1
|
Seyfoori A, Liu K, Caruncho HJ, Walter PB, Akbari M. Tumoroid-On-a-Plate (ToP): Physiologically Relevant Cancer Model Generation and Therapeutic Screening. Adv Healthc Mater 2025; 14:e2402060. [PMID: 39538973 DOI: 10.1002/adhm.202402060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 10/07/2024] [Indexed: 11/16/2024]
Abstract
Employing three-dimensional (3D) in vitro models, including tumor organoids and spheroids, stands pivotal in enhancing cancer therapy. These models bridge the gap between two-dimensional (2D) cell cultures and complex in vivo environments and offer versatile tools for comprehensive studies into cancer progression, drug responses, and tailored therapies. This study introduces the Tumoroid-on-a-Plate (ToP) device, an innovative ope-surface microfluidic platform designed to create predictive 3D models of solid tumors. The ToP device combines tumor mass, stromal cells, and extracellular matrix (ECM) components, to closely replicate the microenvironment of glioblastoma (GBM) and pancreatic adenocarcinoma (PDAC). Using the advanced ToP model and testing various GBM ECM compositions such as collagen and Rreelin within the model, we can assess how specific elements affect GBM invasiveness. The ToP in vitro model also enables screening chemotherapeutics such as temozolomide and iron-chelators in a single and binary treatment setting on the complex ECM-embedded tumoroids to evaluate their toxicity on GBM and PDAC models viability and apoptosis. Furthermore, co-culturing PDAC tumoroids with human-derived fibroblasts reveals the pro-invasive influence of stromal elements on tumor growth and drug response. This research underscores the value of advanced 3D models like ToP in advancing the understanding of cancer complexity and therapy responses.
Collapse
Affiliation(s)
- Amir Seyfoori
- Department of Mechanical Engineering, University of Victoria, Victoria, BC, V8P 5C2, Canada
- Laboratory for Innovations in Micro Engineering (LiME), Department of Mechanical Engineering, University of Victoria, Victoria, BC, V8P 5C2, Canada
- Apricell Biotechnology Inc., Victoria, BC, V8P 1T5, Canada
| | - Kaiwen Liu
- Laboratory for Innovations in Micro Engineering (LiME), Department of Mechanical Engineering, University of Victoria, Victoria, BC, V8P 5C2, Canada
- Apricell Biotechnology Inc., Victoria, BC, V8P 1T5, Canada
| | - Hector J Caruncho
- Division of Medical Sciences, University of Victoria, Victoria, BC, V8P 5C4, Canada
| | - Patrick B Walter
- Department of Biology, University of Victoria, Victoria, BC, V8P 5C2, Canada
| | - Mohsen Akbari
- Department of Mechanical Engineering, University of Victoria, Victoria, BC, V8P 5C2, Canada
- Laboratory for Innovations in Micro Engineering (LiME), Department of Mechanical Engineering, University of Victoria, Victoria, BC, V8P 5C2, Canada
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA, 90064, USA
| |
Collapse
|
2
|
Nazari H, Cho AN, Goss D, Thiery JP, Ebrahimi Warkiani M. Impact of brain organoid-derived sEVs on metastatic adaptation and invasion of breast carcinoma cells through a microphysiological system. LAB ON A CHIP 2024; 24:3434-3455. [PMID: 38888211 DOI: 10.1039/d4lc00296b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2024]
Abstract
Brain metastases are common in triple-negative breast cancer (TNBC), suggesting a complex process of cancer spread. The mechanisms enabling TNBC cell adaptation and proliferation in the brain remain unclear. Small extracellular vesicles (sEVs) play a crucial role in communication between breast carcinoma cells and the brain. However, the lack of relevant models hinders understanding of sEV-mediated communication. The present study assesses the impact of brain organoid-derived sEVs (BO-sEVs) on various behaviours of the MDA-MB-231 cell line, chosen as a representative of TNBC in a 3D microfluidic model. Our results demonstrate that 150-200 nm sEVs expressing CD63, CD9, and CD81 from brain organoid media decrease MDA-MB-231 cell proliferation, enhance their wound-healing capacity, alter their morphology into more mesenchymal mode, and increase their stemness. BO-sEVs led to heightened PD-L1, CD49f, and vimentin levels of expression in MDA-MB-231 cells, suggesting an amplified immunosuppressive, stem-like, and mesenchymal phenotype. Furthermore, these sEVs also induced the expression of neural markers such as GFAP in carcinoma cells. The cytokine antibody profiling array also showed that BO-sEVs enhanced the secretion of MCP-1, IL-6, and IL-8 by MDA-MB-231 cells. Moreover, sEVs significantly enhance the migration and invasion of carcinoma cells toward brain organoids in a 3D organoid-on-a-chip system. Our findings emphasize the potential significance of metastatic site-derived sEVs as pivotal mediators in carcinoma progression and adaptation to the brain microenvironment, thereby unveiling novel therapeutic avenues.
Collapse
Affiliation(s)
- Hojjatollah Nazari
- School of Biomedical Engineering, Faculty of Engineering and IT, University of Technology Sydney, Sydney, NSW, Australia.
| | - Ann-Na Cho
- School of Biomedical Engineering, Faculty of Engineering, The University of Sydney, Sydney, NSW, Australia
| | - Dale Goss
- School of Biomedical Engineering, Faculty of Engineering and IT, University of Technology Sydney, Sydney, NSW, Australia.
| | - Jean Paul Thiery
- UMR 7057 CNRS Matter and Complex Systems, Université Paris Cité, Paris, France
| | - Majid Ebrahimi Warkiani
- School of Biomedical Engineering, Faculty of Engineering and IT, University of Technology Sydney, Sydney, NSW, Australia.
| |
Collapse
|
3
|
Wang C, Nagayach A, Patel H, Dao L, Zhu H, Wasylishen AR, Fan Y, Kendler A, Guo Z. Utilizing human cerebral organoids to model breast cancer brain metastasis in culture. Breast Cancer Res 2024; 26:108. [PMID: 38951862 PMCID: PMC11218086 DOI: 10.1186/s13058-024-01865-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 06/25/2024] [Indexed: 07/03/2024] Open
Abstract
BACKGROUND Metastasis, the spread, and growth of malignant cells at secondary sites within a patient's body, accounts for over 90% of cancer-related mortality. Breast cancer is the most common tumor type diagnosed and the leading cause of cancer lethality in women in the United States. It is estimated that 10-16% breast cancer patients will have brain metastasis. Current therapies to treat patients with breast cancer brain metastasis (BCBM) remain palliative. This is largely due to our limited understanding of the fundamental molecular and cellular mechanisms through which BCBM progresses, which represents a critical barrier for the development of efficient therapies for affected breast cancer patients. METHODS Previous research in BCBM relied on co-culture assays of tumor cells with rodent neural cells or rodent brain slice ex vivo. Given the need to overcome the obstacle for human-relevant host to study cell-cell communication in BCBM, we generated human embryonic stem cell-derived cerebral organoids to co-culture with human breast cancer cell lines. We used MDA-MB-231 and its brain metastatic derivate MDA-MB-231 Br-EGFP, other cell lines of MCF-7, HCC-1806, and SUM159PT. We leveraged this novel 3D co-culture platform to investigate the crosstalk of human breast cancer cells with neural cells in cerebral organoid. RESULTS We found that MDA-MB-231 and SUM159PT breast cancer cells formed tumor colonies in human cerebral organoids. Moreover, MDA-MB-231 Br-EGFP cells showed increased capacity to invade and expand in human cerebral organoids. CONCLUSIONS Our co-culture model has demonstrated a remarkable capacity to discern the brain metastatic ability of human breast cancer cells in cerebral organoids. The generation of BCBM-like structures in organoid will facilitate the study of human tumor microenvironment in culture.
Collapse
Affiliation(s)
- Chenran Wang
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA.
| | - Aarti Nagayach
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA
| | - Harsh Patel
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA
| | - Lan Dao
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Hui Zhu
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Amanda R Wasylishen
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA
| | - Yanbo Fan
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA
| | - Ady Kendler
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA
| | - Ziyuan Guo
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA.
| |
Collapse
|
4
|
Xu D, Hu Z, Wang K, Hu S, Zhou Y, Zhang S, Chen Y, Pan T. Why does HER2-positive breast cancer metastasize to the brain and what can we do about it? Crit Rev Oncol Hematol 2024; 195:104269. [PMID: 38272149 DOI: 10.1016/j.critrevonc.2024.104269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 12/18/2023] [Accepted: 01/19/2024] [Indexed: 01/27/2024] Open
Abstract
Breast cancer is the most frequent malignancy in women. However, in the middle and late stages, some people develop distant metastases, which considerably lower the quality of life and life expectancy. The brain is one of the sites where metastasis frequently happens. According to epidemiological research, brain metastases occur at a late stage in 30-50% of patients with HER2-positive breast cancer, resulting in a poor prognosis. Additionally, few treatments are available for HER2-positive brain metastatic breast cancer, and the mortality rate is remarkable owing to the complexity of the brain's anatomical structure and physiological function. In this review, we described the stages of the brain metastasis of breast cancer, the relationship between the microenvironment and metastatic cancer cells, and the unique molecular and cellular mechanisms. It involves cancer cells migrating, invading, and adhering to the brain; penetrating the blood-brain barrier; interacting with brain cells; and activating signal pathways once inside the brain. Finally, we reviewed current clinically used treatment approaches for brain metastasis in HER2-positive breast cancer; summarized the traditional treatment, targeted treatment, immunotherapy, and other treatment modalities; compared the benefits and drawbacks of each approach; discussed treatment challenges; and emphasized the importance of identifying potential targets to improve patient survival rates and comprehend brain metastasis in breast cancer.
