1
|
Yadav R, Sharma A, Dahiya D, Bal A, Bhatia A. Comparative morphology of tumour microenvironment in claudin-low and claudin-high breast cancers. Pathol Res Pract 2024; 261:155502. [PMID: 39079385 DOI: 10.1016/j.prp.2024.155502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 07/15/2024] [Accepted: 07/27/2024] [Indexed: 08/18/2024]
Abstract
BACKGROUND Claudin-low breast cancers (BCs) exhibit more aggressive behaviour compared to claudin-high types. Claudin-low BCs are often characterized by features such as a higher grade, enrichment of stemness characteristics, and a propensity for metastasis. Tumour microenvironment (TME) defined as the intricate network of surrounding cells, blood vessels, and extracellular matrix components influences the behaviour of cancer cells within the breast tissue. Understanding the TME is crucial for comprehending the aggressive characteristics of claudin-low BCs. METHODS In this study, we have studied the morphology of immune and non-immune TME using Haematoxylin and eosin (H&E)-stained slides of 15 claudin-low and 12 claudin-high tissue samples of BC. RESULTS TME of claudin-low BCs was observed to have a significantly higher frequency of retraction clefts (66.6 %; n = 10/15), immature desmoplastic response (40 %; n = 6/15), higher stromal cellularity (60 %; n = 9/15); and fibroblastic proliferation (53.3 %; n = 8/15) with a low prevalence of elastosis (66.6 %; n = 10/15). The immune microenvironment revealed a higher frequency of total (80 %; n = 12/15) as well as stromal (86.67 %; n = 13/15) and intra-tumoural TILs (60 %; n = 9/15) in them. CONCLUSION The above morphology-based study revealed that claudin-low tumours have unique immune and non-immune TME as compared to claudin-high tumours. Future studies exploring the molecular correlates of each of the above morphological features can help in identifying novel therapeutic targets for the treatment of claudin-low BCs.
Collapse
Affiliation(s)
- Reena Yadav
- Department of Experimental Medicine and Biotechnology, Postgraduate Institute of Medical Education and Research, Chandigarh, India.
| | - Aditti Sharma
- Department of Experimental Medicine and Biotechnology, Postgraduate Institute of Medical Education and Research, Chandigarh, India.
| | - Divya Dahiya
- Department of General Surgery, Postgraduate Institute of Medical Education and Research, Chandigarh, India.
| | - Amanjit Bal
- Department of Histopathology, Postgraduate Institute of Medical Education and Research, Chandigarh, India.
| | - Alka Bhatia
- Department of Experimental Medicine and Biotechnology, Postgraduate Institute of Medical Education and Research, Chandigarh, India.
| |
Collapse
|
2
|
Liu X, Ren B, Fang Y, Ren J, Wang X, Gu M, Zhou F, Xiao R, Luo X, You L, Zhao Y. Comprehensive analysis of bulk and single-cell transcriptomic data reveals a novel signature associated with endoplasmic reticulum stress, lipid metabolism, and liver metastasis in pancreatic cancer. J Transl Med 2024; 22:393. [PMID: 38685045 PMCID: PMC11057100 DOI: 10.1186/s12967-024-05158-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 04/02/2024] [Indexed: 05/02/2024] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is a lethal malignancy with high probability of recurrence and distant metastasis. Liver metastasis is the predominant metastatic mode developed in most pancreatic cancer cases, which seriously affects the overall survival rate of patients. Abnormally activated endoplasmic reticulum stress and lipid metabolism reprogramming are closely related to tumor growth and metastasis. This study aims to construct a prognostic model based on endoplasmic reticulum stress and lipid metabolism for pancreatic cancer, and further explore its correlation with tumor immunity and the possibility of immunotherapy. METHODS Transcriptomic and clinical data are acquired from TCGA, ICGC, and GEO databases. Potential prognostic genes were screened by consistent clustering and WGCNA methods, and the whole cohort was randomly divided into training and testing groups. The prognostic model was constructed by machine learning method in the training cohort and verified in the test, TCGA and ICGC cohorts. The clinical application of this model and its relationship with tumor immunity were analyzed, and the relationship between endoplasmic reticulum stress and intercellular communication was further explored. RESULTS A total of 92 characteristic genes related to endoplasmic reticulum stress, lipid metabolism and liver metastasis were identified in pancreatic cancer. We established and validated a prognostic model for pancreatic cancer with 7 signatures, including ADH1C, APOE, RAP1GAP, NPC1L1, P4HB, SOD2, and TNFSF10. This model is considered to be an independent prognosticator and is a more accurate predictor of overall survival than age, gender, and stage. TIDE score was increased in high-risk group, while the infiltration levels of CD8+ T cells and M1 macrophages were decreased. The number and intensity of intercellular communication were increased in the high ER stress group. CONCLUSIONS We constructed and validated a novel prognostic model for pancreatic cancer, which can also be used as an instrumental variable to predict the prognosis and immune microenvironment. In addition, this study revealed the effect of ER stress on cell-cell communication in the tumor microenvironment.
Collapse
Affiliation(s)
- Xiaohong Liu
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100023, People's Republic of China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, 100023, People's Republic of China
- National Science and Technology Key Infrastructure On Translational Medicine in Peking Union Medical College Hospital, Beijing, 100023, People's Republic of China
| | - Bo Ren
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100023, People's Republic of China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, 100023, People's Republic of China
- National Science and Technology Key Infrastructure On Translational Medicine in Peking Union Medical College Hospital, Beijing, 100023, People's Republic of China
| | - Yuan Fang
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100023, People's Republic of China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, 100023, People's Republic of China
- National Science and Technology Key Infrastructure On Translational Medicine in Peking Union Medical College Hospital, Beijing, 100023, People's Republic of China
| | - Jie Ren
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100023, People's Republic of China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, 100023, People's Republic of China
- National Science and Technology Key Infrastructure On Translational Medicine in Peking Union Medical College Hospital, Beijing, 100023, People's Republic of China
| | - Xing Wang
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100023, People's Republic of China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, 100023, People's Republic of China
- National Science and Technology Key Infrastructure On Translational Medicine in Peking Union Medical College Hospital, Beijing, 100023, People's Republic of China
| | - Minzhi Gu
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100023, People's Republic of China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, 100023, People's Republic of China
- National Science and Technology Key Infrastructure On Translational Medicine in Peking Union Medical College Hospital, Beijing, 100023, People's Republic of China
| | - Feihan Zhou
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100023, People's Republic of China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, 100023, People's Republic of China
- National Science and Technology Key Infrastructure On Translational Medicine in Peking Union Medical College Hospital, Beijing, 100023, People's Republic of China
| | - Ruiling Xiao
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100023, People's Republic of China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, 100023, People's Republic of China
- National Science and Technology Key Infrastructure On Translational Medicine in Peking Union Medical College Hospital, Beijing, 100023, People's Republic of China
| | - Xiyuan Luo
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100023, People's Republic of China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, 100023, People's Republic of China
- National Science and Technology Key Infrastructure On Translational Medicine in Peking Union Medical College Hospital, Beijing, 100023, People's Republic of China
| | - Lei You
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100023, People's Republic of China.
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, 100023, People's Republic of China.
- National Science and Technology Key Infrastructure On Translational Medicine in Peking Union Medical College Hospital, Beijing, 100023, People's Republic of China.
| | - Yupei Zhao
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100023, People's Republic of China.
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, 100023, People's Republic of China.
