1
|
Raaijmakers KTPM, Adema GJ, Bussink J, Ansems M. Cancer-associated fibroblasts, tumor and radiotherapy: interactions in the tumor micro-environment. J Exp Clin Cancer Res 2024; 43:323. [PMID: 39696386 DOI: 10.1186/s13046-024-03251-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 12/04/2024] [Indexed: 12/20/2024] Open
Abstract
Cancer-associated fibroblasts (CAFs) represent a group of genotypically non-malignant stromal cells in the tumor micro-environment (TME) of solid tumors that encompasses up to 80% of the tumor volume. Even though the phenotypic diversity and plasticity of CAFs complicates research, it is well-established that CAFs can affect many aspects of tumor progression, including growth, invasion and therapy resistance. Although anti-tumorigenic properties of CAFs have been reported, the majority of research demonstrates a pro-tumorigenic role for CAFs via (in)direct signaling to cancer cells, immunomodulation and extracellular matrix (ECM) remodeling. Following harsh therapeutic approaches such as radio- and/or chemotherapy, CAFs do not die but rather become senescent. Upon conversion towards senescence, many pro-tumorigenic characteristics of CAFs are preserved or even amplified. Senescent CAFs continue to promote tumor cell therapy resistance, modulate the ECM, stimulate epithelial-to-mesenchymal transition (EMT) and induce immunosuppression. Consequently, CAFs play a significant role in tumor cell survival, relapse and potentially malignant transformation of surviving cancer cells following therapy. Modulating CAF functioning in the TME therefore is a critical area of research. Proposed strategies to enhance therapeutic efficacy include reverting senescent CAFs towards a quiescent phenotype or selectively targeting (non-)senescent CAFs. In this review, we discuss CAF functioning in the TME before and during therapy, with a strong focus on radiotherapy. In the future, CAF functioning in the therapeutic TME should be taken into account when designing treatment plans and new therapeutic approaches.
Collapse
Affiliation(s)
- Kris T P M Raaijmakers
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Gosse J Adema
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Johan Bussink
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Marleen Ansems
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, the Netherlands.
| |
Collapse
|
2
|
Rauth S, Malafa M, Ponnusamy MP, Batra SK. Emerging Trends in Gastrointestinal Cancer Targeted Therapies: Harnessing Tumor Microenvironment, Immune Factors, and Metabolomics Insights. Gastroenterology 2024; 167:867-884. [PMID: 38759843 DOI: 10.1053/j.gastro.2024.05.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 04/23/2024] [Accepted: 05/01/2024] [Indexed: 05/19/2024]
Abstract
Gastrointestinal (GI) cancers are the leading cause of new cancer cases and cancer-related deaths worldwide. The treatment strategies for patients with GI tumors have focused on oncogenic molecular profiles associated with tumor cells. Recent evidence has demonstrated that the tumor cell functions are modulated by its microenvironment, compromising fibroblasts, extracellular matrices, microbiome, immune cells, and the enteric nervous system. Along with the tumor microenvironment components, alterations in key metabolic pathways have emerged as a hallmark of tumor cells. From these perspectives, this review will highlight the functions of different cellular components of the GI tumor microenvironment and their implications for treatment. Furthermore, we discuss the major metabolic reprogramming in GI tumor cells and how understanding metabolic rewiring could lead to new therapeutic strategies. Finally, we briefly summarize the targeted agents currently being studied in GI cancers. Understanding the complex interplay between tumor cell-intrinsic and -extrinsic factors during tumor progression is critical for developing new therapeutic strategies.
Collapse
Affiliation(s)
- Sanchita Rauth
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center at Omaha, Omaha, Nebraska
| | - Mokenge Malafa
- Department of Gastrointestinal Oncology, Moffitt Cancer Center, Tampa, Florida
| | - Moorthy P Ponnusamy
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center at Omaha, Omaha, Nebraska; Fred and Pamela Buffett Cancer Center, Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center at Omaha, Omaha, Nebraska.
| | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center at Omaha, Omaha, Nebraska; Fred and Pamela Buffett Cancer Center, Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center at Omaha, Omaha, Nebraska.
| |
Collapse
|
3
|
Vitorakis N, Gargalionis AN, Papavassiliou KA, Adamopoulos C, Papavassiliou AG. Precision Targeting Strategies in Pancreatic Cancer: The Role of Tumor Microenvironment. Cancers (Basel) 2024; 16:2876. [PMID: 39199647 PMCID: PMC11352254 DOI: 10.3390/cancers16162876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 08/14/2024] [Accepted: 08/17/2024] [Indexed: 09/01/2024] Open
Abstract
Pancreatic cancer demonstrates an ever-increasing incidence over the last years and represents one of the top causes of cancer-associated mortality. Cells of the tumor microenvironment (TME) interact with cancer cells in pancreatic ductal adenocarcinoma (PDAC) tumors to preserve cancer cells' metabolism, inhibit drug delivery, enhance immune suppression mechanisms and finally develop resistance to chemotherapy and immunotherapy. New strategies target TME genetic alterations and specific pathways in cell populations of the TME. Complex molecular interactions develop between PDAC cells and TME cell populations including cancer-associated fibroblasts, myeloid-derived suppressor cells, pancreatic stellate cells, tumor-associated macrophages, tumor-associated neutrophils, and regulatory T cells. In the present review, we aim to fully explore the molecular landscape of the pancreatic cancer TME cell populations and discuss current TME targeting strategies to provide thoughts for further research and preclinical testing.
Collapse
Affiliation(s)
- Nikolaos Vitorakis
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Antonios N Gargalionis
- Department of Clinical Biochemistry, 'Attikon' University General Hospital, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - Kostas A Papavassiliou
- First University Department of Respiratory Medicine, 'Sotiria' Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Christos Adamopoulos
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Athanasios G Papavassiliou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| |
Collapse
|
4
|
Chen M, Chen F, Gao Z, Li X, Hu L, Yang S, Zhao S, Song Z. CAFs and T cells interplay: The emergence of a new arena in cancer combat. Biomed Pharmacother 2024; 177:117045. [PMID: 38955088 DOI: 10.1016/j.biopha.2024.117045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 06/11/2024] [Accepted: 06/25/2024] [Indexed: 07/04/2024] Open
Abstract
The interaction between the immune system and the tumor matrix has a huge impact on the progression and treatment of cancer. This paper summarizes and discusses the crosstalk between T cells and cancer-associated fibroblasts (CAFs). CAFs can also produce inhibitors that counteract the function of T cells and promote tumor immune escape, while T cells can also engage in complex two-way interactions with CAFs through direct cell contact, the exchange of soluble factors such as cytokines, and the remodeling of the extracellular matrix. Precise targeted intervention can effectively reverse tumor-promoting crosstalk between T cells and CAFs, improve anti-tumor immune response, and provide a new perspective for cancer treatment. Therefore, it is important to deeply understand the mechanism of crosstalk between T cells and CAFs. This review aims to outline the underlying mechanisms of these interactions and discuss potential therapeutic strategies that may become fundamental tools in the treatment of cancer, especially hard-to-cure cancers.
Collapse
Affiliation(s)
- Minjie Chen
- Department of Surgery, the Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Fei Chen
- Department of Surgery, the Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Zhaofeng Gao
- Department of Surgery, the Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Xiaoping Li
- Department of Surgery, the Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Lingyu Hu
- Department of Surgery, the Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Shuying Yang
- Department of intensive medicine, The Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China.
| | - Siqi Zhao
- Department of Surgery, the Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China.
| | - Zhengwei Song
- Department of Surgery, the Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China.
| |
Collapse
|
5
|
Lin S, Zhou M, Cheng L, Shuai Z, Zhao M, Jie R, Wan Q, Peng F, Ding S. Exploring the association of POSTN + cancer-associated fibroblasts with triple-negative breast cancer. Int J Biol Macromol 2024; 268:131560. [PMID: 38631570 DOI: 10.1016/j.ijbiomac.2024.131560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Revised: 04/08/2024] [Accepted: 04/10/2024] [Indexed: 04/19/2024]
Abstract
Triple-negative breast cancer (TNBC) is a subtype of breast cancer with a poor prognosis. Cancer-associated fibroblasts (CAFs) play a critical role in regulating TNBC tumor development. This study aimed to identify and characterize a specific subtype of CAFs associated with TNBC. Initially, using high-throughput bulk transcriptomic data in two cohorts, we identified three CAF-related subtypes (CS1, CS2, CS3) in TNBC samples. These three CAFs subtypes were closely linked to the tumor microenvironment. The CS1 subtype exhibited a relatively immune-rich microenvironment and a favourable prognosis, whereas the CS3 subtype displayed an immune-deprived tumor microenvironment and an unfavourable prognosis. Through WGCNA analysis, POSTN was identified as a key biomarker for CAFs associated with TNBC. Then, POSTN+CAFs was identified and characterized. Both POSTN and POSTN+CAFs showed significant positive correlations with stromal molecules HGF and MET at both the transcriptional and protein levels. Specifically co-localized with CAFs in the tumor stromal area, POSTN, produced by POSTN+CAFs, could modulate the HGF-MET axis, serving as a bypass activation pathway to regulate tumor cell proliferation in response to EGFR inhibitor and MET inhibitor. This study underscores the significance of POSTN and POSTN+CAFs as crucial targets for the diagnosis and treatment of TNBC.
Collapse
Affiliation(s)
- Shuangyan Lin
- Department of Cell Biology and Department of Cardiovascular Surgery, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, 57 Zhugan Lane, Hangzhou 310000, Zhejiang, China; Department of Pathology, Zhejiang Hospital, Zhejiang University School of Medicine, 12 Lingyin Rd, Hangzhou 310013, Zhejiang, China
| | - Miaoni Zhou
- Department of Dermatology, Hangzhou Third People's Hospital, Affiliated Hangzhou Dermatology Hospital, Zhejiang University School of Medicine, 38 Xihu Rd, Hangzhou 310009, Zhejiang, China
| | - Liying Cheng
- Jiaxing University Medical College, 899 Shiguang Rd, Jiaxing 314001, Zhejiang, China
| | - Zhifeng Shuai
- Department of Pathology, Zhejiang Hospital, 12 Lingyin Rd, Hangzhou 310013, Zhejiang, China
| | - Mingyuan Zhao
- Department of Pathology, Zhejiang Hospital, 12 Lingyin Rd, Hangzhou 310013, Zhejiang, China
| | - Ruixia Jie
- Department of Pathology, Zhejiang Hospital, 12 Lingyin Rd, Hangzhou 310013, Zhejiang, China
| | - Qun Wan
- Department of Urinary Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou 310003, Zhejiang, China
| | - Fang Peng
- Department of Pathology, Zhejiang Hospital, 12 Lingyin Rd, Hangzhou 310013, Zhejiang, China.
| | - Shiping Ding
- Department of Cell Biology and Department of Cardiovascular Surgery, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, 57 Zhugan Lane, Hangzhou 310000, Zhejiang, China; Department of Cell Biology, Zhejiang University School of Medicine, 866 Yuhangtang Rd, Hangzhou 310058, Zhejiang, China.
| |
Collapse
|
6
|
Zhang Z, Tao J, Qiu J, Cao Z, Huang H, Xiao J, Zhang T. From basic research to clinical application: targeting fibroblast activation protein for cancer diagnosis and treatment. Cell Oncol (Dordr) 2024; 47:361-381. [PMID: 37726505 DOI: 10.1007/s13402-023-00872-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/30/2023] [Indexed: 09/21/2023] Open
Abstract
PURPOSE This study aims to review the multifaceted roles of a membrane protein named Fibroblast Activation Protein (FAP) expressed in tumor tissue, including its molecular functionalities, regulatory mechanisms governing its expression, prognostic significance, and its crucial role in cancer diagnosis and treatment. METHODS Articles that have uncovered the regulatory role of FAP in tumor, as well as its potential utility within clinical realms, spanning diagnosis to therapeutic intervention has been screened for a comprehensive review. RESULTS Our review reveals that FAP plays a pivotal role in solid tumor progression by undertaking a multitude of enzymatic and nonenzymatic roles within the tumor stroma. The exclusive presence of FAP within tumor tissues highlights its potential as a diagnostic marker and therapeutic target. The review also emphasizes the prognostic significance of FAP in predicting tumor progression and patient outcomes. Furthermore, the emerging strategies involving FAPI inhibitor (FAPI) in cancer research and clinical trials for PET/CT diagnosis are discussed. And targeted therapy utilizing FAP including FAPI, chimeric antigen receptor (CAR) T cell therapy, tumor vaccine, antibody-drug conjugates, bispecific T-cell engagers, FAP cleavable prodrugs, and drug delivery system are also introduced. CONCLUSION FAP's intricate interactions with tumor cells and the tumor microenvironment make it a promising target for diagnosis and treatment. Promising strategies such as FAPI offer potential avenues for accurate tumor diagnosis, while multiple therapeutic strategies highlight the prospects of FAP targeting treatments which needs further clinical evaluation.
Collapse
Affiliation(s)
- Zeyu Zhang
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Jinxin Tao
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Jiangdong Qiu
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Zhe Cao
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Hua Huang
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Jianchun Xiao
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Taiping Zhang
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, 100730, China.