Collapse
Affiliation(s)
- Dongyan Xu
- Department of Breast Surgery and Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China; Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Zhengfang Hu
- Beijing Tian Tan Hospital, Capital Medical University, Beijing 100050, China
| | - Kaiyue Wang
- Department of Breast Surgery and Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China; Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Shiyao Hu
- Department of Breast Surgery and Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China; Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Yunxiang Zhou
- Department of Breast Surgery and Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China; Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Shizhen Zhang
- Department of Breast Surgery and Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China; Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Yiding Chen
- Department of Breast Surgery and Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China; Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Tao Pan
- Department of Breast Surgery and Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China; Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China.
| |
Collapse
|
5
|
Feng Y, Hu X, Zhang Y, Wang Y. The Role of Microglia in Brain Metastases: Mechanisms and Strategies. Aging Dis 2024; 15:169-185. [PMID: 37307835 PMCID: PMC10796095 DOI: 10.14336/ad.2023.0514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Accepted: 05/14/2023] [Indexed: 06/14/2023] Open
Abstract
Brain metastases and related complications are one of the major fatal factors in cancer. Patients with breast cancer, lung cancer, and melanoma are at a high risk of developing brain metastases. However, the mechanisms underlying the brain metastatic cascade remain poorly understood. Microglia, one of the major resident macrophages in the brain parenchyma, are involved in multiple processes associated with brain metastasis, including inflammation, angiogenesis, and immune modulation. They also closely interact with metastatic cancer cells, astrocytes, and other immune cells. Current therapeutic approaches against metastatic brain cancers, including small-molecule drugs, antibody-coupled drugs (ADCs), and immune-checkpoint inhibitors (ICIs), have compromised efficacy owing to the impermeability of the blood-brain barrier (BBB) and complex brain microenvironment. Targeting microglia is one of the strategies for treating metastatic brain cancer. In this review, we summarize the multifaceted roles of microglia in brain metastases and highlight them as potential targets for future therapeutic interventions.
Collapse
Affiliation(s)
- Ying Feng
- Department of Medical Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xueqing Hu
- Department of Medical Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yingru Zhang
- Department of Medical Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yan Wang
- Department of Medical Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| |
Collapse
|
6
|
Kurup S, Tan C, Kume T. Cardiac and intestinal tissue conduct developmental and reparative processes in response to lymphangiocrine signaling. Front Cell Dev Biol 2023; 11:1329770. [PMID: 38178871 PMCID: PMC10764504 DOI: 10.3389/fcell.2023.1329770] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Accepted: 12/08/2023] [Indexed: 01/06/2024] Open
Abstract
Lymphatic vessels conduct a diverse range of activities to sustain the integrity of surrounding tissue. Besides facilitating the movement of lymph and its associated factors, lymphatic vessels are capable of producing tissue-specific responses to changes within their microenvironment. Lymphatic endothelial cells (LECs) secrete paracrine signals that bind to neighboring cell-receptors, commencing an intracellular signaling cascade that preludes modifications to the organ tissue's structure and function. While the lymphangiocrine factors and the molecular and cellular mechanisms themselves are specific to the organ tissue, the crosstalk action between LECs and adjacent cells has been highlighted as a commonality in augmenting tissue regeneration within animal models of cardiac and intestinal disease. Lymphangiocrine secretions have been owed for subsequent improvements in organ function by optimizing the clearance of excess tissue fluid and immune cells and stimulating favorable tissue growth, whereas perturbations in lymphatic performance bring about the opposite. Newly published landmark studies have filled gaps in our understanding of cardiac and intestinal maintenance by revealing key players for lymphangiocrine processes. Here, we will expand upon those findings and review the nature of lymphangiocrine factors in the heart and intestine, emphasizing its involvement within an interconnected network that supports daily homeostasis and self-renewal following injury.
Collapse
Affiliation(s)
- Shreya Kurup
- Department of Medicine, Feinberg Cardiovascular and Renal Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
- Honors College, University of Illinois at Chicago, Chicago, IL, United States
| | - Can Tan
- Department of Medicine, Feinberg Cardiovascular and Renal Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Tsutomu Kume
- Department of Medicine, Feinberg Cardiovascular and Renal Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| |
Collapse
|
7
|
Nolan E, Kang Y, Malanchi I. Mechanisms of Organ-Specific Metastasis of Breast Cancer. Cold Spring Harb Perspect Med 2023; 13:a041326. [PMID: 36987584 PMCID: PMC10626265 DOI: 10.1101/cshperspect.a041326] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2023]
Abstract
Cancer metastasis, or the development of secondary tumors in distant tissues, accounts for the vast majority of fatalities in patients with breast cancer. Breast cancer cells show a striking proclivity to metastasize to distinct organs, specifically the lung, liver, bone, and brain, where they face unique environmental pressures and a wide variety of tissue-resident cells that together create a strong barrier for tumor survival and growth. As a consequence, successful metastatic colonization is critically dependent on reciprocal cross talk between cancer cells and host cells within the target organ, a relationship that shapes the formation of a tumor-supportive microenvironment. Here, we discuss the mechanisms governing organ-specific metastasis in breast cancer, focusing on the intricate interactions between metastatic cells and specific niche cells within a secondary organ, and the remarkable adaptations of both compartments that cooperatively support cancer growth. More broadly, we aim to provide a framework for the microenvironmental prerequisites within each distinct metastatic site for successful breast cancer metastatic seeding and outgrowth.
Collapse
Affiliation(s)
- Emma Nolan
- Tumour Host Interaction laboratory, The Francis Crick Institute, NW1 1AT London, United Kingdom
- Auckland Cancer Society Research Centre, University of Auckland, Auckland 1023, New Zealand
| | - Yibin Kang
- Department of Molecular Biology, Princeton University, Princeton, New Jersey 08544, USA
- Ludwig Institute for Cancer Research Princeton Branch, Princeton, New Jersey 08544, USA
| | - Ilaria Malanchi
- Tumour Host Interaction laboratory, The Francis Crick Institute, NW1 1AT London, United Kingdom
| |
Collapse
|
8
|
Qu F, Brough SC, Michno W, Madubata CJ, Hartmann GG, Puno A, Drainas AP, Bhattacharya D, Tomasich E, Lee MC, Yang D, Kim J, Peiris-Pagès M, Simpson KL, Dive C, Preusser M, Toland A, Kong C, Das M, Winslow MM, Pasca AM, Sage J. Crosstalk between small-cell lung cancer cells and astrocytes mimics brain development to promote brain metastasis. Nat Cell Biol 2023; 25:1506-1519. [PMID: 37783795 PMCID: PMC11230587 DOI: 10.1038/s41556-023-01241-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 08/25/2023] [Indexed: 10/04/2023]
Abstract
Brain metastases represent an important clinical problem for patients with small-cell lung cancer (SCLC). However, the mechanisms underlying SCLC growth in the brain remain poorly understood. Here, using intracranial injections in mice and assembloids between SCLC aggregates and human cortical organoids in culture, we found that SCLC cells recruit reactive astrocytes to the tumour microenvironment. This crosstalk between SCLC cells and astrocytes drives the induction of gene expression programmes that are similar to those found during early brain development in neurons and astrocytes. Mechanistically, the brain development factor Reelin, secreted by SCLC cells, recruits astrocytes to brain metastases. These astrocytes in turn promote SCLC growth by secreting neuronal pro-survival factors such as SERPINE1. Thus, SCLC brain metastases grow by co-opting mechanisms involved in reciprocal neuron-astrocyte interactions during brain development. Targeting such developmental programmes activated in this cancer ecosystem may help prevent and treat brain metastases.
Collapse
Affiliation(s)
- Fangfei Qu
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Siqi C Brough
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Wojciech Michno
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Chioma J Madubata
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Griffin G Hartmann
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Alyssa Puno
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Alexandros P Drainas
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Debadrita Bhattacharya
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Erwin Tomasich
- Department of Medicine I, Division of Oncology, Medical University of Vienna, Vienna, Austria
| | - Myung Chang Lee
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Dian Yang
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Jun Kim
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Maria Peiris-Pagès
- Cancer Research UK Cancer Biomarker Centre, Manchester, UK
- Cancer Research UK Manchester Institute, Manchester, UK
| | - Kathryn L Simpson
- Cancer Research UK Cancer Biomarker Centre, Manchester, UK
- Cancer Research UK Manchester Institute, Manchester, UK
| | - Caroline Dive
- Cancer Research UK Cancer Biomarker Centre, Manchester, UK
- Cancer Research UK Manchester Institute, Manchester, UK
| | - Matthias Preusser
- Department of Medicine I, Division of Oncology, Medical University of Vienna, Vienna, Austria
| | - Angus Toland
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Christina Kong
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Millie Das
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Medicine, VA Palo Alto Health Care System, Palo Alto, CA, USA
| | - Monte M Winslow
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Anca M Pasca
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Julien Sage
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA.
