2 and IGF-IR. J Cell Mol Med 2021; 25:9814-9825. [PMID: 34528373 PMCID: PMC8505822 DOI: 10.1111/jcmm.16929] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 08/15/2021] [Accepted: 09/01/2021] [Indexed: 12/11/2022] Open
Abstract
Oral submucous fibrosis (OSF) involves a high risk of malignant transformation and has been implicated in oral cancer. Limited studies have been conducted on the role of OSF in relation to the invasive capabilities and epithelial-mesenchymal transition (EMT) in oral cancer. Herein, we investigated the effects of OSF on the microenvironment of human oral cancer cells. The results showed that the conditioned medium (CM) of fibrotic buccal mucosal fibroblasts (fBMFs) strongly induced the invasion of oral cancer cells and increased the activities of matrix metalloproteinase-2. OSF significantly induced the EMT in oral cancer cells and downregulated epithelial markers, such as E-cadherin, but significantly elevated vimentin, fibronectin, N-cadherin, RhoA, Rac-1 and FAK. Insulin-like growth factor-1 (IGF-1) was elevated in OSF. The protein levels of the IGF-1R were upregulated specifically in fBMF CM treatment for oral cancer cells, and the IGFR gene was confirmed by The Cancer Genome Atlas patient transcriptome data. The Kaplan-Meier curve analysis revealed that patients with oral squamous cell carcinoma and high IGFR expression levels had poorer 5-year survival than those with low IGFR expression (p = 0.004). The fBMF-stimulated EMT cell model may recapture some of the molecular changes during EMT progression in clinical patients with oral cancer.
Collapse
Affiliation(s)
- Pei‐Ni Chen
- Clinical LaboratoryChung Shan Medical University HospitalTaichungTaiwan
- Institute of MedicineChung Shan Medical UniversityTaichungTaiwan
| | - Chiao‐Wen Lin
- Institute of Oral SciencesChung Shan Medical UniversityTaichungTaiwan
- Department of DentistryChung Shan Medical University HospitalTaichungTaiwan
| | - Shun‐Fa Yang
- Institute of MedicineChung Shan Medical UniversityTaichungTaiwan
- Department of Medical ResearchChung Shan Medical University HospitalTaichungTaiwan
| | - Yu‐Chao Chang
- Department of DentistryChung Shan Medical University HospitalTaichungTaiwan
- School of DentistryChung Shan Medical UniversityTaichungTaiwan
| |
Collapse
|
30
|
Franchi-Mendes T, Eduardo R, Domenici G, Brito C. 3D Cancer Models: Depicting Cellular Crosstalk within the Tumour Microenvironment. Cancers (Basel) 2021; 13:4610. [PMID: 34572836 PMCID: PMC8468887 DOI: 10.3390/cancers13184610] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 09/07/2021] [Accepted: 09/08/2021] [Indexed: 12/11/2022] Open
Abstract
The tumour microenvironment plays a critical role in tumour progression and drug resistance processes. Non-malignant cell players, such as fibroblasts, endothelial cells, immune cells and others, interact with each other and with the tumour cells, shaping the disease. Though the role of each cell type and cell communication mechanisms have been progressively studied, the complexity of this cellular network and its role in disease mechanism and therapeutic response are still being unveiled. Animal models have been mainly used, as they can represent systemic interactions and conditions, though they face recognized limitations in translational potential due to interspecies differences. In vitro 3D cancer models can surpass these limitations, by incorporating human cells, including patient-derived ones, and allowing a range of experimental designs with precise control of each tumour microenvironment element. We summarize the role of each tumour microenvironment component and review studies proposing 3D co-culture strategies of tumour cells and non-malignant cell components. Moreover, we discuss the potential of these modelling approaches to uncover potential therapeutic targets in the tumour microenvironment and assess therapeutic efficacy, current bottlenecks and perspectives.
Collapse
Affiliation(s)
- Teresa Franchi-Mendes
- iBET—Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal; (T.F.-M.); (R.E.); (G.D.)
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal
| | - Rodrigo Eduardo
- iBET—Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal; (T.F.-M.); (R.E.); (G.D.)
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal
| | - Giacomo Domenici
- iBET—Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal; (T.F.-M.); (R.E.); (G.D.)
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal
| | - Catarina Brito
- iBET—Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal; (T.F.-M.); (R.E.); (G.D.)