- Clinical Immunology Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| |
Collapse
|
7
|
Gehris J, Ervin C, Hawkins C, Womack S, Churillo AM, Doyle J, Sinusas AJ, Spinale FG. Fibroblast activation protein: Pivoting cancer/chemotherapeutic insight towards heart failure. Biochem Pharmacol 2024; 219:115914. [PMID: 37956895 PMCID: PMC10824141 DOI: 10.1016/j.bcp.2023.115914] [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: 08/25/2023] [Revised: 11/06/2023] [Accepted: 11/07/2023] [Indexed: 11/21/2023]
Abstract
An important mechanism for cancer progression is degradation of the extracellular matrix (ECM) which is accompanied by the emergence and proliferation of an activated fibroblast, termed the cancer associated fibroblast (CAF). More specifically, an enzyme pathway identified to be amplified with local cancer progression and proliferation of the CAF, is fibroblast activation protein (FAP). The development and progression of heart failure (HF) irrespective of the etiology is associated with left ventricular (LV) remodeling and changes in ECM structure and function. As with cancer, HF progression is associated with a change in LV myocardial fibroblast growth and function, and expresses a protein signature not dissimilar to the CAF. The overall goal of this review is to put forward the postulate that scientific discoveries regarding FAP in cancer as well as the development of specific chemotherapeutics could be pivoted to target the emergence of FAP in the activated fibroblast subtype and thus hold translationally relevant diagnostic and therapeutic targets in HF.
Collapse
Affiliation(s)
- John Gehris
- Cell Biology and Anatomy and Cardiovascular Research Center, University of South Carolina School of Medicine and the Columbia VA Health Care System, Columbia, SC, United States
| | - Charlie Ervin
- Cell Biology and Anatomy and Cardiovascular Research Center, University of South Carolina School of Medicine and the Columbia VA Health Care System, Columbia, SC, United States
| | - Charlotte Hawkins
- Cell Biology and Anatomy and Cardiovascular Research Center, University of South Carolina School of Medicine and the Columbia VA Health Care System, Columbia, SC, United States
| | - Sydney Womack
- Cell Biology and Anatomy and Cardiovascular Research Center, University of South Carolina School of Medicine and the Columbia VA Health Care System, Columbia, SC, United States
| | - Amelia M Churillo
- Cell Biology and Anatomy and Cardiovascular Research Center, University of South Carolina School of Medicine and the Columbia VA Health Care System, Columbia, SC, United States
| | - Jonathan Doyle
- Cell Biology and Anatomy and Cardiovascular Research Center, University of South Carolina School of Medicine and the Columbia VA Health Care System, Columbia, SC, United States
| | - Albert J Sinusas
- Yale University Cardiovascular Imaging Center, New Haven CT, United States
| | - Francis G Spinale
- Cell Biology and Anatomy and Cardiovascular Research Center, University of South Carolina School of Medicine and the Columbia VA Health Care System, Columbia, SC, United States.
| |
Collapse
|
8
|
Xu Y, Li W, Lin S, Liu B, Wu P, Li L. Fibroblast diversity and plasticity in the tumor microenvironment: roles in immunity and relevant therapies. Cell Commun Signal 2023; 21:234. [PMID: 37723510 PMCID: PMC10506315 DOI: 10.1186/s12964-023-01204-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 06/22/2023] [Indexed: 09/20/2023] Open
Abstract
Cancer-associated fibroblasts (CAFs), enriched in the tumor stroma, have received increasing attention because of their multifaceted effects on tumorigenesis, development, metastasis, and treatment resistance in malignancies. CAFs contributed to suppressive microenvironment via different mechanisms, while CAFs also exerted some antitumor effects. Therefore, CAFs have been considered promising therapeutic targets for their remarkable roles in malignant tumors. However, patients with malignancies failed to benefit from current CAFs-targeted drugs in many clinical trials, which suggests that further in-depth investigation into CAFs is necessary. Here, we summarize and outline the heterogeneity and plasticity of CAFs mainly by exploring their origin and activation, highlighting the regulation of CAFs in the tumor microenvironment during tumor evolution, as well as the critical roles performed by CAFs in tumor immunity. In addition, we summarize the current immunotherapies targeting CAFs, and conclude with a brief overview of some prospects for the future of CAFs research in the end. Video Abstract.
Collapse
Affiliation(s)
- Yashi Xu
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- National Clinical Research Center for Gynecology and Obstetrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Li
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- National Clinical Research Center for Gynecology and Obstetrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Gynecologic Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Shitong Lin
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- National Clinical Research Center for Gynecology and Obstetrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Binghan Liu
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- National Clinical Research Center for Gynecology and Obstetrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Peng Wu
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
- National Clinical Research Center for Gynecology and Obstetrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Li Li
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
- National Clinical Research Center for Gynecology and Obstetrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Department of Gynecologic Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| |
Collapse
|
9
|
Li Y, Zhao H, Hu S, Zhang X, Chen H, Zheng Q. PET imaging with [ 68Ga]-labeled TGFβ-targeting peptide in a mouse PANC-1 tumor model. Front Oncol 2023; 13:1228281. [PMID: 37781175 PMCID: PMC10540840 DOI: 10.3389/fonc.2023.1228281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 08/24/2023] [Indexed: 10/03/2023] Open
Abstract
Purpose Transforming growth factor β (TGFβ) is upregulated in many types of tumors and plays important roles in tumor microenvironment construction, immune escape, invasion, and metastasis. The therapeutic effect of antibodies and nuclide-conjugated drugs targeting TGFβ has not been ideal. Targeting TGFβ with small-molecule or peptide carriers labeled with diagnostic/therapeutic nuclides is a new development direction. This study aimed to explore and confirm the imaging diagnostic efficiency of TGFβ-targeting peptide P144 coupled with [68Ga] in a PANC-1 tumor model. Procedures TGFβ-targeting inhibitory peptide P144 with stable activity was prepared through peptide synthesis and screening, and P144 was coupled with biological chelator DOTA and labeled with radionuclide [68Ga] to achieve a stable TGFβ-targeting tracer [68Ga]Ga-P144. This tracer was first used for positron emission tomography (PET) molecular imaging study of pancreatic cancer in a mouse PANC-1 tumor model. Results [68Ga]Ga-P144 had a high targeted uptake and relatively long uptake retention time in tumors and lower uptakes in non-target organs and backgrounds. Target pre-blocking experiment with the cold drug P144-DOTA demonstrated that the radioactive uptake with [68Ga]Ga-P144 PET in vivo, especially in tumor tissue, had a high TGFβ-targeting specificity. [68Ga]Ga-P144 PET had ideal imaging efficiency in PANC-1 tumor-bearing mice, with high specificity in vivo and good tumor-targeting effect. Conclusion [68Ga]Ga-P144 has relatively high specificity and tumor-targeted uptake and may be developed as a promising diagnostic tool for TGFβ-positive malignancies.
Collapse
Affiliation(s)
- Yong Li
- Department of Nuclear Medicine, Shenzhen Hospital of Southern Medical University, Shenzhen, China
| | - Hong Zhao
- Department of Nuclear Medicine, Shenzhen People’s Hospital, Shenzhen, China
| | - Shan Hu
- Department of Nuclear Medicine, Shenzhen Hospital of Southern Medical University, Shenzhen, China
| | - Xichen Zhang
- Department of Nuclear Medicine, Shenzhen Hospital of Southern Medical University, Shenzhen, China
| | - Haojian Chen
- Department of Nuclear Medicine, Shenzhen Hospital of Southern Medical University, Shenzhen, China
| | - Qihuang Zheng
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
10
|
Ora M, Soni N, Nazar AH, Dixit M, Singh R, Puri S, Graham MM, Gambhir S. Fibroblast Activation Protein Inhibitor-Based Radionuclide Therapies: Current Status and Future Directions. J Nucl Med 2023:jnumed.123.265594. [PMID: 37268422 DOI: 10.2967/jnumed.123.265594] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 03/30/2023] [Indexed: 06/04/2023] Open
Abstract
Metastatic malignancies have limited management strategies and variable treatment responses. Cancer cells develop beside and depend on the complex tumor microenvironment. Cancer-associated fibroblasts, with their complex interaction with tumor and immune cells, are involved in various steps of tumorigenesis, such as growth, invasion, metastasis, and treatment resistance. Prooncogenic cancer-associated fibroblasts emerged as attractive therapeutic targets. However, clinical trials have achieved suboptimal success. Fibroblast activation protein (FAP) inhibitor-based molecular imaging has shown encouraging results in cancer diagnosis, making them innovative targets for FAP inhibitor-based radionuclide therapies. This review summarizes the results of preclinical and clinical FAP-based radionuclide therapies. We will describe advances and FAP molecule modification in this novel therapy, as well as its dosimetry, safety profile, and efficacy. This summary may guide future research directions and optimize clinical decision-making in this emerging field.
Collapse
Affiliation(s)
- Manish Ora
- Department of Nuclear Medicine, SGPGIMS, Lucknow, India;
| | - Neetu Soni
- Department of Radiology, University of Rochester Medical Center, Rochester, New York
| | | | - Manish Dixit
- Department of Nuclear Medicine, SGPGIMS, Lucknow, India
| | - Rohit Singh
- Division of Hematology-Oncology, University of Vermont Medical Center, Burlington, Vermont; and
| | - Savita Puri
- Department of Radiology, University of Rochester Medical Center, Rochester, New York
| | - Michael M Graham
- Division of Nuclear Medicine, Department of Radiology, University of Iowa Health Care, Iowa City, Iowa
| | | |
Collapse
|
11
|
Bruni S, Mercogliano MF, Mauro FL, Cordo Russo RI, Schillaci R. Cancer immune exclusion: breaking the barricade for a successful immunotherapy. Front Oncol 2023; 13:1135456. [PMID: 37284199 PMCID: PMC10239871 DOI: 10.3389/fonc.2023.1135456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Accepted: 05/10/2023] [Indexed: 06/08/2023] Open
Abstract
Immunotherapy has changed the course of cancer treatment. The initial steps were made through tumor-specific antibodies that guided the setup of an antitumor immune response. A new and successful generation of antibodies are designed to target immune checkpoint molecules aimed to reinvigorate the antitumor immune response. The cellular counterpart is the adoptive cell therapy, where specific immune cells are expanded or engineered to target cancer cells. In all cases, the key for achieving positive clinical resolutions rests upon the access of immune cells to the tumor. In this review, we focus on how the tumor microenvironment architecture, including stromal cells, immunosuppressive cells and extracellular matrix, protects tumor cells from an immune attack leading to immunotherapy resistance, and on the available strategies to tackle immune evasion.
Collapse
|
12
|
Szabo PM, Vajdi A, Kumar N, Tolstorukov MY, Chen BJ, Edwards R, Ligon KL, Chasalow SD, Chow KH, Shetty A, Bolisetty M, Holloway JL, Golhar R, Kidd BA, Hull PA, Houser J, Vlach L, Siemers NO, Saha S. Cancer-associated fibroblasts are the main contributors to epithelial-to-mesenchymal signatures in the tumor microenvironment. Sci Rep 2023; 13:3051. [PMID: 36810872 PMCID: PMC9944255 DOI: 10.1038/s41598-023-28480-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 01/19/2023] [Indexed: 02/24/2023] Open
Abstract
Epithelial-to-mesenchymal transition (EMT) is associated with tumor initiation, metastasis, and drug resistance. However, the mechanisms underlying these associations are largely unknown. We studied several tumor types to identify the source of EMT gene expression signals and a potential mechanism of resistance to immuno-oncology treatment. Across tumor types, EMT-related gene expression was strongly associated with expression of stroma-related genes. Based on RNA sequencing of multiple patient-derived xenograft models, EMT-related gene expression was enriched in the stroma versus parenchyma. EMT-related markers were predominantly expressed by cancer-associated fibroblasts (CAFs), cells of mesenchymal origin which produce a variety of matrix proteins and growth factors. Scores derived from a 3-gene CAF transcriptional signature (COL1A1, COL1A2, COL3A1) were sufficient to reproduce association between EMT-related markers and disease prognosis. Our results suggest that CAFs are the primary source of EMT signaling and have potential roles as biomarkers and targets for immuno-oncology therapies.