- National Science and Technology Key Infrastructure On Translational Medicine in Peking Union Medical College Hospital, Beijing, 100023, People's Republic of China.
| |
Collapse
|
3
|
Gil-Henn H, Girault JA, Lev S. PYK2, a hub of signaling networks in breast cancer progression. Trends Cell Biol 2024; 34:312-326. [PMID: 37586982 DOI: 10.1016/j.tcb.2023.07.006] [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: 05/01/2023] [Revised: 07/17/2023] [Accepted: 07/18/2023] [Indexed: 08/18/2023]
Abstract
Breast cancer (BC) involves complex signaling networks characterized by extensive cross-communication and feedback loops between and within multiple signaling cascades. Many of these signaling pathways are driven by genetic alterations of oncogene and/or tumor-suppressor genes and are influenced by various environmental cues. We describe unique roles of the non-receptor tyrosine kinase (NRTK) PYK2 in signaling integration and feedback looping in BC. PYK2 functions as a signaling hub in various cascades, and its involvement in positive and negative feedback loops enhances signaling robustness, modulates signaling dynamics, and contributes to BC growth, epithelial-to-mesenchymal transition (EMT), stemness, migration, invasion, and metastasis. We also discuss the potential of PYK2 as a therapeutic target in various BC subtypes.
Collapse
Affiliation(s)
- Hava Gil-Henn
- The Azrieli Faculty of Medicine, Bar-Ilan University, Safed 1311502, Israel
| | - Jean-Antoine Girault
- Institut du Fer à Moulin, Institut National de la Santé et de la Recherche Médicale (INSERM) Unité Mixte de Recherche en Santé (UMRS) 1270, Sorbonne Université, 75005 Paris, France
| | - Sima Lev
- Molecular Cell Biology Department, Weizmann Institute of Science, Rehovot 76100, Israel.
| |
Collapse
|
4
|
Alzamami A. Implications of single-cell immune landscape of tumor microenvironment for the colorectal cancer diagnostics and therapy. Med Oncol 2023; 40:352. [PMID: 37950801 DOI: 10.1007/s12032-023-02226-z] [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: 09/11/2023] [Accepted: 10/18/2023] [Indexed: 11/13/2023]
Abstract
Colorectal cancer (CRC) originates from the polyps lining the colon and is among the most common types of cancer. With the increasing popularity of single-cell sequencing technologies, researchers have been able to better understand the immune landscape of colorectal cancer, by analyzing their expression and interactions in detail with the tumor microenvironment (TME) at single-cell level. Since the tumor-immune cell interactions play a critical part in the advancement as well as treatment response in colorectal cancer, the release of inhibitory factors such as T cells are important for recognizing and destroying cancer cells. Such information is vital to identify immunotherapeutic targets for cure and monitoring response to treatments. Therefore, a comprehensive single-cell studies-based overview of key immunogenic agents regulating the TME of CRC is provided in this review. Tumor-associated macrophages can promote tumor growth and resistance to treatment by releasing factors that inhibit the function of other immune cells. Additionally, colorectal cancer cells can express programmed cell death protein 1 and its ligand, which can also inhibit T-cell function. Researchers have found that certain types of immune cells, prominently T cells, natural killer, and dendritic cells, can have a positive impact on the prognosis of colorectal cancer patients. Treatments like immune checkpoint inhibitors and CAR-T therapies that help to release the inhibitory signals from the cancer cells allow the immune cells to function more effectively.
Collapse
Affiliation(s)
- Ahmad Alzamami
- Clinical Laboratory Science Department, College of Applied Medical Sciences, Shaqra University, 11961, Al-Quwayiyah, Saudi Arabia.
| |
Collapse
|
5
|
Entenberg D, Oktay MH, Condeelis JS. Intravital imaging to study cancer progression and metastasis. Nat Rev Cancer 2023; 23:25-42. [PMID: 36385560 PMCID: PMC9912378 DOI: 10.1038/s41568-022-00527-5] [Citation(s) in RCA: 28] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/11/2022] [Indexed: 11/17/2022]
Abstract
Navigation through the bulk tumour, entry into the blood vasculature, survival in the circulation, exit at distant sites and resumption of proliferation are all steps necessary for tumour cells to successfully metastasize. The ability of tumour cells to complete these steps is highly dependent on the timing and sequence of the interactions that these cells have with the tumour microenvironment (TME), including stromal cells, the extracellular matrix and soluble factors. The TME thus plays a major role in determining the overall metastatic phenotype of tumours. The complexity and cause-and-effect dynamics of the TME cannot currently be recapitulated in vitro or inferred from studies of fixed tissue, and are best studied in vivo, in real time and at single-cell resolution. Intravital imaging (IVI) offers these capabilities, and recent years have been a time of immense growth and innovation in the field. Here we review some of the recent advances in IVI of mammalian models of cancer and describe how IVI is being used to understand cancer progression and metastasis, and to develop novel treatments and therapies. We describe new techniques that allow access to a range of tissue and cancer types, novel fluorescent reporters and biosensors that allow fate mapping and the probing of functional and phenotypic states, and the clinical applications that have arisen from applying these techniques, reporters and biosensors to study cancer. We finish by presenting some of the challenges that remain in the field, how to address them and future perspectives.
Collapse
Affiliation(s)
- David Entenberg
- Gruss Lipper Biophotonics Center, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA.
- Integrated Imaging Program, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA.
- Department of Pathology, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA.
| | - Maja H Oktay
- Gruss Lipper Biophotonics Center, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA.
- Integrated Imaging Program, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA.
- Department of Pathology, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA.
- Department of Surgery, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA.
| | - John S Condeelis
- Gruss Lipper Biophotonics Center, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA.
- Integrated Imaging Program, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA.
- Department of Surgery, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA.
- Department of Cell Biology, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA.
| |
Collapse
|
6
|
Peña-Romero AC, Orenes-Piñero E. Dual Effect of Immune Cells within Tumour Microenvironment: Pro- and Anti-Tumour Effects and Their Triggers. Cancers (Basel) 2022; 14:1681. [PMID: 35406451 PMCID: PMC8996887 DOI: 10.3390/cancers14071681] [Citation(s) in RCA: 83] [Impact Index Per Article: 41.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 03/21/2022] [Accepted: 03/23/2022] [Indexed: 02/04/2023] Open
Abstract
Our body is constantly exposed to pathogens or external threats, but with the immune response that our body can develop, we can fight off and defeat possible attacks or infections. Nevertheless, sometimes this threat comes from an internal factor. Situations such as the existence of a tumour also cause our immune system (IS) to be put on alert. Indeed, the link between immunology and cancer is evident these days, with IS being used as one of the important targets for treating cancer. Our IS is able to eliminate those abnormal or damaged cells found in our body, preventing the uncontrolled proliferation of tumour cells that can lead to cancer. However, in several cases, tumour cells can escape from the IS. It has been observed that immune cells, the extracellular matrix, blood vessels, fat cells and various molecules could support tumour growth and development. Thus, the developing tumour receives structural support, irrigation and energy, among other resources, making its survival and progression possible. All these components that accompany and help the tumour to survive and to grow are called the tumour microenvironment (TME). Given the importance of its presence in the tumour development process, this review will focus on one of the components of the TME: immune cells. Immune cells can support anti-tumour immune response protecting us against tumour cells; nevertheless, they can also behave as pro-tumoural cells, thus promoting tumour progression and survival. In this review, the anti-tumour and pro-tumour immunity of several immune cells will be discussed. In addition, the TME influence on this dual effect will be also analysed.
