1
|
Anastasiadou DP, Couturier N, Goel S, Argyris DG, Vodopyanov S, Rivera-Sanchez L, Gonzalez E, Kreger J, Griffen A, Kazakov A, Burt J, Recoder N, Duran CL, Harney AS, Quesnel A, Filippou PS, Lenis VP, Shukla S, Entenberg D, Zintiridou A, Chen X, Eddy RJ, Oktay MH, Condeelis JS, Karagiannis NS, Briceno A, Guzik H, Alon R, DesMarais V, Ioannou G, Gnjatic S, Raynolds DM, Macedo R, Reshef R, Gil-Henn H, MacLean AL, Torres ER, LaFave LM, Lauvau G, Karagiannis GS. Intratumoral CXCL12 Gradients Contextualize Tumor Cell Invasion, Migration and Immune Suppression in Breast Cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.15.618571. [PMID: 39464015 PMCID: PMC11507869 DOI: 10.1101/2024.10.15.618571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Although the CXCL12/CXCR4 pathway has been prior investigated for its prometastatic and immuno- suppressive roles in the tumor microenvironment, evidence on the spatiotemporal regulation of these hallmarks has been lacking. Here, we demonstrate that CXCL12 forms a gradient specifically around cancer cell intravasation doorways, also known as Tumor Microenvironment of Metastasis (TMEM) doorways, thus facilitating the chemotactic translocation of prometastatic tumor cells expressing CXCR4 toward the perivascular TMEM doorways for subsequent entry into peripheral circulation. Fur- thermore, we demonstrate that the CXCL12-rich micro-environment around TMEM doorways may cre- ate immunosuppressive niches, whereby CD8 + T cells, despite being attracted to these regions, often exhibit reduced effector functions, limiting their efficacy. While the CXCL12/CXCR4 pathway can mini- mally influence the overall composition of immune cell populations, it biases the distribution of CD8 + T cells away from TMEM doorways, justifying its prior-established role as immunosuppressive factor for CD8 + T cells. Our research suggests that the complex interactions between CXCL12 and the various tumor and immune cell types contributes not only to the completion of the initial steps of the metastatic cascade, but also offers an immunological "sanctuary" to prometastatic tumor cells homed around TMEM doorways. Overall, our study enhances our current understanding on the mechanisms, via which CXCL12 orchestrates tumor cell behavior and immune dynamics, potentially guiding future thera- peutic strategies to combat breast cancer metastasis and improve anti-tumor immunity.
Collapse
|
2
|
Azimi M, Manavi MS, Afshinpour M, Khorram R, Vafadar R, Rezaei-Tazangi F, Arabzadeh D, Arabzadeh S, Ebrahimi N, Aref AR. Emerging immunologic approaches as cancer anti-angiogenic therapies. Clin Transl Oncol 2024:10.1007/s12094-024-03667-2. [PMID: 39294514 DOI: 10.1007/s12094-024-03667-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 08/07/2024] [Indexed: 09/20/2024]
Abstract
Targeting tumor angiogenesis, the formation of new blood vessels supporting cancer growth and spread, has been an intense focus for therapy development. However, benefits from anti-angiogenic drugs like bevacizumab have been limited by resistance stemming from activation of compensatory pathways. Recent immunotherapy advances have sparked interest in novel immunologic approaches that can induce more durable vascular pruning and overcome limitations of existing angiogenesis inhibitors. This review comprehensively examines these emerging strategies, including modulating tumor-associated macrophages, therapeutic cancer vaccines, engineered nanobodies and T cells, anti-angiogenic cytokines/chemokines, and immunomodulatory drugs like thalidomide analogs. For each approach, the molecular mechanisms, preclinical/clinical data, and potential advantages over conventional drugs are discussed. Innovative therapeutic platforms like nanoparticle delivery systems are explored. Moreover, the importance of combining agents with distinct mechanisms to prevent resistance is evaluated. As tumors hijack angiogenesis for growth, harnessing the immune system's specificity to disrupt this process represents a promising anti-cancer strategy covered by this review.
Collapse
Affiliation(s)
- Mohammadreza Azimi
- Department of Biochemistry, Medical Faculty, Saveh Branch, Islamic Azad University, Saveh, Iran
| | | | - Maral Afshinpour
- Department of Chemistry and Biochemistry, South Dakota State University (SDSU), Brookings, SD, USA
| | - Roya Khorram
- Bone and Joint Diseases Research Center, Department of Orthopedic Surgery, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Reza Vafadar
- Department of Orthopeadic Surgery, Kerman University of Medical Sciences, Kerman, Iran
| | - Fatemeh Rezaei-Tazangi
- Department of Anatomy, School of Medicine, Fasa University of Medical Sciences, Fasa, Iran
| | - Danyal Arabzadeh
- Xi'an Jaiotong University Medical Campus, Xi'an Jaiotong University, Xi'an, Shaanxi Province, China
| | - Sattar Arabzadeh
- Xi'an Jaiotong University Medical Campus, Xi'an Jaiotong University, Xi'an, Shaanxi Province, China
| | - Nasim Ebrahimi
- Genetics Division, Department of Cell and Molecular Biology and Microbiology, Faculty of Science and Technology, University of Isfahan, Isfahan, Iran.
| | - Amir Reza Aref
- Mass General Cancer Center, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
3
|
Toledo B, Zhu Chen L, Paniagua-Sancho M, Marchal JA, Perán M, Giovannetti E. Deciphering the performance of macrophages in tumour microenvironment: a call for precision immunotherapy. J Hematol Oncol 2024; 17:44. [PMID: 38863020 PMCID: PMC11167803 DOI: 10.1186/s13045-024-01559-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 05/21/2024] [Indexed: 06/13/2024] Open
Abstract
Macrophages infiltrating tumour tissues or residing in the microenvironment of solid tumours are known as tumour-associated macrophages (TAMs). These specialized immune cells play crucial roles in tumour growth, angiogenesis, immune regulation, metastasis, and chemoresistance. TAMs encompass various subpopulations, primarily classified into M1 and M2 subtypes based on their differentiation and activities. M1 macrophages, characterized by a pro-inflammatory phenotype, exert anti-tumoural effects, while M2 macrophages, with an anti-inflammatory phenotype, function as protumoural regulators. These highly versatile cells respond to stimuli from tumour cells and other constituents within the tumour microenvironment (TME), such as growth factors, cytokines, chemokines, and enzymes. These stimuli induce their polarization towards one phenotype or another, leading to complex interactions with TME components and influencing both pro-tumour and anti-tumour processes.This review comprehensively and deeply covers the literature on macrophages, their origin and function as well as the intricate interplay between macrophages and the TME, influencing the dual nature of TAMs in promoting both pro- and anti-tumour processes. Moreover, the review delves into the primary pathways implicated in macrophage polarization, examining the diverse stimuli that regulate this process. These stimuli play a crucial role in shaping the phenotype and functions of macrophages. In addition, the advantages and limitations of current macrophage based clinical interventions are reviewed, including enhancing TAM phagocytosis, inducing TAM exhaustion, inhibiting TAM recruitment, and polarizing TAMs towards an M1-like phenotype. In conclusion, while the treatment strategies targeting macrophages in precision medicine show promise, overcoming several obstacles is still necessary to achieve an accessible and efficient immunotherapy.
Collapse
Affiliation(s)
- Belén Toledo
- Department of Health Sciences, University of Jaén, Campus Lagunillas, Jaén, E-23071, Spain
- Department of Medical Oncology, Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam UMC, VU University, Amsterdam, The Netherlands
| | - Linrui Zhu Chen
- Department of Medical Oncology, Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam UMC, VU University, Amsterdam, The Netherlands
| | - María Paniagua-Sancho
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of Granada, Granada, E-18100, Spain
- Instituto de Investigación Sanitaria ibs. GRANADA, Hospitales Universitarios de Granada-Universidad de Granada, Granada, E-18071, Spain
- Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, Granada, E-18016, Spain
- Excellence Research Unit "Modeling Nature" (MNat), University of Granada, Granada, E-18016, Spain
| | - Juan Antonio Marchal
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of Granada, Granada, E-18100, Spain
- Instituto de Investigación Sanitaria ibs. GRANADA, Hospitales Universitarios de Granada-Universidad de Granada, Granada, E-18071, Spain
- Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, Granada, E-18016, Spain
- Excellence Research Unit "Modeling Nature" (MNat), University of Granada, Granada, E-18016, Spain
| | - Macarena Perán
- Department of Health Sciences, University of Jaén, Campus Lagunillas, Jaén, E-23071, Spain.
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of Granada, Granada, E-18100, Spain.
- Excellence Research Unit "Modeling Nature" (MNat), University of Granada, Granada, E-18016, Spain.
| | - Elisa Giovannetti
- Department of Medical Oncology, Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam UMC, VU University, Amsterdam, The Netherlands.
- Cancer Pharmacology Lab, Fondazione Pisana per la Scienza, San Giuliano, Pisa, 56017, Italy.
| |
Collapse
|
4
|
Chen Y, Zhu L, Wang Y, Hu J, Zhang H, Zhu J, Gong W, Liu X, Xiao F, Li X. Tumor-derived mesenchymal progenitor cell-related genes in the regulation of breast cancer proliferation. Gland Surg 2024; 13:325-339. [PMID: 38601284 PMCID: PMC11002474 DOI: 10.21037/gs-23-387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 01/20/2024] [Indexed: 04/12/2024]
Abstract
Background Breast cancer (BC) is one of the most common malignancies worldwide, and its development is affected in various ways by the tumor microenvironment (TME). Tumor-derived mesenchymal progenitor cells (MPCs), as the most important components of the TME, participate in the proliferation and metastasis of BC in several ways. In this study, we aimed to characterize the genes associated with tumor-derived MPCs and determine their effects on BC cells. Methods Tumor-derived MPCs and normal breast tissue-derived mesenchymal stem cells (MSCs) were isolated from tissues specimens of patients with BC. We conducted culture and passage, phenotype identification, proliferation and migration detection, inflammatory factor release detection, and other experiments on isolated MPCs from tumors and MSCs from normal breast tissues. Three paired tumor-derived MPCs and normal breast tissue-derived MSCs were then subjected to transcriptome analysis to determine the expression profiles of the relevant genes, and quantitative real-time polymerase chain reaction (qRT-PCR) was used to further confirm gene expression. Subsequently, the overexpression plasmids were transfected into tumor-derived MPCs, and the expression of various inflammatory factors of tumor-derived MPCs and their proliferation were characterized with a cell viability test reagent (Cell Counting Kit 8). Subsequently, the transfected tumor-derived MPCs were cocultured with BC cells using a conditioned medium coculture method to clarify the role of tumor-derived MSCs in BC. Results Tumor-derived MPCs expressed stem cell characteristics including CD105, CD90, and CD73 and exhibited adipogenic and osteogenic differentiation in vitro. The proliferation of tumor-derived MPCs was significantly lower than that of normal breast tissue-derived MSCs, and the invasive metastatic ability was comparable; however, MPCs were found to release inflammatory factors such as interleukin 6 (IL-6) and transforming growth factor β (TGF-β). Transcriptome analysis showed that stomatin (STOM), collagen and calcium binding EGF domains 1 (CCBE1), and laminin subunit alpha 5 (LAMA5) were significantly upregulated in tumor-derived MPCs. Among them, STOM was highly expressed in tumor-derived MPCs, which mediated the slow proliferation of MPCs and promoted the proliferation of BC cells. Conclusions STOM, CCBE1, and LAMA5 were highly expressed in tumor-derived MPCs, with STOM being found to retard the proliferation of MPCs but promote the proliferation of BC cells. There findings present new possibilities in targeted microenvironmental therapy for BC.
Collapse
Affiliation(s)
- Yizhu Chen
- Medical School of Chinese PLA, Beijing, China
- Department of General Surgery, The First Medical Center of Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
| | - Li Zhu
- Department of General Surgery, The First Medical Center of Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
| | - Yiming Wang
- School of Nursing, Jilin University, Changchun, China
| | - Jia Hu
- Medical School of Chinese PLA, Beijing, China
- Department of General Surgery, The First Medical Center of Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
| | - Hao Zhang
- Medical Research Institute, Hebei Yanda Hospital, Langfang, China
| | - Jingjin Zhu
- Department of General Surgery, The First Medical Center of Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
- School of Medicine, Nankai University, Tianjin, China
| | - Wenye Gong
- Medical School of Chinese PLA, Beijing, China
- Department of General Surgery, The First Medical Center of Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
| | - Xiaohan Liu
- Department of General Surgery, The First Medical Center of Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
- School of Medicine, Nankai University, Tianjin, China
| | - Fengjun Xiao
- Department of Experimental Hematology and Biochemistry, Beijing Institute of Radiation Medicine, Beijing, China
| | - Xiru Li
- Department of General Surgery, The First Medical Center of Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
| |
Collapse
|
5
|
Nair PR, Danilova L, Gómez-de-Mariscal E, Kim D, Fan R, Muñoz-Barrutia A, Fertig EJ, Wirtz D. MLL1 regulates cytokine-driven cell migration and metastasis. SCIENCE ADVANCES 2024; 10:eadk0785. [PMID: 38478601 PMCID: PMC10936879 DOI: 10.1126/sciadv.adk0785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 02/07/2024] [Indexed: 03/17/2024]
Abstract
Cell migration is a critical contributor to metastasis. Cytokine production and its role in cancer cell migration have been traditionally associated with immune cells. We find that the histone methyltransferase Mixed-Lineage Leukemia 1 (MLL1) controls 3D cell migration via cytokines, IL-6, IL-8, and TGF-β1, secreted by the cancer cells themselves. MLL1, with its scaffold protein Menin, controls actin filament assembly via the IL-6/8/pSTAT3/Arp3 axis and myosin contractility via the TGF-β1/Gli2/ROCK1/2/pMLC2 axis, which together regulate dynamic protrusion generation and 3D cell migration. MLL1 also regulates cell proliferation via mitosis-based and cell cycle-related pathways. Mice bearing orthotopic MLL1-depleted tumors exhibit decreased lung metastatic burden and longer survival. MLL1 depletion leads to lower metastatic burden even when controlling for the difference in primary tumor growth rates. Combining MLL1-Menin inhibitor with paclitaxel abrogates tumor growth and metastasis, including preexistent metastasis. These results establish MLL1 as a potent regulator of cell migration and highlight the potential of targeting MLL1 in patients with metastatic disease.