Collapse
Affiliation(s)
- Peter M. Szabo
- grid.419971.30000 0004 0374 8313Bristol Myers Squibb, Princeton, NJ USA ,grid.428458.70000 0004 1792 8104Present Address: Fate Therapeutics, San Diego, CA USA
| | - Amir Vajdi
- grid.65499.370000 0001 2106 9910Dana-Farber Cancer Institute, Boston, MA USA ,grid.417993.10000 0001 2260 0793Present Address: Merck & Co., Inc., Kenilworth, NJ USA
| | | | | | - Benjamin J. Chen
- grid.419971.30000 0004 0374 8313Bristol Myers Squibb, Cambridge, MA USA
| | - Robin Edwards
- grid.419971.30000 0004 0374 8313Bristol Myers Squibb, Princeton, NJ USA ,grid.428496.5Present Address: Daiichi Sankyo, Inc., Princeton, NJ USA
| | - Keith L. Ligon
- grid.65499.370000 0001 2106 9910Dana-Farber Cancer Institute, Boston, MA USA
| | - Scott D. Chasalow
- grid.419971.30000 0004 0374 8313Bristol Myers Squibb, Princeton, NJ USA
| | - Kin-Hoe Chow
- grid.65499.370000 0001 2106 9910Dana-Farber Cancer Institute, Boston, MA USA
| | - Aniket Shetty
- grid.65499.370000 0001 2106 9910Dana-Farber Cancer Institute, Boston, MA USA
| | - Mohan Bolisetty
- grid.419971.30000 0004 0374 8313Bristol Myers Squibb, Princeton, NJ USA
| | - James L. Holloway
- grid.419971.30000 0004 0374 8313Bristol Myers Squibb, Seattle, WA USA
| | - Ryan Golhar
- grid.419971.30000 0004 0374 8313Bristol Myers Squibb, Princeton, NJ USA
| | - Brian A. Kidd
- grid.419971.30000 0004 0374 8313Bristol Myers Squibb, Redwood City, CA USA
| | | | - Jeff Houser
- grid.419971.30000 0004 0374 8313Bristol Myers Squibb, Redwood City, CA USA
| | - Logan Vlach
- grid.419971.30000 0004 0374 8313Bristol Myers Squibb, Redwood City, CA USA ,grid.152326.10000 0001 2264 7217Present Address: Vanderbilt University, Nashville, TN USA
| | - Nathan O. Siemers
- grid.419971.30000 0004 0374 8313Bristol Myers Squibb, Princeton, NJ USA ,Present Address: Fiveprime Group, Monterey, CA USA
| | - Saurabh Saha
- grid.419971.30000 0004 0374 8313Bristol Myers Squibb, Princeton, NJ USA ,Present Address: Centessa Pharmaceuticals, Cambridge, MA USA
| |
Collapse
|
13
|
Dong Y, Zhou H, Alhaskawi A, Wang Z, Lai J, Yao C, Liu Z, Hasan Abdullah Ezzi S, Goutham Kota V, Hasan Abdulla Hasan Abdulla M, Lu H. The Superiority of Fibroblast Activation Protein Inhibitor (FAPI) PET/CT Versus FDG PET/CT in the Diagnosis of Various Malignancies. Cancers (Basel) 2023; 15:cancers15041193. [PMID: 36831535 PMCID: PMC9954090 DOI: 10.3390/cancers15041193] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 02/02/2023] [Accepted: 02/09/2023] [Indexed: 02/16/2023] Open
Abstract
Cancer represents a major cause of death worldwide and is characterized by the uncontrolled proliferation of abnormal cells that escape immune regulation. It is now understood that cancer-associated fibroblasts (CAFs), which express specific fibroblast activation protein (FAP), are critical participants in tumor development and metastasis. Researchers have developed various FAP-targeted probes for imaging of different tumors from antibodies to boronic acid-based inhibitor molecules and determined that quinoline-based FAP inhibitors (FAPIs) are the most appropriate candidate as the radiopharmaceutical for FAPI PET/CT imaging. When applied clinically, FAPI PET/CT yielded satisfactory results. Over the past few years, the utility and effectiveness of tumor detection and staging of FAPI PET/CT have been compared with FDG PET/CT in various aspects, including standardized uptake values (SUVs), rate of absorbance and clearance. This review summarizes the development and clinical application of FAPI PET/CT, emphasizing the diagnosis and management of various tumor types and the future prospects of FAPI imaging.
Collapse
Affiliation(s)
- Yanzhao Dong
- Department of Orthopedics, The First Affiliated Hospital of Zhejiang University, #79 Qingchun Road, Hangzhou 310003, China
| | - Haiying Zhou
- Department of Orthopedics, The First Affiliated Hospital of Zhejiang University, #79 Qingchun Road, Hangzhou 310003, China
| | - Ahmad Alhaskawi
- Department of Orthopedics, The First Affiliated Hospital of Zhejiang University, #79 Qingchun Road, Hangzhou 310003, China
| | - Zewei Wang
- School of Medicine, Zhejiang University, #866 Yuhangtang Road, Hangzhou 310058, China
| | - Jingtian Lai
- School of Medicine, Zhejiang University, #866 Yuhangtang Road, Hangzhou 310058, China
| | - Chengjun Yao
- School of Medicine, Zhejiang University, #866 Yuhangtang Road, Hangzhou 310058, China
| | - Zhenfeng Liu
- Department of Nuclear Medicine, The First Affiliated Hospital of Zhejiang University, #79 Qingchun Road, Hangzhou 310003, China
| | - Sohaib Hasan Abdullah Ezzi
- Department of Orthopaedics, Third Xiangya Hospital of Central South University, #138 Tongzipo Road, Changsha 410013, China
| | - Vishnu Goutham Kota
- School of Medicine, Zhejiang University, #866 Yuhangtang Road, Hangzhou 310058, China
| | | | - Hui Lu
- Department of Orthopedics, The First Affiliated Hospital of Zhejiang University, #79 Qingchun Road, Hangzhou 310003, China
- Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Zhejiang University, #866 Yuhangtang Road, Hangzhou 310058, China
- Correspondence: ; Tel.: +86-0571-87236121
| |
Collapse
|
14
|
Zulaziz N, Chai SJ, Lim KP. The origins, roles and therapies of cancer associated fibroblast in liver cancer. Front Oncol 2023; 13:1151373. [PMID: 37035187 PMCID: PMC10076538 DOI: 10.3389/fonc.2023.1151373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 03/03/2023] [Indexed: 04/11/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the most common form of liver cancer. It is often preceded by chronic inflammation such as liver fibrosis and cirrhosis. Different cell types are believed to give rise to liver-specific cancer associated fibroblast (CAF), these include resident fibroblast, hepatic stellate cell, liver cancer cell, hepatic sinusoidal endothelial cell and mesenchymal stromal cell. The abundance of fibroblasts has contributed to the cancer progression, immune modulation and treatment resistance in HCC. In this review, we discussed the origins, subtypes and roles of cancer associated fibroblasts in HCC. Their specific roles in shaping the tumor microenvironment, facilitating cancer growth, and modulating different immune cell types to confer a permissive environment for cancer growth. CAF is now an attractive therapeutic target for cancer treatment, however specific therapeutic development in HCC is still lacking. Hence, we have included preclinical and clinical development of CAF-specific interventions for other cancer types in this review. However, most CAF-specific therapies have resulted in disappointing clinical outcomes, likely due to the difficulties in differentiating CAF from normal fibroblast. A thorough understanding of the characteristics and functionalities of CAF is warranted to further improve the therapeutic efficacy of anti-CAF therapies.
Collapse
|
15
|
Toledo B, Picon-Ruiz M, Marchal JA, Perán M. Dual Role of Fibroblasts Educated by Tumour in Cancer Behavior and Therapeutic Perspectives. Int J Mol Sci 2022; 23:15576. [PMID: 36555218 PMCID: PMC9778751 DOI: 10.3390/ijms232415576] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 11/25/2022] [Accepted: 12/03/2022] [Indexed: 12/13/2022] Open
Abstract
Tumours are complex systems with dynamic interactions between tumour cells, non-tumour cells, and extracellular components that comprise the tumour microenvironment (TME). The majority of TME's cells are cancer-associated fibroblasts (CAFs), which are crucial in extracellular matrix (ECM) construction, tumour metabolism, immunology, adaptive chemoresistance, and tumour cell motility. CAF subtypes have been identified based on the expression of protein markers. CAFs may act as promoters or suppressors in tumour cells depending on a variety of factors, including cancer stage. Indeed, CAFs have been shown to promote tumour growth, survival and spread, and secretome changes, but they can also slow tumourigenesis at an early stage through mechanisms that are still poorly understood. Stromal-cancer interactions are governed by a variety of soluble factors that determine the outcome of the tumourigenic process. Cancer cells release factors that enhance the ability of fibroblasts to secrete multiple tumour-promoting chemokines, acting on malignant cells to promote proliferation, migration, and invasion. This crosstalk between CAFs and tumour cells has given new prominence to the stromal cells, from being considered as mere physical support to becoming key players in the tumour process. Here, we focus on the concept of cancer as a non-healing wound and the relevance of chronic inflammation to tumour initiation. In addition, we review CAFs heterogeneous origins and markers together with the potential therapeutic implications of CAFs "re-education" and/or targeting tumour progression inhibition.
Collapse
Affiliation(s)
- Belén Toledo
- Department of Health Sciences, University of Jaén, E-23071 Jaén, Spain
| | - Manuel Picon-Ruiz
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of Granada, E-18100 Granada, Spain
- Instituto de Investigación Sanitaria ibs. GRANADA, Hospitales Universitarios de Granada-Universidad de Granada, E-18071 Granada, Spain
- Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, E-18016 Granada, Spain
- Excellence Research Unit “Modeling Nature” (MNat), University of Granada, E-18016 Granada, Spain
| | - Juan Antonio Marchal
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of Granada, E-18100 Granada, Spain
- Instituto de Investigación Sanitaria ibs. GRANADA, Hospitales Universitarios de Granada-Universidad de Granada, E-18071 Granada, Spain
- Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, E-18016 Granada, Spain
- Excellence Research Unit “Modeling Nature” (MNat), University of Granada, E-18016 Granada, Spain
| | - Macarena Perán
- Department of Health Sciences, University of Jaén, E-23071 Jaén, Spain
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of Granada, E-18100 Granada, Spain
- Excellence Research Unit “Modeling Nature” (MNat), University of Granada, E-18016 Granada, Spain
| |
Collapse
|
16
|
Eulberg D, Frömming A, Lapid K, Mangasarian A, Barak A. The prospect of tumor microenvironment-modulating therapeutical strategies. Front Oncol 2022; 12:1070243. [PMID: 36568151 PMCID: PMC9772844 DOI: 10.3389/fonc.2022.1070243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 11/10/2022] [Indexed: 12/13/2022] Open
Abstract
Multiple mechanisms promote tumor prosperity, which does not only depend on cell-autonomous, inherent abnormal characteristics of the malignant cells that facilitate rapid cell division and tumor expansion. The neoplastic tissue is embedded in a supportive and dynamic tumor microenvironment (TME) that nurtures and protects the malignant cells, maintaining and perpetuating malignant cell expansion. The TME consists of different elements, such as atypical vasculature, various innate and adaptive immune cells with immunosuppressive or pro-inflammatory properties, altered extracellular matrix (ECM), activated stromal cells, and a wide range of secreted/stroma-tethered bioactive molecules that contribute to malignancy, directly or indirectly. In this review, we describe the various TME components and provide examples of anti-cancer therapies and novel drugs under development that aim to target these components rather than the intrinsic processes within the malignant cells. Combinatory TME-modulating therapeutic strategies may be required to overcome the resistance to current treatment options and prevent tumor recurrence.
Collapse
|
17
|
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: 97] [Impact Index Per Article: 32.3] [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
|
18
|
Zhu Y, Li X, Wang L, Hong X, Yang J. Metabolic reprogramming and crosstalk of cancer-related fibroblasts and immune cells in the tumor microenvironment. Front Endocrinol (Lausanne) 2022; 13:988295. [PMID: 36046791 PMCID: PMC9421293 DOI: 10.3389/fendo.2022.988295] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 07/25/2022] [Indexed: 12/13/2022] Open
Abstract
It is notorious that cancer cells alter their metabolism to adjust to harsh environments of hypoxia and nutritional starvation. Metabolic reprogramming most often occurs in the tumor microenvironment (TME). TME is defined as the cellular environment in which the tumor resides. This includes surrounding blood vessels, fibroblasts, immune cells, signaling molecules and the extracellular matrix (ECM). It is increasingly recognized that cancer cells, fibroblasts and immune cells within TME can regulate tumor progression through metabolic reprogramming. As the most significant proportion of cells among all the stromal cells that constitute TME, cancer-associated fibroblasts (CAFs) are closely associated with tumorigenesis and progression. Multitudinous studies have shown that CAFs participate in and promote tumor metabolic reprogramming and exert regulatory effects via the dysregulation of metabolic pathways. Previous studies have demonstrated that curbing the substance exchange between CAFs and tumor cells can dramatically restrain tumor growth. Emerging studies suggest that CAFs within the TME have emerged as important determinants of metabolic reprogramming. Metabolic reprogramming also occurs in the metabolic pattern of immune cells. In the meanwhile, immune cell phenotype and functions are metabolically regulated. Notably, immune cell functions influenced by metabolic programs may ultimately lead to alterations in tumor immunity. Despite the fact that multiple previous researches have been devoted to studying the interplays between different cells in the tumor microenvironment, the complicated relationship between CAFs and immune cells and implications of metabolic reprogramming remains unknown and requires further investigation. In this review, we discuss our current comprehension of metabolic reprogramming of CAFs and immune cells (mainly glucose, amino acid, and lipid metabolism) and crosstalk between them that induces immune responses, and we also highlight their contributions to tumorigenesis and progression. Furthermore, we underscore potential therapeutic opportunities arising from metabolism dysregulation and metabolic crosstalk, focusing on strategies targeting CAFs and immune cell metabolic crosstalk in cancer immunotherapy.