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
9
|
Yuzhalin AE, Yu D. Critical functions of extracellular matrix in brain metastasis seeding. Cell Mol Life Sci 2023; 80:297. [PMID: 37728789 PMCID: PMC10511571 DOI: 10.1007/s00018-023-04944-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Revised: 08/16/2023] [Accepted: 08/29/2023] [Indexed: 09/21/2023]
Abstract
Human brain is characterized by extremely sparse extracellular matrix (ECM). Despite its low abundance, the significance of brain ECM in both physiological and pathological conditions should not be underestimated. Brain metastasis is a serious complication of cancer, and recent findings highlighted the contribution of ECM in brain metastasis development. In this review, we provide a comprehensive outlook on how ECM proteins promote brain metastasis seeding. In particular, we discuss (1) disruption of the blood-brain barrier in brain metastasis; (2) role of ECM in modulating brain metastasis dormancy; (3) regulation of brain metastasis seeding by ECM-activated integrin signaling; (4) functions of brain-specific ECM protein reelin in brain metastasis. Lastly, we consider the possibility of targeting ECM for brain metastasis management.
Collapse
Affiliation(s)
- Arseniy E Yuzhalin
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, 6565 MD Anderson Blvd, Unit 108, Houston, TX, 77030, USA
| | - Dihua Yu
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, 6565 MD Anderson Blvd, Unit 108, Houston, TX, 77030, USA.
| |
Collapse
|
10
|
Ma W, Oliveira-Nunes MC, Xu K, Kossenkov A, Reiner BC, Crist RC, Hayden J, Chen Q. Type I interferon response in astrocytes promotes brain metastasis by enhancing monocytic myeloid cell recruitment. Nat Commun 2023; 14:2632. [PMID: 37149684 PMCID: PMC10163863 DOI: 10.1038/s41467-023-38252-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 04/20/2023] [Indexed: 05/08/2023] Open
Abstract
Cancer metastasis to the brain is a significant clinical problem. Metastasis is the consequence of favorable interactions between invaded cancer cells and the microenvironment. Here, we demonstrate that cancer-activated astrocytes create a sustained low-level activated type I interferon (IFN) microenvironment in brain metastatic lesions. We further confirm that the IFN response in astrocytes facilitates brain metastasis. Mechanistically, IFN signaling in astrocytes activates C-C Motif Chemokine Ligand 2 (CCL2) production, which further increases the recruitment of monocytic myeloid cells. The correlation between CCL2 and monocytic myeloid cells is confirmed in clinical brain metastasis samples. Lastly, genetically or pharmacologically inhibiting C-C Motif Chemokine Receptor 2 (CCR2) reduces brain metastases. Our study clarifies a pro-metastatic effect of type I IFN in the brain even though IFN response has been considered to have anti-tumor effects. Moreover, this work expands our understandings on the interactions between cancer-activated astrocytes and immune cells in brain metastasis.
Collapse
Affiliation(s)
- Weili Ma
- Immunology, Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, PA, 19104, USA
| | - Maria Cecília Oliveira-Nunes
- Immunology, Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, PA, 19104, USA
- Carisma Therapeutics, Philadelphia, PA, 19104, USA
| | - Ke Xu
- MD/PhD Program, Boston University School of Medicine, Boston, MA, 02215, USA
| | - Andrew Kossenkov
- Gene Expression & Regulation Program, The Wistar Institute, Philadelphia, PA, 19104, USA
| | - Benjamin C Reiner
- Department of Psychiatry, Perelman School of Medicine, The University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Richard C Crist
- Department of Psychiatry, Perelman School of Medicine, The University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - James Hayden
- Imaging Shared Resource, The Wistar Institute, Philadelphia, PA, 19104, USA
| | - Qing Chen
- Immunology, Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, PA, 19104, USA.
| |
Collapse
|
11
|
Deshpande K, Martirosian V, Nakamura BN, Iyer M, Julian A, Eisenbarth R, Shao L, Attenello F, Neman J. Neuronal exposure induces neurotransmitter signaling and synaptic mediators in tumors early in brain metastasis. Neuro Oncol 2021; 24:914-924. [PMID: 34932815 DOI: 10.1093/neuonc/noab290] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Brain metastases (BM) are responsible for neurological decline and poor overall survival. Although the pro-metastatic roles of glial cells, and the acquisition of neuronal attributes in established BM tumors have been described, there are no studies that investigate the initial interplay between neurons and brain-seeking tumor cells. The aim of this study was to characterize early tumor-neuron interactions and the induced CNS-adaptive changes in tumor cells prior to macro-colonization. METHODS Utilizing pure neuronal cultures and brain-naïve and patient-derived BM tumor cells, we surveyed the early induction of mediators of neurotransmitter (NT) and synaptic signaling in breast and lung tumor cells. Reliance on microenvironmental GABA in breast-to-brain metastatic cells (BBMs) was assessed in vitro and in vivo. RESULTS Co-culture with neurons induces early expression of classical NT receptor genes (HTR4, GRIA2, GRIN2B, GRM4, GRM8, DRD1) and neuronal synaptic mediators (CNR1, EGR2, ARC, NGFR, NRXN1) in breast and lung cancer cells. NT-dependent classification of tumor cells within the neuronal niche shows breast cancer cells become GABAergic responsive brain metastases (GRBMs) and transition from relying on autocrine GABA, to paracrine GABA from adjacent neurons; while autocrine Dopaminergic breast and lung tumor cells persist. In vivo studies confirm reliance on paracrine GABA is an early CNS-acclimation strategy in breast cancer. Moreover, neuronal contact induces early resurgence in Reelin expression in tumor cells through epigenetic activation, facilitating CNS adaptation. CONCLUSION Tumor-neuron interactions allow for CNS-adaptation early in the course of brain metastasis.
Collapse
Affiliation(s)
- Krutika Deshpande
- Department of Neurological Surgery, University of Southern California, Los Angeles, CA, USA.,USC Brain Tumor Center, University of Southern California, Los Angeles, CA, USA.,Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Vahan Martirosian
- Department of Neurological Surgery, University of Southern California, Los Angeles, CA, USA.,USC Brain Tumor Center, University of Southern California, Los Angeles, CA, USA.,Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Brooke Naomi Nakamura
- Department of Medicine, Division of Gastrointestinal and Liver Diseases, University of Southern California, Los Angeles, CA, USA.,USC Brain Tumor Center, University of Southern California, Los Angeles, CA, USA
| | - Mukund Iyer
- Department of Neurological Surgery, University of Southern California, Los Angeles, CA, USA.,USC Brain Tumor Center, University of Southern California, Los Angeles, CA, USA.,Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Alex Julian
- Department of Neurological Surgery, University of Southern California, Los Angeles, CA, USA.,USC Brain Tumor Center, University of Southern California, Los Angeles, CA, USA.,Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Rachel Eisenbarth
- Department of Neurological Surgery, University of Southern California, Los Angeles, CA, USA.,USC Brain Tumor Center, University of Southern California, Los Angeles, CA, USA.,Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Ling Shao
- Department of Medicine, Division of Gastrointestinal and Liver Diseases, University of Southern California, Los Angeles, CA, USA.,Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Frank Attenello
- Department of Neurological Surgery, University of Southern California, Los Angeles, CA, USA.,USC Brain Tumor Center, University of Southern California, Los Angeles, CA, USA.,Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Josh Neman
- Department of Neurological Surgery, University of Southern California, Los Angeles, CA, USA.,USC Brain Tumor Center, University of Southern California, Los Angeles, CA, USA.,Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
12
|
Bhan A, Ansari K, Chen MY, Jandial R. Human induced pluripotent stem cell-derived platelets loaded with lapatinib effectively target HER2+ breast cancer metastasis to the brain. Sci Rep 2021; 11:16866. [PMID: 34654856 PMCID: PMC8521584 DOI: 10.1038/s41598-021-96351-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Accepted: 08/06/2021] [Indexed: 01/09/2023] Open
Abstract
Prognosis of patients with HER2+ breast-to-brain-metastasis (BBM) is dismal even after current standard-of-care treatments, including surgical resection, whole-brain radiation, and systemic chemotherapy. Radiation and systemic chemotherapies can also induce cytotoxicity, leading to significant side effects. Studies indicate that donor-derived platelets can serve as immune-compatible drug carriers that interact with and deliver drugs to cancer cells with fewer side effects, making them a promising therapeutic option with enhanced antitumor activity. Moreover, human induced pluripotent stem cells (hiPSCs) provide a potentially renewable source of clinical-grade transfusable platelets that can be drug-loaded to complement the supply of donor-derived platelets. Here, we describe methods for ex vivo generation of megakaryocytes (MKs) and functional platelets from hiPSCs (hiPSC-platelets) in a scalable fashion. We then loaded hiPSC-platelets with lapatinib and infused them into BBM tumor-bearing NOD/SCID mouse models. Such treatment significantly increased intracellular lapatinib accumulation in BBMs in vivo, potentially via tumor cell-induced activation/aggregation. Lapatinib-loaded hiPSC-platelets exhibited normal morphology and function and released lapatinib pH-dependently. Importantly, lapatinib delivery to BBM cells via hiPSC-platelets inhibited tumor growth and prolonged survival of tumor-bearing mice. Overall, use of lapatinib-loaded hiPSC-platelets effectively reduced adverse effects of free lapatinib and enhanced its therapeutic efficacy, suggesting that they represent a novel means to deliver chemotherapeutic drugs as treatment for BBM.