Collapse
Affiliation(s)
| | - Esteban Orenes-Piñero
- Department of Biochemistry and Molecular Biology-A, University of Murcia, 30120 Murcia, Spain;
| |
Collapse
|
7
|
Mazza C, Gaydou V, Eymard JC, Birembaut P, Untereiner V, Côté JF, Brocheriou I, Coeffic D, Villena P, Larré S, Vuiblet V, Piot O. Identification of Neoadjuvant Chemotherapy Response in Muscle-Invasive Bladder Cancer by Fourier-Transform Infrared Micro-Imaging. Cancers (Basel) 2021; 14:cancers14010021. [PMID: 35008184 PMCID: PMC8750189 DOI: 10.3390/cancers14010021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 12/09/2021] [Accepted: 12/17/2021] [Indexed: 11/24/2022] Open
Abstract
Simple Summary Assessing the tumor response to chemotherapy is a paramount predictive step to improve patient care. Infrared spectroscopy probes the chemical composition of samples, and in combination with statistical multivariate processing, presents the capacity to highlight subtle molecular alterations associated with malignancy characteristics. Microscopic infrared imaging of tissue samples reveals spectral heterogeneity within histological structures, providing a new approach to characterize tumoral heterogeneity. We have taken advantage of the analytical capabilities of mid-infrared spectral imaging to implement a classification model to predict the response of a tumor to chemotherapy. Our development was demonstrated in muscle-invasive bladder cancer (MIBC) by comparing samples from responders and non-responders to neoadjuvant chemotherapy. Abstract Background: Neoadjuvant chemotherapy (NAC) improves survival in responder patients. However, for non-responders, the treatment represents an ineffective exposure to chemotherapy and its potential adverse events. Predicting the response to treatment is a major issue in the therapeutic management of patients, particularly for patients with muscle-invasive bladder cancer. Methods: Tissue samples of trans-urethral resection of bladder tumor collected at the diagnosis time, were analyzed by mid-infrared imaging. A sequence of spectral data processing was implemented for automatic recognition of informative pixels and scoring each pixel according to a continuous scale (from 0 to 10) associated with the response to NAC. The ground truth status of the responder or non-responder was based on histopathological examination of the samples. Results: Although the TMA spots of tumors appeared histologically homogeneous, the infrared approach highlighted spectral heterogeneity. Both the quantification of this heterogeneity and the scoring of the NAC response at the pixel level were used to construct sensitivity and specificity maps from which decision criteria can be extracted to classify cancerous samples. Conclusions: This proof-of-concept appears as the first to evaluate the potential of the mid-infrared approach for the prediction of response to neoadjuvant chemotherapy in MIBC tissues.
Collapse
Affiliation(s)
- Camille Mazza
- Jean Godinot Institute, 51100 Reims, France; (C.M.); (J.-C.E.)
| | - Vincent Gaydou
- BioSpecT (Translational BioSpectroscopy) EA 7506, SFR Santé, Université de Reims Champagne-Ardenne, 51100 Reims, France; (V.G.); (S.L.)
| | | | - Philippe Birembaut
- Department of Biopathology, University Hospital of Reims, 51100 Reims, France;
| | - Valérie Untereiner
- Cellular and Tissular Imaging Platform (PICT), Université de Reims Champagne-Ardenne, 51100 Reims, France;
| | - Jean-François Côté
- Department of Biopathology, Hôpital de la Pitié-Salpêtrière, APHP, 51100 Paris, France; (J.-F.C.); (I.B.)
| | - Isabelle Brocheriou
- Department of Biopathology, Hôpital de la Pitié-Salpêtrière, APHP, 51100 Paris, France; (J.-F.C.); (I.B.)
| | - David Coeffic
- Polyclinique Courlancy, 51100 Reims, France; (D.C.); (P.V.)
| | | | - Stéphane Larré
- BioSpecT (Translational BioSpectroscopy) EA 7506, SFR Santé, Université de Reims Champagne-Ardenne, 51100 Reims, France; (V.G.); (S.L.)
- Department of Urology, University Hospital of Reims, 51100 Reims, France
| | - Vincent Vuiblet
- BioSpecT (Translational BioSpectroscopy) EA 7506, SFR Santé, Université de Reims Champagne-Ardenne, 51100 Reims, France; (V.G.); (S.L.)
- Department of Biopathology, University Hospital of Reims, 51100 Reims, France;
- Correspondence: (V.V.); (O.P.)
| | - Olivier Piot
- BioSpecT (Translational BioSpectroscopy) EA 7506, SFR Santé, Université de Reims Champagne-Ardenne, 51100 Reims, France; (V.G.); (S.L.)
- Cellular and Tissular Imaging Platform (PICT), Université de Reims Champagne-Ardenne, 51100 Reims, France;
- Correspondence: (V.V.); (O.P.)
| |
Collapse
|
8
|
Diehl V, Huber LS, Trebicka J, Wygrecka M, Iozzo RV, Schaefer L. The Role of Decorin and Biglycan Signaling in Tumorigenesis. Front Oncol 2021; 11:801801. [PMID: 34917515 PMCID: PMC8668865 DOI: 10.3389/fonc.2021.801801] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 11/09/2021] [Indexed: 12/12/2022] Open
Abstract
The complex and adaptive nature of malignant neoplasm constitute a major challenge for the development of effective anti-oncogenic therapies. Emerging evidence has uncovered the pivotal functions exerted by the small leucine-rich proteoglycans, decorin and biglycan, in affecting tumor growth and progression. In their soluble forms, decorin and biglycan act as powerful signaling molecules. By receptor-mediated signal transduction, both proteoglycans modulate key processes vital for tumor initiation and progression, such as autophagy, inflammation, cell-cycle, apoptosis, and angiogenesis. Despite of their structural homology, these two proteoglycans interact with distinct cell surface receptors and thus modulate distinct signaling pathways that ultimately affect cancer development. In this review, we summarize growing evidence for the complex roles of decorin and biglycan signaling in tumor biology and address potential novel therapeutic implications.
Collapse
Affiliation(s)
- Valentina Diehl
- Institute of Pharmacology and Toxicology, Goethe University, Frankfurt, Germany
| | - Lisa Sophie Huber
- Institute of Pharmacology and Toxicology, Goethe University, Frankfurt, Germany
| | - Jonel Trebicka
- Department of Internal Medicine I, Goethe University, Frankfurt, Germany
| | - Malgorzata Wygrecka
- Center for Infection and Genomics of the Lung, Member of the German Center for Lung Research, University of Giessen and Marburg Lung Center, Giessen, Germany
| | - Renato V. Iozzo
- Department of Pathology, Anatomy and Cell Biology and the Translational Cellular Oncology Program, Sidney Kimmel Cancer Center, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA, United States
| | - Liliana Schaefer
- Institute of Pharmacology and Toxicology, Goethe University, Frankfurt, Germany
| |
Collapse
|
9
|
Modular microenvironment components reproduce vascular dynamics de novo in a multi-scale agent-based model. Cell Syst 2021; 12:795-809.e9. [PMID: 34139155 DOI: 10.1016/j.cels.2021.05.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 11/06/2020] [Accepted: 05/11/2021] [Indexed: 12/24/2022]
Abstract
Cells do not exist in isolation; they continuously act within and react to their environment. And this environment is not static; it continuously adapts and responds to cells. Here, we investigate how vascular structure and function impact emergent cell population behavior using an agent-based model (ABM). Our ABM enables researchers to "mix and match" cell agents, subcellular modules, and microenvironment components ranging from simple nutrient sources to complex, realistic vascular architectures that accurately capture hemodynamics. We use this ABM to highlight the bilateral relationship between cells and nearby vasculature, demonstrate the effect of vascular structure on environmental heterogeneity, and emphasize the non-linear, non-intuitive relationship between vascular function and the behavior of cell populations over time. Our ABM is well suited to characterizing in vitro and in vivo studies, with applications from basic science to translational synthetic biology and medicine. The model is freely available at https://github.com/bagherilab/ARCADE. A record of this paper's transparent peer review process is included in the supplemental information.
Collapse
|
10
|
Giorello MB, Borzone FR, Labovsky V, Piccioni FV, Chasseing NA. Cancer-Associated Fibroblasts in the Breast Tumor Microenvironment. J Mammary Gland Biol Neoplasia 2021; 26:135-155. [PMID: 33398516 DOI: 10.1007/s10911-020-09475-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 11/30/2020] [Indexed: 12/11/2022] Open
Abstract
Years of investigation have shed light on a theory in which breast tumor epithelial cells are under the effect of the stromal microenvironment. This review aims to discuss recent findings concerning the phenotypic and functional characteristics of cancer associated fibroblasts (CAFs) and their involvement in tumor evolution, as well as their potential implications for anti-cancer therapy. In this manuscript, we reviewed that CAFs play a fundamental role in initiation, growth, invasion, and metastasis of breast cancer, and also serve as biomarkers in the clinical diagnosis, therapy, and prognosis of this disease.