Collapse
Affiliation(s)
- Yifei Zhu
- School of Medicine, Southeast University, Nanjing, China
| | - Xinyan Li
- School of Medicine, Southeast University, Nanjing, China
| | - Lei Wang
- School of Medicine, Southeast University, Nanjing, China
| | - Xiwei Hong
- School of Medicine, Southeast University, Nanjing, China
| | - Jie Yang
- Department of General surgery, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China
| |
Collapse
|
19
|
Greifenstein L, Kramer CS, Moon ES, Rösch F, Klega A, Landvogt C, Müller C, Baum RP. From Automated Synthesis to In Vivo Application in Multiple Types of Cancer-Clinical Results with [ 68Ga]Ga-DATA 5m.SA.FAPi. Pharmaceuticals (Basel) 2022; 15:1000. [PMID: 36015148 PMCID: PMC9415298 DOI: 10.3390/ph15081000] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 07/29/2022] [Accepted: 08/09/2022] [Indexed: 11/16/2022] Open
Abstract
Radiolabeled FAPI (fibroblast activation protein inhibitors) recently gained attention as widely applicable imaging and potential therapeutic compounds targeting CAF (cancer-associated fibroblasts) or DAF (disease-associated fibroblasts in benign disorders). Moreover, the use of FAPI has distinct advantages compared to FDG (e.g., increased sensitivity in regions with high glucose metabolism, no need for fasting, and rapid imaging). In this study, we wanted to evaluate the radiochemical synthesis and the clinical properties of the new CAF-targeting tracer [68Ga]Ga-DATA5m.SA.FAPi. The compound consists of a (radio)chemically easy to use hybrid chelate DATA.SA, which can be labeled at low temperatures, making it an interesting molecule for 'instant kit-type' labeling, and a squaric acid moiety that provides distinct advantages for synthesis and radiolabeling. Our work demonstrates that automatic synthesis of the FAP inhibitor [68Ga]Ga-DATA5m.SA.FAPi is feasible and reproducible, providing convenient access to this new hybrid chelator-based tracer. Our studies demonstrated the diagnostic usability of [68Ga]Ga-DATA5m.SA.FAPi for the unambiguous detection of cancer-associated fibroblasts of various carcinomas and their metastases (NSCLC, liposarcoma, parotid tumors, prostate cancer, and pancreas adenocarcinoma), while physiological uptake in brain, liver, intestine, bone, and lungs was very low.
Collapse
Affiliation(s)
- Lukas Greifenstein
- CURANOSTICUM Wiesbaden-Frankfurt, Center for Advanced Radiomolecular Precision Oncology, 65191 Wiesbaden, Germany
| | - Carsten S. Kramer
- CURANOSTICUM Wiesbaden-Frankfurt, Center for Advanced Radiomolecular Precision Oncology, 65191 Wiesbaden, Germany
| | - Euy Sung Moon
- Department of Chemistry–TRIGA, Institute of Nuclear Chemistry, Johannes Gutenberg University, 55128 Mainz, Germany
| | - Frank Rösch
- Department of Chemistry–TRIGA, Institute of Nuclear Chemistry, Johannes Gutenberg University, 55128 Mainz, Germany
| | - Andre Klega
- CURANOSTICUM Wiesbaden-Frankfurt, Center for Advanced Radiomolecular Precision Oncology, 65191 Wiesbaden, Germany
| | - Christian Landvogt
- CURANOSTICUM Wiesbaden-Frankfurt, Center for Advanced Radiomolecular Precision Oncology, 65191 Wiesbaden, Germany
| | - Corinna Müller
- CURANOSTICUM Wiesbaden-Frankfurt, Center for Advanced Radiomolecular Precision Oncology, 65191 Wiesbaden, Germany
| | - Richard P. Baum
- CURANOSTICUM Wiesbaden-Frankfurt, Center for Advanced Radiomolecular Precision Oncology, 65191 Wiesbaden, Germany
| |
Collapse
|
20
|
Meng Y, Yu J, Zhu M, Zhou J, Li N, Liu F, Zhang H, Fang X, Li J, Feng X, Wang L, Jiang H, Lu J, Shao C, Bian Y. CT radiomics signature: a potential biomarker for fibroblast activation protein expression in patients with pancreatic ductal adenocarcinoma. Abdom Radiol (NY) 2022; 47:2822-2834. [PMID: 35451626 DOI: 10.1007/s00261-022-03512-6] [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: 12/13/2021] [Revised: 03/25/2022] [Accepted: 03/25/2022] [Indexed: 01/18/2023]
Abstract
PURPOSE To develop and validate a radiomics model to predict fibroblast activation protein (FAP) expression in patients with pancreatic ductal adenocarcinoma (PDAC). METHODS This retrospective study included consecutive 152 patients with PDAC who underwent MDCT scan and surgical resection from January 2017 to December 2017 (training set) and from January 2018 to April 2018 (validation set). In the training set, 1409 portal radiomic features were extracted from each patient's preoperative imaging. Optimal features were selected using the least absolute shrinkage and selection operator (LASSO) logistic regression algorithm, whereupon the extreme gradient boosting (XGBoost) was developed using the radiomics features. The performance of the XGBoost classifier performance was assessed by its calibration, discrimination, and clinical usefulness. RESULTS The patients were divided into FAP-low (n = 91; 59.87%) and FAP-high (n = 61; 40.13%) groups according to the optimal FAP cutoff (45.71%). Patients in the FAP-low group showed longer survival. The XGBoost classifier comprised 13 selected radiomics features and showed good discrimination in the training set [area under the curve (AUC), 0.97] and the validation set (AUC, 0.75). It also performed well in the calibration test and decision-curve analysis, demonstrating its potential clinical value. CONCLUSIONS The XGBoost classifier based on CT radiomics in the portal venous phase can non-invasively predict FAP expression and may help to improve clinical decision-making in patients with PDAC.
Collapse
Affiliation(s)
- Yinghao Meng
- Department of Radiology, Changhai Hospital, Naval Medical University, Shanghai, China
- Department of Radiology, No. 971 Hospital of Navy, Qingdao, Shandong, China
| | - Jieyu Yu
- Department of Radiology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Mengmeng Zhu
- Department of Radiology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Jian Zhou
- Department of Radiology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Na Li
- Department of Radiology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Fang Liu
- Department of Radiology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Hao Zhang
- Department of Radiology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Xu Fang
- Department of Radiology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Jing Li
- Department of Radiology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Xiaocheng Feng
- Department of Radiology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Li Wang
- Department of Radiology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Hui Jiang
- Department of Pathology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Jianping Lu
- Department of Radiology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Chengwei Shao
- Department of Radiology, Changhai Hospital, Naval Medical University, Shanghai, China.
- Department of Radiology, Changhai Hospital, 168 Changhai Road, Shanghai, 200433, China.
| | - Yun Bian
- Department of Radiology, Changhai Hospital, Naval Medical University, Shanghai, China.
- Department of Radiology, Changhai Hospital, 168 Changhai Road, Shanghai, 200433, China.
| |
Collapse
|
21
|
Papait A, Romoli J, Stefani FR, Chiodelli P, Montresor MC, Agoni L, Silini AR, Parolini O. Fight the Cancer, Hit the CAF! Cancers (Basel) 2022; 14:cancers14153570. [PMID: 35892828 PMCID: PMC9330284 DOI: 10.3390/cancers14153570] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 07/19/2022] [Accepted: 07/19/2022] [Indexed: 11/16/2022] Open
Abstract
Simple Summary In the last 20 years, the tumor microenvironment (TME) has raised an increasing interest from the therapeutic point of view. Indeed, different strategies targeting either the endothelial or the immune component have been implemented. Furthermore, cancer-associated fibroblasts (CAF) have attracted even more interest due to their ability to prime the TME in order to favor tumor progression and metastasis. This current review provides a comprehensive overview on the latest discoveries regarding CAF, more specifically on their complex characterization and on preclinical studies and clinical trials that target CAF within the TME. Abstract The tumor microenvironment (TME) is comprised of different cellular components, such as immune and stromal cells, which co-operate in unison to promote tumor progression and metastasis. In the last decade, there has been an increasing focus on one specific component of the TME, the stromal component, often referred to as Cancer-Associated Fibroblasts (CAF). CAF modulate the immune response and alter the composition of the extracellular matrix with a decisive impact on the response to immunotherapies and conventional chemotherapy. The most recent publications based on single-cell analysis have underlined CAF heterogeneity and the unique plasticity that strongly impact the TME. In this review, we focus not only on the characterization of CAF based on the most recent findings, but also on their impact on the immune system. We also discuss clinical trials and preclinical studies where targeting CAF revealed controversial results. Therefore, future efforts should focus on understanding the functional properties of individual subtypes of CAF, taking into consideration the peculiarities of each pathological context.
Collapse
Affiliation(s)
- Andrea Papait
- Department of Life Science and Public Health, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (A.P.); (J.R.)
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, 00168 Rome, Italy
| | - Jacopo Romoli
- Department of Life Science and Public Health, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (A.P.); (J.R.)
- Centro di Ricerca E. Menni, Fondazione Poliambulanza Istituto Ospedaliero, 25124 Brescia, Italy; (F.R.S.); (P.C.); (A.R.S.)
| | - Francesca Romana Stefani
- Centro di Ricerca E. Menni, Fondazione Poliambulanza Istituto Ospedaliero, 25124 Brescia, Italy; (F.R.S.); (P.C.); (A.R.S.)
| | - Paola Chiodelli
- Centro di Ricerca E. Menni, Fondazione Poliambulanza Istituto Ospedaliero, 25124 Brescia, Italy; (F.R.S.); (P.C.); (A.R.S.)
| | | | - Lorenzo Agoni
- Obstetrics and Gynecology Unit, Fondazione Poliambulanza Istituto Ospedaliero, 25124 Brescia, Italy;
| | - Antonietta Rosa Silini
- Centro di Ricerca E. Menni, Fondazione Poliambulanza Istituto Ospedaliero, 25124 Brescia, Italy; (F.R.S.); (P.C.); (A.R.S.)
| | - Ornella Parolini
- Department of Life Science and Public Health, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (A.P.); (J.R.)
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, 00168 Rome, Italy
- Correspondence: ; Tel.: +39-0630154464
| |
Collapse
|
22
|
Duhen T, Gough MJ, Leidner RS, Stanton SE. Development and therapeutic manipulation of the head and neck cancer tumor environment to improve clinical outcomes. FRONTIERS IN ORAL HEALTH 2022; 3:902160. [PMID: 35937775 PMCID: PMC9354490 DOI: 10.3389/froh.2022.902160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 06/27/2022] [Indexed: 11/13/2022] Open
Abstract
The clinical response to cancer therapies involves the complex interplay between the systemic, tumoral, and stromal immune response as well as the direct impact of treatments on cancer cells. Each individual's immunological and cancer histories are different, and their carcinogen exposures may differ. This means that even though two patients with oral tumors may carry an identical mutation in TP53, they are likely to have different pre-existing immune responses to their tumors. These differences may arise due to their distinct accessory mutations, genetic backgrounds, and may relate to clinical factors including previous chemotherapy exposure and concurrent medical comorbidities. In isolation, their cancer cells may respond similarly to cancer therapy, but due to their baseline variability in pre-existing immune responses, patients can have different responses to identical therapies. In this review we discuss how the immune environment of tumors develops, the critical immune cell populations in advanced cancers, and how immune interventions can manipulate the immune environment of patients with pre-malignancies or advanced cancers to improve therapeutic outcomes.
Collapse
Affiliation(s)
| | - Michael J. Gough
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, OR, United States
| | | | | |
Collapse
|
23
|
Xiang X, Niu YR, Wang ZH, Ye LL, Peng WB, Zhou Q. Cancer-associated fibroblasts: Vital suppressors of the immune response in the tumor microenvironment. Cytokine Growth Factor Rev 2022; 67:35-48. [DOI: 10.1016/j.cytogfr.2022.07.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 07/13/2022] [Accepted: 07/15/2022] [Indexed: 12/17/2022]
|
24
|
Feng B, Wu J, Shen B, Jiang F, Feng J. Cancer-associated fibroblasts and resistance to anticancer therapies: status, mechanisms, and countermeasures. Cancer Cell Int 2022; 22:166. [PMID: 35488263 PMCID: PMC9052457 DOI: 10.1186/s12935-022-02599-7] [Citation(s) in RCA: 87] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 04/19/2022] [Indexed: 12/12/2022] Open
Abstract
Cancer-associated fibroblasts (CAFs) are critical components of the tumor microenvironment (TME) with diverse functions such as extracellular matrix (ECM) remodeling, modulation of metabolism and angiogenesis, and crosstalk with both cancer cells and infiltrating immune cells by production of growth factors, cytokines, and chemokines. Within the TME milieu, CAFs exhibit morphological and functional transitions with relatively specific markers and hold tremendous potential to facilitate tumorigenesis, development, and resistance towards multiple therapeutic strategies including chemotherapy, radiotherapy, targeted therapy, anti-angiogenesis therapy, immunotherapy, and endocrine therapy. Accordingly, CAFs themselves and the downstream effectors and/or signaling pathways are potential targets for optimizing the sensitivity of anti-cancer therapies. This review aims to provide a detailed landscape of the role that CAFs play in conferring therapeutic resistance in different cancers and the underlying mechanisms. The translational and therapeutic perspectives of CAFs in the individualized treatment of malignant tumors are also discussed.