Collapse
Affiliation(s)
- Arunoday Bhan
- Division of Neurosurgery, Beckman Research Institute, City of Hope Medical Center, 1500 E. Duarte Rd, Duarte, CA, 91010, USA.
| | - Khairul Ansari
- Division of Neurosurgery, Beckman Research Institute, City of Hope Medical Center, 1500 E. Duarte Rd, Duarte, CA, 91010, USA
- Celcuity LLC, Minneapolis, MN, 55446, USA
| | - Mike Y Chen
- Division of Neurosurgery, Beckman Research Institute, City of Hope Medical Center, 1500 E. Duarte Rd, Duarte, CA, 91010, USA
| | - Rahul Jandial
- Division of Neurosurgery, Beckman Research Institute, City of Hope Medical Center, 1500 E. Duarte Rd, Duarte, CA, 91010, USA
| |
Collapse
|
13
|
Joe NS, Hodgdon C, Kraemer L, Redmond KJ, Stearns V, Gilkes DM. A common goal to CARE: Cancer Advocates, Researchers, and Clinicians Explore current treatments and clinical trials for breast cancer brain metastases. NPJ Breast Cancer 2021; 7:121. [PMID: 34521857 PMCID: PMC8440644 DOI: 10.1038/s41523-021-00326-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Accepted: 08/09/2021] [Indexed: 02/08/2023] Open
Abstract
Breast cancer is the most commonly diagnosed cancer in women worldwide. Approximately one-tenth of all patients with advanced breast cancer develop brain metastases resulting in an overall survival rate of fewer than 2 years. The challenges lie in developing new approaches to treat, monitor, and prevent breast cancer brain metastasis (BCBM). This review will provide an overview of BCBM from the integrated perspective of clinicians, researchers, and patient advocates. We will summarize the current management of BCBM, including diagnosis, treatment, and monitoring. We will highlight ongoing translational research for BCBM, including clinical trials and improved detection methods that can become the mainstay for BCBM treatment if they demonstrate efficacy. We will discuss preclinical BCBM research that focuses on the intrinsic properties of breast cancer cells and the influence of the brain microenvironment. Finally, we will spotlight emerging studies and future research needs to improve survival outcomes and preserve the quality of life for patients with BCBM.
Collapse
Affiliation(s)
- Natalie S Joe
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Cellular and Molecular Medicine Program, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Christine Hodgdon
- INSPIRE (Influencing Science through Patient-Informed Research & Education) Advocacy Program, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - Kristin J Redmond
- Department of Radiation Oncology and Molecular Radiation Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Vered Stearns
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- INSPIRE (Influencing Science through Patient-Informed Research & Education) Advocacy Program, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Daniele M Gilkes
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Cellular and Molecular Medicine Program, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- INSPIRE (Influencing Science through Patient-Informed Research & Education) Advocacy Program, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
14
|
Ansari KI, Bhan A, Saotome M, Tyagi A, De Kumar B, Chen C, Takaku M, Jandial R. Autocrine GM-CSF signaling contributes to growth of HER2+ breast leptomeningeal carcinomatosis. Cancer Res 2021; 81:4723-4735. [PMID: 34247146 DOI: 10.1158/0008-5472.can-21-0259] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 05/29/2021] [Accepted: 07/08/2021] [Indexed: 11/16/2022]
Abstract
Leptomeningeal carcinomatosis (LC) occurs when tumor cells spread to the cerebrospinal fluid-containing leptomeninges surrounding the brain and spinal cord. LC is an ominous complication of cancer with a dire prognosis. Although any malignancy can spread to the leptomeninges, breast cancer, particularly the HER2+ subtype, is its most common origin. HER2+ breast LC (HER2+ LC) remains incurable, with few treatment options, and the molecular mechanisms underlying proliferation of HER2+ breast cancer cells in the acellular, protein, and cytokine-poor leptomeningeal environment remain elusive. Therefore, we sought to characterize signaling pathways that drive HER2+ LC development as well as those that restrict its growth to leptomeninges. Primary HER2+ LC patient-derived ("Lepto") cell lines in co-culture with various central nervous system (CNS) cell types revealed that oligodendrocyte progenitor cells (OPC), the largest population of dividing cells in the CNS, inhibited HER2+ LC growth in vitro and in vivo, thereby limiting the spread of HER2+ LC beyond the leptomeninges. Cytokine array-based analyses identified Lepto cell-secreted granulocyte-macrophage colony-stimulating factor (GM-CSF) as an oncogenic autocrine driver of HER2+ LC growth. Liquid chromatography-tandem mass spectrometry-based analyses revealed that the OPC-derived protein TPP1 proteolytically degrades GM-CSF, decreasing GM-CSF signaling and leading to suppression of HER2+ LC growth and limiting its spread. Lastly, intrathecal delivery of neutralizing anti-GM-CSF antibodies and a pan-Aurora kinase inhibitor (CCT137690) synergistically inhibited GM-CSF and suppressed activity of GM-CSF effectors, reducing HER2+ LC growth in vivo. Thus, OPC suppress GM-CSF-driven growth of HER2+ LC in the leptomeningeal environment, providing a potential targetable axis.
Collapse
|
15
|
Oliveira-Barros EGD, Branco LC, Da Costa NM, Nicolau-Neto P, Palmero C, Pontes B, Ferreira do Amaral R, Alves-Leon SV, Marcondes de Souza J, Romão L, Fernandes PV, Martins I, Takiya CM, Ribeiro Pinto LF, Palumbo A, Nasciutti LE. GLIPR1 and SPARC expression profile reveals a signature associated with prostate Cancer Brain metastasis. Mol Cell Endocrinol 2021; 528:111230. [PMID: 33675864 DOI: 10.1016/j.mce.2021.111230] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 02/23/2021] [Accepted: 02/24/2021] [Indexed: 02/07/2023]
Abstract
Despite advances in treatment of lethal prostate cancer, the incidence of prostate cancer brain metastases is increasing. In this sense, we analyzed the molecular profile, as well as the functional consequences involved in the reciprocal interactions between prostate tumor cells and human astrocytes. We observed that the DU145 cells, but not the LNCaP cells or the RWPE-1 cells, exhibited more pronounced, malignant and invasive phenotypes along their interactions with astrocytes. Moreover, global gene expression analysis revealed several genes that were differently expressed in our co-culture models with the overexpression of GLIPR1 and SPARC potentially representing a molecular signature associated with the invasion of central nervous system by prostate malignant cells. Further, these results were corroborated by immunohistochemistry and in silico analysis. Thus, we conjecture that the data here presented may increase the knowledge about the molecular mechanisms associated with the invasion of CNS by prostate malignant cells.
Collapse
Affiliation(s)
- Eliane Gouvêa de Oliveira-Barros
- Programa de Pesquisa Em Biologia Celular e do Desenvolvimento, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro (UFRJ), Cidade Universitária-Ilha do Fundão, Rio de Janeiro, CEP 21941-902, Brazil; Laboratório de Biologia Celular, Departamento de Biologia, Instituto de Ciências Biológicas, Universidade Federal de Juiz de Fora (UFJF), Rua José Lourenço Kelmer-Campus, São Pedro, Juiz de Fora, CEP: 36036-900, Brazil.
| | - Luíza Castello Branco
- Programa de Pesquisa Em Biologia Celular e do Desenvolvimento, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro (UFRJ), Cidade Universitária-Ilha do Fundão, Rio de Janeiro, CEP 21941-902, Brazil.
| | - Nathalia Meireles Da Costa
- Programa de Carcinogênese Molecular, Centro de Pesquisas, Instituto Nacional de Câncer (INCA), Rua André Cavalcanti, 37-Centro, Rio de Janeiro, CEP 20231-050, Brazil.
| | - Pedro Nicolau-Neto
- Programa de Carcinogênese Molecular, Centro de Pesquisas, Instituto Nacional de Câncer (INCA), Rua André Cavalcanti, 37-Centro, Rio de Janeiro, CEP 20231-050, Brazil.
| | - Celia Palmero
- Programa de Pesquisa Em Biologia Celular e do Desenvolvimento, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro (UFRJ), Cidade Universitária-Ilha do Fundão, Rio de Janeiro, CEP 21941-902, Brazil; UFRJ/Polo Macaé, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.