Collapse
Affiliation(s)
- Praful R. Nair
- Institute for Nanobiotechnology, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Ludmila Danilova
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Estibaliz Gómez-de-Mariscal
- Bioengineering and Aerospace Engineering Department, Universidad Carlos III de Madrid, 28911 Leganés, and Instituto de Investigación Sanitaria Gregorio Marañón, 28007 Madrid, Spain
- Optical Cell Biology Group, Instituto Gulbenkian de Ciência, R. Q.ta Grande 6 2780, 2780-156 Oeiras, Portugal
| | - Dongjoo Kim
- Department of Biomedical Engineering, Yale University, New Haven, CT 06520, USA
| | - Rong Fan
- Department of Biomedical Engineering, Yale University, New Haven, CT 06520, USA
| | - Arrate Muñoz-Barrutia
- Bioengineering and Aerospace Engineering Department, Universidad Carlos III de Madrid, 28911 Leganés, and Instituto de Investigación Sanitaria Gregorio Marañón, 28007 Madrid, Spain
| | - Elana J. Fertig
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
- Department of Applied Mathematics and Statistics, Johns Hopkins University Whiting School of Engineering, Baltimore, MD 21218, USA
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21218, USA
| | - Denis Wirtz
- Institute for Nanobiotechnology, The Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
- Department of Chemical and Biomolecular Engineering, Johns Hopkins Physical Sciences-Oncology Center, The Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| |
Collapse
|
6
|
Lu X, Wang X, Cheng H, Wang X, Liu C, Tan X. Anti-triple-negative breast cancer metastasis efficacy and molecular mechanism of the STING agonist for innate immune pathway. Ann Med 2023; 55:2210845. [PMID: 37162544 PMCID: PMC10173802 DOI: 10.1080/07853890.2023.2210845] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/11/2023] Open
Abstract
BACKGROUND With high recurrence and metastatic rates, triple-negative breast cancer (TNBC) has few therapy choices. The innate immune stimulator of interferon genes protein (STING) pathway has emerged as a critical foundation for improving anticancer immunotherapy. Although 2',3'-cGAMP has been shown to have therapeutic potential as a STING agonist in subcutaneous solid tumour treatments in mice, the effect of cGAMP in metastatic malignancies has received less attention. METHODS Bioluminescence imaging technology was applied to monitor TNBC tumour cell metastasis in living mice. Serum biochemical test and blood routine examination of mice were used to demonstrate cGAMP administration had no toxicity. The activation of DCs and CD8+ T cells was demonstrated by flow cytometry. The pharmacological mechanism of cGAMP for suppressing breast tumour metastasis was also explored. RESULTS cGAMP treatment substantially suppressed tumour development and metastasis without adverse effects. cGAMP activated the cGAS-STING-IRF3 pathway, which modified the tumour immune milieu to reverse the Epithelial-Mesenchymal Transition (EMT) and PI3K/AKT pathways and prevent tumour metastasis. It was postulated and proven that cGAMP had a pharmacological mechanism for reducing breast tumour metastasis. CONCLUSION The findings suggest that cGAMP could be useful in the immunotherapy of immune-insensitive metastatic breast cancer.
Collapse
Affiliation(s)
- Xing Lu
- Department of Chemistry & Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Xiang Wang
- Department of Chemistry & Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Hao Cheng
- Department of Chemistry & Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Xiaoqing Wang
- Department of Chemistry & Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Chang Liu
- Department of Chemistry & Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Xiangshi Tan
- Department of Chemistry & Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| |
Collapse
|
7
|
Basak U, Sarkar T, Mukherjee S, Chakraborty S, Dutta A, Dutta S, Nayak D, Kaushik S, Das T, Sa G. Tumor-associated macrophages: an effective player of the tumor microenvironment. Front Immunol 2023; 14:1295257. [PMID: 38035101 PMCID: PMC10687432 DOI: 10.3389/fimmu.2023.1295257] [Citation(s) in RCA: 24] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 10/23/2023] [Indexed: 12/02/2023] Open
Abstract
Cancer progression is primarily caused by interactions between transformed cells and the components of the tumor microenvironment (TME). TAMs (tumor-associated macrophages) make up the majority of the invading immune components, which are further categorized as anti-tumor M1 and pro-tumor M2 subtypes. While M1 is known to have anti-cancer properties, M2 is recognized to extend a protective role to the tumor. As a result, the tumor manipulates the TME in such a way that it induces macrophage infiltration and M1 to M2 switching bias to secure its survival. This M2-TAM bias in the TME promotes cancer cell proliferation, neoangiogenesis, lymphangiogenesis, epithelial-to-mesenchymal transition, matrix remodeling for metastatic support, and TME manipulation to an immunosuppressive state. TAMs additionally promote the emergence of cancer stem cells (CSCs), which are known for their ability to originate, metastasize, and relapse into tumors. CSCs also help M2-TAM by revealing immune escape and survival strategies during the initiation and relapse phases. This review describes the reasons for immunotherapy failure and, thereby, devises better strategies to impair the tumor-TAM crosstalk. This study will shed light on the understudied TAM-mediated tumor progression and address the much-needed holistic approach to anti-cancer therapy, which encompasses targeting cancer cells, CSCs, and TAMs all at the same time.
Collapse
Affiliation(s)
- Udit Basak
- Division of Molecular Medicine, Bose Institute, Kolkata, India
| | - Tania Sarkar
- Division of Molecular Medicine, Bose Institute, Kolkata, India
| | - Sumon Mukherjee
- Division of Molecular Medicine, Bose Institute, Kolkata, India
| | | | - Apratim Dutta
- Division of Molecular Medicine, Bose Institute, Kolkata, India
| | - Saikat Dutta
- Division of Molecular Medicine, Bose Institute, Kolkata, India
| | - Debadatta Nayak
- Central Council for Research in Homeopathy (CCRH), New Delhi, India
| | - Subhash Kaushik
- Central Council for Research in Homeopathy (CCRH), New Delhi, India
| | - Tanya Das
- Division of Molecular Medicine, Bose Institute, Kolkata, India
| | - Gaurisankar Sa
- Division of Molecular Medicine, Bose Institute, Kolkata, India
| |
Collapse
|
8
|
Jang BY, Shin MK, Han DH, Sung JS. Curcumin Disrupts a Positive Feedback Loop between ADMSCs and Cancer Cells in the Breast Tumor Microenvironment via the CXCL12/CXCR4 Axis. Pharmaceutics 2023; 15:2627. [PMID: 38004606 PMCID: PMC10675183 DOI: 10.3390/pharmaceutics15112627] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 11/10/2023] [Accepted: 11/13/2023] [Indexed: 11/26/2023] Open
Abstract
Adipose tissue has a significant impact on breast cancer initiation and progression owing to its substantial proportion in the breast. Adipose-derived mesenchymal stem cells (ADMSCs) are major players in the breast tumor microenvironment (TME) as they interact with cancer cells. The intricate interaction between ADMSCs and cancer cells not only drives the differentiation of ADMSCs into cancer-associated fibroblasts (CAFs) but also the metastasis of cancer cells, which is attributed to the CXCL12/CXCR4 axis. We investigated the effects of curcumin, a flavonoid known for CXCL12/CXCR4 axis inhibition, on breast TME by analyzing whether it can disrupt the ADMSC-cancer positive loop. Using MCF7 breast cancer cell-derived conditioned medium (MCF7-CM), we induced ADMSC transformation and verified that curcumin diminished the phenotypic change, inhibiting CAF marker expression. Additionally, curcumin suppressed the CXCL12/CXCR4 axis and its downstream signaling both in ADMSCs and MCF7 cells. The CM from ADMSCs, whose ADMSC-to-CAF transformation was repressed by the curcumin treatment, inhibited the positive feedback loop between ADMSCs and MCF7 as well as epithelial-mesenchymal transition in MCF7. Our study showed that curcumin is a potent anti-cancer agent that can remodel the breast TME, thereby restricting the ADMSC-cancer positive feedback loop associated with the CXCL12/CXCR4 axis.
Collapse
Affiliation(s)
| | | | | | - Jung-Suk Sung
- Department of Life Science, Dongguk University-Seoul, Goyang 10326, Republic of Korea; (B.-Y.J.); (M.K.S.); (D.-H.H.)
| |
Collapse
|
9
|
Dhandapani H, Siddiqui A, Karadkar S, Tayalia P. In Vitro 3D Spheroid Model Preserves Tumor Microenvironment of Hot and Cold Breast Cancer Subtypes. Adv Healthc Mater 2023; 12:e2300164. [PMID: 37141121 DOI: 10.1002/adhm.202300164] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 04/11/2023] [Indexed: 05/05/2023]
Abstract
Dynamic interaction of cancer, immune, and stromal cells with extracellular matrix components modulates and resists the response of standard care therapies. To mimic this, an in vitro 3D spheroid model is designed using liquid overlay method to simulate hot (MDA-MB-231) and cold (MCF-7) breast tumor microenvironment (TME). This study shows increased mesenchymal phenotype, stemness, and suppressive microenvironment in MDA-MB-231-spheroids upon exposure to doxorubicin. Intriguingly, the presence of human dermal fibroblasts enhances cancer-associated fibroblast phenotype in MDA-MB-231-spheroids through increased expression of CXCL12 and FSP-1, leading to higher infiltration of immune cells (THP-1 monocytes). However, a suppressive TME is observed in both subtypes, as seen by upregulation of M2-macrophage-specific CD68 and CD206 markers. Specifically, increased PDL-1 expressing tumor-associated macrophages along with FoxP3 expressing T regulatory cells are found in MDA-MB-231-spheroids when cultured with peripheral blood mononuclear cells. Further, it is found that the addition of 1-methyl-tryptophan, a potent indoleamine-2,3-dioxygenase-1 inhibitor, subsides the suppressive phenotype by decreasing the M2 polarization via downregulation of tryptophan metabolism and IL10 expression, particularly in MCF-7 triculture spheroids. Thus, the in vitro 3D spheroid model of TME can be utilized in therapeutics to validate immunomodulatory drugs for various breast cancer subtypes.
Collapse
Affiliation(s)
- Hemavathi Dhandapani
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai, Maharashtra, 400076, India
| | - Armaan Siddiqui
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai, Maharashtra, 400076, India
| | - Shivam Karadkar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai, Maharashtra, 400076, India
| | - Prakriti Tayalia
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai, Maharashtra, 400076, India
| |
Collapse
|
10
|
The Chemokine Receptor CCR1 Mediates Microglia Stimulated Glioma Invasion. Int J Mol Sci 2023; 24:ijms24065136. [PMID: 36982211 PMCID: PMC10049042 DOI: 10.3390/ijms24065136] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 02/22/2023] [Accepted: 03/02/2023] [Indexed: 03/10/2023] Open
Abstract
Glioblastoma multiforme (GBM) is the most aggressive form of adult brain tumor which is highly resistant to conventional treatment and therapy. Glioma cells are highly motile resulting in infiltrative tumors with poorly defined borders. Another hallmark of GBM is a high degree of tumor macrophage/microglia infiltration. The level of these tumor-associated macrophages/microglia (TAMs) correlates with higher malignancy and poorer prognosis. We previously demonstrated that inhibition of TAM infiltration into glioma tumors with the CSF-1R antagonist pexidartinib (PLX3397) can inhibit glioma cell invasion in-vitro and in-vivo. In this study, we demonstrate an important role for the chemokine receptor CCR1 in mediating microglia/TAM stimulated glioma invasion. Using two structurally distinct CCR1 antagonists, including a novel inhibitor “MG-1-5”, we were able to block microglial activated GL261 glioma cell invasion in a dose dependent manner. Interestingly, treatment of a murine microglia cell line with glioma conditioned media resulted in a strong induction of CCR1 gene and protein expression. This induction was attenuated by inhibition of CSF-1R. In addition, glioma conditioned media treatment of microglia resulted in a rapid upregulation of gene expression of several CCR1 ligands including CCL3, CCL5, CCL6 and CCL9. These data support the existence of tumor stimulated autocrine loop within TAMs which ultimately mediates tumor cell invasion.
Collapse
|
11
|
Organotropism of breast cancer metastasis: A comprehensive approach to the shared gene network. GENE REPORTS 2023. [DOI: 10.1016/j.genrep.2023.101749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
|
12
|
Anang V, Singh A, Kottarath SK, Verma C. Receptors of immune cells mediates recognition for tumors. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2023; 194:219-267. [PMID: 36631194 DOI: 10.1016/bs.pmbts.2022.09.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Over the last few decades, the immune system has been steered toward eradication of cancer cells with the help of cancer immunotherapy. T cells, B cells, monocytes/macrophages, dendritic cells, T-reg cells, and natural killer (NK) cells are some of the numerous immune cell types that play a significant part in cancer cell detection and reduction of inflammation, and the antitumor response. Briefly stated, chimeric antigen receptors, adoptive transfer and immune checkpoint modulators are currently the subjects of research focus for successful immunotherapy-based treatments for a variety of cancers. This chapter discusses ongoing investigations on the mechanisms and recent developments by which receptors of immune cells especially that of lymphocytes and monocytes/macrophages regulate the detection of immune system leading to malignancies. We will also be looking into the treatment strategies based on these mechanisms.
Collapse
Affiliation(s)
- Vandana Anang
- International Center for Genetic Engineering and Biotechnology (ICGEB), New Delhi, India
| | | | - Sarat Kumar Kottarath
- Department of Experimental Therapeutics, MD Anderson Cancer Center, Huston, TX, United States.
| | - Chaitenya Verma
- Department of Pathology, Wexner Medical Center, Ohio State University, Columbus, OH, United States.
| |
Collapse
|
13
|
García-Chico C, López-Ortiz S, Peñín-Grandes S, Pinto-Fraga J, Valenzuela PL, Emanuele E, Ceci C, Graziani G, Fiuza-Luces C, Lista S, Lucia A, Santos-Lozano A. Physical Exercise and the Hallmarks of Breast Cancer: A Narrative Review. Cancers (Basel) 2023; 15:324. [PMID: 36612320 PMCID: PMC9818971 DOI: 10.3390/cancers15010324] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 12/26/2022] [Accepted: 12/27/2022] [Indexed: 01/06/2023] Open
Abstract
Growing evidence suggests that, among the different molecular/cellular pathophysiological mechanisms associated with cancer, there are 14 hallmarks that play a major role, including: (i) sustaining proliferative signaling, (ii) evading growth suppressors, (iii) activating invasion and metastasis, (iv) enabling replicative immortality, (v) inducing angiogenesis, (vi) resisting cell death, (vii) reprogramming energy metabolism, (viii) evading immune destruction, (ix) genome instability and mutations, (x) tumor-promoting inflammation, (xi) unlocking phenotypic plasticity, (xii) nonmutational epigenetic reprogramming, (xiii) polymorphic microbiomes, and (xiv) senescent cells. These hallmarks are also associated with the development of breast cancer, which represents the most prevalent tumor type in the world. The present narrative review aims to describe, for the first time, the effects of physical activity/exercise on these hallmarks. In summary, an active lifestyle, and particularly regular physical exercise, provides beneficial effects on all major hallmarks associated with breast cancer, and might therefore help to counteract the progression of the disease or its associated burden.