Collapse
Affiliation(s)
- Bing Feng
- Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, 42 Baiziting, Nanjing, 210009, China
| | - Jianzhong Wu
- Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, 42 Baiziting, Nanjing, 210009, China
| | - Bo Shen
- Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, 42 Baiziting, Nanjing, 210009, China
| | - Feng Jiang
- Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, 42 Baiziting, Nanjing, 210009, China.
| | - Jifeng Feng
- Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, 42 Baiziting, Nanjing, 210009, China.
| |
Collapse
|
25
|
Stott MC, Oldfield L, Hale J, Costello E, Halloran CM. Recent advances in understanding pancreatic cancer. Fac Rev 2022; 11:9. [PMID: 35509672 PMCID: PMC9022729 DOI: 10.12703/r/11-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an intractable cancer and a leading cause of cancer deaths worldwide. Over 90% of patients die within 1 year of diagnosis. Deaths from PDAC are increasing and it remains a cancer of substantial unmet need. A number of factors contribute to its poor prognosis: namely, late presentation, early metastases and limited systemic therapy options because of chemoresistance. A variety of research approaches underway are aimed at improving patient survival. Here, we review high-risk groups and efforts for early detection. We examine recent developments in the understanding of complex molecular and metabolic alterations which accompany PDAC. We explore artificial intelligence and biological targets for therapy and examine the role of tumour stroma and the immune microenvironment. We also review recent developments with respect to the PDAC microbiome. It is hoped that current research efforts will translate into earlier diagnosis, improvements in treatment and better outcomes for patients.
Collapse
Affiliation(s)
- Martyn C Stott
- Department of Molecular & Clinical Cancer Medicine, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Sherrington Building, Liverpool, UK
| | - Lucy Oldfield
- Department of Molecular & Clinical Cancer Medicine, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Sherrington Building, Liverpool, UK
| | - Jessica Hale
- Department of Molecular & Clinical Cancer Medicine, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Sherrington Building, Liverpool, UK
| | - Eithne Costello
- Department of Molecular & Clinical Cancer Medicine, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Sherrington Building, Liverpool, UK
| | - Christopher M Halloran
- Department of Molecular & Clinical Cancer Medicine, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Sherrington Building, Liverpool, UK
| |
Collapse
|
26
|
Song P, Pan Q, Sun Z, Zou L, Yang L. Fibroblast activation protein alpha: Comprehensive detection methods for drug target and tumor marker. Chem Biol Interact 2022; 354:109830. [PMID: 35104486 DOI: 10.1016/j.cbi.2022.109830] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 12/22/2021] [Accepted: 01/21/2022] [Indexed: 11/25/2022]
Abstract
Fibroblast activation protein alpha (FAP-α, EC3.4.2. B28), a type II transmembrane proteolytic enzyme for the serine protease peptidase family. It is underexpressed in normal tissues but increased significantly in disease states, especially in neoplasm, which is a potential biomarker to turmor diagnosis. The inhibition of FAP-α activity will retard tumor formation, which is expected to be a promising tumor therapeutic target. At present, although the FAP-α expression detection methods has diversification, a superlative detection means is necessary for the clinical diagnosis. This review covers the discovery and the latest advances in FAP-α, as well as the future research prospects. The tissue distribution, structural characteristics, small-molecule ligands and structure-activity relationship of major inhibitors of FAP-α were summarized in this review. Furthermore, a variety of detection methods including traditional detection methods and emerging probes detection were classified and compared, and the design strategy and kinetic parameters of these FAP-α probe substrates were summarized. In addition, these comprehensive information provides a series of practical and reliable assays for the optimal design principles of FAP-α probes, promoting the application of FAP-α as a disease marker in diagnosis, and a drug target in drug design.
Collapse
Affiliation(s)
- Peifang Song
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Quisha Pan
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | | | - Liwei Zou
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Ling Yang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
27
|
Tagirasa R, Yoo E. Role of Serine Proteases at the Tumor-Stroma Interface. Front Immunol 2022; 13:832418. [PMID: 35222418 PMCID: PMC8873516 DOI: 10.3389/fimmu.2022.832418] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 01/24/2022] [Indexed: 01/19/2023] Open
Abstract
During tumor development, invasion and metastasis, the intimate interaction between tumor and stroma shapes the tumor microenvironment and dictates the fate of tumor cells. Stromal cells can also influence anti-tumor immunity and response to immunotherapy. Understanding the molecular mechanisms that govern this complex and dynamic interplay, thus is important for cancer diagnosis and therapy. Proteolytic enzymes that are expressed and secreted by both cancer and stromal cells play important roles in modulating tumor-stromal interaction. Among, several serine proteases such as fibroblast activation protein, urokinase-type plasminogen activator, kallikrein-related peptidases, and granzymes have attracted great attention owing to their elevated expression and dysregulated activity in the tumor microenvironment. This review highlights the role of serine proteases that are mainly derived from stromal cells in tumor progression and associated theranostic applications.
Collapse
|
28
|
Verhulst E, Garnier D, De Meester I, Bauvois B. Validating Cell Surface Proteases as Drug Targets for Cancer Therapy: What Do We Know, and Where Do We Go? Cancers (Basel) 2022; 14:624. [PMID: 35158891 PMCID: PMC8833564 DOI: 10.3390/cancers14030624] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 01/20/2022] [Accepted: 01/21/2022] [Indexed: 02/06/2023] Open
Abstract
Cell surface proteases (also known as ectoproteases) are transmembrane and membrane-bound enzymes involved in various physiological and pathological processes. Several members, most notably dipeptidyl peptidase 4 (DPP4/CD26) and its related family member fibroblast activation protein (FAP), aminopeptidase N (APN/CD13), a disintegrin and metalloprotease 17 (ADAM17/TACE), and matrix metalloproteinases (MMPs) MMP2 and MMP9, are often overexpressed in cancers and have been associated with tumour dysfunction. With multifaceted actions, these ectoproteases have been validated as therapeutic targets for cancer. Numerous inhibitors have been developed to target these enzymes, attempting to control their enzymatic activity. Even though clinical trials with these compounds did not show the expected results in most cases, the field of ectoprotease inhibitors is growing. This review summarizes the current knowledge on this subject and highlights the recent development of more effective and selective drugs targeting ectoproteases among which small molecular weight inhibitors, peptide conjugates, prodrugs, or monoclonal antibodies (mAbs) and derivatives. These promising avenues have the potential to deliver novel therapeutic strategies in the treatment of cancers.
Collapse
Affiliation(s)
- Emile Verhulst
- Laboratory of Medical Biochemistry, Department of Pharmaceutical Sciences, University of Antwerp, 2000 Antwerp, Belgium; (E.V.); (I.D.M.)
| | - Delphine Garnier
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, Cell Death and Drug Resistance in Lymphoproliferative Disorders Team, F-75006 Paris, France;
| | - Ingrid De Meester
- Laboratory of Medical Biochemistry, Department of Pharmaceutical Sciences, University of Antwerp, 2000 Antwerp, Belgium; (E.V.); (I.D.M.)
| | - Brigitte Bauvois
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, Cell Death and Drug Resistance in Lymphoproliferative Disorders Team, F-75006 Paris, France;
| |
Collapse
|
29
|
Multiplex Quantitative Analysis of Tumor-Infiltrating Lymphocytes, Cancer-Associated Fibroblasts, and CD200 in Pancreatic Cancer. Cancers (Basel) 2021; 13:cancers13215501. [PMID: 34771664 PMCID: PMC8583434 DOI: 10.3390/cancers13215501] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Revised: 10/27/2021] [Accepted: 10/29/2021] [Indexed: 11/16/2022] Open
Abstract
Pancreatic cancer is marked by a desmoplastic tumor microenvironment and low tumor immunogenicity, making it difficult for immunotherapy drugs to improve outcomes for patients. Tumor-infiltrating lymphocytes (TILs) and cancer-associated fibroblasts (CAFs) are seen in the tumor microenvironment of patients with pancreatic ductal adenocarcinoma (PDAC). In this work, we sought to characterize the expression levels and potential prognostic value of TILs (CD4, CD8, and CD20) and CAFs (Thy-1, FAP, and SMA) in a large retrospective cohort of PDAC patients. Additionally, we investigated the expression levels and prognostic significance of CD200, an immunoinhibitory protein that has shown interest as a potential target for immune checkpoint blockade. We measured the expression levels of these seven proteins with multiplexed immunofluorescence staining and quantitative immunofluorescence (QIF). We found CD8 and FAP to be independent predictors of progression-free survival and overall survival. CD200 was found to be heterogeneously expressed in both the tumor and stromal compartments of PDAC, with the majority of patients having positive stromal expression and negative tumor expression. This work demonstrates the potential clinical utility of CD8 and FAP in PDAC patients, and it sheds light on the expression patterns of CD200 in pancreatic cancer as the protein is being tested as a target for immune checkpoint blockade.
Collapse
|
30
|
Fibroblasts as immune regulators in infection, inflammation and cancer. Nat Rev Immunol 2021; 21:704-717. [PMID: 33911232 DOI: 10.1038/s41577-021-00540-z] [Citation(s) in RCA: 260] [Impact Index Per Article: 65.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/11/2021] [Indexed: 02/07/2023]
Abstract
In chronic infection, inflammation and cancer, the tissue microenvironment controls how local immune cells behave, with tissue-resident fibroblasts emerging as a key cell type in regulating activation or suppression of an immune response. Fibroblasts are heterogeneous cells, encompassing functionally distinct populations, the phenotypes of which differ according to their tissue of origin and type of inciting disease. Their immunological properties are also diverse, ranging from the maintenance of a potent inflammatory environment in chronic inflammation to promoting immunosuppression in malignancy, and encapsulating and incarcerating infectious agents within tissues. In this Review, we compare the mechanisms by which fibroblasts control local immune responses, as well as the factors regulating their inflammatory and suppressive profiles, in different tissues and pathological settings. This cross-disease perspective highlights the importance of tissue context in determining fibroblast-immune cell interactions, as well as potential therapeutic avenues to exploit this knowledge for the benefit of patients with chronic infection, inflammation and cancer.
Collapse
|
31
|
Truong LH, Pauklin S. Pancreatic Cancer Microenvironment and Cellular Composition: Current Understandings and Therapeutic Approaches. Cancers (Basel) 2021; 13:5028. [PMID: 34638513 PMCID: PMC8507722 DOI: 10.3390/cancers13195028] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 10/01/2021] [Accepted: 10/06/2021] [Indexed: 12/15/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) remains one of the most lethal human solid tumors, despite great efforts in improving therapeutics over the past few decades. In PDAC, the distinct characteristic of the tumor microenvironment (TME) is the main barrier for developing effective treatments. PDAC TME is characterized by a dense stroma, cancer-associated fibroblasts, and immune cells populations that crosstalk to the subpopulations of neoplastic cells that include cancer stem cells (CSCs). The heterogeneity in TME is also exhibited in the diversity and dynamics of acellular components, including the Extracellular matrix (ECM), cytokines, growth factors, and secreted ligands to signaling pathways. These contribute to drug resistance, metastasis, and relapse in PDAC. However, clinical trials targeting TME components have often reported unexpected results and still have not benefited patients. The failures in those trials and various efforts to understand the PDAC biology demonstrate the highly heterogeneous and multi-faceted TME compositions and the complexity of their interplay within TME. Hence, further functional and mechanistic insight is needed. In this review, we will present a current understanding of PDAC biology with a focus on the heterogeneity in TME and crosstalk among its components. We also discuss clinical challenges and the arising therapeutic opportunities in PDAC research.
Collapse
Affiliation(s)
| | - Siim Pauklin
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Old Road, University of Oxford, Oxford OX3 7LD, UK;
| |
Collapse
|
32
|
Xin L, Gao J, Zheng Z, Chen Y, Lv S, Zhao Z, Yu C, Yang X, Zhang R. Fibroblast Activation Protein-α as a Target in the Bench-to-Bedside Diagnosis and Treatment of Tumors: A Narrative Review. Front Oncol 2021; 11:648187. [PMID: 34490078 PMCID: PMC8416977 DOI: 10.3389/fonc.2021.648187] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 07/28/2021] [Indexed: 12/13/2022] Open
Abstract
Fibroblast activation protein-α (FAP) is a type II integral serine protease that is specifically expressed by activated fibroblasts. Cancer-associated fibroblasts (CAFs) in the tumor stroma have an abundant and stable expression of FAP, which plays an important role in promoting tumor growth, invasion, metastasis, and immunosuppression. For example, in females with a high incidence of breast cancer, CAFs account for 50–70% of the cells in the tumor’s microenvironment. CAF overexpression of FAP promotes tumor development and metastasis by influencing extracellular matrix remodeling, intracellular signaling, angiogenesis, epithelial-to-mesenchymal transition, and immunosuppression. This review discusses the basic biological characteristics of FAP and its applications in the diagnosis and treatment of various cancers. We review the emerging basic and clinical research data regarding the use of nanomaterials that target FAP.