| | - Bruno Pontes
- Programa de Pesquisa Em Biologia Celular e do Desenvolvimento, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro (UFRJ), Cidade Universitária-Ilha do Fundão, Rio de Janeiro, CEP 21941-902, Brazil; Centro Nacional de Biologia Estrutural e Bioimagem (CENABIO), Universidade Federal do Rio de Janeiro (UFRJ), Cidade Universitária-Ilha do Fundão, Rio de Janeiro, CEP 21941-902, Brazil.
| | - Rackele Ferreira do Amaral
- Programa de Pesquisa Em Biologia Celular e do Desenvolvimento, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro (UFRJ), Cidade Universitária-Ilha do Fundão, Rio de Janeiro, CEP 21941-902, Brazil.
| | - Soniza Vieira Alves-Leon
- Hospital Universitário Clementino Fraga Filho (HUCFF), Universidade Federal do Rio de Janeiro (UFRJ), Cidade Universitária-Ilha do Fundão, Rio de Janeiro, CEP 21941-902, Brazil.
| | - Jorge Marcondes de Souza
- Hospital Universitário Clementino Fraga Filho (HUCFF), Universidade Federal do Rio de Janeiro (UFRJ), Cidade Universitária-Ilha do Fundão, Rio de Janeiro, CEP 21941-902, Brazil.
| | - Luciana Romão
- Programa de Pesquisa Em Biologia Celular e do Desenvolvimento, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro (UFRJ), Cidade Universitária-Ilha do Fundão, Rio de Janeiro, CEP 21941-902, Brazil.
| | - Priscila Valverde Fernandes
- Divisão de Patologia, Instituto Nacional de Câncer (INCA), Rua Cordeiro da Graça, 156 - Santo Cristo, Rio de Janeiro, CEP: 20220 -040, Brazil.
| | - Ivanir Martins
- Divisão de Patologia, Instituto Nacional de Câncer (INCA), Rua Cordeiro da Graça, 156 - Santo Cristo, Rio de Janeiro, CEP: 20220 -040, Brazil.
| | - Christina Maeda Takiya
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro (UFRJ), Cidade Universitária-Ilha do Fundão, Rio de Janeiro, CEP 21941-902, Brazil.
| | - Luis Felipe Ribeiro Pinto
- Programa de Carcinogênese Molecular, Centro de Pesquisas, Instituto Nacional de Câncer (INCA), Rua André Cavalcanti, 37-Centro, Rio de Janeiro, CEP 20231-050, Brazil.
| | - Antonio Palumbo
- Programa de Pesquisa Em Biologia Celular e do Desenvolvimento, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro (UFRJ), Cidade Universitária-Ilha do Fundão, Rio de Janeiro, CEP 21941-902, Brazil.
| | - Luiz Eurico Nasciutti
- Programa de Pesquisa Em Biologia Celular e do Desenvolvimento, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro (UFRJ), Cidade Universitária-Ilha do Fundão, Rio de Janeiro, CEP 21941-902, Brazil.
| |
Collapse
|
16
|
Bhan A, Ansari KI, Chen MY, Jandial R. Inhibition of Jumonji Histone Demethylases Selectively Suppresses HER2 + Breast Leptomeningeal Carcinomatosis Growth via Inhibition of GMCSF Expression. Cancer Res 2021; 81:3200-3214. [PMID: 33941612 DOI: 10.1158/0008-5472.can-20-3317] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 03/17/2021] [Accepted: 04/28/2021] [Indexed: 11/16/2022]
Abstract
HER2+ breast leptomeningeal carcinomatosis (HER2+ LC) occurs when tumor cells spread to cerebrospinal fluid-containing leptomeninges surrounding the brain and spinal cord, a complication with a dire prognosis. HER2+ LC remains incurable, with few treatment options. Currently, much effort is devoted toward development of therapies that target mutations. However, targeting epigenetic or transcriptional states of HER2+ LC tumors might efficiently target HER2+ LC growth via inhibition of oncogenic signaling; this approach remains promising but is less explored. To test this possibility, we established primary HER2+ LC (Lepto) cell lines from nodular HER2+ LC tissues. These lines are phenotypically CD326+CD49f-, confirming that they are derived from HER2+ LC tumors, and express surface CD44+CD24-, a cancer stem cell (CSC) phenotype. Like CSCs, Lepto lines showed greater drug resistance and more aggressive behavior compared with other HER2+ breast cancer lines in vitro and in vivo. Interestingly, the three Lepto lines overexpressed Jumonji domain-containing histone lysine demethylases KDM4A/4C. Treatment with JIB04, a selective inhibitor of Jumonji demethylases, or genetic loss of function of KDM4A/4C induced apoptosis and cell-cycle arrest and reduced Lepto cell viability, tumorsphere formation, regrowth, and invasion in vitro. JIB04 treatment of patient-derived xenograft mouse models in vivo reduced HER2+ LC tumor growth and prolonged animal survival. Mechanistically, KDM4A/4C inhibition downregulated GMCSF expression and prevented GMCSF-dependent Lepto cell proliferation. Collectively, these results establish KDM4A/4C as a viable therapeutic target in HER2+ LC and spotlight the benefits of targeting the tumorigenic transcriptional network. SIGNIFICANCE: HER2+ LC tumors overexpress KDM4A/4C and are sensitive to the Jumonji demethylase inhibitor JIB04, which reduces the viability of primary HER2+ LC cells and increases survival in mouse models.
Collapse
Affiliation(s)
- Arunoday Bhan
- Division of Neurosurgery, Beckman Research Institute, City of Hope, Duarte, California
| | - Khairul I Ansari
- Division of Neurosurgery, Beckman Research Institute, City of Hope, Duarte, California.,Celcuity, Minneapolis, Minnesota
| | - Mike Y Chen
- Division of Neurosurgery, Beckman Research Institute, City of Hope, Duarte, California
| | - Rahul Jandial
- Division of Neurosurgery, Beckman Research Institute, City of Hope, Duarte, California.
| |
Collapse
|
17
|
Srinivasan ES, Tan AC, Anders CK, Pendergast AM, Sipkins DA, Ashley DM, Fecci PE, Khasraw M. Salting the Soil: Targeting the Microenvironment of Brain Metastases. Mol Cancer Ther 2021; 20:455-466. [PMID: 33402399 PMCID: PMC8041238 DOI: 10.1158/1535-7163.mct-20-0579] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 08/31/2020] [Accepted: 12/31/2020] [Indexed: 12/14/2022]
Abstract
Paget's "seed and soil" hypothesis of metastatic spread has acted as a foundation of the field for over a century, with continued evolution as mechanisms of the process have been elucidated. The central nervous system (CNS) presents a unique soil through this lens, relatively isolated from peripheral circulation and immune surveillance with distinct cellular and structural composition. Research in primary and metastatic brain tumors has demonstrated that this tumor microenvironment (TME) plays an essential role in the growth of CNS tumors. In each case, the cancerous cells develop complex and bidirectional relationships that reorganize the local TME and reprogram the CNS cells, including endothelial cells, pericytes, astrocytes, microglia, infiltrating monocytes, and lymphocytes. These interactions create a structurally and immunologically permissive TME with malignant processes promoting positive feedback loops and systemic consequences. Strategies to interrupt interactions with the native CNS components, on "salting the soil," to create an inhospitable environment are promising in the preclinical setting. This review aims to examine the general and specific pathways thus far investigated in brain metastases and related work in glioma to identify targetable mechanisms that may have general application across the spectrum of intracranial tumors.
Collapse
Affiliation(s)
- Ethan S Srinivasan
- Duke Brain and Spine Metastases Center, Duke University, Durham, North Carolina
| | - Aaron C Tan
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore
| | - Carey K Anders
- Duke Brain and Spine Metastases Center, Duke University, Durham, North Carolina
| | | | - Dorothy A Sipkins
- Duke Brain and Spine Metastases Center, Duke University, Durham, North Carolina
| | - David M Ashley
- Duke Brain and Spine Metastases Center, Duke University, Durham, North Carolina
| | - Peter E Fecci
- Duke Brain and Spine Metastases Center, Duke University, Durham, North Carolina
| | - Mustafa Khasraw
- Duke Brain and Spine Metastases Center, Duke University, Durham, North Carolina.