Collapse
Affiliation(s)
- Celia García-Chico
- i+HeALTH, Miguel de Cervantes European University, 27038 Valladolid, Spain
| | - Susana López-Ortiz
- i+HeALTH, Miguel de Cervantes European University, 27038 Valladolid, Spain
| | - Saúl Peñín-Grandes
- i+HeALTH, Miguel de Cervantes European University, 27038 Valladolid, Spain
| | - José Pinto-Fraga
- i+HeALTH, Miguel de Cervantes European University, 27038 Valladolid, Spain
| | - Pedro L. Valenzuela
- Research Institute of the Hospital 12 de Octubre (‘Imas12’ [PaHerg Group]), 28041 Madrid, Spain
- Department of Systems Biology, University of Alcalá, 28871 Madrid, Spain
| | | | - Claudia Ceci
- Departmental Faculty of Medicine, Saint Camillus International University of Health and Medical Sciences, 00133 Rome, Italy
- Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Grazia Graziani
- Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Carmen Fiuza-Luces
- Research Institute of the Hospital 12 de Octubre (‘Imas12’ [PaHerg Group]), 28041 Madrid, Spain
| | - Simone Lista
- i+HeALTH, Miguel de Cervantes European University, 27038 Valladolid, Spain
| | - Alejandro Lucia
- Research Institute of the Hospital 12 de Octubre (‘Imas12’ [PaHerg Group]), 28041 Madrid, Spain
- Faculty of Sport Sciences, Universidad Europea de Madrid, 28670 Madrid, Spain
| | - Alejandro Santos-Lozano
- i+HeALTH, Miguel de Cervantes European University, 27038 Valladolid, Spain
- Research Institute of the Hospital 12 de Octubre (‘Imas12’ [PaHerg Group]), 28041 Madrid, Spain
| |
Collapse
|
14
|
Assessment of MRI to estimate metastatic dissemination risk and prometastatic effects of chemotherapy. NPJ Breast Cancer 2022; 8:101. [PMID: 36056005 PMCID: PMC9440218 DOI: 10.1038/s41523-022-00463-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 07/11/2022] [Indexed: 11/10/2022] Open
Abstract
Metastatic dissemination in breast cancer is regulated by specialized intravasation sites called “tumor microenvironment of metastasis” (TMEM) doorways, composed of a tumor cell expressing the actin-regulatory protein Mena, a perivascular macrophage, and an endothelial cell, all in stable physical contact. High TMEM doorway number is associated with an increased risk of distant metastasis in human breast cancer and mouse models of breast carcinoma. Here, we developed a novel magnetic resonance imaging (MRI) methodology, called TMEM Activity-MRI, to detect TMEM-associated vascular openings that serve as the portal of entry for cancer cell intravasation and metastatic dissemination. We demonstrate that TMEM Activity-MRI correlates with primary tumor TMEM doorway counts in both breast cancer patients and mouse models, including MMTV-PyMT and patient-derived xenograft models. In addition, TMEM Activity-MRI is reduced in mouse models upon treatment with rebastinib, a specific and potent TMEM doorway inhibitor. TMEM Activity-MRI is an assay that specifically measures TMEM-associated vascular opening (TAVO) events in the tumor microenvironment, and as such, can be utilized in mechanistic studies investigating molecular pathways of cancer cell dissemination and metastasis. Finally, we demonstrate that TMEM Activity-MRI increases upon treatment with paclitaxel in mouse models, consistent with prior observations that chemotherapy enhances TMEM doorway assembly and activity in human breast cancer. Our findings suggest that TMEM Activity-MRI is a promising precision medicine tool for localized breast cancer that could be used as a non-invasive test to determine metastatic risk and serve as an intermediate pharmacodynamic biomarker to monitor therapeutic response to agents that block TMEM doorway-mediated dissemination.
Collapse
|
15
|
Zhou X, Wang X, Sun Q, Zhang W, Liu C, Ma W, Sun C. Natural compounds: A new perspective on targeting polarization and infiltration of tumor-associated macrophages in lung cancer. Biomed Pharmacother 2022; 151:113096. [PMID: 35567987 DOI: 10.1016/j.biopha.2022.113096] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 04/29/2022] [Accepted: 05/04/2022] [Indexed: 11/02/2022] Open
Abstract
With the development in tumor immunology, people are gradually understanding the complexity and diversity of the tumor microenvironment immune status and its important effect on tumors. Tumor-associated macrophages (TAMs), an important part of the tumor immune microenvironment, have a double effect on tumor growth and metastasis. Many studies have focused on lung cancer, especially non-small cell lung cancer and other "hot tumors" with typical inflammatory characteristics. The polarization and infiltration of TAMs is an important mechanism in the occurrence and development of malignant tumors, such as lung cancer, and in the tumor immune microenvironment. Therapeutic drugs designed for these reasons are key to targeting TAMs in the treatment of lung cancer. A large number of reports have suggested that natural compounds have a strong potential of affecting immunity by targeting the polarization and infiltration of TAMs to improve the immune microenvironment of lung cancer and exert a natural antitumor effect. This paper discusses the infiltration and polarization effects of natural compounds on lung cancer TAMs, provides a detailed classification and systematic review of natural compounds, and summarizes the bias of different kinds of natural compounds by affecting their antitumor mechanism of TAMs, with the aim of providing new perspectives and potential therapeutic drugs for targeted macrophages in the treatment of lung cancer.
Collapse
Affiliation(s)
- Xintong Zhou
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xiaomin Wang
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Qi Sun
- College of Acupuncture and Massage, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Wenfeng Zhang
- School of Traditional Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Cun Liu
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Wenzhe Ma
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Changgang Sun
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, China; College of Chinese Medicine, Weifang Medical University, Weifang, China; Qingdao Academy of Chinese Medical Sciences, Shandong University of Traditional Chinese Medicine, Qingdao, China.
| |
Collapse
|
16
|
Hozhabri H, Moghaddam MM, Moghaddam MM, Mohammadian A. A comprehensive bioinformatics analysis to identify potential prognostic biomarkers among CC and CXC chemokines in breast cancer. Sci Rep 2022; 12:10374. [PMID: 35725915 PMCID: PMC9209453 DOI: 10.1038/s41598-022-14610-2] [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: 12/02/2021] [Accepted: 05/06/2022] [Indexed: 11/09/2022] Open
Abstract
Breast cancer (BC) is a major human health problem due to its increasing incidence and mortality rate. CC and CXC chemokines are associated with tumorigenesis and the progression of many cancers. Since the prognostic values of CC and CXC families' expression in various types of cancers are becoming increasingly evident, we aimed to conduct a comprehensive bioinformatics analysis elucidating the prognostic values of the CC and CXC families in BC. Therefore, TCGA, UALCAN, Kaplan–Meier plotter, bc-GenExMiner, cBioPortal, STRING, Enrichr, and TIMER were utilized for analysis. We found that high levels of CCL4/5/14/19/21/22 were associated with better OS and RFS, while elevated expression of CCL24 was correlated with shorter OS in BC patients. Also, high levels of CXCL9/13 indicated longer OS, and enhanced expression of CXCL12/14 was linked with better OS and RFS in BC patients. Meanwhile, increased transcription levels of CXCL8 were associated with worse OS and RFS in BC patients. In addition, our results showed that CCL5, CCL8, CCL14, CCL20, CCL27, CXCL4, and CXCL14 were notably correlated with the clinical outcomes of BC patients. Our findings provide a new point of view that may help the clinical application of CC and CXC chemokines as prognostic biomarkers in BC.
Collapse
Affiliation(s)
- Hossein Hozhabri
- Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran.
| | | | - Madiheh Mazaheri Moghaddam
- Department of Genetics and Molecular Medicine, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Ali Mohammadian
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
17
|
Masih M, Agarwal S, Kaur R, Gautam PK. Role of chemokines in breast cancer. Cytokine 2022; 155:155909. [PMID: 35597171 DOI: 10.1016/j.cyto.2022.155909] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 05/02/2022] [Accepted: 05/09/2022] [Indexed: 02/06/2023]
Abstract
Chemokines belong to a family of chemoattractant cytokines and are well known to have an essential role in various cancer aetiologies. Multiplesubsets of immune cells are recruited and enrolled into the tumor microenvironment through interactions between chemokines and their specific receptors. These populations and their interactions have a distinct impact on tumor growth, progression, and treatment outcomes. While it is clear that many chemokines and their cognate receptors can be detected in breast and other cancers, the role of each chemokine and receptor has yet to be determined. This review focuses on the main chemokines that play a crucial role in the tumor microenvironment, emphasizing breast cancer. We have also discussed the techniques used to identify the chemokines and their future implication in the early diagnosis of cancer. In-depth knowledge of chemokines and their role in breast cancer progression can provide specific targets for breast cancer biotherapy.
Collapse
Affiliation(s)
- Marilyn Masih
- Department of Biochemistry, AIIMS, New Delhi -110029, India.
| | - Sonam Agarwal
- Department of Biochemistry, AIIMS, New Delhi -110029, India.
| | - Rupinder Kaur
- Department of Biochemistry, AIIMS, New Delhi -110029, India.
| | | |
Collapse
|
18
|
Szulc-Kielbik I, Kielbik M. Tumor-Associated Macrophages: Reasons to Be Cheerful, Reasons to Be Fearful. EXPERIENTIA SUPPLEMENTUM (2012) 2022; 113:107-140. [PMID: 35165862 DOI: 10.1007/978-3-030-91311-3_4] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Tumor microenvironment (TME) is a complex and constantly evolving entity that consists not only of cancer cells, but also of resident host cells and immune-infiltrating cells, among which macrophages are significant components, due to their diversity of functions through which they can influence the immune response against tumor cells. Macrophages present in tumor environment are termed as tumor-associated macrophages (TAMs). They are strongly plastic cells, and depending on the TME stimuli (i.e., cytokines, chemokines), TAMs polarize to antitumoral (M1-like TAMs) or protumoral (M2-like TAMs) phenotype. Both types of TAMs differ in the surface receptors' expression, activation of intracellular signaling pathways, and ability of production and various metabolites release. At the early stage of tumor formation, TAMs are M1-like phenotype, and they are able to eliminate tumor cells, i.e., by reactive oxygen species formation or by presentation of cancer antigens to other effector immune cells. However, during tumor progression, TAMs M2-like phenotype is dominating. They mainly contribute to angiogenesis, stromal remodeling, enhancement of tumor cells migration and invasion, and immunosuppression. This wide variety of TAMs' functions makes them an excellent subject for use in developing antitumor therapies which mainly is based on three strategies: TAMs' elimination, reprograming, or recruitment inhibition.
Collapse
Affiliation(s)
| | - Michal Kielbik
- Institute of Medical Biology, Polish Academy of Sciences, Lodz, Poland.
| |
Collapse
|
19
|
CSF1R Inhibition Combined with GM-CSF Reprograms Macrophages and Disrupts Protumoral Interplays with AML Cells. Cancers (Basel) 2021; 13:cancers13215289. [PMID: 34771453 PMCID: PMC8582394 DOI: 10.3390/cancers13215289] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 10/04/2021] [Accepted: 10/15/2021] [Indexed: 12/20/2022] Open
Abstract
Relapse is a major issue in acute myeloid leukemia (AML) and while the contribution of gene mutations in developing drug resistance is well established, little is known on the role of macrophages (MΦs) in an AML cell microenvironment. We examined whether myeloblasts could educate MΦs to adopt a protumoral orientation supporting myeloblast survival and resistance to therapy. Flow cytometry analyses demonstrated that M2-like CD163+ MΦs are abundantly present, at diagnosis, in the bone marrow of AML patients. We showed that myeloblasts, or their conditioned medium, polarize monocytes to M2-like CD163+ MΦs, induce the secretion of many protumoral factors, and promote myeloblast survival and proliferation as long as close intercellular contacts are maintained. Importantly, pharmacologic inhibition of the CSF1 receptor (CSF1R), in the presence of GM-CSF, reprogrammed MΦ polarization to an M1-like orientation, induced the secretion of soluble factors with antitumoral activities, reduced protumoral agonists, and promoted the apoptosis of myeloblasts interacting with MΦs. Furthermore, myeloblasts, which became resistant to venetoclax or midostaurin during their interplay with protumoral CD163+ MΦs, regained sensitivity to these targeted therapies following CSF1R inhibition in the presence of GM-CSF. These data reveal a crucial role of CD163+ MΦ interactions with myeloblasts that promote myeloblast survival and identify CSF1R inhibition as a novel target for AML therapy.
Collapse
|
20
|
Waza AA, Tarfeen N, Majid S, Hassan Y, Mir R, Rather MY, Shah NUD. Metastatic Breast Cancer, Organotropism and Therapeutics: A Review. Curr Cancer Drug Targets 2021; 21:813-828. [PMID: 34365922 DOI: 10.2174/1568009621666210806094410] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Revised: 05/21/2021] [Accepted: 05/24/2021] [Indexed: 11/22/2022]
Abstract
The final stage of breast cancer involves spreading breast cancer cells to the vital organs like the brain, liver lungs and bones in the process called metastasis. Once the target organ is overtaken by the metastatic breast cancer cells, its usual function is compromised causing organ dysfunction and death. Despite the significant research on breast cancer metastasis, it's still the main culprit of breast cancer-related deaths. Exploring the complex molecular pathways associated with the initiation and progression of breast cancer metastasis could lead to the discovery of more effective ways of treating the devastating phenomenon. The present review article highlights the recent advances to understand the complexity associated with breast cancer metastases, organotropism and therapeutic advances.