Collapse
Affiliation(s)
- Lei Xin
- Department of Radiology, Affiliated Tumor Hospital of Shanxi Medical University, Taiyuan, China
| | - Jinfang Gao
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Taiyuan, China
| | - Ziliang Zheng
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan, China
| | - Yiyou Chen
- Department of Radiology, Affiliated Tumor Hospital of Shanxi Medical University, Taiyuan, China
| | - Shuxin Lv
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Taiyuan, China
| | - Zhikai Zhao
- Department of Radiology, Affiliated Tumor Hospital of Shanxi Medical University, Taiyuan, China
| | - Chunhai Yu
- Department of Radiology, Affiliated Tumor Hospital of Shanxi Medical University, Taiyuan, China
| | - Xiaotang Yang
- Department of Radiology, Affiliated Tumor Hospital of Shanxi Medical University, Taiyuan, China
| | - Ruiping Zhang
- Department of Radiology, Affiliated Tumor Hospital of Shanxi Medical University, Taiyuan, China
| |
Collapse
|
33
|
Monjazeb AM, Schalper KA, Villarroel-Espindola F, Nguyen A, Shiao SL, Young K. Effects of Radiation on the Tumor Microenvironment. Semin Radiat Oncol 2021; 30:145-157. [PMID: 32381294 DOI: 10.1016/j.semradonc.2019.12.004] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
A malignant tumor consists of malignant cells as well as a wide array of normal host tissues including stroma, vasculature, and immune infiltrate. The interaction between cancer and these host tissues is critical as these host tissues play a variety of roles in supporting or resisting disease progression. Radiotherapy (RT) has direct effects on malignant cells, but, also, critically important effects on these other components of the tumor microenvironment (TME). Given the growing role of immune checkpoint inhibitors and other immunotherapy strategies, understanding how RT affects the TME, particularly the immune compartment, is essential to advance RT in this new era of cancer therapy. The interactions between RT and the TME are complex, affecting the innate and adaptive arms of the immune system. RT can induce both proinflammatory effects and immune suppressive effects that can either promote or impede antitumor immunity. It is likely that the initial proinflammatory effects of RT eventually lead to rebound immune-suppression as chronic inflammation sets in. The exact kinetics and nature of how RT changes the TME likely depends on timing, dose, fractionation, site irradiated, and tumor type. With increased understanding of the effects of RT on the TME, in the future it is likely that we will be able to personalize RT by varying the dose, site, and timing of intervention to generate the desired response to partner with immunotherapy strategies.
Collapse
Affiliation(s)
- Arta M Monjazeb
- UC Davis Comprehensive Cancer Center, Department of Radiation Oncology, Sacramento, CA.
| | - Kurt A Schalper
- Yale University School of Medicine, Department of Pathology, New Haven, CT
| | | | - Anthony Nguyen
- Cedars-Sinai Medical Center, Department of Radiation Oncology, Los Angeles, CA
| | - Stephen L Shiao
- Cedars-Sinai Medical Center, Department of Radiation Oncology, Los Angeles, CA
| | - Kristina Young
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, OR; Radiation Oncology Division, The Oregon Clinic, Portland, OR
| |
Collapse
|
34
|
Kasperska A, Borowczak J, Szczerbowski K, Stec E, Ahmadi N, Szylber Ł. Current challenges in targeting tumor desmoplasia to improve the efficacy of immunotherapy. Curr Cancer Drug Targets 2021; 21:919-931. [PMID: 34525931 DOI: 10.2174/1568009621666210825101456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 05/31/2021] [Accepted: 06/07/2021] [Indexed: 11/22/2022]
Abstract
Desmoplasia is crucial for the development, progression and treatment of immune-resistant malignancies. and treatment of immune-resistant malignancies. Targeting desmoplasia-related metabolic pathways appears to be an interesting approach to expand our stock of disposable anti-tumor agents.CXCL12/CXCR4 axis inhibition reduces fibrosis, alleviates immunosuppression and significantly enhances the efficacy of PD-1 immunotherapy. CD40L substitute therapy may increase the activity of T-cells, downregulate CD40+, prolong patients' survival and prevent cancer progression. Although FAPα antagonists used in preclinical models did not lead to permanent cure, an alleviation of immune-resistance, modification of desmoplasia and a decrease in angiogenesis were observed. Targeting DDR2 may enhance the effect of anti-PD-1 treatment in multiple neoplasm cell lines and has the ability to overcome the adaptation to BRAF-targeted therapy in melanoma. Reprogramming desmoplasia could potentially cooperate not only with present treatment, but also other potential therapeutic targets. We present the most promising metabolic pathways related to desmoplasia and discuss the emerging strategies to improve the efficacy of immunotherapy.
Collapse
Affiliation(s)
- Anna Kasperska
- Department of Clinical Pathomorphology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun. Poland
| | - Jędrzej Borowczak
- Department of Clinical Pathomorphology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun. Poland
| | - Krzysztof Szczerbowski
- Department of Clinical Pathomorphology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun. Poland
| | - Ewa Stec
- Department of Clinical Pathomorphology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun. Poland
| | - Navid Ahmadi
- Department of Cardiothoracic Surgery, Royal Papworth Hospital, Cambridge. United Kingdom
| | - Łukasz Szylber
- Department of Clinical Pathomorphology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun. Poland
| |
Collapse
|
35
|
Xu X, Deng Z, Dehghani H, Iordachita I, Lim M, Wong JW, Wang KKH. Quantitative Bioluminescence Tomography-guided Conformal Irradiation for Preclinical Radiation Research. Int J Radiat Oncol Biol Phys 2021; 111:1310-1321. [PMID: 34411639 PMCID: PMC8602741 DOI: 10.1016/j.ijrobp.2021.08.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 07/16/2021] [Accepted: 08/05/2021] [Indexed: 10/31/2022]
Abstract
PURPOSE Widely used cone beam computed tomography (CBCT)-guided irradiators in preclinical radiation research are limited to localize soft tissue target because of low imaging contrast. Knowledge of target volume is a fundamental need for radiation therapy (RT). Without such information to guide radiation, normal tissue can be overirradiated, introducing experimental uncertainties. This led us to develop high-contrast quantitative bioluminescence tomography (QBLT) for guidance. The use of a 3-dimensional bioluminescence signal, related to cell viability, for preclinical radiation research is one step toward biology-guided RT. METHODS AND MATERIALS Our QBLT system enables multiprojection and multispectral bioluminescence imaging to maximize input data for the tomographic reconstruction. Accurate quantification of spectrum and dynamic change of in vivo signal were also accounted for the QBLT. A spectral-derivative method was implemented to eliminate the modeling of the light propagation from animal surface to detector. We demonstrated the QBLT capability of guiding conformal RT using a bioluminescent glioblastoma (GBM) model in vivo. A threshold was determined to delineate QBLT reconstructed gross target volume (GTVQBLT), which provides the best overlap between the GTVQBLT and CBCT contrast labeled GBM (GTV), used as the ground truth for GBM volume. To account for the uncertainty of GTVQBLT in target positioning and volume delineation, a margin was determined and added to the GTVQBLT to form a QBLT planning target volume (PTVQBLT) for guidance. RESULTS The QBLT can reconstruct in vivo GBM with localization accuracy within 1 mm. A 0.5-mm margin was determined and added to GTVQBLT to form PTVQBLT, largely improving tumor coverage from 75.0% (0 mm margin) to 97.9% in average, while minimizing normal tissue toxicity. With the goal of prescribed dose 5 Gy covering 95% of PTVQBLT, QBLT-guided 7-field conformal RT can effectively irradiate 99.4 ± 1.0% of GTV. CONCLUSIONS The QBLT provides a unique opportunity for investigators to use biologic information for target delineation, guiding conformal irradiation, and reducing normal tissue involvement, which is expected to increase reproducibility of scientific discovery.
Collapse
Affiliation(s)
- Xiangkun Xu
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University, Baltimore, Maryland; Biomedical Imaging and Radiation Technology Laboratory, Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Zijian Deng
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University, Baltimore, Maryland; Biomedical Imaging and Radiation Technology Laboratory, Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Hamid Dehghani
- School of Computer Science, University of Birmingham, Birmingham, West Midlands, United Kingdom
| | - Iulian Iordachita
- Laboratory for Computational Sensing and Robotics, Johns Hopkins University, Baltimore, Maryland
| | - Michael Lim
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University, Baltimore, Maryland; Department of Neurosurgery, Stanford University, Stanford, California
| | - John W Wong
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University, Baltimore, Maryland
| | - Ken Kang-Hsin Wang
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University, Baltimore, Maryland; Biomedical Imaging and Radiation Technology Laboratory, Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, Texas.
| |
Collapse
|
36
|
Maia A, Wiemann S. Cancer-Associated Fibroblasts: Implications for Cancer Therapy. Cancers (Basel) 2021; 13:3526. [PMID: 34298736 PMCID: PMC8307167 DOI: 10.3390/cancers13143526] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 07/09/2021] [Accepted: 07/12/2021] [Indexed: 12/12/2022] Open
Abstract
Tumour cells do not exist as an isolated entity. Instead, they are surrounded by and closely interact with cells of the environment they are emerged in. The tumour microenvironment (TME) is not static and several factors, including cancer cells and therapies, have been described to modulate several of its components. Fibroblasts are key elements of the TME with the capacity to influence tumour progression, invasion and response to therapy, which makes them attractive targets in cancer treatment. In this review, we focus on fibroblasts and their numerous roles in the TME with a special attention to recent findings describing their heterogeneity and role in therapy response. Furthermore, we explore how different therapies can impact these cells and their communication with cancer cells. Finally, we highlight potential strategies targeting this cell type that can be employed for improving patient outcome.
Collapse
Affiliation(s)
- Ana Maia
- German Cancer Research Center (DKFZ), Division of Molecular Genome Analysis, Im Neuenheimer Feld 580, 69120 Heidelberg, Germany
| | - Stefan Wiemann
- German Cancer Research Center (DKFZ), Division of Molecular Genome Analysis, Im Neuenheimer Feld 580, 69120 Heidelberg, Germany
| |
Collapse
|
37
|
Belhabib I, Zaghdoudi S, Lac C, Bousquet C, Jean C. Extracellular Matrices and Cancer-Associated Fibroblasts: Targets for Cancer Diagnosis and Therapy? Cancers (Basel) 2021; 13:3466. [PMID: 34298680 PMCID: PMC8303391 DOI: 10.3390/cancers13143466] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 06/25/2021] [Accepted: 07/05/2021] [Indexed: 12/12/2022] Open
Abstract
Solid cancer progression is dictated by neoplastic cell features and pro-tumoral crosstalks with their microenvironment. Stroma modifications, such as fibroblast activation into cancer-associated fibroblasts (CAFs) and extracellular matrix (ECM) remodeling, are now recognized as critical events for cancer progression and as potential therapeutic or diagnostic targets. The recent appreciation of the key, complex and multiple roles of the ECM in cancer and of the CAF diversity, has revolutionized the field and raised innovative but challenging questions. Here, we rapidly present CAF heterogeneity in link with their specific ECM remodeling features observed in cancer, before developing each of the impacts of such ECM modifications on tumor progression (survival, angiogenesis, pre-metastatic niche, chemoresistance, etc.), and on patient prognosis. Finally, based on preclinical studies and recent results obtained from clinical trials, we highlight key mechanisms or proteins that are, or may be, used as potential therapeutic or diagnostic targets, and we report and discuss benefits, disappointments, or even failures, of recently reported stroma-targeting strategies.
Collapse
Affiliation(s)
| | | | | | | | - Christine Jean
- Centre de Recherche en Cancérologie de Toulouse (CRCT), INSERM U1037, Université Toulouse III Paul Sabatier, ERL5294 CNRS, 31037 Toulouse, France; (I.B.); (S.Z.); (C.L.); (C.B.)
| |
Collapse
|
38
|
Mhaidly R, Mechta‐Grigoriou F. Role of cancer-associated fibroblast subpopulations in immune infiltration, as a new means of treatment in cancer. Immunol Rev 2021; 302:259-272. [PMID: 34013544 PMCID: PMC8360036 DOI: 10.1111/imr.12978] [Citation(s) in RCA: 127] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 04/26/2021] [Accepted: 04/27/2021] [Indexed: 12/12/2022]
Abstract
The tumor microenvironment (TME) has been identified as one of the driving factors of tumor progression and invasion. Within this microenvironment, cancer-associated fibroblasts (CAF) have multiple tumor-promoting functions and play key roles in drug resistance, through multiple mechanisms, including extracellular matrix (ECM) remodeling, production of growth factors, cytokines, and chemokines, and modulation of metabolism and angiogenesis. More recently, a growing body of evidence has shown that CAF also modulate immune cell activity and suppress anti-tumor immune response. In this review, we describe the current knowledge on CAF heterogeneity in terms of identity and functions. Moreover, we analyze how distinct CAF subpopulations differentially interact with immune cells, with a particular focus on T lymphocytes. We address how specific CAF subsets contribute to cancer progression through induction of an immunosuppressive microenvironment. Finally, we highlight potential therapeutic strategies for targeting CAF subpopulations in cancer.