| |
Collapse
|
18
|
Ndoye A, Miskin RP, DiPersio CM. Integrin α3β1 Represses Reelin Expression in Breast Cancer Cells to Promote Invasion. Cancers (Basel) 2021; 13:cancers13020344. [PMID: 33477804 PMCID: PMC7832892 DOI: 10.3390/cancers13020344] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 01/13/2021] [Accepted: 01/15/2021] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Breast cancer remains the second leading cause of cancer-related deaths in women, and about 1 in 8 women in the United States develops invasive breast cancer in her lifetime. Integrin α3β1 has been linked to breast cancer progression, but mechanisms whereby it promotes tumor invasion remain unclear. The goal of our study was to determine how α3β1 drives invasion, towards exploiting this integrin as a therapeutic target for breast cancer. We found that α3β1 represses the expression of Reelin, a secreted glycoprotein that inhibits invasion and for which loss of expression is associated with poor prognosis in breast cancer. We also show that increased Reelin expression following RNAi-mediated suppression of α3β1 causes a significant decrease in breast cancer cell invasion. Our findings demonstrate a critical role for α3β1 in promoting cell invasion through repression of Reelin, highlighting the potential value of this integrin as a therapeutic target for breast cancer. Abstract Integrin α3β1, a cell adhesion receptor for certain laminins, is known to promote breast tumor growth and invasion. Our previous gene microarray study showed that the RELN gene, which encodes the extracellular glycoprotein Reelin, was upregulated in α3β1-deficient (i.e., α3 knockdown) MDA-MB-231 cells. In breast cancer, reduced RELN expression is associated with increased invasion and poor prognosis. In this study we demonstrate that α3β1 represses RELN expression to enhance breast cancer cell invasion. RELN mRNA was significantly increased upon RNAi-mediated α3 knockdown in two triple-negative breast cancer cell lines, MDA-MB-231 and SUM159. Modulation of baseline Reelin levels altered invasive potential, where enhanced Reelin expression in MDA-MB-231 cells reduced invasion, while RNAi-mediated suppression of Reelin in SUM159 cells increased invasion. Moreover, treatment of α3β1-expressing MDA-MB-231 cells with culture medium that was conditioned by α3 knockdown MDA-MB-231 cells led to decreased invasion. RNAi-mediated suppression of Reelin in α3 knockdown MDA-MB-231 cells mitigated this effect of conditioned-medium, identifying secreted Reelin as an inhibitor of cell invasion. These results demonstrate a novel role for α3β1 in repressing Reelin in breast cancer cells to promote invasion, supporting this integrin as a potential therapeutic target.
Collapse
Affiliation(s)
- Abibatou Ndoye
- Department of Surgery, Albany Medical College, Albany, 12208 NY, USA;
| | | | - C. Michael DiPersio
- Department of Surgery, Albany Medical College, Albany, 12208 NY, USA;
- Department of Molecular & Cellular Physiology, Albany Medical College, Albany, 12208 NY, USA
- Correspondence:
| |
Collapse
|
19
|
Li JM, Yang F, Li J, Yuan WQ, Wang H, Luo YQ. Reelin Promotes Cisplatin Resistance by Induction of Epithelial-Mesenchymal Transition via p38/GSK3β/Snail Signaling in Non-Small Cell Lung Cancer. Med Sci Monit 2020; 26:e925298. [PMID: 32764530 PMCID: PMC7433388 DOI: 10.12659/msm.925298] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Background Emerging evidence suggests the involvement of Reelin in chemoresistance in various cancers. However, its function in cisplatin (DDP) sensitivity of non-small cell lung cancer (NSCLC) needs to be investigated. Material/Methods Reelin expression in cisplatin-sensitive A549 cells and cisplatin-resistant NSCLC (A549/DDP) cells was analyzed by western blot analysis. qRT-PCR, western blotting, immunofluorescence, CCK-8 assays, Annexin V/propidium iodide apoptosis assay, and Transwell migration assays were carried out to determine the function of Reelin on DDP resistance. Results Reelin was markedly increased in A549/DDP cells relative to A549 cells. Knockdown of Reelin enhanced DDP chemosensitivity of A549/DDP cells, whereas overexpression of Reelin enhanced DDP resistance of A549, H1299, and H460 cells. Reelin induced DDP resistance in NSCLC cells via facilitating epithelial-mesenchymal transition (EMT). Furthermore, Reelin modulated p38/GSK3β signal transduction and promoted Snail (EMT-associated transcription factor) expression. Suppression of p38/Snail reversed Reelin-induced EMT and resistance of NSCLC cells to DDP. Conclusions These data indicated that Reelin induces DDP resistance of NSCLC by regulation of the p38/GSK3β/Snail/EMT signaling pathway and provide evidence that Reelin suppression can be an effective strategy to suppress DDP resistance in NSCLC.
Collapse
Affiliation(s)
- Ji-Min Li
- Department of Laboratory Medicine, The Affiliated Anhui Provincial Hospital of Anhui Medical University, Hefei, Anhui, China (mainland)
| | - Fang Yang
- Department of Laboratory Medicine, The Affiliated Anhui Provincial Hospital of Anhui Medical University, Hefei, Anhui, China (mainland)
| | - Juan Li
- Department of Blood Transfusion, The Affiliated Anhui Provincial Hospital of Anhui Medical University, Hefei, Anhui, China (mainland)
| | - Wei-Qi Yuan
- Department of Laboratory Medicine, The Affiliated Anhui Provincial Hospital of Anhui Medical University, Hefei, Anhui, China (mainland)
| | - Hao Wang
- Department of Laboratory Medicine, The Affiliated Anhui Provincial Hospital of Anhui Medical University, Hefei, Anhui, China (mainland)
| | - Yi-Qin Luo
- Department of Laboratory Medicine, The Affiliated Anhui Provincial Hospital of Anhui Medical University, Hefei, Anhui, China (mainland)
| |
Collapse
|
20
|
Li X, Fan W, Yao A, Song H, Ge Y, Yan M, Shan Y, Zhang C, Li P, Jia L. Downregulation of reelin predicts poor prognosis for glioma. Biomark Med 2020; 14:651-663. [PMID: 32613843 DOI: 10.2217/bmm-2019-0609] [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] [Indexed: 12/15/2022] Open
Abstract
Aim: In the present study, we studied the relationship between RELN and prognosis in glioma. Materials & methods: Expression profiles and methylation data of RELN were obtained from bioinformatic datasets. Correlations between RELN and clinicopathological features and overall survival were respectively assessed using chi-square test and Kaplan-Meier analysis. Results: RELN was downregulated in glioma, and its downregulation correlated well with glioma malignancy and overall survival. Meanwhile, hypermethylation of RELN was significantly correlated with low RELN expression. Additionally, gene set enrichment analysis demonstrated that low expression of RELN correlated with many key cancer pathways, possibly highlighting the importance of RELN in carcinogenesis of brain. Conclusion: RELN may serve as a potential prognostic marker and promising target molecule for new therapy of glioma.
Collapse
Affiliation(s)
- Xueli Li
- Department of Medical Genetics & Cell Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, China
| | - Wange Fan
- Department of Medical Genetics & Cell Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, China
| | - Anhui Yao
- Department of Neurosurgery, The General Hospital of PLA, Beijing, China.,Department of Neurosurgery, 988th Hospital of Chinese People's Liberation Army, Zhengzhou, Henan Province, PR China
| | - Huiling Song
- Department of Medical Genetics & Cell Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, China
| | - Yunxiao Ge
- Department of Medical Genetics & Cell Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, China
| | - Mengyao Yan
- Department of Medical Genetics & Cell Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, China
| | - Yubo Shan
- Department of Medical Genetics & Cell Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, China
| | - Chujie Zhang
- Department of Medical Genetics & Cell Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, China
| | - Pu Li
- Department of Medical Genetics & Cell Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, China
| | - Liyun Jia
- Department of Medical Genetics & Cell Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, China
| |
Collapse
|
21
|
Abstract
Brain metastases are associated with poor prognosis irrespective of the primary tumor they originate from. Current treatments for brain metastases are palliative, and patients with symptomatic brain metastasis have a one-year survival of <20%. Lung cancer, breast cancer, and melanoma have higher incidences of brain metastases compared with other types of cancers. However, it is not very clear why some cancers metastasize to the brain more frequently than others. Studies thus far suggest that brain-specific tropism of certain types of cancers is defined by a winning combination of the following factors: unique genetic subtypes of primary tumors or its subclones enabling detachment, dissemination, blood-brain barrier penetration, plus proliferation and survival in hypoxic low-glucose microenvironment; specific transcriptomic and epigenetic changes of colony-forming metastatic cells, allowing their outgrowth; favorable metastasis-permissive microenvironment of the brain created by interactions of cancer cells and cells in the brain through triggering inflammation, recruiting myeloid-derived suppressor cells, and promoting metabolic adaptation; immunosuppression resulting in the failure of adaptive immune response to recognize or kill cancer cells in the brain. Here, we briefly review recent advances in understanding brain metastasis organotropism and outline directions for future research.