Collapse
Affiliation(s)
- Ajaz Ahmad Waza
- Multidisciplinary Research Unit (MRU), Government Medical College (GMC) Srinagar, J & K, 190010. India
| | - Najeebul Tarfeen
- Centre of Research for Development, University of Kashmir, Srinagar 190006 . India
| | - Sabhiya Majid
- Department of Biochemistry, Government Medical College (GMC) Srinagar, J & K, 190010. India
| | - Yasmeena Hassan
- Division of Nursing, Sher-i-Kashmir Institute of Medical Sciences (SKIMS), Soura, Srinagar, J & K. India
| | - Rashid Mir
- Department of Medical Lab Technology, Faculty of Applied Medical Sciences, University of Tabuk, Kingdom of Saudi Arabia, Tabuk. Saudi Arabia
| | - Mohd Younis Rather
- Multidisciplinary Research Unit (MRU), Government Medical College (GMC) Srinagar, J & K, 190010. India
| | - Naseer Ue Din Shah
- Centre of Research for Development, University of Kashmir, Srinagar 190006 . India
| |
Collapse
|
21
|
Macrophage Polarization States in the Tumor Microenvironment. Int J Mol Sci 2021; 22:ijms22136995. [PMID: 34209703 PMCID: PMC8268869 DOI: 10.3390/ijms22136995] [Citation(s) in RCA: 690] [Impact Index Per Article: 230.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 06/09/2021] [Accepted: 06/25/2021] [Indexed: 12/13/2022] Open
Abstract
The M1/M2 macrophage paradigm plays a key role in tumor progression. M1 macrophages are historically regarded as anti-tumor, while M2-polarized macrophages, commonly deemed tumor-associated macrophages (TAMs), are contributors to many pro-tumorigenic outcomes in cancer through angiogenic and lymphangiogenic regulation, immune suppression, hypoxia induction, tumor cell proliferation, and metastasis. The tumor microenvironment (TME) can influence macrophage recruitment and polarization, giving way to these pro-tumorigenic outcomes. Investigating TME-induced macrophage polarization is critical for further understanding of TAM-related pro-tumor outcomes and potential development of new therapeutic approaches. This review explores the current understanding of TME-induced macrophage polarization and the role of M2-polarized macrophages in promoting tumor progression.
Collapse
|
22
|
Asiry S, Kim G, Filippou PS, Sanchez LR, Entenberg D, Marks DK, Oktay MH, Karagiannis GS. The Cancer Cell Dissemination Machinery as an Immunosuppressive Niche: A New Obstacle Towards the Era of Cancer Immunotherapy. Front Immunol 2021; 12:654877. [PMID: 33927723 PMCID: PMC8076861 DOI: 10.3389/fimmu.2021.654877] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Accepted: 03/23/2021] [Indexed: 12/13/2022] Open
Abstract
Although cancer immunotherapy has resulted in unpreceded survival benefits to subsets of oncology patients, accumulating evidence from preclinical animal models suggests that the immunosuppressive tumor microenvironment remains a detrimental factor limiting benefit for many patient subgroups. Recent efforts on lymphocyte-mediated immunotherapies are primarily focused on eliminating cancer foci at primary and metastatic sites, but few studies have investigated the impact of these therapies on the highly complex process of cancer cell dissemination. The metastatic cascade involves the directional streaming of invasive/migratory tumor cells toward specialized blood vessel intravasation gateways, called TMEM doorways, to the peripheral circulation. Importantly, this process occurs under the auspices of a specialized tumor microenvironment, herewith referred to as "Dissemination Trajectory", which is supported by an ample array of tumor-associated macrophages (TAMs), skewed towards an M2-like polarization spectrum, and which is also vital for providing microenvironmental cues for cancer cell invasion, migration and stemness. Based on pre-existing evidence from preclinical animal models, this article outlines the hypothesis that dissemination trajectories do not only support the metastatic cascade, but also embody immunosuppressive niches, capable of providing transient and localized immunosubversion cues to the migratory/invasive cancer cell subpopulation while in the act of departing from a primary tumor. So long as these dissemination trajectories function as "immune deserts", the migratory tumor cell subpopulation remains efficient in evading immunological destruction and seeding metastatic sites, despite administration of cancer immunotherapy and/or other cytotoxic treatments. A deeper understanding of the molecular and cellular composition, as well as the signaling circuitries governing the function of these dissemination trajectories will further our overall understanding on TAM-mediated immunosuppression and will be paramount for the development of new therapeutic strategies for the advancement of optimal cancer chemotherapies, immunotherapies, and targeted therapies.
Collapse
Affiliation(s)
- Saeed Asiry
- Department of Pathology, Montefiore Medical Center, Albert Einstein College of Medicine, New York City, NY, United States
| | - Gina Kim
- Department of Surgery, Montefiore Medical Center, Albert Einstein College of Medicine, New York City, NY, United States
| | - Panagiota S. Filippou
- School of Health and Life Sciences, Teesside University, Middlesbrough, United Kingdom
- National Horizons Centre, Teesside University, Darlington, United Kingdom
| | - Luis Rivera Sanchez
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, New York City, NY, United States
| | - David Entenberg
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, New York City, NY, United States
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, New York City, NY, United States
- Integrated Imaging Program, Albert Einstein College of Medicine, New York City, NY, United States
| | - Douglas K. Marks
- Department of Medicine, NYU Long Island School of Medicine, Mineola, NY, United States
| | - Maja H. Oktay
- Department of Pathology, Montefiore Medical Center, Albert Einstein College of Medicine, New York City, NY, United States
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, New York City, NY, United States
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, New York City, NY, United States
- Integrated Imaging Program, Albert Einstein College of Medicine, New York City, NY, United States
| | - George S. Karagiannis
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, New York City, NY, United States
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, New York City, NY, United States
- Integrated Imaging Program, Albert Einstein College of Medicine, New York City, NY, United States
| |
Collapse
|
23
|
Su F, Daquinag AC, Ahn S, Saha A, Dai Y, Zhao Z, DiGiovanni J, Kolonin MG. Progression of prostate carcinoma is promoted by adipose stromal cell-secreted CXCL12 signaling in prostate epithelium. NPJ Precis Oncol 2021; 5:26. [PMID: 33753872 PMCID: PMC7985375 DOI: 10.1038/s41698-021-00160-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 02/23/2021] [Indexed: 02/07/2023] Open
Abstract
Aggressiveness of carcinomas is linked with tumor recruitment of adipose stromal cells (ASC), which is increased in obesity. ASC promote cancer through molecular pathways not fully understood. Here, we demonstrate that epithelial-mesenchymal transition (EMT) in prostate tumors is promoted by obesity and suppressed upon pharmacological ASC depletion in HiMyc mice, a spontaneous genetic model of prostate cancer. CXCL12 expression in tumors was associated with ASC recruitment and localized to stromal cells expressing platelet-derived growth factor receptors Pdgfra and Pdgfrb. The role of this chemokine secreted by stromal cells in cancer progression was further investigated by using tissue-specific knockout models. ASC deletion of CXCL12 gene in the Pdgfr + lineages suppressed tumor growth and EMT, indicating stroma as the key source of CXCL12. Clinical sample analysis revealed that CXCL12 expression by peritumoral adipose stroma is increased in obesity, and that the correlating increase in Pdgfr/CXCL12 expression in the tumor is linked with decreased survival of patients with prostate carcinoma. Our study establishes ASC as the source of CXCL12 driving tumor aggressiveness and outlines an approach to treatment of carcinoma progression.
Collapse
Affiliation(s)
- Fei Su
- The Brown Foundation Institute of Molecular Medicine for the Prevention of Disease, The University of Texas Health Sciences Center at Houston, Houston, TX, USA
| | - Alexes C Daquinag
- The Brown Foundation Institute of Molecular Medicine for the Prevention of Disease, The University of Texas Health Sciences Center at Houston, Houston, TX, USA
| | - Songyeon Ahn
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX, USA
| | - Achinto Saha
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX, USA
- Livestrong Cancer Institutes, Dell Medical School, The University of Texas at Austin, Austin, TX, USA
| | - Yulin Dai
- Center for Precision Health, School of Biomedical Informatics, University of Texas Health Science Center, Houston, TX, USA
| | - Zhongming Zhao
- Center for Precision Health, School of Biomedical Informatics, University of Texas Health Science Center, Houston, TX, USA
| | - John DiGiovanni
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX, USA
- Livestrong Cancer Institutes, Dell Medical School, The University of Texas at Austin, Austin, TX, USA
| | - Mikhail G Kolonin
- The Brown Foundation Institute of Molecular Medicine for the Prevention of Disease, The University of Texas Health Sciences Center at Houston, Houston, TX, USA.
| |
Collapse
|
24
|
Yarso KY, Bellynda M, Azmiardi A, Wasita B, Heriyanto DS, Astuti I, Hakimi M, Aryandono T. Chemotherapy Negates the Effect of SDF1 mRNA to Distant Metastasis and Poor Overall Survival in Breast Cancer Patients. Asian Pac J Cancer Prev 2021; 22:757-766. [PMID: 33773539 PMCID: PMC8286657 DOI: 10.31557/apjcp.2021.22.3.757] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Indexed: 01/06/2023] Open
Abstract
OBJECTIVE Investigate the effect of SDF1a, nuclear, and cytoplasmic CXCR4 breast cancer tissue on metastasis and overall survival in patients with complete-chemotherapy and no-chemotherapy. METHODS Cohort ambidirectional design was employed with survival analysis that followed the patient's diagnosis until obtaining the outcome, distant metastasis, or death. We analyzed samples in three groups (all-patient, no-chemotherapy, and complete-chemotherapy groups). Breast cancer cell nuclear and cytoplasm expressions of CXCR4 protein were examined using immunohistochemistry. Amplification of mRNA SDF1a of breast cancer tissue was examined using rtPCR on 131 samples from the same initial paraffin block. RESULTS In the distant metastasis and Overall Survival (OS) analysis, there was no correlation between cytoplasmic and nuclear CXCR4 in all-patient, no-chemotherapy, and complete-chemotherapy groups. SDF1a was significantly correlated to shorter distant metastasis and poor OS in the all-patient (p=0.004 and p=0.04, respectively) and no-chemotherapy group (p=0.008 and p=0.026, respectively). However, in the complete-chemotherapy group, SDF1a was not correlated to either metastasis (p=0.527) or OS (p=0.993), advanced stage demonstrated a strong association on shorter distant metastatic in no-chemotherapy (p=0.021) and complete-chemotherapy group (p=0.004) and also poor OS in both groups (p=0.006 and p=0.002, respectively). The hormone receptor showed a protective effect on the no-chemotherapy group's OS (p= 0.019). Meanwhile, not undergoing chemotherapy was associated with poor OS in the all-patient group (p= 0.011). CONCLUSION SDF1a mRNA amplification has a significant correlation with the occurrence of metastasis and OS in all-patient and no-chemotherapy group. Undergoing chemotherapy negates the effect of SDF1a for distant metastasis and OS.
Collapse
Affiliation(s)
- Kristanto Yuli Yarso
- Department of Surgery, Oncology Division, Medical Faculty, Sebelas Maret University, Indonesia
| | - Monica Bellynda
- Department of Surgery, Medical Faculty, Sebelas Maret University, Indonesia
| | - Akhmad Azmiardi
- Department of Public Health, Faculty of Public Health, Veteran Bangun Nusantara Sukoharjo University, Indonesia
| | - Brian Wasita
- Department of Anatomical Pathology, Medical Faculty, Sebelas Maret University, Indonesia
| | - Didik Setyo Heriyanto
- Department of Anatomical Pathology, Medical Faculty, Gadjah Mada University, Indonesia
| | - Indwiani Astuti
- Department of Pharmacology and Therapy, Medical Faculty, Gadjah Mada University, Indonesia
| | - Mohammad Hakimi
- Department of Obstetrics and Gynecology, Medical Faculty, Gadjah Mada University, Indonesia
| | - Teguh Aryandono
- Department of Surgery, Oncology Division, Medical Faculty, Gadjah Mada University, Indonesia
| |
Collapse
|
25
|
The Signaling Duo CXCL12 and CXCR4: Chemokine Fuel for Breast Cancer Tumorigenesis. Cancers (Basel) 2020; 12:cancers12103071. [PMID: 33096815 PMCID: PMC7590182 DOI: 10.3390/cancers12103071] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 10/05/2020] [Accepted: 10/18/2020] [Indexed: 12/16/2022] Open
Abstract
Simple Summary Breast cancer remains the most common malignancy in women. In this review, we explore the role of the CXCL12/CXCR4 pathway in breast cancer. We show that the CXCL12/CXCR4 cascade is involved in nearly every aspect of breast cancer tumorigenesis including proliferation, cell motility and distant metastasis. Moreover, we summarize current knowledge about the CXCL12/CXCR4-targeted therapies. Due to the critical roles of this pathway in breast cancer and other malignancies, we believe that audiences in different fields will find this overview helpful. Abstract The CXCL12/CXCR4 signaling pathway has emerged in the recent years as a key player in breast cancer tumorigenesis. This pathway controls many aspects of breast cancer development including cancer cell proliferation, motility and metastasis to all target organs. Moreover, the CXCL12/CXCR4 cascade affects both immune and stromal cells, creating tumor-supporting microenvironment. In this review, we examine state-of-the-art knowledge about detrimental roles of the CXCL12/CXCR4 signaling, discuss its therapeutic potential and suggest further research directions beneficial both for basic research and personalized medicine in breast cancer.