Collapse
Affiliation(s)
- María Belén Giorello
- Laboratorio de Inmunohematología (IBYME) - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Biología y Medicina Experimental, Vuelta de Obligado 2490, CP, 1428, Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina.
| | - Francisco Raúl Borzone
- Laboratorio de Inmunohematología (IBYME) - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Biología y Medicina Experimental, Vuelta de Obligado 2490, CP, 1428, Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - Vivian Labovsky
- Laboratorio de Inmunohematología (IBYME) - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Biología y Medicina Experimental, Vuelta de Obligado 2490, CP, 1428, Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - Flavia Valeria Piccioni
- Laboratorio de Oncología Molecular y Nuevos Blancos Terapéuticos (IBYME) y Laboratorio de Inmunohematología (IBYME) - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Biología y Medicina Experimental, Vuelta de Obligado 2490, CP, 1428, Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - Norma Alejandra Chasseing
- Laboratorio de Inmunohematología (IBYME) - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Biología y Medicina Experimental, Vuelta de Obligado 2490, CP, 1428, Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina.
| |
Collapse
|
11
|
Peng L, Zhang Y, Wang Z. Immune Responses against Disseminated Tumor Cells. Cancers (Basel) 2021; 13:2515. [PMID: 34063848 PMCID: PMC8196619 DOI: 10.3390/cancers13112515] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 05/17/2021] [Accepted: 05/19/2021] [Indexed: 02/07/2023] Open
Abstract
Most cancer-related deaths are a consequence of metastases, a series of linear events, notably the invasion-metastasis cascade. The current understanding of cancer immune surveillance derives from studies in primary tumors, but disseminated cancer cells acquire mutations and, in some cases, appear to progress independently after spreading from primary sites. An early step in this process is micrometastatic dissemination. As such, the equilibrium between the immune system and disseminated cancer cells controls the fate of the cancer. Immune checkpoint inhibitors (ICIs) exhibit significant clinical activity in patients, but the efficacy of ICIs depends on both the tumor and its microenvironment. Data often suggest that disseminated cancer cells are not adequately targeted by the immune system. In this review, we summarize the main basic findings of immune responses against disseminated tumor cells and their organ-specific characteristics. Such studies may provide new directions for cancer immune therapy.
Collapse
Affiliation(s)
- Ling Peng
- Department of Pulmonary and Critical Care Medicine, Zhejiang Provincial People’s Hospital, Hangzhou 310014, China
| | - Yongchang Zhang
- Lung Cancer and Gastrointestinal Unit, Department of Medical Oncology, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Changsha 410013, China;
| | - Zibing Wang
- Department of Immunotherapy, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou 450008, China
| |
Collapse
|
12
|
Immune Checkpoint Inhibition in Metastatic Colorectal Cancer Harboring Microsatellite Instability or Mismatch Repair Deficiency. Cancers (Basel) 2021; 13:cancers13051149. [PMID: 33800202 PMCID: PMC7962453 DOI: 10.3390/cancers13051149] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 03/02/2021] [Accepted: 03/04/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Microsatellite instability (MSI) is a molecular indicator of defective DNA mismatch repair (dMMR). MSI/dMMR status is observed in approximately 5% of metastatic colorectal cancers (mCRC) but 10–18% of localized colorectal cancers. MSI/dMMR status is a major predictive biomarker for the efficacy of immune checkpoint inhibitors (ICIs). This review presents the current and future challenges of ICIs for patients with MSI/dMMR colorectal cancer. Abstract Microsatellite instability (MSI) is a tumor phenotype related to a deficient DNA mismatch repair system (dMMR). This phenotype, observed in 5% of metastatic mCRC but 10–18% of localized CRC, is associated with high tumor mutational burden with highly immunogenic neoantigens. It has emerged as a major predictive biomarker for the efficacy of ICIs. In this review, we will present a comprehensive overview of the literature concerning the efficacy of ICIs in MSI/dMMR mCRC, with a focus on new developments in first-line metastatic setting. Then, we will present current and future challenges of immuno-oncology for patients with MSI/dMMR metastatic CRC.
Collapse
|
13
|
Holstein E, Dittmann A, Kääriäinen A, Pesola V, Koivunen J, Pihlajaniemi T, Naba A, Izzi V. The Burden of Post-Translational Modification (PTM)-Disrupting Mutations in the Tumor Matrisome. Cancers (Basel) 2021; 13:1081. [PMID: 33802493 PMCID: PMC7959462 DOI: 10.3390/cancers13051081] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 02/25/2021] [Accepted: 02/26/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND To evaluate the occurrence of mutations affecting post-translational modification (PTM) sites in matrisome genes across different tumor types, in light of their genomic and functional contexts and in comparison with the rest of the genome. METHODS This study spans 9075 tumor samples and 32 tumor types from The Cancer Genome Atlas (TCGA) Pan-Cancer cohort and identifies 151,088 non-silent mutations in the coding regions of the matrisome, of which 1811 affecting known sites of hydroxylation, phosphorylation, N- and O-glycosylation, acetylation, ubiquitylation, sumoylation and methylation PTM. RESULTS PTM-disruptive mutations (PTMmut) in the matrisome are less frequent than in the rest of the genome, seem independent of cell-of-origin patterns but show dependence on the nature of the matrisome protein affected and the background PTM types it generally harbors. Also, matrisome PTMmut are often found among structural and functional protein regions and in proteins involved in homo- and heterotypic interactions, suggesting potential disruption of matrisome functions. CONCLUSIONS Though quantitatively minoritarian in the spectrum of matrisome mutations, PTMmut show distinctive features and damaging potential which might concur to deregulated structural, functional, and signaling networks in the tumor microenvironment.
Collapse
Affiliation(s)
- Elisa Holstein
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, FI-90014 Oulu, Finland; (E.H.); (A.D.); (A.K.); (V.P.); (J.K.); (T.P.)
| | - Annalena Dittmann
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, FI-90014 Oulu, Finland; (E.H.); (A.D.); (A.K.); (V.P.); (J.K.); (T.P.)
| | - Anni Kääriäinen
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, FI-90014 Oulu, Finland; (E.H.); (A.D.); (A.K.); (V.P.); (J.K.); (T.P.)
| | - Vilma Pesola
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, FI-90014 Oulu, Finland; (E.H.); (A.D.); (A.K.); (V.P.); (J.K.); (T.P.)
| | - Jarkko Koivunen
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, FI-90014 Oulu, Finland; (E.H.); (A.D.); (A.K.); (V.P.); (J.K.); (T.P.)
| | - Taina Pihlajaniemi
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, FI-90014 Oulu, Finland; (E.H.); (A.D.); (A.K.); (V.P.); (J.K.); (T.P.)
| | - Alexandra Naba
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, IL 60612, USA;
- University of Illinois Cancer Center, Chicago, IL 60612, USA
| | - Valerio Izzi
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, FI-90014 Oulu, Finland; (E.H.); (A.D.); (A.K.); (V.P.); (J.K.); (T.P.)