Collapse
Affiliation(s)
- Rana Mhaidly
- Institut CurieStress and Cancer LaboratoryEquipe labelisée Ligue Nationale Contre le CancerPSL Research UniversityParisFrance
- U830, InsermParisFrance
| | - Fatima Mechta‐Grigoriou
- Institut CurieStress and Cancer LaboratoryEquipe labelisée Ligue Nationale Contre le CancerPSL Research UniversityParisFrance
- U830, InsermParisFrance
| |
Collapse
|
39
|
Gorchs L, Kaipe H. Interactions between Cancer-Associated Fibroblasts and T Cells in the Pancreatic Tumor Microenvironment and the Role of Chemokines. Cancers (Basel) 2021; 13:2995. [PMID: 34203869 PMCID: PMC8232575 DOI: 10.3390/cancers13122995] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 06/10/2021] [Accepted: 06/14/2021] [Indexed: 01/18/2023] Open
Abstract
Less than 10% of patients diagnosed with pancreatic ductal adenocarcinoma (PDAC) survive 5 years or more, making it one of the most fatal cancers. Accumulation of T cells in pancreatic tumors is associated with better prognosis, but immunotherapies to enhance the anti-tumor activity of infiltrating T cells are failing in this devastating disease. Pancreatic tumors are characterized by a desmoplastic stroma, which mainly consists of activated cancer-associated fibroblasts (CAFs). Pancreatic CAFs have emerged as important regulators of the tumor microenvironment by contributing to immune evasion through the release of chemokines, cytokines, and growth factors, which alters T-cell migration, differentiation and cytotoxic activity. However, recent discoveries have also revealed that subsets of CAFs with diverse functions can either restrain or promote tumor progression. Here, we discuss our current knowledge about the interactions between CAFs and T cells in PDAC and summarize different therapy strategies targeting the CAF-T cell axis with focus on CAF-derived soluble immunosuppressive factors and chemokines. Identifying the functions of different CAF subsets and understanding their roles in T-cell trafficking within the tumor may be fundamental for the development of an effective combinational treatment for PDAC.
Collapse
Affiliation(s)
- Laia Gorchs
- Department of Laboratory Medicine, Karolinska Institutet, 14152 Stockholm, Sweden
| | - Helen Kaipe
- Department of Laboratory Medicine, Karolinska Institutet, 14152 Stockholm, Sweden
- Clinical Immunology and Transfusion Medicine, Karolinska University Hospital, 14152 Stockholm, Sweden
| |
Collapse
|
40
|
Desbois M, Wang Y. Cancer-associated fibroblasts: Key players in shaping the tumor immune microenvironment. Immunol Rev 2021; 302:241-258. [PMID: 34075584 DOI: 10.1111/imr.12982] [Citation(s) in RCA: 101] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 04/14/2021] [Accepted: 04/30/2021] [Indexed: 12/18/2022]
Abstract
Cancer immunotherapies have rapidly changed the therapeutic landscape for cancer. Nevertheless, most of the patients show innate or acquired resistance to these therapies. Studies conducted in recent years have highlighted an emerging role of cancer-associated fibroblasts (CAFs) in immune regulation that shapes the tumor immune microenvironment (TIME) and influences response to cancer immunotherapies. In this review, we outline recent advances in the understanding of phenotypic and functional heterogeneity of CAFs. We will focus on emerging roles of CAFs in shaping the TIME, especially under a framework of tumor immunity continuum, and discuss current and future CAF-targeting therapeutic strategies in particular in the context of optimizing the success of immunotherapies.
Collapse
Affiliation(s)
- Mélanie Desbois
- Department of Oncology Biomarker Development, Genentech, Inc., South San Francisco, CA, USA
| | - Yulei Wang
- Department of Oncology Biomarker Development, Genentech, Inc., South San Francisco, CA, USA
| |
Collapse
|
41
|
Wang S, Cui J, Zhang K, Gu J, Zheng Y, Zhang B, Shi L. [SP13786 Inhibits the Migration and Invasion of Lung Adenocarcinoma Cell A549
by Supressing Stat3-EMT via CAFs Exosomes]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2021; 24:384-393. [PMID: 34024061 PMCID: PMC8246397 DOI: 10.3779/j.issn.1009-3419.2021.104.07] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
背景与目的 成纤维细胞活化蛋白(fibroblast activation protein, FAP)是肿瘤相关成纤维细胞(cancer-associated fibroblasts, CAFs)的表面标志物之一,与CAFs的恶性表征关系密切,SP13786是FAP的特异性小分子抑制剂。本研究探讨SP13786作用于CAFs后,CAFs外泌体(exosomes, exo)对A549细胞迁移、侵袭的影响与机制。 方法 原代提取CAFs和癌旁成纤维细胞(peri-tumer fibroblasts, PTFs);MTT实验检测不同浓度SP13786对CAFs增殖的影响;聚合物沉淀法提取PTFs-exo、CAFs-exo以及CAFs+SP13786-exo。将A549细胞设对照组、PTFs组、CAFs组及CAFs+SP13786组并分别以等体积的DMEM、PTFs-exo、CAFs-exo及CAFs+SP13786-exo孵育细胞。激光共聚焦实验检测A549细胞摄取外泌体的情况;免疫荧光、免疫组化和Western blot方法检测α平滑肌肌动蛋白(alpha-smooth muscle actin, α-SMA)、FAP在PTFs和CAFs中的表达及E-cadherin、N-cadherin、Slug、Stat3、P-Stat3在各组A549细胞中的表达;划痕实验和Transwell实验检测各组细胞的迁移和侵袭能力。 结果 免疫荧光、免疫组化和Western blot结果均显示α-SMA、FAP在CAFs中高表达,在PTFs中低表达(P < 0.05),表明从肺腺癌组织和癌旁组织中分别成功获得了CAFs和PTFs。MTT实验测得SP13786对于CAFs细胞的半数抑制浓度(50% inhibitory concentration, IC50)约为3.3 nmol/L。免疫组化和Western blot结果显示与CAFs组相比,CAFs+SP13786组的α-SMA与FAP的表达显著降低(P < 0.05),说明抑制FAP可以显著降低CAFs的恶性表征。激光共聚焦结果显示外泌体能够被A549细胞所摄取。划痕实验与Transwell实验显示SP13786可抑制CAFs-exo对A549细胞迁移和侵袭的促进作用(P < 0.05)。与CAFs组比较,SP13786组A549细胞E-cadherin表达增多,N-cadherin与Slug表达降低(P < 0.05);免疫荧光与Western blot显示SP13786组A549细胞的P-Stat3较CAFs组明显降低(P < 0.05),而总Stat3无显著差异。Stat3的特异性抑制剂WP1066明显抑制CAFs组A549细胞上皮间质转化(epithelial-mesenchymal transition, EMT),P-Stat3显著降低(P < 0.05),而加入WP1066后再加入SP13786-exo,P-Stat3未见进一步减低,EMT的抑制亦未见显著变化(P > 0.05)。 结论 FAP的小分子特异性抑制剂SP13786通过影响CAFs外泌体间接抑制A549细胞的迁移、侵袭,其可能机制是抑制Stat3的磷酸化从而影响A549细胞的EMT。
Collapse
Affiliation(s)
- Shushu Wang
- Shandong Province Key Laboratory of Applied Pharmacology, Weifang Medical University, Weifang 261053, China
| | - Jiayu Cui
- College of Clinical Medicine, Weifang Medical University, Weifang 261053, China
| | - Kaijia Zhang
- Shandong Province Key Laboratory of Applied Pharmacology, Weifang Medical University, Weifang 261053, China
| | - Jinhua Gu
- College of Clinical Medicine, Weifang Medical University, Weifang 261053, China
| | - Yuanhang Zheng
- College of Clinical Medicine, Weifang Medical University, Weifang 261053, China
| | - Baogang Zhang
- College of Clinical Medicine, Weifang Medical University, Weifang 261053, China
| | - Lihong Shi
- Shandong Province Key Laboratory of Applied Pharmacology, Weifang Medical University, Weifang 261053, China
| |
Collapse
|
42
|
Raskov H, Orhan A, Gaggar S, Gögenur I. Cancer-Associated Fibroblasts and Tumor-Associated Macrophages in Cancer and Cancer Immunotherapy. Front Oncol 2021; 11:668731. [PMID: 34094963 PMCID: PMC8172975 DOI: 10.3389/fonc.2021.668731] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 04/14/2021] [Indexed: 02/06/2023] Open
Abstract
Our understanding of the tumor microenvironment (TME), including the interplay between tumor cells, stromal cells, immune cells, and extracellular matrix components, is mandatory for the innovation of new therapeutic approaches in cancer. The cell-cell communication within the TME plays a pivotal role in the evolution and progression of cancer. Cancer-associated fibroblasts (CAF) and tumor-associated macrophages (TAM) are major cell populations in the stroma of all solid tumors and often exert protumorigenic functions; however, the origin and precise functions of CAF and TAM are still incompletely understood. CAF and TAM hold significant potential as therapeutic targets to improve outcomes in oncology when combined with existing therapies. The regulation of CAF/TAM communication and/or their differentiation could be of high impact for improving the future targeted treatment strategies. Nevertheless, there is much scope for research and innovation in this field with regards to the development of novel drugs. In this review, we elaborate on the current knowledge on CAF and TAM in cancer and cancer immunotherapy. Additionally, by focusing on their heterogenous functions in different stages and types of cancer, we explore their role as potential therapeutic targets and highlight certain aspects of their functions that need further research.
Collapse
Affiliation(s)
- Hans Raskov
- Center for Surgical Science, Zealand University Hospital, Køge, Denmark
| | - Adile Orhan
- Center for Surgical Science, Zealand University Hospital, Køge, Denmark.,Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Shruti Gaggar
- Center for Surgical Science, Zealand University Hospital, Køge, Denmark
| | - Ismail Gögenur
- Center for Surgical Science, Zealand University Hospital, Køge, Denmark.,Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
43
|
Zhang H, Jiang H, Zhu L, Li J, Ma S. Cancer-associated fibroblasts in non-small cell lung cancer: Recent advances and future perspectives. Cancer Lett 2021; 514:38-47. [PMID: 34019960 DOI: 10.1016/j.canlet.2021.05.009] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 05/07/2021] [Accepted: 05/11/2021] [Indexed: 12/24/2022]
Abstract
Non-small cell lung cancer (NSCLC) constitutes the majority of lung cancer, which is the leading cause of cancer-related deaths in the world. Nearly 70% of NSCLC patients were diagnosed at advanced stage with only 15% of five-year survival rate. Cancer-associated fibroblasts (CAFs) are the major component of tumor microenvironment and account for almost 70% of the cells in tumor tissues. By the crosstalk with cancer cells, CAFs reprogrammed cancer cell metabolism, remodeled extracellular matrix (ECM) and created a supportive niche for cancer stem cells. CAFs lead collective invasion of tumor cells and shape tumor immune microenvironment, promoting tumor metastasis and immune escape. In this review, we have summarized the progress of studies regarding CAFs influences on NSCLC in recent five years from the aspects of cell growth, metabolism, therapy resistance, invasion and metastasis and immune suppression. We have discussed the involved mechanisms and implications for the development of anti-NSCLC therapies. The current strategies of CAFs targeting and elimination have also been generalized. Only better understanding of the molecular biology of CAFs may contribute to the development of novel anti-NSCLC strategies.
Collapse
Affiliation(s)
- Hongfang Zhang
- Hangzhou Cancer Institution, Affiliated Hangzhou Cancer Hospital, Zhejiang University School of Medicine, Hangzhou, 310002, China; Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Hong Jiang
- Department of Cardiothoracic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Lucheng Zhu
- Department of Thoracic Oncology, Affiliated Hangzhou Cancer Hospital, Zhejiang University School of Medicine, Hangzhou, 310002, China
| | - Jiawei Li
- Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Shenglin Ma
- Hangzhou Cancer Institution, Affiliated Hangzhou Cancer Hospital, Zhejiang University School of Medicine, Hangzhou, 310002, China; Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China; Zhejiang University Cancer Center, Hangzhou, 310058, China.
| |
Collapse
|
44
|
Kaur J, Singh P, Enzler T, Sahai V. Emerging antibody therapies for pancreatic adenocarcinoma: a review of recent phase 2 trials. Expert Opin Emerg Drugs 2021; 26:103-129. [PMID: 33734833 DOI: 10.1080/14728214.2021.1905795] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Introduction: Pancreatic adenocarcinoma is now the third-leading cause of cancer-related deaths in the US which can be attributed to rising incidence, diagnosis at advanced stages and early development of metastasis. Systemic therapy remains palliative with early development of resistance possibly related to the constitutive activation of 'undruggable' KRAS, immunosuppressive microenvironment, and intense desmoplasia. The advancements in molecular biology has led to the development and investigation of targeted and immune therapeutics.Areas covered: This study provides a comprehensive review of the literature to further the understanding of molecular targets with their respective antibody-based therapies in clinical development in pancreatic cancer. PubMed was systematically searched for English-language articles discussing antibody-based therapies under phase 2 clinical trial investigation in pancreatic adenocarcinoma.Expert opinion: PDAC remains highly resistant to chemotherapy with no significant improvement in survival for patients with advanced or metastatic cancer. Unfortunately, the majority of the antibody-based targeted and immune therapeutics have failed to meet their primary efficacy endpoints in early phase trials. However, there are a few promising antibody-based drugs with intriguing preliminary data that merit further investigation, while many more continue to be developed and investigated preclinically, and in early phase trials.