Collapse
|
26
|
Abu-Thuraia A, Goyette MA, Boulais J, Delliaux C, Apcher C, Schott C, Chidiac R, Bagci H, Thibault MP, Davidson D, Ferron M, Veillette A, Daly RJ, Gingras AC, Gratton JP, Côté JF. AXL confers cell migration and invasion by hijacking a PEAK1-regulated focal adhesion protein network. Nat Commun 2020; 11:3586. [PMID: 32681075 PMCID: PMC7368075 DOI: 10.1038/s41467-020-17415-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 06/28/2020] [Indexed: 01/08/2023] Open
Abstract
Aberrant expression of receptor tyrosine kinase AXL is linked to metastasis. AXL can be activated by its ligand GAS6 or by other kinases, but the signaling pathways conferring its metastatic activity are unknown. Here, we define the AXL-regulated phosphoproteome in breast cancer cells. We reveal that AXL stimulates the phosphorylation of a network of focal adhesion (FA) proteins, culminating in faster FA disassembly. Mechanistically, AXL phosphorylates NEDD9, leading to its binding to CRKII which in turn associates with and orchestrates the phosphorylation of the pseudo-kinase PEAK1. We find that PEAK1 is in complex with the tyrosine kinase CSK to mediate the phosphorylation of PAXILLIN. Uncoupling of PEAK1 from AXL signaling decreases metastasis in vivo, but not tumor growth. Our results uncover a contribution of AXL signaling to FA dynamics, reveal a long sought-after mechanism underlying AXL metastatic activity, and identify PEAK1 as a therapeutic target in AXL positive tumors. AXL receptor tyrosine kinase has a role in metastasis but the mechanism is unclear. In this study, the authors show that AXL activation can control focal adhesion dynamics via PEAK1 and that AXL-mediated PEAK1 phosphorylation is required for metastasis of triple negative breast cancer cells in vivo.
Collapse
Affiliation(s)
- Afnan Abu-Thuraia
- Montreal Clinical Research Institute (IRCM), Montréal, QC, H2W 1R7, Canada.,Molecular Biology Programs, Université de Montréal, Montréal, QC, H3T 1J4, Canada
| | - Marie-Anne Goyette
- Montreal Clinical Research Institute (IRCM), Montréal, QC, H2W 1R7, Canada.,Molecular Biology Programs, Université de Montréal, Montréal, QC, H3T 1J4, Canada
| | - Jonathan Boulais
- Montreal Clinical Research Institute (IRCM), Montréal, QC, H2W 1R7, Canada
| | - Carine Delliaux
- Montreal Clinical Research Institute (IRCM), Montréal, QC, H2W 1R7, Canada
| | - Chloé Apcher
- Montreal Clinical Research Institute (IRCM), Montréal, QC, H2W 1R7, Canada.,Molecular Biology Programs, Université de Montréal, Montréal, QC, H3T 1J4, Canada
| | - Céline Schott
- Montreal Clinical Research Institute (IRCM), Montréal, QC, H2W 1R7, Canada.,Molecular Biology Programs, Université de Montréal, Montréal, QC, H3T 1J4, Canada
| | - Rony Chidiac
- Department of Pharmacology and Physiology, Université de Montréal, Montréal, QC, H3C 3J7, Canada
| | - Halil Bagci
- Montreal Clinical Research Institute (IRCM), Montréal, QC, H2W 1R7, Canada.,Department of Anatomy and Cell Biology, McGill University, Montréal, QC, H3A 0C7, Canada.,Institute of Biochemistry, ETH Zürich, Otto-Stern-Weg 3, 8093, Zürich, Switzerland
| | | | - Dominique Davidson
- Montreal Clinical Research Institute (IRCM), Montréal, QC, H2W 1R7, Canada
| | - Mathieu Ferron
- Montreal Clinical Research Institute (IRCM), Montréal, QC, H2W 1R7, Canada.,Molecular Biology Programs, Université de Montréal, Montréal, QC, H3T 1J4, Canada.,Division of Experimental Medicine, McGill University, Montréal, QC, H4A 3J1, Canada
| | - André Veillette
- Montreal Clinical Research Institute (IRCM), Montréal, QC, H2W 1R7, Canada.,Molecular Biology Programs, Université de Montréal, Montréal, QC, H3T 1J4, Canada
| | - Roger J Daly
- Cancer Program, Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, 3800, Australia
| | - Anne-Claude Gingras
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, M5G 1X5, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Jean-Philippe Gratton
- Department of Pharmacology and Physiology, Université de Montréal, Montréal, QC, H3C 3J7, Canada
| | - Jean-François Côté
- Montreal Clinical Research Institute (IRCM), Montréal, QC, H2W 1R7, Canada. .,Molecular Biology Programs, Université de Montréal, Montréal, QC, H3T 1J4, Canada. .,Department of Anatomy and Cell Biology, McGill University, Montréal, QC, H3A 0C7, Canada. .,Department of Biochemistry and Molecular Medicine, Université de Montréal, Montréal, QC, H3C 3J7, Canada.
| |
Collapse
|
27
|
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
|
28
|
Elliot A, Myllymäki H, Feng Y. Inflammatory Responses during Tumour Initiation: From Zebrafish Transgenic Models of Cancer to Evidence from Mouse and Man. Cells 2020; 9:cells9041018. [PMID: 32325966 PMCID: PMC7226149 DOI: 10.3390/cells9041018] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Revised: 04/08/2020] [Accepted: 04/14/2020] [Indexed: 12/12/2022] Open
Abstract
The zebrafish is now an important model organism for cancer biology studies and provides unique and complementary opportunities in comparison to the mammalian equivalent. The translucency of zebrafish has allowed in vivo live imaging studies of tumour initiation and progression at the cellular level, providing novel insights into our understanding of cancer. Here we summarise the available transgenic zebrafish tumour models and discuss what we have gleaned from them with respect to cancer inflammation. In particular, we focus on the host inflammatory response towards transformed cells during the pre-neoplastic stage of tumour development. We discuss features of tumour-associated macrophages and neutrophils in mammalian models and present evidence that supports the idea that these inflammatory cells promote early stage tumour development and progression. Direct live imaging of tumour initiation in zebrafish models has shown that the intrinsic inflammation induced by pre-neoplastic cells is tumour promoting. Signals mediating leukocyte recruitment to pre-neoplastic cells in zebrafish correspond to the signals that mediate leukocyte recruitment in mammalian tumours. The activation state of macrophages and neutrophils recruited to pre-neoplastic cells in zebrafish appears to be heterogenous, as seen in mammalian models, which provides an opportunity to study the plasticity of innate immune cells during tumour initiation. Although several potential mechanisms are described that might mediate the trophic function of innate immune cells during tumour initiation in zebrafish, there are several unknowns that are yet to be resolved. Rapid advancement of genetic tools and imaging technologies for zebrafish will facilitate research into the mechanisms that modulate leukocyte function during tumour initiation and identify targets for cancer prevention.
Collapse
Affiliation(s)
| | | | - Yi Feng
- Correspondence: ; Tel.: +44-(0)131-242-6685
| |
Collapse
|
29
|
Pelon F, Bourachot B, Kieffer Y, Magagna I, Mermet-Meillon F, Bonnet I, Costa A, Givel AM, Attieh Y, Barbazan J, Bonneau C, Fuhrmann L, Descroix S, Vignjevic D, Silberzan P, Parrini MC, Vincent-Salomon A, Mechta-Grigoriou F. Cancer-associated fibroblast heterogeneity in axillary lymph nodes drives metastases in breast cancer through complementary mechanisms. Nat Commun 2020; 11:404. [PMID: 31964880 PMCID: PMC6972713 DOI: 10.1038/s41467-019-14134-w] [Citation(s) in RCA: 230] [Impact Index Per Article: 57.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 12/17/2019] [Indexed: 01/05/2023] Open
Abstract
Although fibroblast heterogeneity is recognized in primary tumors, both its characterization in and its impact on metastases remain unknown. Here, combining flow cytometry, immunohistochemistry and RNA-sequencing on breast cancer samples, we identify four Cancer-Associated Fibroblast (CAF) subpopulations in metastatic lymph nodes (LN). Two myofibroblastic subsets, CAF-S1 and CAF-S4, accumulate in LN and correlate with cancer cell invasion. By developing functional assays on primary cultures, we demonstrate that these subsets promote metastasis through distinct functions. While CAF-S1 stimulate cancer cell migration and initiate an epithelial-to-mesenchymal transition through CXCL12 and TGFβ pathways, highly contractile CAF-S4 induce cancer cell invasion in 3-dimensions via NOTCH signaling. Patients with high levels of CAFs, particularly CAF-S4, in LN at diagnosis are prone to develop late distant metastases. Our findings suggest that CAF subset accumulation in LN is a prognostic marker, suggesting that CAF subsets could be examined in axillary LN at diagnosis.
Collapse
Affiliation(s)
- Floriane Pelon
- Institut Curie, Stress and Cancer Laboratory, Equipe labélisée par la Ligue Nationale contre le Cancer, PSL Research University, 26, rue d'Ulm, F-75005, Paris, France
- Inserm, U830, 26, rue d'Ulm, F-75005, Paris, France
| | - Brigitte Bourachot
- Institut Curie, Stress and Cancer Laboratory, Equipe labélisée par la Ligue Nationale contre le Cancer, PSL Research University, 26, rue d'Ulm, F-75005, Paris, France
- Inserm, U830, 26, rue d'Ulm, F-75005, Paris, France
| | - Yann Kieffer
- Institut Curie, Stress and Cancer Laboratory, Equipe labélisée par la Ligue Nationale contre le Cancer, PSL Research University, 26, rue d'Ulm, F-75005, Paris, France
- Inserm, U830, 26, rue d'Ulm, F-75005, Paris, France
| | - Ilaria Magagna
- Institut Curie, Stress and Cancer Laboratory, Equipe labélisée par la Ligue Nationale contre le Cancer, PSL Research University, 26, rue d'Ulm, F-75005, Paris, France
- Inserm, U830, 26, rue d'Ulm, F-75005, Paris, France
| | - Fanny Mermet-Meillon
- Analysis of Transduction Pathway, Institut Curie, Inserm, U830, PSL Research University, 26 rue d'Ulm, F-75005, Paris, France
| | - Isabelle Bonnet
- Institut Curie, Biology-inspired Physics at MesoScales Laboratory, Equipe labélisée par la Ligue Nationale contre le Cancer, CNRS UMR168, PSL Research University, Sorbonne Université, 26, rue d'Ulm, F-75005, Paris, France
| | - Ana Costa
- Institut Curie, Stress and Cancer Laboratory, Equipe labélisée par la Ligue Nationale contre le Cancer, PSL Research University, 26, rue d'Ulm, F-75005, Paris, France
- Inserm, U830, 26, rue d'Ulm, F-75005, Paris, France
| | - Anne-Marie Givel
- Institut Curie, Stress and Cancer Laboratory, Equipe labélisée par la Ligue Nationale contre le Cancer, PSL Research University, 26, rue d'Ulm, F-75005, Paris, France
- Inserm, U830, 26, rue d'Ulm, F-75005, Paris, France
| | - Youmna Attieh
- Institut Curie, Cell Migration and Invasion, UMR144, PSL Research University, 26, rue d'Ulm, F-75005, Paris, France
| | - Jorge Barbazan
- Institut Curie, Cell Migration and Invasion, UMR144, PSL Research University, 26, rue d'Ulm, F-75005, Paris, France
| | - Claire Bonneau
- Institut Curie, Stress and Cancer Laboratory, Equipe labélisée par la Ligue Nationale contre le Cancer, PSL Research University, 26, rue d'Ulm, F-75005, Paris, France
- Inserm, U830, 26, rue d'Ulm, F-75005, Paris, France
| | - Laetitia Fuhrmann
- Department of Pathology, Institut Curie Hospital, 26, rue d'Ulm, F-75248, Paris, France
| | - Stéphanie Descroix
- Institut Curie, Laboratoire Physico Chimie Curie, Institut Pierre-Gilles de Gennes, CNRS UMR168, 75005, Paris, France
| | - Danijela Vignjevic
- Institut Curie, Cell Migration and Invasion, UMR144, PSL Research University, 26, rue d'Ulm, F-75005, Paris, France
| | - Pascal Silberzan
- Institut Curie, Biology-inspired Physics at MesoScales Laboratory, Equipe labélisée par la Ligue Nationale contre le Cancer, CNRS UMR168, PSL Research University, Sorbonne Université, 26, rue d'Ulm, F-75005, Paris, France
| | - Maria Carla Parrini
- Analysis of Transduction Pathway, Institut Curie, Inserm, U830, PSL Research University, 26 rue d'Ulm, F-75005, Paris, France
| | - Anne Vincent-Salomon
- Department of Pathology, Institut Curie Hospital, 26, rue d'Ulm, F-75248, Paris, France
| | - Fatima Mechta-Grigoriou
- Institut Curie, Stress and Cancer Laboratory, Equipe labélisée par la Ligue Nationale contre le Cancer, PSL Research University, 26, rue d'Ulm, F-75005, Paris, France.
- Inserm, U830, 26, rue d'Ulm, F-75005, Paris, France.
| |
Collapse
|
30
|
Current Strategies to Target Tumor-Associated-Macrophages to Improve Anti-Tumor Immune Responses. Cells 2019; 9:cells9010046. [PMID: 31878087 PMCID: PMC7017001 DOI: 10.3390/cells9010046] [Citation(s) in RCA: 198] [Impact Index Per Article: 39.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 12/17/2019] [Accepted: 12/20/2019] [Indexed: 12/13/2022] Open
Abstract
: Established evidence demonstrates that tumor-infiltrating myeloid cells promote rather than stop-cancer progression. Tumor-associated macrophages (TAMs) are abundantly present at tumor sites, and here they support cancer proliferation and distant spreading, as well as contribute to an immune-suppressive milieu. Their pro-tumor activities hamper the response of cancer patients to conventional therapies, such as chemotherapy or radiotherapy, and also to immunotherapies based on checkpoint inhibition. Active research frontlines of the last years have investigated novel therapeutic strategies aimed at depleting TAMs and/or at reprogramming their tumor-promoting effects, with the goal of re-establishing a favorable immunological anti-tumor response within the tumor tissue. In recent years, numerous clinical trials have included pharmacological strategies to target TAMs alone or in combination with other therapies. This review summarizes the past and current knowledge available on experimental tumor models and human clinical studies targeting TAMs for cancer treatment.
Collapse
|
31
|
Nguyen KTP, Druhan LJ, Avalos BR, Zhai L, Rauova L, Nesmelova IV, Dréau D. CXCL12-CXCL4 heterodimerization prevents CXCL12-driven breast cancer cell migration. Cell Signal 2019; 66:109488. [PMID: 31785332 DOI: 10.1016/j.cellsig.2019.109488] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 11/24/2019] [Accepted: 11/26/2019] [Indexed: 02/07/2023]
Abstract
Despite improvements in cancer early detection and treatment, metastatic breast cancer remains deadly. Current therapeutic approaches have very limited efficacy in patients with triple negative breast cancer. Among the many mechanisms associated that contribute to cancer progression, signaling through the CXCL12-CXCR4 is an essential step in cancer cell migration. We previously demonstrated the formation of CXCL12-CXCL4 heterodimers (Carlson et al., 2013). Here, we investigated whether CXCL12-CXCL4 heterodimers alter tumor cell migration. CXCL12 alone dose-dependently promoted the MDA-MB 231 cell migration (p < .05), which could be prevented by blocking the CXCR4 receptor. The addition of CXCL4 inhibited the CXCL12-induced cell migration (p < .05). Using NMR spectroscopy, we identified the CXCL4-CXCL12 binding interface. Moreover, we generated a CXCL4-derived peptide homolog of the binding interface that mimicked the activity of native CXCL4 protein. These results confirm the formation of CXCL12-CXCL4 heterodimers and their inhibitory effects on the migration of breast tumors cells. These findings suggest that specific peptides mimicking heterodimerization of CXCL12 might prevent breast cancer cell migration.