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal
- The Discoveries Centre for Regenerative and Precision Medicine, Lisbon Campus, Av. da República, 2780-157 Oeiras, Portugal
| |
Collapse
|
31
|
Zhou Y, Zhou H, Shi J, Guan A, Zhu Y, Hou Z, Li R. Decreased m6A Modification of CD34/CD276(B7-H3) Leads to Immune Escape in Colon Cancer. Front Cell Dev Biol 2021; 9:715674. [PMID: 34307389 PMCID: PMC8297592 DOI: 10.3389/fcell.2021.715674] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 06/21/2021] [Indexed: 12/21/2022] Open
Abstract
Previous studies have reported that m6a modification promotes tumor immune escape by affecting tumor microenvironment (TME). Due to the complexity of TME, a single biomarker is insufficient to describe the complex biological characteristics of tumor and its microenvironment. Therefore, it is more meaningful to explore a group of effective biomarkers reflecting different characteristics of cancer to evaluate the biological characteristics of solid tumors. Here, the immune gene CD34/CD276 with different m6A peak was obtained by m6A sequencing (MeRIP-seq) of colon cancer (CRC)clinical samples and combined with MsIgDB database, which was used to perform cluster analysis on TCGA-COAD level 3 data. The CD34/CD276 as a molecular marker for CRC prognosis was confirmed by survival analysis and immunohistochemical assay. Further bioinformatics analysis was carried out to analyze the molecular mechanism of CD34/CD276 affecting the TME through m6a-dependent down-regulation and ultimately promoting immune escape of CRC.
Collapse
Affiliation(s)
- Yiran Zhou
- Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, First Department of General Surgery, Yan'an Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Haodong Zhou
- Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, First Department of General Surgery, Yan'an Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Jianlin Shi
- Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Department of Thoracic Surgery, Yan'an Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Aoran Guan
- Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, First Department of General Surgery, Yan'an Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yankun Zhu
- Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, First Department of General Surgery, Yan'an Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Zongliu Hou
- Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming, China
| | - Ruhong Li
- Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, First Department of General Surgery, Yan'an Affiliated Hospital of Kunming Medical University, Kunming, China
| |
Collapse
|
32
|
ECM Remodeling in Squamous Cell Carcinoma of the Aerodigestive Tract: Pathways for Cancer Dissemination and Emerging Biomarkers. Cancers (Basel) 2021. [DOI: 10.3390/cancers13112759
expr 955442319 + 839973387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023] Open
Abstract
Squamous cell carcinomas (SCC) include a number of different types of tumors developing in the skin, in hollow organs, as well as the upper aerodigestive tract (UADT) including the head and neck region and the esophagus which will be dealt with in this review. These tumors are often refractory to current therapeutic approaches with poor patient outcome. The most important prognostic determinant of SCC tumors is the presence of distant metastasis, significantly correlating with low patient survival rates. Rapidly emerging evidence indicate that the extracellular matrix (ECM) composition and remodeling profoundly affect SSC metastatic dissemination. In this review, we will summarize the current knowledge on the role of ECM and its remodeling enzymes in affecting the growth and dissemination of UADT SCC. Taken together, these published evidence suggest that a thorough analysis of the ECM composition in the UADT SCC microenvironment may help disclosing the mechanism of resistance to the treatments and help defining possible targets for clinical intervention.
Collapse
|
33
|
ECM Remodeling in Squamous Cell Carcinoma of the Aerodigestive Tract: Pathways for Cancer Dissemination and Emerging Biomarkers. Cancers (Basel) 2021; 13:cancers13112759. [PMID: 34199373 PMCID: PMC8199582 DOI: 10.3390/cancers13112759] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/27/2021] [Accepted: 05/28/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Local and distant metastasis of patients affected by squamous cell carcinoma of the upper aerodigestive tract predicts poor prognosis. In the latest years, the introduction of new therapeutic approaches, including targeted and immune therapies, has improved the overall survival. However, a large number of these patients do not benefit from these treatments. Thus, the identification of suitable prognostic and predictive biomarkers, as well as the discovery of new therapeutic targets have emerged as a crucial clinical need. In this context, the extracellular matrix represents a suitable target for the development of such therapeutic tools. In fact, the extracellular matrix is composed by complex molecules able to interact with a plethora of receptors and growth factors, thus modulating the dynamic crosstalk between cancer cells and the tumor microenvironment. In this review, we summarize the current knowledge of the role of the extracellular matrix in affecting squamous cell carcinoma growth and dissemination. Despite extracellular matrix is known to affect the development of many cancer types, only a restricted number of these molecules have been recognized to impact on squamous cell carcinoma progression. Thus, we consider that a thorough analysis of these molecules may be key to develop new potential therapeutic targets/biomarkers. Abstract Squamous cell carcinomas (SCC) include a number of different types of tumors developing in the skin, in hollow organs, as well as the upper aerodigestive tract (UADT) including the head and neck region and the esophagus which will be dealt with in this review. These tumors are often refractory to current therapeutic approaches with poor patient outcome. The most important prognostic determinant of SCC tumors is the presence of distant metastasis, significantly correlating with low patient survival rates. Rapidly emerging evidence indicate that the extracellular matrix (ECM) composition and remodeling profoundly affect SSC metastatic dissemination. In this review, we will summarize the current knowledge on the role of ECM and its remodeling enzymes in affecting the growth and dissemination of UADT SCC. Taken together, these published evidence suggest that a thorough analysis of the ECM composition in the UADT SCC microenvironment may help disclosing the mechanism of resistance to the treatments and help defining possible targets for clinical intervention.