- Faculty of Medicine, University of Oulu, FI-90014 Oulu, Finland
- Finnish Cancer Institute, 00130 Helsinki, Finland
| |
Collapse
|
14
|
Hachim MY, Hachim IY, Talaat IM, Yakout NM, Hamoudi R. M1 Polarization Markers Are Upregulated in Basal-Like Breast Cancer Molecular Subtype and Associated With Favorable Patient Outcome. Front Immunol 2020; 11:560074. [PMID: 33304345 PMCID: PMC7701279 DOI: 10.3389/fimmu.2020.560074] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 10/16/2020] [Indexed: 12/29/2022] Open
Abstract
Background Breast cancer heterogeneity is an essential element that plays a role in the therapy response variability and the patient's outcome. This highlights the need for more precise subtyping methods that focus not only on tumor cells but also investigate the profile of stromal cells as well as immune cells. Objectives To mine publicly available transcriptomic breast cancer datasets and reanalyze their transcriptomic profiling using unsupervised clustering in order to identify novel subsets in molecular subtypes of breast cancer, then explore the stromal and immune cells profile in each subset using bioinformatics and systems immunology approaches. Materials and Methods Transcriptomic data from 1,084 breast cancer patients obtained from The Cancer Genome Atlas (TCGA) database were extracted and subjected to unsupervised clustering using a recently described, multi-step algorithm called Iterative Clustering and Guide-gene Selection (ICGS). For each cluster, the stromal and immune profile was investigated using ESTIMATE and CIBERSORT analytical tool. Clinical outcomes and differentially expressed genes of the characterized clusters were identified and validated in silico and in vitro in a cohort of 80 breast cancer samples by immunohistochemistry. Results Seven unique sub-clusters showed distinct molecular and clinical profiles between the well-known breast cancer subtypes. Those unsupervised clusters identified more homogenous subgroups in each of the classical subtypes with a different prognostic profile. Immune profiling of the identified clusters showed that while the classically activated macrophages (M1) are correlated with the more aggressive basal-like breast cancer subtype, the alternatively activated macrophages (M2) showed a higher level of infiltration in luminal A and luminal B subtypes. Indeed, patients with higher levels of M1 expression showed less advanced disease and better patient outcomes presented as prolonged overall survival. Moreover, the M1 high basal-like breast cancer group showed a higher expression of interferon-gamma induced chemokines and guanylate-binding proteins (GBPs) involved in immunity against microbes. Conclusion Adding immune profiling using transcriptomic data can add precision for diagnosis and prognosis and can cluster patients according to the available modalities of therapy in a more personalized approach.
Collapse
Affiliation(s)
- Mahmood Yaseen Hachim
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates
| | - Ibrahim Yaseen Hachim
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates
- Clinical Sciences Department, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Iman M. Talaat
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates
- Clinical Sciences Department, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
- Pathology Department, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Nada M. Yakout
- Pathology Department, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Rifat Hamoudi
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates
- Clinical Sciences Department, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
- Division of Surgery and Interventional Science, University College London, London, United Kingdom
| |
Collapse
|
15
|
Velu P, Vijayalakshmi A, Vinothkumar V. Inhibiting the PI3K/Akt, NF-κB signalling pathways with syringic acid for attenuating the development of oral squamous cell carcinoma cells SCC131. J Pharm Pharmacol 2020; 72:1595-1606. [PMID: 32790092 DOI: 10.1111/jphp.13350] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 07/15/2020] [Indexed: 12/25/2022]
Abstract
OBJECTIVES To evaluate the anti-inflammatory and antiproliferative effect of syringic acid (SRA) on oral squamous cell carcinoma (OSCC) SCC131 cells via suppression of NF-κB-induced PI3K/Akt signalling pathway. METHODS The present study assesses the anticancer effects of SRA alongside human oral cancer (HOC) SCC131 cells through the fabrication of reactive oxygen species (ROS) and activated apoptosis. DAPI and Rh-123 staining were used to assess the apoptotic nuclear characteristic, mitochondrial membrane potential, cell adhesion and migration by fluorescence microscope with SRA treatment. KEY FINDINGS Syringic acid inhibits cell viability (IC50 values of 25 µm), adhesion, migration and induced apoptosis. MTT assay demonstrated SRA-induced apoptotic events, inhibition of invasion and angiogenic signalling in SCC131 cell line. Furthermore, SRA treated with SCC131 cells suppresses the protein expression of inflammatory, angiogenesis and PI3K/Akt signalling pathways. It is suggested that SRA prevents the translocation of NF-κB and PI3K/Akt activated products to the nucleus, thereby suppressing angiogenesis via downregulation of vascular endothelial growth factor. CONCLUSIONS Therefore, addition of SRA to SCC131 cells may provide a promising natural therapeutic strategy against squamous cell carcinomas with potential application in clinical analysis.
Collapse
Affiliation(s)
- Periyannan Velu
- Department of Biochemistry and Biotechnology, Faculty of Science, Annamalai University, Chidambaram, Tamil Nadu, India
| | - Annamalai Vijayalakshmi
- Department of Biochemistry and Biotechnology, Faculty of Science, Annamalai University, Chidambaram, Tamil Nadu, India
| | - Veerasamy Vinothkumar
- Department of Biochemistry and Biotechnology, Faculty of Science, Annamalai University, Chidambaram, Tamil Nadu, India
| |
Collapse
|
16
|
Izzi V, Davis MN, Naba A. Pan-Cancer Analysis of the Genomic Alterations and Mutations of the Matrisome. Cancers (Basel) 2020; 12:E2046. [PMID: 32722287 PMCID: PMC7463652 DOI: 10.3390/cancers12082046] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 07/17/2020] [Accepted: 07/22/2020] [Indexed: 12/13/2022] Open
Abstract
The extracellular matrix (ECM) is a master regulator of all cellular functions and a major component of the tumor microenvironment. We previously defined the "matrisome" as the ensemble of genes encoding ECM proteins and proteins modulating ECM structure or function. While compositional and biomechanical changes in the ECM regulate cancer progression, no study has investigated the genomic alterations of matrisome genes in cancers and their consequences. Here, mining The Cancer Genome Atlas (TCGA) data, we found that copy number alterations and mutations are frequent in matrisome genes, even more so than in the rest of the genome. We also found that these alterations are predicted to significantly impact gene expression and protein function. Moreover, we identified matrisome genes whose mutational burden is an independent predictor of survival. We propose that studying genomic alterations of matrisome genes will further our understanding of the roles of this compartment in cancer progression and will lead to the development of innovative therapeutic strategies targeting the ECM.
Collapse
Affiliation(s)
- Valerio Izzi
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, FI-90014 Oulu, Finland;
- Finnish Cancer Institute, 00130 Helsinki, Finland
| | - Martin N. Davis
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, IL 60612, USA;
| | - Alexandra Naba
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, IL 60612, USA;
- University of Illinois Cancer Center, Chicago, IL 60612, USA
| |
Collapse
|
17
|
Borriello L, Karagiannis GS, Duran CL, Coste A, Oktay MH, Entenberg D, Condeelis JS. The role of the tumor microenvironment in tumor cell intravasation and dissemination. Eur J Cell Biol 2020; 99:151098. [PMID: 32800278 DOI: 10.1016/j.ejcb.2020.151098] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 06/15/2020] [Accepted: 06/29/2020] [Indexed: 01/11/2023] Open
Abstract
Metastasis, a process that requires tumor cell dissemination followed by tumor growth, is the primary cause of death in cancer patients. An essential step of tumor cell dissemination is intravasation, a process by which tumor cells cross the blood vessel endothelium and disseminate to distant sites. Studying this process is of utmost importance given that intravasation in the primary tumor, as well as the secondary and tertiary metastases, is the key step in the systemic spread of tumor cells, and that this process continues even after removal of the primary tumor. High-resolution intravital imaging of the tumor microenvironment of breast carcinoma has revealed that tumor cell intravasation exclusively occurs at doorways, termed "Tumor MicroEnvironment of Metastasis" (TMEM), composed of three different cell types: a Tie2high/VEGFhigh perivascular macrophage, a Mena overexpressing tumor cell, and an endothelial cell, all in direct contact. In this review article, we discuss the interactions between these cell types, the subsequent signaling events which lead to tumor cell intravasation, and the role of invadopodia in supporting tumor cell invasion and dissemination. We end our review by discussing how the knowledge acquired from the use of intravital imaging is now leading to new clinical trials targeting tumor cell dissemination and preventing metastatic progression.