Collapse
Affiliation(s)
- Khanh T P Nguyen
- Department of Biological Sciences, UNC Charlotte, Charlotte, NC, United States of America
| | - Lawrence J Druhan
- Department of Hematologic Oncology & Blood Disorders, Levine Cancer Institute, Atrium Health, Charlotte, NC, United States of America; Center for Biomedical Engineering and Science, UNC Charlotte, Charlotte, NC, United States of America
| | - Belinda R Avalos
- Department of Hematologic Oncology & Blood Disorders, Levine Cancer Institute, Atrium Health, Charlotte, NC, United States of America; Center for Biomedical Engineering and Science, UNC Charlotte, Charlotte, NC, United States of America
| | - Li Zhai
- Department of Pediatrics, The Children's Hospital of Philadelphia, PA, United States of America
| | - Lubica Rauova
- Department of Pediatrics, The Children's Hospital of Philadelphia, PA, United States of America; Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States of America
| | - Irina V Nesmelova
- Center for Biomedical Engineering and Science, UNC Charlotte, Charlotte, NC, United States of America; Department of Physics and Optical Science, UNC Charlotte, Charlotte, NC, United States of America
| | - Didier Dréau
- Department of Biological Sciences, UNC Charlotte, Charlotte, NC, United States of America; Center for Biomedical Engineering and Science, UNC Charlotte, Charlotte, NC, United States of America.
| |
Collapse
|
32
|
Shatnawi A, Malkaram SA, Fandy T, Tsouko E. Identification of the inhibitor of growth protein 4 (ING4) as a potential target in prostate cancer therapy. Mol Cell Biochem 2019; 464:153-167. [PMID: 31773467 DOI: 10.1007/s11010-019-03657-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 11/16/2019] [Indexed: 02/02/2023]
Abstract
INhibitor of Growth protein 4 (ING4) is a potential chromatin modifier that has been implicated in several cancer-related processes. However, the role of ING4 in prostate cancer (PC) is largely unknown. This study aimed to assess ING4's role in global transcriptional regulation in PC cells to identify potential cellular processes associated with ING4 loss. RNA-Seq using next-generation sequencing (NGS) was used to identify altered genes in LNCaP PC cells following ING4 depletion. Ingenuity pathways analysis (IPA®) was applied to the data to highlight candidates, ING4-regulated pathways, networks and cellular processes. Selected genes were validated using RT-qPCR. RNA-Seq of LNCaP cells revealed a total of 159 differentially expressed genes (fold change ≥ 1.5 or ≤ - 1.5, FDR ≤ 0.05) following ING4 knockdown. RT-qPCR used to validate the expression level of selected genes was in agreement with RNA-Seq results. Key genes, unique pathways, and biological networks were identified using IPA® analysis. This is the first report of global gene regulation in PC cells by ING4. The resultant differential expression profile revealed the potential role of ING4 in PC pathogenesis possibly through modulation of key genes, pathways and biological networks that are central drivers of the disease. Collectively, these findings shed light on a novel transcriptional regulator of PC that ultimately may influence the disease progression and as a potential target in the disease therapy.
Collapse
Affiliation(s)
- Aymen Shatnawi
- Department of Pharmaceutical and Administrative Sciences, University of Charleston School of Pharmacy, 2300 MacCorkle Ave SE, Charleston, WV, 25304, USA.
| | - Sridhar A Malkaram
- Department of Mathematics and Computer Sciences, West Virginia State University, W729, Wallace Hall, Institute, WV, 25112, USA
| | - Tamer Fandy
- Department of Pharmaceutical and Administrative Sciences, University of Charleston School of Pharmacy, 2300 MacCorkle Ave SE, Charleston, WV, 25304, USA
| | - Efrosini Tsouko
- Department of Orthopedic Surgery, Baylor College of Medicine, Houston, TX, 77030, USA
| |
Collapse
|
33
|
Kielbassa K, Vegna S, Ramirez C, Akkari L. Understanding the Origin and Diversity of Macrophages to Tailor Their Targeting in Solid Cancers. Front Immunol 2019; 10:2215. [PMID: 31611871 PMCID: PMC6773830 DOI: 10.3389/fimmu.2019.02215] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 09/02/2019] [Indexed: 12/20/2022] Open
Abstract
Tumor-associated macrophages (TAMs) are a major component of the tumor immune microenvironment (TIME) and are associated with a poor prognostic factor in several cancers. TAMs promote tumor growth by facilitating immunosuppression, angiogenesis, and inflammation, and can promote tumor recurrence post-therapeutic intervention. Major TAM-targeted therapies include depletion, reprogramming, as well as disrupting the balance of macrophage recruitment and their effector functions. However, intervention-targeting macrophages have been challenging, since TAM populations are highly plastic and adaptation or resistance to these approaches often arise. Defining a roadmap of macrophage dynamics in the TIME related to tissue and tumor type could represent exploitable vulnerabilities related to their altered functions in cancer malignancy. Here, we review multiple macrophage-targeting strategies in brain, liver, and lung cancers, which all emerge in tissues rich in resident macrophages. We discuss the successes and failures of these therapeutic approaches as well as the potential of personalized macrophage-targeting treatments in combination therapies.
Collapse
Affiliation(s)
| | | | | | - Leila Akkari
- Division of Tumour Biology and Immunology, Oncode Institute, Netherlands Cancer Institute, Amsterdam, Netherlands
| |
Collapse
|
34
|
Kang S, Kang K, Chae A, Kim YK, Jang H, Min DH. Fucoidan-coated coral-like Pt nanoparticles for computed tomography-guided highly enhanced synergistic anticancer effect against drug-resistant breast cancer cells. NANOSCALE 2019; 11:15173-15183. [PMID: 31380881 DOI: 10.1039/c9nr04495g] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
Chemotherapy, the most commonly applied cancer treatment, often causes unexpected failure due to multidrug resistance (MDR). To overcome MDR, we have designed a platform to realize a combinational synergistic effect of a natural bioactive product (fucoidan), anticancer small compound (doxorubicin), and photothermal nanocarrier (Pt nanoparticle) to treat drug-resistant breast cancer cells. Especially, fucoidan, a sulfated, polysaccharide-structured, therapeutic biopolymer, has been recently recognized as a potential anticancer compound; however, its cancer-inhibiting efficacy has been regarded as low owing to its insufficient level in serum following its conventional oral ingestion. To enhance its potency, fucoidan was applied as a biocompatible surfactant and surface-coating biopolymer in nanocarrier synthesis to manufacture coral-like, fucoidan-coated Pt nanoparticles with a rough surface morphology by a one-pot method. As a result, the biological-thermo-chemo trimodal combination treatment showed excellent therapeutic efficiency against the MDR breast cancer cell MCF-7 ADR both in vitro and in vivo, and the computed tomography contrast effect was also confirmed from the constituent element Pt. Beyond universal application in drug delivery and photothermal therapy, the present approach of applying a MDR modulating/anticancer natural product from nanoparticle synthesis to theranostics will contribute greatly to maximizing their potential through interdisciplinary convergence in the near future.
Collapse
Affiliation(s)
- Seounghun Kang
- Department of Chemistry, Seoul National University, Seoul 08826, Republic of Korea.
| | | | | | | | | | | |
Collapse
|
35
|
Chatterjee S, Bhat V, Berdnikov A, Liu J, Zhang G, Buchel E, Safneck J, Marshall AJ, Murphy LC, Postovit LM, Raouf A. Paracrine Crosstalk between Fibroblasts and ER + Breast Cancer Cells Creates an IL1β-Enriched Niche that Promotes Tumor Growth. iScience 2019; 19:388-401. [PMID: 31419632 PMCID: PMC6706609 DOI: 10.1016/j.isci.2019.07.034] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Revised: 03/16/2019] [Accepted: 07/18/2019] [Indexed: 12/30/2022] Open
Abstract
Breast cancer-induced activated fibroblasts support tumor progression. However, the role of normal fibroblasts in tumor progression remains controversial. In this study, we used modified patient-derived organoid cultures and demonstrate that constitutively secreted cytokines from normal breast fibroblasts initiate a paracrine signaling mechanism with estrogen receptor-positive (ER+) breast cancer cells, which results in the creation of an interleukin (IL)-1β-enriched microenvironment. We found that this paracrine signaling mechanism is shared between normal and activated fibroblasts. Interestingly, we observed that in reconstructed tumor microenvironment containing autologous ER+ breast cancer cells, activated fibroblasts, and immune cells, tamoxifen is more effective in reducing tumor cell proliferation when this paracrine signaling is blocked. Our findings then suggest that ER+ tumor cells could create a growth-promoting environment without activating stromal fibroblasts and that in breast-conserving surgeries, normal fibroblasts could be a significant modulator of tumor recurrence by enhancing the proliferation of residual breast cancer cells in the tumor-adjacent breast tissue. Normal fibroblast-cancer cell interaction promotes tumor progression Paracrine signaling common to normal and activated fibroblasts promotes drug resistance Fibroblast-secreted factors create an IL1β-enriched niche for ER+ breast cancer cell growth
Collapse
Affiliation(s)
- Sumanta Chatterjee
- Department of Immunology, Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0T5, Canada; Research Institute of Oncology & Hematology, CancerCareManitoba, Winnipeg, MB R3E 0V9, Canada
| | - Vasudeva Bhat
- Department of Immunology, Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0T5, Canada; Research Institute of Oncology & Hematology, CancerCareManitoba, Winnipeg, MB R3E 0V9, Canada
| | - Alexei Berdnikov
- Department of Surgery, Section of Plastic Surgery, Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3A 1M5, Canada
| | - Jiahui Liu
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2E1, Canada
| | - Guihua Zhang
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2E1, Canada
| | - Edward Buchel
- Department of Surgery, Section of Plastic Surgery, Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3A 1M5, Canada
| | - Janice Safneck
- Department of Pathology, Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 3P5, Canada
| | - Aaron J Marshall
- Department of Immunology, Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0T5, Canada
| | - Leigh C Murphy
- Research Institute of Oncology & Hematology, CancerCareManitoba, Winnipeg, MB R3E 0V9, Canada; Department of Biochemistry and Medical Genetics, Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Lynne-Marie Postovit
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2E1, Canada; Department of Obstetrics and Gynecology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2E1, Canada
| | - Afshin Raouf
- Department of Immunology, Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0T5, Canada; Research Institute of Oncology & Hematology, CancerCareManitoba, Winnipeg, MB R3E 0V9, Canada.
| |
Collapse
|
36
|
Zhang J, Chen J, Wo D, Yan H, Liu P, Ma E, Li L, Zheng L, Chen D, Yu Z, Liang C, Peng J, Ren DN, Zhu W. LRP6 Ectodomain Prevents SDF-1/CXCR4-Induced Breast Cancer Metastasis to Lung. Clin Cancer Res 2019; 25:4832-4845. [PMID: 31010839 DOI: 10.1158/1078-0432.ccr-18-3557] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 03/04/2019] [Accepted: 04/15/2019] [Indexed: 11/16/2022]
Abstract
PURPOSE Lung metastasis is an important cause of breast cancer-related deaths, in which SDF-1/CXCR4 signaling pathway plays a critical role. Single transmembrane protein LRP6 is viewed as an oncogene via activating the Wnt/β-catenin signaling pathway. Our work aims to investigate the relationship between SDF-1/CXCR4 and LRP6 in breast cancer lung metastasis. EXPERIMENTAL DESIGN We examined the expressions and functions of SDF-1/CXCR4 and LRP6 as well as their relationship in breast cancer in vitro and in vivo. RESULTS LRP6 ectodomain (LRP6N) directly bound to CXCR4 and competitively prevented SDF-1 binding to CXCR4. LRP6N prevented SDF-1/CXCR4-induced metastasis to lung and prolonged survival in mice bearing breast tumors, whereas LRP6 knockdown activated SDF-1/CXCR4 signal transduction and promoted lung metastasis and tumor death. Furthermore, patients with breast cancer with high CXCR4 expression had poor prognosis, which was exacerbated by low LRP6 expression but improved by high LRP6 expression. Interestingly, a secreted LRP6N was found in the serum of mice and humans, which was downregulated by the onset of cancer metastasis in both mice bearing breast cancer as well as in patients with breast cancer. CONCLUSIONS LRP6N might be a promising diagnostic marker for the early detection of breast cancer metastasis as well as an inhibitor of SDF-1/CXCR4-induced breast cancer metastasis. LRP6N also provides an interesting link between Wnt signaling and SDF-1/CXCR4 signaling, the two key pathways involved in cancer development.