Collapse
Affiliation(s)
- Arseniy E Yuzhalin
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Dihua Yu
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| |
Collapse
|
22
|
Ansari KI, Bhan A, Liu X, Chen MY, Jandial R. Astrocytic IGFBP2 and CHI3L1 in cerebrospinal fluid drive cortical metastasis of HER2+breast cancer. Clin Exp Metastasis 2020; 37:401-412. [PMID: 32279122 DOI: 10.1007/s10585-020-10032-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Accepted: 04/02/2020] [Indexed: 12/17/2022]
Abstract
The brain is often reported as the first site of recurrence among breast cancer patients overexpressing human epidermal growth factor receptor 2 (HER2). Although most HER2+tumors metastasize to the subcortical region of the brain, a subset develops in the cortical region. We hypothesize that factors in cerebrospinal fluid (CSF) play a critical role in the adaptation, proliferation, and establishment of cortical metastases. We established novel cell lines using patient biopsies to model breast cancer cortical and subcortical metastases. We assessed the localization and growth of these cells in vivo and proliferation and apoptosis in vitro under various conditions. Proteomic analysis of human CSF identified astrocyte-derived factors that support the proliferation of cortical metastases, and we used neutralizing antibodies to test the effects of inhibiting these factors both in vivo and in vitro. The cortical breast cancer brain metastatic cells exhibited greater proliferation than subcortical breast cancer brain metastatic cells in CSF containing several growth factors that nourish both the CNS and tumor cells. Specifically, the astrocytic paracrine factors IGFBP2 and CHI3LI promoted the proliferation of cortical metastatic cells and the formation of metastatic lesions. Disruption of these factors suppressed astrocyte-tumor cell interactions in vitro and the growth of cortical tumors in vivo. Our findings suggest that inhibition of IGFBP2 and CHI3LI signaling, in addition to existing treatment modalities, may be an effective therapeutic strategy targeting breast cancer cortical metastasis.
Collapse
Affiliation(s)
- Khairul I Ansari
- Division of Neurosurgery, Beckman Research Institute, City of Hope, 1500 E. Duarte Road, Duarte, CA, 91010, USA.
- Celcuity, 16305 36th Ave N, Suite 100, Minneapolis, MN, 55446, USA.
| | - Arunoday Bhan
- Division of Neurosurgery, Beckman Research Institute, City of Hope, 1500 E. Duarte Road, Duarte, CA, 91010, USA
| | - Xueli Liu
- Division of Biostatistics, Beckman Research Institute, City of Hope, Duarte, CA, USA
| | - Mike Y Chen
- Division of Neurosurgery, Beckman Research Institute, City of Hope, 1500 E. Duarte Road, Duarte, CA, 91010, USA
| | - Rahul Jandial
- Division of Neurosurgery, Beckman Research Institute, City of Hope, 1500 E. Duarte Road, Duarte, CA, 91010, USA.
| |
Collapse
|
23
|
Mikulska-Ruminska K, Strzelecki J, Nowak W. Dynamics, nanomechanics and signal transduction in reelin repeats. Sci Rep 2019; 9:18974. [PMID: 31831824 PMCID: PMC6908669 DOI: 10.1038/s41598-019-55461-8] [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: 08/09/2019] [Accepted: 11/27/2019] [Indexed: 12/04/2022] Open
Abstract
Reelin is a large glycoprotein controlling brain development and cell adhesion. It regulates the positioning of neurons, as well as neurotransmission and memory formation. Perturbations in reelin signaling are linked to psychiatric disorders. Reelin participates in signal transduction by binding to the lipoprotein receptors VLDLR and ApoER2 through its central region. This part is rich in repeating BNR-EGF-BNR modules. We used standard molecular dynamics, steered molecular dynamics, and perturbation response scanning computational methods to characterize unique dynamical properties of reelin modules involved in signaling. Each module has specific sensors and effectors arranged in a similar topology. In the modules studied, disulfide bridges play a protective role, probably making both selective binding and protease activity of reelin possible. Results of single reelin molecule stretching by atomic force microscopy provide the first data on the mechanical stability of individual reelin domains. The forces required for partial unfolding of the modules studied are below 60 pN.
Collapse
Affiliation(s)
- Karolina Mikulska-Ruminska
- Institute of Physics, Faculty of Physics, Astronomy and Informatics, Nicolaus Copernicus University, Grudziadzka 5, 87-100, Torun, Poland.
| | - Janusz Strzelecki
- Institute of Physics, Faculty of Physics, Astronomy and Informatics, Nicolaus Copernicus University, Grudziadzka 5, 87-100, Torun, Poland
| | - Wieslaw Nowak
- Institute of Physics, Faculty of Physics, Astronomy and Informatics, Nicolaus Copernicus University, Grudziadzka 5, 87-100, Torun, Poland.
| |
Collapse
|
24
|
Fecci PE, Champion CD, Hoj J, McKernan CM, Goodwin CR, Kirkpatrick JP, Anders CK, Pendergast AM, Sampson JH. The Evolving Modern Management of Brain Metastasis. Clin Cancer Res 2019; 25:6570-6580. [PMID: 31213459 PMCID: PMC8258430 DOI: 10.1158/1078-0432.ccr-18-1624] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 04/23/2019] [Accepted: 06/13/2019] [Indexed: 12/11/2022]
Abstract
The incidence of brain metastases is increasing as cancer therapies improve and patients live longer, providing new challenges to the multidisciplinary teams that care for these patients. Brain metastatic cancer cells possess unique characteristics that allow them to penetrate the blood-brain barrier, colonize the brain parenchyma, and persist in the intracranial environment. In addition, brain metastases subvert the innate and adaptive immune system, permitting evasion of the antitumor immune response. Better understanding of the above mechanisms will allow for development and delivery of more effective therapies for brain metastases. In this review, we outline the molecular mechanisms underlying development, survival, and immunosuppression of brain metastases. We also discuss current and emerging treatment strategies, including surgery, radiation, disease-specific and mutation-targeted systemic therapy, and immunotherapy.
Collapse
Affiliation(s)
- Peter E Fecci
- Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina
- Duke Center for Brain and Spinal Metastases, Duke University Medical Center, Durham, North Carolina
| | - Cosette D Champion
- Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina
- Duke Center for Brain and Spinal Metastases, Duke University Medical Center, Durham, North Carolina
| | - Jacob Hoj
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, North Carolina
| | - Courtney M McKernan
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, North Carolina
| | - C Rory Goodwin
- Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina
- Duke Center for Brain and Spinal Metastases, Duke University Medical Center, Durham, North Carolina
| | - John P Kirkpatrick
- Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina
- Duke Center for Brain and Spinal Metastases, Duke University Medical Center, Durham, North Carolina
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina
| | - Carey K Anders
- Duke Cancer Institute, Division of Medical Oncology, Duke University Medical Center, Durham, North Carolina
| | - Ann Marie Pendergast
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, North Carolina
| | - John H Sampson
- Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina.
- Duke Center for Brain and Spinal Metastases, Duke University Medical Center, Durham, North Carolina
| |
Collapse
|
25
|
Zou Y, Watters A, Cheng N, Perry CE, Xu K, Alicea GM, Parris JLD, Baraban E, Ray P, Nayak A, Xu X, Herlyn M, Murphy ME, Weeraratna AT, Schug ZT, Chen Q. Polyunsaturated Fatty Acids from Astrocytes Activate PPARγ Signaling in Cancer Cells to Promote Brain Metastasis. Cancer Discov 2019; 9:1720-1735. [PMID: 31578185 DOI: 10.1158/2159-8290.cd-19-0270] [Citation(s) in RCA: 97] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 08/19/2019] [Accepted: 09/27/2019] [Indexed: 01/21/2023]
Abstract
Brain metastasis, the most lethal form of melanoma and carcinoma, is the consequence of favorable interactions between the invading cancer cells and the brain cells. Peroxisome proliferator-activated receptor γ (PPARγ) has ambiguous functions in cancer development, and its relevance in advanced brain metastasis remains unclear. Here, we demonstrate that astrocytes, the unique brain glial cells, activate PPARγ in brain metastatic cancer cells. PPARγ activation enhances cell proliferation and metastatic outgrowth in the brain. Mechanistically, astrocytes have a high content of polyunsaturated fatty acids that act as "donors" of PPARγ activators to the invading cancer cells. In clinical samples, PPARγ signaling is significantly higher in brain metastatic lesions. Notably, systemic administration of PPARγ antagonists significantly reduces brain metastatic burden in vivo. Our study clarifies a prometastatic role for PPARγ signaling in cancer metastasis in the lipid-rich brain microenvironment and argues for the use of PPARγ blockade to treat brain metastasis. SIGNIFICANCE: Brain-tropic cancer cells take advantage of the lipid-rich brain microenvironment to facilitate their proliferation by activating PPARγ signaling. This protumor effect of PPARγ in advanced brain metastases is in contrast to its antitumor function in carcinogenesis and early metastatic steps, indicating that PPARγ has diverse functions at different stages of cancer development.This article is highlighted in the In This Issue feature, p. 1631.