Collapse
|
34
|
Correa-Gallegos D, Jiang D, Rinkevich Y. Fibroblasts as confederates of the immune system. Immunol Rev 2021; 302:147-162. [PMID: 34036608 DOI: 10.1111/imr.12972] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 04/28/2021] [Accepted: 04/28/2021] [Indexed: 12/18/2022]
Abstract
Fibroblastic stromal cells are as diverse, in origin and function, as the niches they fashion in the mammalian body. This cellular variety impacts the spectrum of responses elicited by the immune system. Fibroblast influence on the immune system keeps evolving our perspective on fibroblast roles and functions beyond just a passive structural part of organs. This review discusses the foundations of fibroblastic stromal-immune crosstalk, under the scope of stromal heterogeneity as a basis for tissue-specific tutoring of the immune system. Focusing on the skin as a relevant immunological organ, we detail the complex interactions between distinct fibroblast populations and immune cells that occur during homeostasis, injury repair, scarring, and disease. We further review the relevance of fibroblastic stromal cell heterogeneity and how this heterogeneity is central to regulate the immune system from its inception during embryonic development into adulthood.
Collapse
Affiliation(s)
- Donovan Correa-Gallegos
- Institute of Lung Biology and Disease, Comprehensive Pneumology Center, Helmholtz Zentrum München, Munich, Germany
| | - Dongsheng Jiang
- Institute of Lung Biology and Disease, Comprehensive Pneumology Center, Helmholtz Zentrum München, Munich, Germany
| | - Yuval Rinkevich
- Institute of Regenerative Biology and Medicine, Helmholtz Zentrum München, Munich, Germany
| |
Collapse
|
35
|
Collagen molecular phenotypic switch between non-neoplastic and neoplastic canine mammary tissues. Sci Rep 2021; 11:8659. [PMID: 33883562 PMCID: PMC8060395 DOI: 10.1038/s41598-021-87380-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 03/23/2021] [Indexed: 01/24/2023] Open
Abstract
In spite of major advances over the past several decades in diagnosis and treatment, breast cancer remains a global cause of morbidity and premature death for both human and veterinary patients. Due to multiple shared clinicopathological features, dogs provide an excellent model of human breast cancer, thus, a comparative oncology approach may advance our understanding of breast cancer biology and improve patient outcomes. Despite an increasing awareness of the critical role of fibrillar collagens in breast cancer biology, tumor-permissive collagen features are still ill-defined. Here, we characterize the molecular and morphological phenotypes of type I collagen in canine mammary gland tumors. Canine mammary carcinoma samples contained longer collagen fibers as well as a greater population of wider fibers compared to non-neoplastic and adenoma samples. Furthermore, the total number of collagen cross-links enriched in the stable hydroxylysine-aldehyde derived cross-links was significantly increased in neoplastic mammary gland samples compared to non-neoplastic mammary gland tissue. The mass spectrometric analyses of type I collagen revealed that in malignant mammary tumor samples, lysine residues, in particular those in the telopeptides, were markedly over-hydroxylated in comparison to non-neoplastic mammary tissue. The extent of glycosylation of hydroxylysine residues was comparable among the groups. Consistent with these data, expression levels of genes encoding lysyl hydroxylase 2 (LH2) and its molecular chaperone FK506-binding protein 65 were both significantly increased in neoplastic samples. These alterations likely lead to an increase in the LH2-mediated stable collagen cross-links in mammary carcinoma that may promote tumor cell metastasis in these patients.