Collapse
Affiliation(s)
- Jiankang Zhang
- Clinical and Translational Research Center, Research Institute of Heart Failure Shanghai East Hospital, Key Laboratory of Arrhythmias of Ministry of Education, Tongji University School of Medicine, Shanghai, China
| | - Jinxiao Chen
- Department of Plastic and Reconstructive Surgery, Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Da Wo
- Department of Plastic and Reconstructive Surgery, Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Hongwei Yan
- Clinical and Translational Research Center, Research Institute of Heart Failure Shanghai East Hospital, Key Laboratory of Arrhythmias of Ministry of Education, Tongji University School of Medicine, Shanghai, China
| | - Peng Liu
- Clinical and Translational Research Center, Research Institute of Heart Failure Shanghai East Hospital, Key Laboratory of Arrhythmias of Ministry of Education, Tongji University School of Medicine, Shanghai, China
| | - En Ma
- Clinical and Translational Research Center, Research Institute of Heart Failure Shanghai East Hospital, Key Laboratory of Arrhythmias of Ministry of Education, Tongji University School of Medicine, Shanghai, China
| | - Limei Li
- Clinical and Translational Research Center, Research Institute of Heart Failure Shanghai East Hospital, Key Laboratory of Arrhythmias of Ministry of Education, Tongji University School of Medicine, Shanghai, China
| | - Liang Zheng
- Clinical and Translational Research Center, Research Institute of Heart Failure Shanghai East Hospital, Key Laboratory of Arrhythmias of Ministry of Education, Tongji University School of Medicine, Shanghai, China
| | - Daxin Chen
- Fujian Key Laboratory of Integrative Medicine on Geriatric, Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
| | - Zuoren Yu
- Clinical and Translational Research Center, Research Institute of Heart Failure Shanghai East Hospital, Key Laboratory of Arrhythmias of Ministry of Education, Tongji University School of Medicine, Shanghai, China
| | - Chunli Liang
- Department of Surgery East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jun Peng
- Fujian Key Laboratory of Integrative Medicine on Geriatric, Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China.
| | - Dan-Ni Ren
- Fujian Key Laboratory of Integrative Medicine on Geriatric, Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China.
| | - Weidong Zhu
- Clinical and Translational Research Center, Research Institute of Heart Failure Shanghai East Hospital, Key Laboratory of Arrhythmias of Ministry of Education, Tongji University School of Medicine, Shanghai, China.
| |
Collapse
|
37
|
Blocking CXCR4 alleviates desmoplasia, increases T-lymphocyte infiltration, and improves immunotherapy in metastatic breast cancer. Proc Natl Acad Sci U S A 2019; 116:4558-4566. [PMID: 30700545 DOI: 10.1073/pnas.1815515116] [Citation(s) in RCA: 256] [Impact Index Per Article: 51.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Metastatic breast cancers (mBCs) are largely resistant to immune checkpoint blockade, but the mechanisms remain unclear. Primary breast cancers are characterized by a dense fibrotic stroma, which is considered immunosuppressive in multiple malignancies, but the stromal composition of breast cancer metastases and its role in immunosuppression are largely unknown. Here we show that liver and lung metastases of human breast cancers tend to be highly fibrotic, and unlike primary breast tumors, they exclude cytotoxic T lymphocytes (CTLs). Unbiased analysis of the The Cancer Genome Atlas database of human breast tumors revealed a set of genes that are associated with stromal T-lymphocyte exclusion. Among these, we focused on CXCL12 as a relevant target based on its known roles in immunosuppression in other cancer types. We found that the CXCL12 receptor CXCR4 is highly expressed in both human primary tumors and metastases. To gain insight into the role of the CXCL12/CXCR4 axis, we inhibited CXCR4 signaling pharmacologically and found that plerixafor decreases fibrosis, alleviates solid stress, decompresses blood vessels, increases CTL infiltration, and decreases immunosuppression in murine mBC models. By deleting CXCR4 in αSMA+ cells, we confirmed that these immunosuppressive effects are dependent on CXCR4 signaling in αSMA+ cells, which include cancer-associated fibroblasts as well as other cells such as pericytes. Accordingly, CXCR4 inhibition more than doubles the response to immune checkpoint blockers in mice bearing mBCs. These findings demonstrate that CXCL12/CXCR4-mediated desmoplasia in mBC promotes immunosuppression and is a potential target for overcoming therapeutic resistance to immune checkpoint blockade in mBC patients.
Collapse
|
38
|
Mao SPH, Park M, Cabrera RM, Christin JR, Karagiannis GS, Oktay MH, Zaiss DMW, Abrams SI, Guo W, Condeelis JS, Kenny PA, Segall JE. Loss of amphiregulin reduces myoepithelial cell coverage of mammary ducts and alters breast tumor growth. Breast Cancer Res 2018; 20:131. [PMID: 30367629 PMCID: PMC6203982 DOI: 10.1186/s13058-018-1057-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Accepted: 10/02/2018] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Amphiregulin (AREG), a ligand of the epidermal growth factor receptor, is not only essential for proper mammary ductal development, but also associated with breast cancer proliferation and growth. In the absence of AREG, mammary ductal growth is stunted and fails to expand. Furthermore, suppression of AREG expression in estrogen receptor-positive breast tumor cells inhibits in-vitro and in-vivo growth. METHODS We crossed AREG-null (AREG-/-) mice with the murine luminal B breast cancer model, MMTV-PyMT (PyMT), to generate spontaneous breast tumors that lack AREG (AREG-/- PyMT). We evaluated tumor growth, cytokeratin-8 (K8)-positive luminal cells, cytokeratin-14 (K14)-positive myoepithelial cells, and expression of AREG, Ki67, and PyMT. Primary myoepithelial cells from nontumor-bearing AREG+/+ mice underwent fluorescence-activated cell sorting and were adapted to culture for in-vitro coculture studies with AT-3 cells, a cell line derived from C57Bl/6 PyMT mammary tumors. RESULTS Intriguingly, PyMT-induced lesions progress more rapidly in AREG-/- mice than in AREG+/+ mice. Quantification of K8+ luminal and K14+ myoepithelial cells in non-PyMT AREG-/- mammary glands showed fewer K14+ cells and a thinner myoepithelial layer. Study of AT-3 cells indicated that coculture with myoepithelial cells or exposure to AREG, epidermal growth factor, or basic fibroblast growth factor can suppress PyMT expression. Late-stage AREG-/- PyMT tumors are significantly less solid in structure, with more areas of papillary and cystic growth. Papillary areas appear to be both less proliferative and less necrotic. In The Cancer Genome Atlas database, luminal-B invasive papillary carcinomas have lower AREG expression than luminal B invasive ductal carcinomas. CONCLUSIONS Our study has revealed a previously unknown role of AREG in myoepithelial cell development and PyMT expression. AREG expression is essential for proper myoepithelial coverage of mammary ducts. Both AREG and myoepithelial cells can suppress PyMT expression. We find that lower AREG expression is associated with invasive papillary breast cancer in both the MMTV-PyMT model and human breast cancer.
Collapse
MESH Headings
- Amphiregulin/genetics
- Amphiregulin/metabolism
- Animals
- Antigens, Polyomavirus Transforming/genetics
- Antigens, Polyomavirus Transforming/metabolism
- Cell Line, Tumor
- Cell Proliferation
- Epithelial Cells/pathology
- Epithelial Cells/virology
- Female
- Humans
- Mammary Glands, Animal/cytology
- Mammary Glands, Animal/pathology
- Mammary Neoplasms, Experimental/genetics
- Mammary Neoplasms, Experimental/pathology
- Mammary Neoplasms, Experimental/virology
- Mammary Tumor Virus, Mouse/genetics
- Mammary Tumor Virus, Mouse/pathogenicity
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Neoplasm Invasiveness/pathology
- Polyomavirus/genetics
- Polyomavirus/immunology
Collapse
Affiliation(s)
- Serena P. H. Mao
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, 1301 Morris Park Avenue, Bronx, NY 10461 USA
| | - Minji Park
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, 1301 Morris Park Avenue, Bronx, NY 10461 USA
| | - Ramon M. Cabrera
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, 1301 Morris Park Avenue, Bronx, NY 10461 USA
| | - John R. Christin
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY 10461 USA
| | - George S. Karagiannis
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, 1301 Morris Park Avenue, Bronx, NY 10461 USA
- Gruss Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY 10461 USA
- Integrated Imaging Program, Albert Einstein College of Medicine, Bronx, NY 10461 USA
| | - Maja H. Oktay
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, 1301 Morris Park Avenue, Bronx, NY 10461 USA
- Gruss Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY 10461 USA
- Integrated Imaging Program, Albert Einstein College of Medicine, Bronx, NY 10461 USA
| | - Dietmar M. W. Zaiss
- Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh, UK
| | - Scott I. Abrams
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263 USA
| | - Wenjun Guo
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY 10461 USA
| | - John S. Condeelis
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, 1301 Morris Park Avenue, Bronx, NY 10461 USA
- Gruss Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY 10461 USA
- Integrated Imaging Program, Albert Einstein College of Medicine, Bronx, NY 10461 USA
| | - Paraic A. Kenny
- Kabara Cancer Research Institute, Gundersen Medical Foundation, La Crosse, WI 54601 USA
| | - Jeffrey E. Segall
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, 1301 Morris Park Avenue, Bronx, NY 10461 USA
- Gruss Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY 10461 USA
| |
Collapse
|
39
|
Fu Z, Zhang P, Luo H, Huang H, Wang F. CXCL12 modulates the radiosensitivity of cervical cancer by regulating CD44. Mol Med Rep 2018; 18:5101-5108. [PMID: 30320394 DOI: 10.3892/mmr.2018.9554] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 07/13/2018] [Indexed: 11/05/2022] Open
Abstract
The aim of the present study was to investigate the regulation of stromal cell‑derived factor 1 (CXCL12) in the radioresistance of cervical cancer, which was upregulated in tumors in our previous study. A CCK‑8 assay was used to detect cell viability. Flow cytometry was used to measure cell apoptosis and the expression levels of CD44 and CXCR4. ELISA was performed to measure the expression level of CXCL12 protein and CXCL12 mRNA was detected by reverse transcription‑quantitative polymerase chain reaction assays. Cell viability and apoptosis were determined with or without treatment with CXCL12 small interfering (si)RNA to examine the function of CXCL12 in Hela cells. The expression level of CD44 antigen (CD44) and C‑X‑C chemokine receptor type 4 (CXCR4) were measured using flow cytometry in the presence of CXCL12 and irradiation. In the present study, it was demonstrated that inhibition of CXCL12 reduced cell viability and increased cellular apoptosis in Hela cells treated with irradiation. Following treatment with CXCL12 siRNA, the expression level of CD44 was downregulated and the expression level of CXCR4 was upregulated. This effect of regulation additionally occurred in the presence of irradiation. In conclusion, the present data demonstrated that CXCL12 served an important role in the radioresistance of cervical cancer, suggestinh a novel therapeutic target.
Collapse
Affiliation(s)
- Zhichao Fu
- Department of Radiation Oncology, Dongfang Hospital, Xiamen University, Fuzhou, Fujian 350025, P.R. China
| | - Pei Zhang
- Department of Radiation Medicine, Faculty of Naval Medicine, Second Military Medical University, Shanghai 200433, P.R. China
| | - Huachun Luo
- Department of Radiation Oncology, Fuzhou General Hospital of Nanjing Military Command, Fuzhou, Fujian 350025, P.R. China
| | - Huijuan Huang
- Department of Gynaecology and Obstetrics, Dongfang Hospital, Xiamen University, Fuzhou, Fujian 350025, P.R. China
| | - Fengmei Wang
- Department of Gynaecology and Obstetrics, Dongfang Hospital, Xiamen University, Fuzhou, Fujian 350025, P.R. China
| |
Collapse
|
40
|
Lee NH, Nikfarjam M, He H. Functions of the CXC ligand family in the pancreatic tumor microenvironment. Pancreatology 2018; 18:705-716. [PMID: 30078614 DOI: 10.1016/j.pan.2018.07.011] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 07/06/2018] [Accepted: 07/30/2018] [Indexed: 02/06/2023]
Abstract
Therapeutic resistance is the major contributor to the poor prognosis of and low survival from pancreatic cancer (PC). Cancer progression is a complex process reliant on interactions between the tumor and the tumor microenvironment (TME). Members of the CXCL family of chemokines are present in the pancreatic TME and seem to play a vital role in regulating PC progression. As pancreatic tumors interact with the TME and with PC stem cells (CSCs), determining the roles of specific members of the CXCL family is vital to the development of improved therapies. This review highlights the roles of selected CXCLs in the interactions between pancreatic tumor and its stroma, and in CSC phenotypes, which can be used to identify potential treatment targets.
Collapse
Affiliation(s)
- Nien-Hung Lee
- Department of Surgery, University of Melbourne, Austin Health, Melbourne, Victoria, Australia
| | - Mehrdad Nikfarjam
- Department of Surgery, University of Melbourne, Austin Health, Melbourne, Victoria, Australia
| | - Hong He
- Department of Surgery, University of Melbourne, Austin Health, Melbourne, Victoria, Australia.
| |
Collapse
|
41
|
Liu H, Li Z, Deng M, Liu Q, Zhang T, Guo W, Li P, Qiao W. Prognostic and clinicopathological value of CXCL12/SDF1 expression in breast cancer: A meta-analysis. Clin Chim Acta 2018; 484:72-80. [DOI: 10.1016/j.cca.2018.05.041] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 05/19/2018] [Accepted: 05/21/2018] [Indexed: 12/21/2022]
|
42
|
Chia K, Mazzolini J, Mione M, Sieger D. Tumor initiating cells induce Cxcr4-mediated infiltration of pro-tumoral macrophages into the brain. eLife 2018; 7:e31918. [PMID: 29465400 PMCID: PMC5821457 DOI: 10.7554/elife.31918] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 01/31/2018] [Indexed: 12/28/2022] Open
Abstract
It is now clear that microglia and macrophages are present in brain tumors, but whether or how they affect initiation and development of tumors is not known. Exploiting the advantages of the zebrafish (Danio rerio) model, we showed that macrophages and microglia respond immediately upon oncogene activation in the brain. Overexpression of human AKT1 within neural cells of larval zebrafish led to a significant increase in the macrophage and microglia populations. By using a combination of transgenic and mutant zebrafish lines, we showed that this increase was caused by the infiltration of peripheral macrophages into the brain mediated via Sdf1b-Cxcr4b signaling. Intriguingly, confocal live imaging reveals highly dynamic interactions between macrophages/microglia and pre-neoplastic cells, which do not result in phagocytosis of pre-neoplastic cells. Finally, depletion of macrophages and microglia resulted in a significant reduction of oncogenic cell proliferation. Thus, macrophages and microglia show tumor promoting functions already during the earliest stages of the developing tumor microenvironment.