Collapse
Affiliation(s)
- Yongkang Zou
- Immunology, Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, Pennsylvania
| | - Andrea Watters
- Immunology, Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, Pennsylvania
| | - Nan Cheng
- Immunology, Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, Pennsylvania
| | - Caroline E Perry
- Molecular and Cellular Oncogenesis Program, The Wistar Institute, Philadelphia, Pennsylvania
| | - Ke Xu
- MD/PhD Program, Boston University School of Medicine, Boston, Massachusetts
| | - Gretchen M Alicea
- Immunology, Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, Pennsylvania
| | - Joshua L D Parris
- Molecular and Cellular Oncogenesis Program, The Wistar Institute, Philadelphia, Pennsylvania
| | - Ezra Baraban
- Departments of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Pulak Ray
- Delaware Neurosurgical Group, ChristianaCare Health System, Newark, Delaware
| | - Anupma Nayak
- Departments of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Xiaowei Xu
- Departments of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Meenhard Herlyn
- Melanoma Research Center, The Wistar Institute, Philadelphia, Pennsylvania.,Melanoma Research Center, The Wistar Institute, Philadelphia, Pennsylvania
| | - Maureen E Murphy
- Molecular and Cellular Oncogenesis Program, The Wistar Institute, Philadelphia, Pennsylvania
| | - Ashani T Weeraratna
- Immunology, Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, Pennsylvania
| | - Zachary T Schug
- Molecular and Cellular Oncogenesis Program, The Wistar Institute, Philadelphia, Pennsylvania
| | - Qing Chen
- Immunology, Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, Pennsylvania.
| |
Collapse
|
26
|
Alsereihi R, Schulten HJ, Bakhashab S, Saini K, Al-Hejin AM, Hussein D. Leveraging the Role of the Metastatic Associated Protein Anterior Gradient Homologue 2 in Unfolded Protein Degradation: A Novel Therapeutic Biomarker for Cancer. Cancers (Basel) 2019; 11:cancers11070890. [PMID: 31247903 PMCID: PMC6678570 DOI: 10.3390/cancers11070890] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2019] [Revised: 06/18/2019] [Accepted: 06/21/2019] [Indexed: 12/15/2022] Open
Abstract
Effective diagnostic, prognostic and therapeutic biomarkers can help in tracking disease progress, predict patients’ survival, and considerably affect the drive for successful clinical management. The present review aims to determine how the metastatic-linked protein anterior gradient homologue 2 (AGR2) operates to affect cancer progression, and to identify associated potential diagnostic, prognostic and therapeutic biomarkers, particularly in central nervous system (CNS) tumors. Studies that show a high expression level of AGR2, and associate the protein expression with the resilience to chemotherapeutic treatments or with poor cancer survival, are reported. The primary protein structures of the seven variants of AGR2, including their functional domains, are summarized. Based on experiments in various biological models, this review shows an orchestra of multiple molecules that regulate AGR2 expression, including a feedback loop with p53. The AGR2-associated molecular functions and pathways including genomic integrity, proliferation, apoptosis, angiogenesis, adhesion, migration, stemness, and inflammation, are detailed. In addition, the mechanisms that can enable the rampant oncogenic effects of AGR2 are clarified. The different strategies used to therapeutically target AGR2-positive cancer cells are evaluated in light of the current evidence. Moreover, novel associated pathways and clinically relevant deregulated genes in AGR2 high CNS tumors are identified using a meta-analysis approach.
Collapse
Affiliation(s)
- Reem Alsereihi
- Neurooncology Translational Group, King Fahd Medical Research Center, King Abdulaziz University, P.O. Box 80216, Jeddah, 21589, Saudi Arabia.
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, P.O. Box 80203, Jeddah 21589, Saudi Arabia.
| | - Hans-Juergen Schulten
- Center of Excellence in Genomic Medicine Research, King Abdulaziz University, P.O. Box 80216, Jeddah 21589, Saudi Arabia.
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia.
| | - Sherin Bakhashab
- Center of Excellence in Genomic Medicine Research, King Abdulaziz University, P.O. Box 80216, Jeddah 21589, Saudi Arabia.
- Biochemistry Department, King Abdulaziz University, P.O. Box 80218, Jeddah 21589, Saudi Arabia.
| | - Kulvinder Saini
- School of Biotechnology, Eternal University, Baru Sahib-173101, Himachal Pradesh, India.
| | - Ahmed M Al-Hejin
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, P.O. Box 80203, Jeddah 21589, Saudi Arabia.
- Microbiology Unit, King Fahad Medical Research Center, King Abdulaziz University, P.O. Box 80216, Jeddah 21589, Saudi Arabia.
| | - Deema Hussein
- Neurooncology Translational Group, King Fahd Medical Research Center, King Abdulaziz University, P.O. Box 80216, Jeddah, 21589, Saudi Arabia.
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia.
| |
Collapse
|
27
|
Doron H, Pukrop T, Erez N. A Blazing Landscape: Neuroinflammation Shapes Brain Metastasis. Cancer Res 2019; 79:423-436. [PMID: 30679177 DOI: 10.1158/0008-5472.can-18-1805] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 09/22/2018] [Accepted: 11/16/2018] [Indexed: 12/18/2022]
Abstract
Brain metastases are more common than primary CNS tumors and confer grave prognosis on patients, as existing treatments have very limited efficacy. The tumor microenvironment has a central role in facilitating tumorigenesis and metastasis. In recent years, there has been much progress in our understanding of the functional role of the brain metastatic microenvironment. In this review, we discuss the latest advances in brain metastasis research, with special emphasis on the role of the brain microenvironment and neuroinflammation, integrating insights from comparable findings in neuropathologies and primary CNS tumors. In addition, we overview findings on the formation of a hospitable metastatic niche and point out the major gaps in knowledge toward developing new therapeutics that will cotarget the stromal compartment in an effort to improve the treatment and prevention of brain metastases.
Collapse
Affiliation(s)
- Hila Doron
- Department of Pathology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Tobias Pukrop
- Department of Internal Medicine III, Hematology and Medical Oncology, University Hospital Regensburg, Regensburg, Germany
| | - Neta Erez
- Department of Pathology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
28
|
Wilhelm I, Fazakas C, Molnár K, Végh AG, Haskó J, Krizbai IA. Foe or friend? Janus-faces of the neurovascular unit in the formation of brain metastases. J Cereb Blood Flow Metab 2018; 38:563-587. [PMID: 28920514 PMCID: PMC5888855 DOI: 10.1177/0271678x17732025] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Revised: 07/13/2017] [Accepted: 08/21/2017] [Indexed: 12/11/2022]
Abstract
Despite the potential obstacle represented by the blood-brain barrier for extravasating malignant cells, metastases are more frequent than primary tumors in the central nervous system. Not only tightly interconnected endothelial cells can hinder metastasis formation, other cells of the brain microenvironment (like astrocytes and microglia) can also be very hostile, destroying the large majority of metastatic cells. However, malignant cells that are able to overcome these harmful mechanisms may benefit from the shielding and even support provided by cerebral endothelial cells, astrocytes and microglia, rendering the brain a sanctuary site against anti-tumor strategies. Thus, cells of the neurovascular unit have a Janus-faced attitude towards brain metastatic cells, being both destructive and protective. In this review, we present the main mechanisms of brain metastasis formation, including those involved in extravasation through the brain vasculature and survival in the cerebral environment.
Collapse
Affiliation(s)
- Imola Wilhelm
- Institute of Biophysics, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary
- Institute of Life Sciences, Vasile Goldiş Western University of Arad, Arad, Romania
| | - Csilla Fazakas
- Institute of Biophysics, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary
| | - Kinga Molnár
- Institute of Biophysics, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary
| | - Attila G Végh
- Institute of Biophysics, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary
| | - János Haskó
- Institute of Biophysics, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary
| | - István A Krizbai
- Institute of Biophysics, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary
- Institute of Life Sciences, Vasile Goldiş Western University of Arad, Arad, Romania
| |
Collapse
|
29
|
Kabraji S, Ni J, Lin NU, Xie S, Winer EP, Zhao JJ. Drug Resistance in HER2-Positive Breast Cancer Brain Metastases: Blame the Barrier or the Brain? Clin Cancer Res 2018; 24:1795-1804. [PMID: 29437794 DOI: 10.1158/1078-0432.ccr-17-3351] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Revised: 01/06/2018] [Accepted: 02/01/2018] [Indexed: 12/11/2022]
Abstract
The brain is the most common site of first metastasis for patients with HER2-positive breast cancer treated with HER2-targeting drugs. However, the development of effective therapies for breast cancer brain metastases (BCBM) is limited by an incomplete understanding of the mechanisms governing drug sensitivity in the central nervous system. Pharmacodynamic data from patients and in vivo models suggest that inadequate drug penetration across the "blood-tumor" barrier is not the whole story. Using HER2-positive BCBMs as a case study, we highlight recent data from orthotopic brain metastasis models that implicate brain-specific drug resistance mechanisms in BCBMs and suggest a translational research paradigm to guide drug development for treatment of BCBMs. Clin Cancer Res; 24(8); 1795-804. ©2018 AACR.
Collapse
Affiliation(s)
- Sheheryar Kabraji
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, Massachusetts. .,Department of Cancer Biology, Dana Farber Cancer Institute, Boston, Massachusetts
| | - Jing Ni
- Department of Cancer Biology, Dana Farber Cancer Institute, Boston, Massachusetts.,Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts
| | - Nancy U Lin
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, Massachusetts
| | - Shaozhen Xie
- Department of Cancer Biology, Dana Farber Cancer Institute, Boston, Massachusetts.,Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts
| | - Eric P Winer
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, Massachusetts
| | - Jean J Zhao
- Department of Cancer Biology, Dana Farber Cancer Institute, Boston, Massachusetts. .,Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|