Collapse
Affiliation(s)
- Lucia Borriello
- Department of Anatomy and Structural Biology, Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, USA; Gruss-Lipper Biophotonics Center, Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, USA.
| | - George S Karagiannis
- Department of Anatomy and Structural Biology, Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, USA; Gruss-Lipper Biophotonics Center, Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, USA; Integrated Imaging Program, Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, USA
| | - Camille L Duran
- Department of Anatomy and Structural Biology, Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, USA; Gruss-Lipper Biophotonics Center, Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, USA; Integrated Imaging Program, Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, USA
| | - Anouchka Coste
- Department of Anatomy and Structural Biology, Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, USA; Gruss-Lipper Biophotonics Center, Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, USA; Department of Surgery, Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, USA
| | - Maja H Oktay
- Department of Anatomy and Structural Biology, Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, USA; Gruss-Lipper Biophotonics Center, Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, USA; Integrated Imaging Program, Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, USA; Department of Pathology, Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, USA
| | - David Entenberg
- Department of Anatomy and Structural Biology, Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, USA; Gruss-Lipper Biophotonics Center, Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, USA; Integrated Imaging Program, Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, USA.
| | - John S Condeelis
- Department of Anatomy and Structural Biology, Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, USA; Gruss-Lipper Biophotonics Center, Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, USA; Integrated Imaging Program, Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, USA; Department of Surgery, Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, USA.
| |
Collapse
|
18
|
Bo X, Wang J, Wang C, Nan L, Gao Z, Xin Y, Li M, Shen S, Liu H, Ni X, Suo T, Zhang D, Lu P, Wang Y, Liu H. High infiltration of mast cells is associated with improved response to adjuvant chemotherapy in gallbladder cancer. Cancer Sci 2020; 111:817-825. [PMID: 31925976 PMCID: PMC7060478 DOI: 10.1111/cas.14302] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 12/09/2019] [Accepted: 12/27/2019] [Indexed: 12/18/2022] Open
Abstract
Recent studies have reported that tumor‐infiltrating mast cells (TIM) play an important role in tumor regression, but the effect of TIM in gallbladder cancer (GBC) remains unclear. The present study aims to investigate the prognostic value of TIM in GBC patients and its responsiveness to gemcitabine‐based adjuvant chemotherapy (ACT). A total of 298 GBC patients from Zhongshan Hospital were recruited for this study. TIM infiltration was measured by immunohistochemical staining. Accumulation of TIM is significantly associated with prolonged overall survival in GBC patients. The benefit from gemcitabine‐based ACT was superior among patients with high infiltration of TIM with GBC. Multivariate analysis identified TIM infiltration as an independent prognostic factor for overall survival. A heatmap showed that TIM‐activated gene signatures were positively correlated with CD8+ T cells' gene signatures. Gene set enrichment analysis (GSEA) suggested that TIM was related to multiple T cell‐related processes and signaling pathways, including the interferon gamma signaling pathway and the leukocyte migration signaling pathway. It was confirmed that CD8+ T cell infiltration was positively correlated with high TIM infiltration in tissue microarray (TMA), suggesting that TIM infiltration was linked to the immune surveillance in GBC. TIM can be used as an independent prognostic factor and a predictor of therapeutic response of gemcitabine‐based ACT in GBC patients, which may mediate immune surveillance by recruiting and activating CD8+ T cells in GBC.
Collapse
Affiliation(s)
- Xiaobo Bo
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.,Biliary tract disease Institute, Fudan University, Shanghai, China
| | - Jie Wang
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.,Biliary tract disease Institute, Fudan University, Shanghai, China
| | - Changcheng Wang
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.,Biliary tract disease Institute, Fudan University, Shanghai, China
| | - Lingxi Nan
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.,Biliary tract disease Institute, Fudan University, Shanghai, China
| | - Zhihui Gao
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.,Biliary tract disease Institute, Fudan University, Shanghai, China
| | - Yanlei Xin
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.,Biliary tract disease Institute, Fudan University, Shanghai, China
| | - Min Li
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.,Biliary tract disease Institute, Fudan University, Shanghai, China
| | - Sheng Shen
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.,Biliary tract disease Institute, Fudan University, Shanghai, China
| | - Han Liu
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.,Biliary tract disease Institute, Fudan University, Shanghai, China
| | - Xiaoling Ni
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.,Biliary tract disease Institute, Fudan University, Shanghai, China
| | - Tao Suo
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.,Biliary tract disease Institute, Fudan University, Shanghai, China
| | - Dexiang Zhang
- General Surgery Department, Zhongshan-Xuhui Hospital Affiliated to Fudan University, Shanghai, China
| | - Pinxiang Lu
- General Surgery Department, Zhongshan-Xuhui Hospital Affiliated to Fudan University, Shanghai, China
| | - Yueqi Wang
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.,Biliary tract disease Institute, Fudan University, Shanghai, China
| | - Houbao Liu
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.,Biliary tract disease Institute, Fudan University, Shanghai, China
| |
Collapse
|
19
|
Chatterjee A, Jana S, Chatterjee S, Wastall LM, Mandal G, Nargis N, Roy H, Hughes TA, Bhattacharyya A. MicroRNA-222 reprogrammed cancer-associated fibroblasts enhance growth and metastasis of breast cancer. Br J Cancer 2019; 121:679-689. [PMID: 31481734 PMCID: PMC6889135 DOI: 10.1038/s41416-019-0566-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Revised: 07/25/2019] [Accepted: 08/15/2019] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Cancer-associated fibroblasts (CAFs) are known to impact on tumour behaviour, but the mechanisms controlling this are poorly understood. METHODS Breast normal fibroblasts (NFs) or CAFs were isolated from cancers by laser microdissection or were cultured. Fibroblasts were transfected to manipulate miR-222 or Lamin B receptor (LBR). The fibroblast-conditioned medium was collected and used to treat epithelial BC lines MDA-MB-231 and MDA-MB-157. Migration, invasion, proliferation or senescence was assessed using transwell, MTT or X-gal assays, respectively. RESULTS MiR-222 was upregulated in CAFs as compared with NFs. Ectopic miR-222 expression in NFs induced CAF-like expression profiles, while miR-222 knockdown in CAFs inhibited CAF phenotypes. LBR was identified as a direct miR-222 target, and was functionally relevant since LBR knockdown phenocopied miR-222 overexpression and LBR overexpression phenocopied miR-222 knockdown. MiR-222 overexpression, or LBR knockdown, was sufficient to induce NFs to show the CAF characteristics of enhanced migration, invasion and senescence, and furthermore, the conditioned medium from these fibroblasts induced increased BC cell migration and invasion. The reverse manipulations in CAFs inhibited these behaviours in fibroblasts, and inhibited paracrine influences on BC cells. CONCLUSION MiR-222/LBR have key roles in controlling pro-progression influences of CAFs in BC. This pathway may present therapeutic opportunities to inhibit CAF-induced cancer progression.
Collapse
Affiliation(s)
- Annesha Chatterjee
- Immunology Laboratory, Department of Zoology, University of Calcutta, Kolkata, West Bengal, India
| | - Samir Jana
- Immunology Laboratory, Department of Zoology, University of Calcutta, Kolkata, West Bengal, India
| | - Soumya Chatterjee
- Immunology Laboratory, Department of Zoology, University of Calcutta, Kolkata, West Bengal, India
| | - Laura M Wastall
- Department of Cellular Pathology, St James's University Hospital, Leeds, UK
| | - Gunjan Mandal
- Immunology Laboratory, Department of Zoology, University of Calcutta, Kolkata, West Bengal, India
| | - Nelofar Nargis
- Immunology Laboratory, Department of Zoology, University of Calcutta, Kolkata, West Bengal, India
| | - Himansu Roy
- Department of Surgery, Medical College, Kolkata, West Bengal, India
| | | | - Arindam Bhattacharyya
- Immunology Laboratory, Department of Zoology, University of Calcutta, Kolkata, West Bengal, India.
| |
Collapse
|
20
|
Zhang X, Xing XX, Cui JF. Invadopodia formation: An important step in matrix stiffness-regulated tumor invasion and metastasis. Shijie Huaren Xiaohua Zazhi 2019; 27:589-597. [DOI: 10.11569/wcjd.v27.i9.589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Highly motile and invasive abilities are symbolic features of metastatic tumor cells. Being a critical molecular event for maintaining the highly migratory and invasive capabilities of tumor cells, invadopodia formation undoubtedly determines the progression of tumor invasion and metastasis. Growing numbers of studies suggest that increased matrix stiffness, as a notable property of physical mechanics in solid tumors, participates in the regulation of tumor invasion and metastasis via different molecular mechanisms. However, to date the relevant mechanisms of matrix stiffness-induced invadopodia formation and activity in tumor cells remain largely unclear. This paper is to make a review on the structure and function of invadopodia, the stages and inductive factors of invadopodia formation, the regulatory mechanisms of matrix stiffness-induced invadopodia formation and so on, with an aim to reveal the important roles of invadopodia in matrix stiffness-regulated tumor invasion and metastasis.