Collapse
Affiliation(s)
- Kelda Chia
- Centre for Discovery Brain SciencesUniversity of EdinburghEdinburghUnited Kingdom
| | - Julie Mazzolini
- Centre for Discovery Brain SciencesUniversity of EdinburghEdinburghUnited Kingdom
| | - Marina Mione
- Centre for Integrative Biology (CIBIO)University of TrentoTrentoItaly
| | - Dirk Sieger
- Centre for Discovery Brain SciencesUniversity of EdinburghEdinburghUnited Kingdom
| |
Collapse
|
43
|
ALÇIĞIR ME, ANADOL E, GÜLTİKEN N, KARAKAŞ ALKAN K, ALKAN H, KANCA H. Mikst Orjinli Köpek Meme Tümörlerinde CXC Chemokine Ligand 12 ve G Protein Coupled Receptor 30 Ekspresyonları. ACTA ACUST UNITED AC 2018. [DOI: 10.30607/kvj.368924] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
44
|
Rowson-Hodel A, Wald J, Hatakeyama J, O’Neal W, Stonebraker J, VanderVorst K, Saldana M, Borowsky A, Sweeney C, Carraway K. Membrane Mucin Muc4 promotes blood cell association with tumor cells and mediates efficient metastasis in a mouse model of breast cancer. Oncogene 2018; 37:197-207. [PMID: 28892049 PMCID: PMC5930013 DOI: 10.1038/onc.2017.327] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Revised: 06/21/2017] [Accepted: 08/04/2017] [Indexed: 12/11/2022]
Abstract
Mucin-4 (Muc4) is a large cell surface glycoprotein implicated in the protection and lubrication of epithelial structures. Previous studies suggest that aberrantly expressed Muc4 can influence the adhesiveness, proliferation, viability and invasiveness of cultured tumor cells, as well as the growth rate and metastatic efficiency of xenografted tumors. Although it has been suggested that one of the major mechanisms by which Muc4 potentiates tumor progression is via its engagement of the ErbB2/HER2 receptor tyrosine kinase, other mechanisms exist and remain to be delineated. Moreover, the requirement for endogenous Muc4 for tumor growth progression has not been previously explored in the context of gene ablation. To assess the contribution of endogenous Muc4 to mammary tumor growth properties, we first created a genetically engineered mouse line lacking functional Muc4 (Muc4ko), and then crossed these animals with the NDL (Neu DeLetion mutant) model of ErbB2-induced mammary tumorigenesis. We observed that Muc4ko animals are fertile and develop normally, and adult mice exhibit no overt tissue abnormalities. In tumor studies, we observed that although some markers of tumor growth such as vascularity and cyclin D1 expression are suppressed, primary mammary tumors from Muc4ko/NDL female mice exhibit similar latencies and growth rates as Muc4wt/NDL animals. However, the presence of lung metastases is markedly suppressed in Muc4ko/NDL mice. Interestingly, histological analysis of lung lesions from Muc4ko/NDL mice revealed a reduced association of disseminated cells with platelets and white blood cells. Moreover, isolated cells derived from Muc4ko/NDL tumors interact with fewer blood cells when injected directly into the vasculature or diluted into blood from wild type mice. We further observed that blood cells more efficiently promote the viability of non-adherent Muc4wt/NDL cells than Muc4ko/NDL cells. Together, our observations suggest that Muc4 may facilitate metastasis by promoting the association of circulating tumor cells with blood cells to augment tumor cell survival in circulation.
Collapse
Affiliation(s)
- A.R. Rowson-Hodel
- Department of Biochemistry and Molecular Medicine, and UC Davis Comprehensive Cancer Center, University of California, Davis, School of Medicine, Sacramento, California, USA
| | - J.H. Wald
- Department of Biochemistry and Molecular Medicine, and UC Davis Comprehensive Cancer Center, University of California, Davis, School of Medicine, Sacramento, California, USA
| | - J. Hatakeyama
- Department of Biochemistry and Molecular Medicine, and UC Davis Comprehensive Cancer Center, University of California, Davis, School of Medicine, Sacramento, California, USA
| | - W.K. O’Neal
- Marsico Lung Institute/UNC Cystic Fibrosis Research Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - J.R. Stonebraker
- Marsico Lung Institute/UNC Cystic Fibrosis Research Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - K. VanderVorst
- Department of Biochemistry and Molecular Medicine, and UC Davis Comprehensive Cancer Center, University of California, Davis, School of Medicine, Sacramento, California, USA
| | - M.J. Saldana
- Department of Biochemistry and Molecular Medicine, and UC Davis Comprehensive Cancer Center, University of California, Davis, School of Medicine, Sacramento, California, USA
| | - A.D. Borowsky
- Department of Pathology and Laboratory Medicine, University of California, Davis, School of Medicine, Sacramento, California, USA
| | - C. Sweeney
- Department of Biochemistry and Molecular Medicine, and UC Davis Comprehensive Cancer Center, University of California, Davis, School of Medicine, Sacramento, California, USA
| | - K.L. Carraway
- Department of Biochemistry and Molecular Medicine, and UC Davis Comprehensive Cancer Center, University of California, Davis, School of Medicine, Sacramento, California, USA
| |
Collapse
|
45
|
Jin L, Han B, Siegel E, Cui Y, Giuliano A, Cui X. Breast cancer lung metastasis: Molecular biology and therapeutic implications. Cancer Biol Ther 2018; 19:858-868. [PMID: 29580128 PMCID: PMC6300341 DOI: 10.1080/15384047.2018.1456599] [Citation(s) in RCA: 172] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Revised: 03/18/2018] [Accepted: 03/19/2018] [Indexed: 02/05/2023] Open
Abstract
Distant metastasis accounts for the vast majority of deaths in patients with cancer. Breast cancer exhibits a distinct metastatic pattern commonly involving bone, liver, lung, and brain. Breast cancer can be divided into different subtypes based on gene expression profiles, and different breast cancer subtypes show preference to distinct organ sites of metastasis. Luminal breast tumors tend to metastasize to bone while basal-like breast cancer (BLBC) displays a lung tropism of metastasis. However, the mechanisms underlying this organ-specific pattern of metastasis still remain to be elucidated. In this review, we will summarize the recent advances regarding the molecular signaling pathways as well as the therapeutic strategies for treating breast cancer lung metastasis.
Collapse
Affiliation(s)
- Liting Jin
- Department of Breast Surgery, Hubei Cancer Hospital, Wuhan, China
| | - Bingchen Han
- Department of Surgery, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Emily Siegel
- Department of Surgery, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Yukun Cui
- Laboratory for Breast Cancer Diagnosis and Treatment, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Armando Giuliano
- Department of Surgery, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Xiaojiang Cui
- Department of Surgery, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- CONTACT Xiaojiang Cui Cedars-Sinai Medical Center, 8700 Beverly Blvd, Davis Building 2065, Los Angeles, CA 90048
| |
Collapse
|
46
|
Qian BZ. Inflammation fires up cancer metastasis. Semin Cancer Biol 2017; 47:170-176. [PMID: 28838845 DOI: 10.1016/j.semcancer.2017.08.006] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Revised: 08/08/2017] [Accepted: 08/09/2017] [Indexed: 12/16/2022]
Abstract
Metastatic disease is the major challenge of cancer that accounts for over 90% of total cancer lethality. Mounting clinical and preclinical data now indicate that inflammation, a potent immune and repair response, is indispensable for metastasis. In this review we describe our current understanding of how major inflammatory cells contribute to metastatic cascade with a focus on the primary tumour. We also discuss exciting new directions for future research and novel therapeutic approaches to tackle metastatic disease through targeting inflammation.
Collapse
Affiliation(s)
- Bin-Zhi Qian
- University of Edinburgh and MRC Centre for Reproductive Health, EH16 4TJ, Edinburgh, United Kingdom; Edinburgh Cancer Research UK Centre, EH16 4TJ, Edinburgh, United Kingdom.
| |
Collapse
|
47
|
BRMS1 gene expression may be associated with clinico-pathological features of breast cancer. Biosci Rep 2017; 37:BSR20170672. [PMID: 28533425 PMCID: PMC5563535 DOI: 10.1042/bsr20170672] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Revised: 05/18/2017] [Accepted: 05/22/2017] [Indexed: 01/17/2023] Open
Abstract
Our aim is to investigate whether or not the breast cancer metastasis suppressor 1 (BRMS1) gene expression is directly linked to clinico-pathological features of breast cancer. Following a stringent inclusion and exclusion criteria, case–control studies with associations between BRMS1 and breast cancer were selected from articles obtained by way of searches conducted through an electronic database. All statistical analyses were performed with Stata 12.0 (Stata Corp, College Station, TX, U.S.A.). Ultimately, 1,263 patients with breast cancer were found in a meta-analysis retrieved from a total that included 12 studies. Results of our meta-analysis suggested that BRMS1 protein in breast cancer tissues was significantly lower in comparison with normal breast tissues (odds ratio, OR = 0.08, 95% confidence interval (CI) = 0.04–0.15). The BRMS1 protein in metastatic breast cancer tissue was decreased than from that was found in non-metastatic breast cancer tissue (OR = 0.20, 95%CI = 0.13–0.29), and BRMS1 protein in tumor-node-metastasis (TNM) stages 1 and 2 was found to be higher than TNM stages 3 and 4 (OR = 4.62, 95%CI = 2.77–7.70). BRMS1 protein in all three major types of breast cancer was lower than that of control tissues respectively. We also found strong correlations between BRMS1 mRNA levels and TNM stage and tumor size. The results our meta-analysis showed that reduction in BRMS1 expression level was linked directly to clinico-pathological features of breast cancer significantly; therefore, suggesting the loss of expression or reduced levels of BRMS1 is potentially a strong indicator of the metastatic capacity of breast cancer with poor prognosis.
Collapse
|
48
|
Karagiannis GS, Pastoriza JM, Wang Y, Harney AS, Entenberg D, Pignatelli J, Sharma VP, Xue EA, Cheng E, D'Alfonso TM, Jones JG, Anampa J, Rohan TE, Sparano JA, Condeelis JS, Oktay MH. Neoadjuvant chemotherapy induces breast cancer metastasis through a TMEM-mediated mechanism. Sci Transl Med 2017; 9:eaan0026. [PMID: 28679654 PMCID: PMC5592784 DOI: 10.1126/scitranslmed.aan0026] [Citation(s) in RCA: 326] [Impact Index Per Article: 46.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Accepted: 06/13/2017] [Indexed: 12/11/2022]
Abstract
Breast cancer cells disseminate through TIE2/MENACalc/MENAINV-dependent cancer cell intravasation sites, called tumor microenvironment of metastasis (TMEM), which are clinically validated as prognostic markers of metastasis in breast cancer patients. Using fixed tissue and intravital imaging of a PyMT murine model and patient-derived xenografts, we show that chemotherapy increases the density and activity of TMEM sites and Mena expression and promotes distant metastasis. Moreover, in the residual breast cancers of patients treated with neoadjuvant paclitaxel after doxorubicin plus cyclophosphamide, TMEM score and its mechanistically connected MENAINV isoform expression pattern were both increased, suggesting that chemotherapy, despite decreasing tumor size, increases the risk of metastatic dissemination. Chemotherapy-induced TMEM activity and cancer cell dissemination were reversed by either administration of the TIE2 inhibitor rebastinib or knockdown of the MENA gene. Our results indicate that TMEM score increases and MENA isoform expression pattern changes with chemotherapy and can be used in predicting prometastatic changes in response to chemotherapy. Furthermore, inhibitors of TMEM function may improve clinical benefits of chemotherapy in the neoadjuvant setting or in metastatic disease.
Collapse
Affiliation(s)
- George S Karagiannis
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
- Integrated Imaging Program, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Jessica M Pastoriza
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Department of Surgery, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY 10467, USA
| | - Yarong Wang
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Integrated Imaging Program, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Allison S Harney
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Integrated Imaging Program, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Department of Radiology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - David Entenberg
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Integrated Imaging Program, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Jeanine Pignatelli
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Ved P Sharma
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Emily A Xue
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Esther Cheng
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Timothy M D'Alfonso
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Joan G Jones
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Integrated Imaging Program, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Department of Pathology, Montefiore Medical Center, Bronx, NY 10467, USA
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Jesus Anampa
- Department of Oncology, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY 10467, USA
| | - Thomas E Rohan
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Joseph A Sparano
- Department of Oncology, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY 10467, USA
| | - John S Condeelis
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
- Integrated Imaging Program, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Maja H Oktay
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Department of Pathology, Montefiore Medical Center, Bronx, NY 10467, USA
| |
Collapse
|
49
|
Sawa-Wejksza K, Kandefer-Szerszeń M. Tumor-Associated Macrophages as Target for Antitumor Therapy. Arch Immunol Ther Exp (Warsz) 2017; 66:97-111. [PMID: 28660349 PMCID: PMC5851686 DOI: 10.1007/s00005-017-0480-8] [Citation(s) in RCA: 147] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Accepted: 05/15/2017] [Indexed: 12/20/2022]
Abstract
It is well known that the microenvironment of solid tumors is rich in inflammatory cells that influence tumor growth and development. Macrophages, called tumor-associated macrophages (TAMs), are the most abundant immune cell population present in tumor tissue. Several studies have demonstrated that the density of TAMs is associated with a poor prognosis and positively correlates with tumor growth. Several studies have proved that TAMs may activate and protect tumor stem cells, stimulate their proliferation as well as promote angiogenesis and metastasis. Furthermore, TAMs-derived cytokines and other proteins, such as CCL-17, CCL-22, TGF-β, IL-10, arginase 1, and galectin-3, make a significant contribution to immunosuppression. Since TAMs influence various aspects of cancer progression, there are many attempts to use them as a target for immunotherapy. The numerous studies have shown that the primary tumor growth and the number of metastatic sites can be significantly decreased by decreasing the population of macrophages in tumor tissue, for example, by blocking recruitment of monocytes or eliminating TAMs already present in the tumor tissue. Moreover, there are attempts at reprogramming TAMs into proinflammatory M1 macrophages or neutralizing the protumoral products of TAMs. Another approach uses TAMs for anticancer drug delivery into the tumor environment. In this review, we would like to summarize the clinical and preclinical trials that were focused on macrophages as a target for anticancer therapies.
Collapse
Affiliation(s)
- Katarzyna Sawa-Wejksza
- Department of Virology and Immunology, Institute of Microbiology and Biotechnology, Maria Curie-Skłodowska University, Akademicka 19, 20-033, Lublin, Poland.
| | - Martyna Kandefer-Szerszeń
- Department of Virology and Immunology, Institute of Microbiology and Biotechnology, Maria Curie-Skłodowska University, Akademicka 19, 20-033, Lublin, Poland
| |
Collapse
|
50
|
King J, Mir H, Singh S. Association of Cytokines and Chemokines in Pathogenesis of Breast Cancer. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2017; 151:113-136. [DOI: 10.1016/bs.pmbts.2017.07.003] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|