Collapse
|
36
|
Amos SE, Choi YS. The Cancer Microenvironment: Mechanical Challenges of the Metastatic Cascade. Front Bioeng Biotechnol 2021; 9:625859. [PMID: 33644019 PMCID: PMC7907606 DOI: 10.3389/fbioe.2021.625859] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 01/18/2021] [Indexed: 12/12/2022] Open
Abstract
The metastatic cascade presents a significant challenge to patient survival in the fight against cancer. As metastatic cells disseminate and colonize a secondary site, stepwise exposure to microenvironment-specific mechanical stimuli influences and protects successful metastasis. Following cancerous transformation and associated cell recruitment, the tumor microenvironment (TME) becomes a mechanically complex niche, owing to changes in extracellular matrix (ECM) stiffness and architecture. The ECM mechanically reprograms the cancer cell phenotype, priming cells for invasion. 2D and 3D hydrogel-based culture platforms approximate these environmental variables and permit investigations into tumor-dependent shifts in malignancy. Following TME modification, malignant cells must invade the local ECM, driven toward blood, and lymph vessels by sensing biochemical and biophysical gradients. Microfluidic chips recreate cancer-modified ECM tracks, empowering studies into modes of confined motility. Intravasation and extravasation consist of complex cancer-endothelial interactions that modify an otherwise submicron-scale migration. Perfused microfluidic platforms facilitate the physiological culture of endothelial cells and thus enhance the translatability of basic research into metastatic transendothelial migration. These platforms also shed light on the poorly understood circulating tumor cell, which defies adherent cell norms by surviving the shear stress of blood flow and avoiding anoikis. Metastatic cancers possess the plasticity to adapt to new mechanical conditions, permitting their invasiveness, and ensuring their survival against anomalous stimuli. Here, we review the cellular mechanics of metastasis in the context of current in vitro approaches. Advances that further expose the mechanisms underpinning the phenotypic fluidity of metastatic cancers remain central to the development of novel interventions targeting cancer.
Collapse
Affiliation(s)
| | - Yu Suk Choi
- School of Human Sciences, The University of Western Australia, Perth, WA, Australia
| |
Collapse
|
37
|
Barrett RL, Puré E. Cancer-associated fibroblasts and their influence on tumor immunity and immunotherapy. eLife 2020; 9:57243. [PMID: 33370234 PMCID: PMC7769568 DOI: 10.7554/elife.57243] [Citation(s) in RCA: 201] [Impact Index Per Article: 40.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 12/12/2020] [Indexed: 12/15/2022] Open
Abstract
Fibroblasts play an essential role in organogenesis and the integrity of tissue architecture and function. Growth in most solid tumors is dependent upon remodeling 'stroma', composed of cancer-associated fibroblasts (CAFs) and extracellular matrix (ECM), which plays a critical role in tumor initiation, progression, metastasis, and therapeutic resistance. Recent studies have clearly established that the potent immunosuppressive activity of stroma is a major mechanism by which stroma can promote tumor progression and confer resistance to immune-based therapies. Herein, we review recent advances in identifying the stroma-dependent mechanisms that regulate cancer-associated inflammation and antitumor immunity, in particular, the interactions between fibroblasts and immune cells. We also review the potential mechanisms by which stroma can confer resistance to immune-based therapies for solid tumors and current advancements in stroma-targeted therapies.