Collapse
Affiliation(s)
- Xi Zhang
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Xiao-Xia Xing
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Jie-Feng Cui
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| |
Collapse
|
21
|
Shen J, Sun X, Pan B, Cao S, Cao J, Che D, Liu F, Zhang S, Yu Y. IL-17 induces macrophages to M2-like phenotype via NF-κB. Cancer Manag Res 2018; 10:4217-4228. [PMID: 30323677 PMCID: PMC6177522 DOI: 10.2147/cmar.s174899] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Background Tumor-associated macrophage (TAM) is emerging as one of the important complications in cancer promotion. Interleukin-17 (IL-17), a potent pro-inflammatory cytokine, plays an active role in promoting M2 macrophage differentiation (TAMs are M2-like phenotypes). In this study, we aimed to evaluate that IL-17 stimulates key phenotypic and functional signatures of M2 macrophages associated with cancer progression in non-small-cell lung cancer (NSCLC) patients. Patients and methods The markers and cytokines of M2 macrophages were detected in THP-1-derived macrophages and mouse peritoneal macrophages treated with IL-17. The activation of nuclear factor kappa B (NF-κB) and nuclear localization of p65 in IL-17-treated cells were investigated. The BAY11-7082 inhibitor and the siRNA of p65 were used to block the NF-κB activation. A total of 85 patients who underwent surgery for histologically verified NSCLC were enrolled in this study. The expression of IL-17 and M2 macrophage markers were assessed by immunostaining. Survivals were estimated using the Kaplan–Meier method. Results The CD163 and CD206 cell surface markers and transforming growth factor beta (TGF-β), vascular endothelial growth factor (VEGF) and IL-10 of M2 macrophages were significantly increased in IL-17-treated THP-1-derived macrophages in a dose-dependent manner. IL-17 increased the mRNA levels of Arginase I and Fizz1, the phosphorylation of IkBα and nuclear localization of p65 (a subunit of NF-κB). The BAY11-7082 abrogated IL-17-induced CD206 and CD163 expression, TGF-β, VEGF, IL-10, Arginase I and Fizz1 expression and p65 nuclear translocation. Further experiments showed that IL-17 induced the expression of CD206, CD163, Arginase I, Fizz1 and Ym1 in mouse peritoneal macrophages that were inhibited by siRNA of p65. The immunostaining experiments on human NSCLC tissues indicated that high IL-17 expression was significantly correlated with CD163 and c-Maf. The intratumoral IL-17+ CD163+ c-Maf+ cells were associated with NSCLC progression. Conclusion IL-17 stimulated macrophages to M2-like phenotypes via NF-κB activation. IL-17 may be a potential therapeutic target for NSCLC.
Collapse
Affiliation(s)
- Jing Shen
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, People's Republic of China,
| | - Xin Sun
- Department of Cardiology, The First Affiliated Hospital, Cardiovascular Institute, Harbin Medical University, Harbin, People's Republic of China
| | - Bo Pan
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, People's Republic of China,
| | - Shoubo Cao
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, People's Republic of China,
| | - Jingyan Cao
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, People's Republic of China,
| | - Dehai Che
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, People's Republic of China,
| | - Fang Liu
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, People's Republic of China,
| | - Shuai Zhang
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, People's Republic of China,
| | - Yan Yu
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, People's Republic of China,
| |
Collapse
|
22
|
Socovich AM, Naba A. The cancer matrisome: From comprehensive characterization to biomarker discovery. Semin Cell Dev Biol 2018; 89:157-166. [PMID: 29964200 DOI: 10.1016/j.semcdb.2018.06.005] [Citation(s) in RCA: 104] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 06/18/2018] [Accepted: 06/27/2018] [Indexed: 02/07/2023]
Abstract
Tumor progression and dissemination critically depend on support from the tumor microenvironment, the ensemble of cellular and acellular components surrounding and interacting with tumor cells. The extracellular matrix (ECM), the complex scaffolding of hundreds of proteins organizing cells in tissues, is a major component of the tumor microenvironment. It orchestrates cellular processes including proliferation, migration, and invasion, that are highly dysregulated during cancer progression. Alterations in ECM abundance, integrity, and mechanical properties have been correlated with poorer prognosis for cancer patients. Yet the ECM proteome, or "matrisome," of tumors remained until recently largely unexplored. This review will present the recent developments in computational and proteomic technologies that have allowed the comprehensive characterization of the ECM of different tumor types and microenvironmental niches. These approaches have resulted in the definition of protein signatures distinguishing tumors from normal tissues, tumors of different stages, primary from secondary tumors, and tumors from other diseased states such as fibrosis. Moreover, recent studies have demonstrated that the levels of expression of certain genes encoding ECM and ECM-associated proteins is prognostic of cancer patient survival and can thus serve as biomarkers. Last, proteomic studies have permitted the identification of novel ECM proteins playing functional roles in cancer progression. Such proteins have the potential to be exploited as therapeutic targets.
Collapse
Affiliation(s)
- Alexandra M Socovich
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, IL, USA
| | - Alexandra Naba
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, IL, USA; University of Illinois Cancer Center, Chicago, IL, USA.
| |
Collapse
|
23
|
Zhang D, Li L, Jiang H, Li Q, Wang-Gillam A, Yu J, Head R, Liu J, Ruzinova MB, Lim KH. Tumor-Stroma IL1β-IRAK4 Feedforward Circuitry Drives Tumor Fibrosis, Chemoresistance, and Poor Prognosis in Pancreatic Cancer. Cancer Res 2018; 78:1700-1712. [PMID: 29363544 PMCID: PMC5890818 DOI: 10.1158/0008-5472.can-17-1366] [Citation(s) in RCA: 120] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Revised: 12/13/2017] [Accepted: 01/19/2018] [Indexed: 12/22/2022]
Abstract
Targeting the desmoplastic stroma of pancreatic ductal adenocarcinoma (PDAC) holds promise to augment the effect of chemotherapy, but success in the clinic has thus far been limited. Preclinical mouse models suggest that near-depletion of cancer-associated fibroblasts (CAF) carries a risk of accelerating PDAC progression, underscoring the need to concurrently target key signaling mechanisms that drive the malignant attributes of both CAF and PDAC cells. We previously reported that inhibition of IL1 receptor-associated kinase 4 (IRAK4) suppresses NFκB activity and promotes response to chemotherapy in PDAC cells. In this study, we report that CAF in PDAC tumors robustly express activated IRAK4 and NFκB. IRAK4 expression in CAF promoted NFκB activity, drove tumor fibrosis, and supported PDAC cell proliferation, survival, and chemoresistance. Cytokine array analysis of CAF and microarray analysis of PDAC cells identified IL1β as a key cytokine that activated IRAK4 in CAF. Targeting IRAK4 or IL1β rendered PDAC tumors less fibrotic and more sensitive to gemcitabine. In clinical specimens of human PDAC, high stromal IL1β expression associated strongly with poor overall survival. Together, our studies establish a tumor-stroma IL1β-IRAK4 feedforward signal that can be therapeutically disrupted to increase chemotherapeutic efficacy in PDAC.Significance: Targeting the IL1β-IRAK4 signaling pathway potentiates the effect of chemotherapy in pancreatic cancer. Cancer Res; 78(7); 1700-12. ©2018 AACR.