Collapse
Affiliation(s)
- Jasmeet Kaur
- Department of Internal Medicine, Saint Joseph Mercy Oakland Hospital, Pontiac, MI, USA
| | - Paramveer Singh
- Division of Hematology and Oncology, Department of Internal Medicine, Wayne State University, Detroit, MI, USA
| | - Thomas Enzler
- Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Vaibhav Sahai
- Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
45
|
Koustoulidou S, Hoorens MWH, Dalm SU, Mahajan S, Debets R, Seimbille Y, de Jong M. Cancer-Associated Fibroblasts as Players in Cancer Development and Progression and Their Role in Targeted Radionuclide Imaging and Therapy. Cancers (Basel) 2021; 13:1100. [PMID: 33806468 PMCID: PMC7961537 DOI: 10.3390/cancers13051100] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 02/19/2021] [Accepted: 02/20/2021] [Indexed: 12/12/2022] Open
Abstract
Cancer Associated Fibroblasts (CAFs) form a major component of the tumour microenvironment, they have a complex origin and execute diverse functions in tumour development and progression. As such, CAFs constitute an attractive target for novel therapeutic interventions that will aid both diagnosis and treatment of various cancers. There are, however, a few limitations in reaching successful translation of CAF targeted interventions from bench to bedside. Several approaches targeting CAFs have been investigated so far and a few CAF-targeting tracers have successfully been developed and applied. This includes tracers targeting Fibroblast Activation Protein (FAP) on CAFs. A number of FAP-targeting tracers have shown great promise in the clinic. In this review, we summarize our current knowledge of the functional heterogeneity and biology of CAFs in cancer. Moreover, we highlight the latest developments towards theranostic applications that will help tumour characterization, radioligand therapy and staging in cancers with a distinct CAF population.
Collapse
Affiliation(s)
- Sofia Koustoulidou
- Department of Radiology and Nuclear Medicine, Erasmus University Medical Center, Dr. Molewaterplein 40, 3015 GD Rotterdam, The Netherlands; (M.W.H.H.); (S.U.D.); (Y.S.); (M.d.J.)
| | - Mark W. H. Hoorens
- Department of Radiology and Nuclear Medicine, Erasmus University Medical Center, Dr. Molewaterplein 40, 3015 GD Rotterdam, The Netherlands; (M.W.H.H.); (S.U.D.); (Y.S.); (M.d.J.)
| | - Simone U. Dalm
- Department of Radiology and Nuclear Medicine, Erasmus University Medical Center, Dr. Molewaterplein 40, 3015 GD Rotterdam, The Netherlands; (M.W.H.H.); (S.U.D.); (Y.S.); (M.d.J.)
| | - Shweta Mahajan
- Department of Medical Oncology, Erasmus University Medical Center, Dr. Molewaterplein 40, 3015 GD Rotterdam, The Netherlands; (R.D.); (S.M.)
| | - Reno Debets
- Department of Medical Oncology, Erasmus University Medical Center, Dr. Molewaterplein 40, 3015 GD Rotterdam, The Netherlands; (R.D.); (S.M.)
| | - Yann Seimbille
- Department of Radiology and Nuclear Medicine, Erasmus University Medical Center, Dr. Molewaterplein 40, 3015 GD Rotterdam, The Netherlands; (M.W.H.H.); (S.U.D.); (Y.S.); (M.d.J.)
| | - Marion de Jong
- Department of Radiology and Nuclear Medicine, Erasmus University Medical Center, Dr. Molewaterplein 40, 3015 GD Rotterdam, The Netherlands; (M.W.H.H.); (S.U.D.); (Y.S.); (M.d.J.)
| |
Collapse
|
46
|
Ghahremanifard P, Chanda A, Bonni S, Bose P. TGF-β Mediated Immune Evasion in Cancer-Spotlight on Cancer-Associated Fibroblasts. Cancers (Basel) 2020; 12:cancers12123650. [PMID: 33291370 PMCID: PMC7762018 DOI: 10.3390/cancers12123650] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 12/02/2020] [Accepted: 12/02/2020] [Indexed: 12/16/2022] Open
Abstract
Various components of the tumor microenvironment (TME) play a critical role in promoting tumorigenesis, progression, and metastasis. One of the primary functions of the TME is to stimulate an immunosuppressive environment around the tumor through multiple mechanisms including the activation of the transforming growth factor-beta (TGF-β) signaling pathway. Cancer-associated fibroblasts (CAFs) are key cells in the TME that regulate the secretion of extracellular matrix (ECM) components under the influence of TGF-β. Recent reports from our group and others have described an ECM-related and CAF-associated novel gene signature that can predict resistance to immune checkpoint blockade (ICB). Importantly, studies have begun to test whether targeting some of these CAF-associated components can be used as a combinatorial approach with ICB. This perspective summarizes recent advances in our understanding of CAF and TGF-β-regulated immunosuppressive mechanisms and ways to target such signaling in cancer.
Collapse
Affiliation(s)
- Parisa Ghahremanifard
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada; (P.G.); (A.C.); (S.B.)
- Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Ayan Chanda
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada; (P.G.); (A.C.); (S.B.)
- Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Shirin Bonni
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada; (P.G.); (A.C.); (S.B.)
- Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Pinaki Bose
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada; (P.G.); (A.C.); (S.B.)
- Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
- Department of Oncology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
- Ohlson Research Initiative, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4Z6, Canada
- Correspondence: ; Tel.: +1-403-220-8507; Fax: +1-403-270-3145
| |
Collapse
|
47
|
Hilmi M, Nicolle R, Bousquet C, Neuzillet C. Cancer-Associated Fibroblasts: Accomplices in the Tumor Immune Evasion. Cancers (Basel) 2020; 12:cancers12102969. [PMID: 33066357 PMCID: PMC7602282 DOI: 10.3390/cancers12102969] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 10/04/2020] [Accepted: 10/12/2020] [Indexed: 12/14/2022] Open
Abstract
Simple Summary A growing number of studies suggest that cancer-associated fibroblasts (CAFs) modulate both myeloid and lymphoid cells through secretion of molecules (i.e., chemical function) and production of the extracellular matrix (ECM), i.e., physical function. Even though targeting functions CAFs is a relevant strategy, published clinical trials solely aimed at targeting the stroma showed disappointing results, despite being based on solid preclinical evidence. Our review dissects the interactions between CAFs and immune cells and explains how a deeper understanding of CAF subpopulations is the cornerstone to propose relevant therapies that will ultimately improve survival of patients with cancer. Abstract Cancer-associated fibroblasts (CAFs) are prominent cells within the tumor microenvironment, by communicating with other cells within the tumor and by secreting the extracellular matrix components. The discovery of the immunogenic role of CAFs has made their study particularly attractive due to the potential applications in the field of cancer immunotherapy. Indeed, CAFs are highly involved in tumor immune evasion by physically impeding the immune system and interacting with both myeloid and lymphoid cells. However, CAFs do not represent a single cell entity but are divided into several subtypes with different functions that may be antagonistic. Considering that CAFs are orchestrators of the tumor microenvironment and modulate immune cells, targeting their functions may be a promising strategy. In this review, we provide an overview of (i) the mechanisms involved in immune regulation by CAFs and (ii) the therapeutic applications of CAFs modulation to improve the antitumor immune response and the efficacy of immunotherapy.
Collapse
Affiliation(s)
- Marc Hilmi
- Department of Medical Oncology, Curie Institute, University of Versailles Saint-Quentin, 92210 Saint-Cloud, France;
- GERCOR, 151 rue du Faubourg Saint-Antoine, 75011 Paris, France
- Correspondence: ; Tel.: +33-06-8547-3027
| | - Rémy Nicolle
- Programme Cartes d’Identité des Tumeurs (CIT), Ligue Nationale Contre Le Cancer, 75013 Paris, France;
| | - Corinne Bousquet
- Cancer Research Center of Toulouse (CRCT), INSERM UMR 1037, University Toulouse III Paul Sabatier, ERL5294 CNRS, 31000 Toulouse, France;
| | - Cindy Neuzillet
- Department of Medical Oncology, Curie Institute, University of Versailles Saint-Quentin, 92210 Saint-Cloud, France;
- GERCOR, 151 rue du Faubourg Saint-Antoine, 75011 Paris, France
- Institut Curie, Cell Migration and Invasion, UMR144, PSL Research University, 26, rue d’Ulm, F-75005 Paris, France
| |
Collapse
|
48
|
Ho WJ, Jaffee EM, Zheng L. The tumour microenvironment in pancreatic cancer - clinical challenges and opportunities. Nat Rev Clin Oncol 2020; 17:527-540. [PMID: 32398706 PMCID: PMC7442729 DOI: 10.1038/s41571-020-0363-5] [Citation(s) in RCA: 661] [Impact Index Per Article: 132.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/26/2020] [Indexed: 12/17/2022]
Abstract
Metastatic pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal solid tumours despite the use of multi-agent conventional chemotherapy regimens. Such poor outcomes have fuelled ongoing efforts to exploit the tumour microenvironment (TME) for therapy, but strategies aimed at deconstructing the surrounding desmoplastic stroma and targeting the immunosuppressive pathways have largely failed. In fact, evidence has now shown that the stroma is multi-faceted, which illustrates the complexity of exploring features of the TME as isolated targets. In this Review, we describe ways in which the PDAC microenvironment has been targeted and note the current understanding of the clinical outcomes that have unexpectedly contradicted preclinical observations. We also consider the more sophisticated therapeutic strategies under active investigation - multi-modal treatment approaches and exploitation of biologically integrated targets - which aim to remodel the TME against PDAC.
Collapse
Affiliation(s)
- Won Jin Ho
- Sidney Kimmel Comprehensive Cancer Center, The Skip Viragh Pancreatic Cancer Center for Clinical Research and Care, and The Bloomberg-Kimmel Institute for Immunotherapy at Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Elizabeth M Jaffee
- Sidney Kimmel Comprehensive Cancer Center, The Skip Viragh Pancreatic Cancer Center for Clinical Research and Care, and The Bloomberg-Kimmel Institute for Immunotherapy at Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Lei Zheng
- Sidney Kimmel Comprehensive Cancer Center, The Skip Viragh Pancreatic Cancer Center for Clinical Research and Care, and The Bloomberg-Kimmel Institute for Immunotherapy at Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
49
|
Sperb N, Tsesmelis M, Wirth T. Crosstalk between Tumor and Stromal Cells in Pancreatic Ductal Adenocarcinoma. Int J Mol Sci 2020; 21:E5486. [PMID: 32752017 PMCID: PMC7432853 DOI: 10.3390/ijms21155486] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 07/22/2020] [Accepted: 07/29/2020] [Indexed: 12/14/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) remains a lethal cancer. The poor prognosis calls for a more detailed understanding of disease biology in order to pave the way for the development of effective therapies. Typically, the pancreatic tumor is composed of a minority of malignant cells within an excessive tumor microenvironment (TME) consisting of extracellular matrix (ECM), fibroblasts, immune cells, and endothelial cells. Research conducted in recent years has particularly focused on cancer-associated fibroblasts (CAFs) which represent the most prominent cellular component of the desmoplastic stroma. Here, we review the complex crosstalk between CAFs, tumor cells, and other components of the TME, and illustrate how these interactions drive disease progression. We also discuss the emerging field of CAF heterogeneity, their tumor-supportive versus tumor-suppressive capacity, and the consequences for designing stroma-targeted therapies in the future.
Collapse
Affiliation(s)
| | | | - Thomas Wirth
- Institute of Physiological Chemistry, University of Ulm, 89081 Ulm, Germany; (N.S.); (M.T.)
| |
Collapse
|
50
|
Pancreatic Cancer Associated Fibroblasts (CAF): Under-Explored Target for Pancreatic Cancer Treatment. Cancers (Basel) 2020; 12:cancers12051347. [PMID: 32466266 PMCID: PMC7281461 DOI: 10.3390/cancers12051347] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 05/12/2020] [Accepted: 05/19/2020] [Indexed: 02/07/2023] Open
Abstract
Pancreatic cancer is the 4th leading cause of cancer deaths in the United States. The pancreatic cancer phenotype is primarily a consequence of oncogenes disturbing the resident pancreas parenchymal cell repair program. Many solid tumor types including pancreatic cancer have severe tumor fibrosis called desmoplasia. Desmoplastic stroma is coopted by the tumor as a support structure and CAFs aid in tumor growth, invasion, and metastases. This stroma is caused by cancer associated fibroblasts (CAFs), which lay down extensive connective tissue in and around the tumor cells. CAFs represent a heterogeneous population of cells that produce various paracrine molecules such as transforming growth factor-beta (TGF-beta) and platelet derived growth factors (PDGFs) that aid tumor growth, local invasion, and development of metastases. The hard, fibrotic shell of desmoplasia serves as a barrier to the infiltration of both chemo- and immunotherapy drugs and host immune cells to the tumor. Although there have been recent improvements in chemotherapy and surgical techniques for management of pancreatic cancer, the majority of patients will die from this disease. Therefore, new treatment strategies are clearly needed. CAFs represent an under-explored potential therapeutic target. This paper discusses what we know about the role of CAFs in pancreatic cancer cell growth, invasion, and metastases. Additionally, we present different strategies that are being and could be explored as anti-CAF treatments for pancreatic cancer.
Collapse
|