Collapse
Affiliation(s)
| | - Ellen Puré
- University of Pennsylvania, Philadelphia, United States
| |
Collapse
|
38
|
Zhang GL, Gutter-Kapon L, Ilan N, Batool T, Singh K, Digre A, Luo Z, Sandler S, Shaked Y, Sanderson RD, Wang XM, Li JP, Vlodavsky I. Significance of host heparanase in promoting tumor growth and metastasis. Matrix Biol 2020; 93:25-42. [PMID: 32534153 PMCID: PMC7704762 DOI: 10.1016/j.matbio.2020.06.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Revised: 06/03/2020] [Accepted: 06/03/2020] [Indexed: 02/08/2023]
Abstract
Heparanase, the sole heparan sulfate degrading endoglycosidase, regulates multiple biological activities that enhance tumor growth, angiogenesis and metastasis. Much of the impact of heparanase on tumor progression is related to its function in mediating tumor-host crosstalk, priming the tumor microenvironment to better support tumor growth and metastasis. We have utilized mice over-expressing (Hpa-tg) heparanase to reveal the role of host heparanase in tumor initiation, growth and metastasis. While in wild type mice tumor development in response to DMBA carcinogenesis was restricted to the mammary gland, Hpa-tg mice developed tumors also in their lungs and liver, associating with reduced survival of the tumor-bearing mice. Consistently, xenograft tumors (lymphoma, melanoma, lung carcinoma, pancreatic carcinoma) transplanted in Hpa-tg mice exhibited accelerated tumor growth and shorter survival of the tumor-bearing mice compared with wild type mice. Hpa-tg mice were also more prone to the development of metastases following intravenous or subcutaneous injection of tumor cells. In some models, the growth advantage was associated with infiltration of heparanase-high host cells into the tumors. However, in other models, heparanase-high host cells were not detected in the primary tumor, implying that the growth advantage in Hpa-tg mice is due to systemic factors. Indeed, we found that plasma from Hpa-tg mice enhanced tumor cell migration and invasion attributed to increased levels of pro-tumorigenic factors (i.e., RANKL, SPARC, MIP-2) in the plasma of Hpa-Tg vs. wild type mice. Furthermore, tumor aggressiveness and short survival time were demonstrated in wild type mice transplanted with bone marrow derived from Hpa-tg but not wild type mice. These results were attributed, among other factors, to upregulation of pro-tumorigenic (i.e., IL35+) and downregulation of anti-tumorigenic (i.e., IFN-γ+) T-cell subpopulations in the spleen, lymph nodes and blood of Hpa-tg vs. wild type mice and their increased infiltration into the primary tumor. Collectively, our results emphasize the significance of host heparanase in mediating the pro-tumorigenic and pro-metastatic interactions between the tumor cells and the host tumor microenvironment, immune cells and systemic factors.
Collapse
Affiliation(s)
- Gan-Lin Zhang
- Oncology Department, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China; Department of Medical Biochemistry and Microbiology, SciLifeLab Uppsala, The Biomedical Center, University of Uppsala, Uppsala, Sweden
| | - Lilach Gutter-Kapon
- Technion Integrated Cancer Center, Rappaport Faculty of Medicine, P. O. Box 9649, Technion, Haifa 31096, Israel
| | - Neta Ilan
- Technion Integrated Cancer Center, Rappaport Faculty of Medicine, P. O. Box 9649, Technion, Haifa 31096, Israel
| | - Tahira Batool
- Department of Medical Biochemistry and Microbiology, SciLifeLab Uppsala, The Biomedical Center, University of Uppsala, Uppsala, Sweden
| | - Kailash Singh
- Department of Cell Biology, University of Uppsala, Uppsala, Sweden
| | - Andreas Digre
- Department of Medical Biochemistry and Microbiology, SciLifeLab Uppsala, The Biomedical Center, University of Uppsala, Uppsala, Sweden
| | - Zhengkang Luo
- Department of Cell Biology, University of Uppsala, Uppsala, Sweden
| | - Stellan Sandler
- Department of Cell Biology, University of Uppsala, Uppsala, Sweden
| | - Yuval Shaked
- Technion Integrated Cancer Center, Rappaport Faculty of Medicine, P. O. Box 9649, Technion, Haifa 31096, Israel
| | - Ralph D Sanderson
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Xiao-Min Wang
- Oncology Department, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China.
| | - Jin-Ping Li
- Oncology Department, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China; Department of Medical Biochemistry and Microbiology, SciLifeLab Uppsala, The Biomedical Center, University of Uppsala, Uppsala, Sweden.
| | - Israel Vlodavsky
- Technion Integrated Cancer Center, Rappaport Faculty of Medicine, P. O. Box 9649, Technion, Haifa 31096, Israel.
| |
Collapse
|
39
|
Modulation of Immune Infiltration of Ovarian Cancer Tumor Microenvironment by Specific Subpopulations of Fibroblasts. Cancers (Basel) 2020; 12:cancers12113184. [PMID: 33138184 PMCID: PMC7692816 DOI: 10.3390/cancers12113184] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 10/20/2020] [Accepted: 10/24/2020] [Indexed: 12/15/2022] Open
Abstract
Tumor immune infiltration plays a key role in the progression of solid tumors, including ovarian cancer, and immunotherapies are rapidly emerging as effective treatment modalities. However, the role of cancer-associated fibroblasts (CAFs), a predominant stromal constituent, in determining the tumor-immune microenvironment and modulating efficacy of immunotherapies remains poorly understood. We have conducted an extensive bioinformatic analysis of our and other publicly available ovarian cancer datasets (GSE137237, GSE132289 and GSE71340), to determine the correlation of fibroblast subtypes within the tumor microenvironment (TME) with the characteristics of tumor-immune infiltration. We identified (1) four functional modules of CAFs in ovarian cancer that are associated with the TME and metastasis of ovarian cancer, (2) immune-suppressive function of the collagen 1,3,5-expressing CAFs in primary ovarian cancer and omental metastases, and (3) consistent positive correlations between the functional modules of CAFs with anti-immune response genes and negative correlation with pro-immune response genes. Our study identifies a specific fibroblast subtype, fibroblast functional module (FFM)2, in the ovarian cancer tumor microenvironment that can potentially modulate a tumor-promoting immune microenvironment, which may be detrimental toward the effectiveness of ovarian cancer immunotherapies.