Collapse
Affiliation(s)
- Daoxiang Zhang
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Lin Li
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Hongmei Jiang
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Qiong Li
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri
- Department of Laboratory Medicine, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Andrea Wang-Gillam
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Jinsheng Yu
- Department of Genetics, Genome Technology Access Center, Washington University School of Medicine, Saint Louis, Missouri
| | - Richard Head
- Department of Genetics, Genome Technology Access Center, Washington University School of Medicine, Saint Louis, Missouri
| | - Jingxia Liu
- Division of Public Health Sciences, Department of Surgery, Washington University School of Medicine, St. Louis, Missouri
| | - Marianna B Ruzinova
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri
| | - Kian-Huat Lim
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri.
| |
Collapse
|
24
|
Aguirre-Ghiso JA, Sosa MS. Emerging Topics on Disseminated Cancer Cell Dormancy and the Paradigm of Metastasis. ANNUAL REVIEW OF CANCER BIOLOGY-SERIES 2018. [DOI: 10.1146/annurev-cancerbio-030617-050446] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Julio A. Aguirre-Ghiso
- Division of Hematology and Medical Oncology, Department of Medicine, Department of Otolaryngology, Tisch Cancer Institute, and Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Maria Soledad Sosa
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
25
|
Lamprecht S, Sigal-Batikoff I, Shany S, Abu-Freha N, Ling E, Delinasios GJ, Moyal-Atias K, Delinasios JG, Fich A. Teaming Up for Trouble: Cancer Cells, Transforming Growth Factor-β1 Signaling and the Epigenetic Corruption of Stromal Naïve Fibroblasts. Cancers (Basel) 2018; 10:cancers10030061. [PMID: 29495500 PMCID: PMC5876636 DOI: 10.3390/cancers10030061] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2017] [Revised: 01/28/2018] [Accepted: 02/21/2018] [Indexed: 12/22/2022] Open
Abstract
It is well recognized that cancer cells subvert the phenotype of stromal naïve fibroblasts and instruct the neighboring cells to sustain their growth agenda. The mechanisms underpinning the switch of fibroblasts to cancer-associated fibroblasts (CAFs) are the focus of intense investigation. One of the most significant hallmarks of the biological identity of CAFs is that their tumor-promoting phenotype is stably maintained during in vitro and ex vivo propagation without the continual interaction with the adjacent cancer cells. In this review, we discuss robust evidence showing that the master cytokine Transforming Growth Factor-β1 (TGFβ-1) is a prime mover in reshaping, via epigenetic switches, the phenotype of stromal fibroblasts to a durable state. We also examine, in detail, the pervasive involvement of TGFβ-1 signaling from both cancer cells and CAFs in fostering cancer development, taking colorectal cancer (CRC) as a paradigm of human neoplasia. Finally, we review the stroma-centric anticancer therapeutic approach focused on CAFs—the most abundant cell population of the tumor microenvironment (TME)—as target cells.
Collapse
Affiliation(s)
- Sergio Lamprecht
- Department of Clinical Biochemistry and Pharmacology, Ben Gurion University of the Negev, Beersheva 8410500, Israel.
- Faculty of Health Sciences, Ben Gurion University of the Negev, Beersheva 8410500, Israel.
- Institute of Gastroenterology and Hepatology, Soroka University Medical Center, Beersheva 8410100, Israel.
| | - Ina Sigal-Batikoff
- Department of Clinical Biochemistry and Pharmacology, Ben Gurion University of the Negev, Beersheva 8410500, Israel.
- Faculty of Health Sciences, Ben Gurion University of the Negev, Beersheva 8410500, Israel.
- Institute of Gastroenterology and Hepatology, Soroka University Medical Center, Beersheva 8410100, Israel.
| | - Shraga Shany
- Department of Clinical Biochemistry and Pharmacology, Ben Gurion University of the Negev, Beersheva 8410500, Israel.
- Faculty of Health Sciences, Ben Gurion University of the Negev, Beersheva 8410500, Israel.
| | - Naim Abu-Freha
- Faculty of Health Sciences, Ben Gurion University of the Negev, Beersheva 8410500, Israel.
- Institute of Gastroenterology and Hepatology, Soroka University Medical Center, Beersheva 8410100, Israel.
| | - Eduard Ling
- Faculty of Health Sciences, Ben Gurion University of the Negev, Beersheva 8410500, Israel.
- Pediatrics Department B, Soroka University Medical Center, Beersheva 8410100, Israel.
| | - George J Delinasios
- International Institute of Anticancer Research, Kapandriti, Athens 19014, Greece.
| | - Keren Moyal-Atias
- Faculty of Health Sciences, Ben Gurion University of the Negev, Beersheva 8410500, Israel.
- Institute of Gastroenterology and Hepatology, Soroka University Medical Center, Beersheva 8410100, Israel.
| | - John G Delinasios
- International Institute of Anticancer Research, Kapandriti, Athens 19014, Greece.
| | - Alexander Fich
- Faculty of Health Sciences, Ben Gurion University of the Negev, Beersheva 8410500, Israel.
- Institute of Gastroenterology and Hepatology, Soroka University Medical Center, Beersheva 8410100, Israel.
| |
Collapse
|
26
|
Lichtenstein AV. Genetic Mosaicism and Cancer: Cause and Effect. Cancer Res 2018; 78:1375-1378. [DOI: 10.1158/0008-5472.can-17-2769] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Revised: 10/14/2017] [Accepted: 01/09/2018] [Indexed: 11/16/2022]
|
27
|
Dietz AC, Wayne AS. Cells to prevent/treat relapse following allogeneic stem cell transplantation. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2017; 2017:708-715. [PMID: 29222325 PMCID: PMC6142604 DOI: 10.1182/asheducation-2017.1.708] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Relapse of cancer remains one of the primary causes of treatment failure and mortality after allogeneic hematopoietic stem cell transplantation (HSCT). A multitude of approaches have been used in the management of posttransplant relapse. This review focuses on recent data with cellular therapies designed to treat or prevent posttransplant relapse of hematologic malignancies, although many of these therapeutic approaches also have applications to solid tumors and in the nontransplant setting. Currently available cell therapies include second transplant, natural killer cells, monocyte-derived dendritic cell vaccines, and lymphocytes via donor lymphocyte infusion, antigen-primed cytotoxic T lymphocytes, cytokine-induced killer cells, marrow-infiltrating lymphocytes, and chimeric antigen receptor T cells. These treatment options offer the prospect for improved relapse-free survival after HSCT.
Collapse
Affiliation(s)
- Andrew C. Dietz
- Children’s Center for Cancer and Blood Diseases, Division of Hematology, Oncology, and Blood and Marrow Transplantation, Children’s Hospital Los Angeles, Los Angeles, CA; and
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Alan S. Wayne
- Children’s Center for Cancer and Blood Diseases, Division of Hematology, Oncology, and Blood and Marrow Transplantation, Children’s Hospital Los Angeles, Los Angeles, CA; and
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA
| |
Collapse
|
28
|
Tumor Cell Invadopodia: Invasive Protrusions that Orchestrate Metastasis. Trends Cell Biol 2017; 27:595-607. [PMID: 28412099 DOI: 10.1016/j.tcb.2017.03.003] [Citation(s) in RCA: 256] [Impact Index Per Article: 36.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 03/09/2017] [Accepted: 03/13/2017] [Indexed: 12/26/2022]
Abstract
Invadopodia are a subset of invadosomes that are implicated in the integration of signals from the tumor microenvironment to support tumor cell invasion and dissemination. Recent progress has begun to define how tumor cells regulate the plasticity necessary for invadopodia to assemble and function efficiently in the different microenvironments encountered during dissemination in vivo. Exquisite mapping by many laboratories of the pathways involved in integrating diverse invadopodium initiation signals, from growth factors, to extracellular matrix (ECM) and cell-cell contact in the tumor microenvironment, has led to insight into the molecular basis of this plasticity. Here, we integrate this new information to discuss how the invadopodium is an important conductor that orchestrates tumor cell dissemination during metastasis.
Collapse
|