Collapse
|
40
|
Dawson MR, Xuan B, Hsu J, Ghosh D. Force balancing ACT-IN the tumor microenvironment: Cytoskeletal modifications in cancer and stromal cells to promote malignancy. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2020; 360:1-31. [PMID: 33962748 DOI: 10.1016/bs.ircmb.2020.09.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The tumor microenvironment is a complex milieu that dictates the growth, invasion, and metastasis of cancer cells. Both cancer and stromal cells in the tumor tissue encounter and adapt to a variety of extracellular factors, and subsequently contribute and drive the progression of the disease to more advanced stages. As the disease progresses, a small population of cancer cells becomes more invasive through a complex process known as epithelial-mesenchymal transition, and nearby stromal cells assume a carcinoma associated fibroblast phenotype characterized by enhanced migration, cell contractility, and matrix secretion with the ability to reorganize extracellular matrices. As cells transition into more malignant phenotypes their biophysical properties, controlled by the organization of cytoskeletal proteins, are altered. Actin and its associated proteins are essential modulators and facilitators of these changes. As the cells respond to the cues in the microenvironment, actin driven mechanical forces inside and outside the cells also evolve. Recent advances in biophysical techniques have enabled us to probe these actin driven changes in cancer and stromal cells and demarcate their role in driving changes in the microenvironment. Understanding the underlying biophysical mechanisms that drive cancer progression could provide critical insight on novel therapeutic approaches in the fight against cancer.
Collapse
Affiliation(s)
- Michelle R Dawson
- Department of Molecular Pharmacology, Physiology, and Biotechnology, Brown University, Providence, RI, United States; Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI, United States; Brown University, Center for Biomedical Engineering, Providence, RI, United States.
| | - Botai Xuan
- Department of Molecular Pharmacology, Physiology, and Biotechnology, Brown University, Providence, RI, United States
| | - Jeffrey Hsu
- Department of Molecular Pharmacology, Physiology, and Biotechnology, Brown University, Providence, RI, United States
| | - Deepraj Ghosh
- Department of Molecular Pharmacology, Physiology, and Biotechnology, Brown University, Providence, RI, United States
| |
Collapse
|
41
|
DeLeon-Pennell KY, Barker TH, Lindsey ML. Fibroblasts: The arbiters of extracellular matrix remodeling. Matrix Biol 2020; 91-92:1-7. [PMID: 32504772 PMCID: PMC7434687 DOI: 10.1016/j.matbio.2020.05.006] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 05/29/2020] [Accepted: 05/29/2020] [Indexed: 12/12/2022]
Abstract
Extracellular matrix (ECM) is the foundation on which all cells and organs converge to orchestrate normal physiological functions. In the setting of pathology, the ECM is modified to incorporate additional roles, with modifications including turnover of existing ECM and deposition of new ECM. The fibroblast is center stage in coordinating both normal tissue homeostasis and response to disease. Understanding how fibroblasts work under normal conditions and are activated in response to injury or stress will provide mechanistic insight that triggers discovery of new therapeutic treatments for a wide range of disease. We highlight here fibroblast roles in the cancer, lung, and heart as example systems where fibroblasts are major contributors to homeostasis and pathology.
Collapse
Affiliation(s)
- Kristine Y DeLeon-Pennell
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, and Research Service, Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC 29425, USA
| | - Thomas H Barker
- Department of Biomedical Engineering, University of Virginia, 415 Lane Road, Charlottesville, VA 22903, USA
| | - Merry L Lindsey
- Department of Cellular and Integrative Physiology, Center for Heart and Vascular Research, University of Nebraska Medical Center, 985850 Nebraska Medical Center, Omaha, NE 68198-5850, USA; and Research Service, Nebraska-Western Iowa Health Care System, Omaha, NE 68105; Research Service, Nebraska-Western Iowa Health Care System, Omaha, NE 68105.
| |
Collapse
|