1
|
Hiraga T. Immune microenvironment of cancer bone metastasis. Bone 2025; 191:117328. [PMID: 39549899 DOI: 10.1016/j.bone.2024.117328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 11/06/2024] [Accepted: 11/08/2024] [Indexed: 11/18/2024]
Abstract
Bone is a common and frequent site of metastasis in cancer patients, leading to a significant reduction in quality of life and increased mortality. Bone marrow, the primary site of hematopoiesis, also serves as both a primary and secondary lymphoid organ. It harbors and supports a diverse array of immune cells, thereby creating a distinct immune microenvironment. These immune cells engage in a range of activities, including anti-tumor, pro-tumor, or a combination of both, which influence the development and progression of bone metastases. Rapid advances in cancer immunotherapy have underscored its potential to eradicate bone metastases. However, clinical outcomes have not yet met expectations. To improve the efficacy of immunotherapy, it is crucial to gain a comprehensive and in-depth understanding of the immune microenvironment within bone metastases. This review provides an overview of the current understanding of the role of different immune cells, their anti-tumor and pro-tumor activities, and their overall contribution to bone metastasis.
Collapse
Affiliation(s)
- Toru Hiraga
- Department of Histology and Cell Biology, Matsumoto Dental University, Shiojiri, Nagano, Japan.
| |
Collapse
|
2
|
Qian BZ, Ma RY. Immune Microenvironment in Breast Cancer Metastasis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1464:413-432. [PMID: 39821036 DOI: 10.1007/978-3-031-70875-6_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2025]
Abstract
Metastatic disease is the final stage of breast cancer that accounts for vast majority of patient death. Mounting data over recent years strongly support the critical roles of the immune microenvironment in determining breast cancer metastasis. The latest single-cell studies provide further molecular evidence illustrating the heterogeneity of this immune microenvironment. This chapter summarizes major discoveries on the role of various immune cells in metastasis progression and discusses future research opportunities. Studies investigating immune heterogeneity within primary breast cancer and across different metastasis target organs can potentially lead to more precise treatment strategies with improved efficacy.
Collapse
Affiliation(s)
- Bin-Zhi Qian
- Department of Oncology, Fudan University Shanghai Cancer Center, Zhangjiang-Fudan International Innovation Center, Shanghai Medical College, The Human Phenome Institute, Fudan University, Shanghai, China.
| | - Ruo-Yu Ma
- Department of Oncology, Fudan University Shanghai Cancer Center, Zhangjiang-Fudan International Innovation Center, Shanghai Medical College, The Human Phenome Institute, Fudan University, Shanghai, China
| |
Collapse
|
3
|
Lenart NA, Rao SS. Cell-cell interactions mediating primary and metastatic breast cancer dormancy. Cancer Metastasis Rev 2024; 44:6. [PMID: 39585533 DOI: 10.1007/s10555-024-10223-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 10/18/2024] [Indexed: 11/26/2024]
Abstract
Breast cancer remains one of the leading causes of death in women around the world. A majority of deaths from breast cancer occur due to cancer cells colonizing distant organ sites. When colonizing these distant organ sites, breast cancer cells have been known to enter into a state of dormancy for extended periods of time. However, the mechanisms that promote dormancy as well as dormant-to-proliferative switch are not fully understood. The tumor microenvironment plays a key role in mediating cancer cell phenotype including regulation of the dormant state. In this review, we highlight cell-cell interactions in the tumor microenvironment mediating breast cancer dormancy at the primary and metastatic sites. Specifically, we describe how immune cells from the lymphoid lineage, tumor-associated myeloid lineage cells, and stromal cells of non-hematopoietic origin as well as tissue resident stromal cells impact dormancy vs. proliferation in breast cancer cells as well as the associated mechanisms. In addition, we highlight the importance of developing model systems and the associated considerations that will be critical in unraveling the mechanisms that promote primary and metastatic breast cancer dormancy mediated via cell-cell interactions.
Collapse
Affiliation(s)
- Nicholas A Lenart
- Department of Chemical and Biological Engineering, The University of Alabama, Tuscaloosa, AL, 35487-0203, USA
| | - Shreyas S Rao
- Department of Chemical and Biological Engineering, The University of Alabama, Tuscaloosa, AL, 35487-0203, USA.
| |
Collapse
|
4
|
Batista IA, Machado JC, Melo SA. Advances in exosomes utilization for clinical applications in cancer. Trends Cancer 2024; 10:947-968. [PMID: 39168775 DOI: 10.1016/j.trecan.2024.07.010] [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: 03/28/2024] [Revised: 07/04/2024] [Accepted: 07/25/2024] [Indexed: 08/23/2024]
Abstract
Exosomes are regarded as having transformative potential for clinical applications. Exosome-based liquid biopsies offer a noninvasive method for early cancer detection and real-time disease monitoring. Clinical trials are underway to validate the efficacy of exosomal biomarkers for enhancing diagnostic accuracy and predicting treatment responses. Additionally, engineered exosomes are being developed as targeted drug delivery systems that can navigate the bloodstream to deliver therapeutic agents to tumor sites, thus enhancing treatment efficacy while minimizing systemic toxicity. Exosomes also exhibit immunomodulatory properties, which are being harnessed to boost antitumor immune responses. In this review, we detail the latest advances in clinical trials and research studies, underscoring the potential of exosomes to revolutionize cancer care.
Collapse
Affiliation(s)
- Inês A Batista
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal; Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Porto, Portugal
| | - José C Machado
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal; Departamento de Patologia, Faculdade de Medicina, Universidade do Porto, Porto, Portugal; P.CCC Porto Comprehensive Cancer Centre, Raquel Seruca, Portugal
| | - Sonia A Melo
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal; Departamento de Patologia, Faculdade de Medicina, Universidade do Porto, Porto, Portugal; P.CCC Porto Comprehensive Cancer Centre, Raquel Seruca, Portugal.
| |
Collapse
|
5
|
Das Gupta A, Park J, Sorrells JE, Kim H, Krawczynska N, Pradeep D, Wang Y, Vidana Gamage HE, Nelczyk AT, Boppart SA, Boppart MD, Nelson ER. 27-Hydroxycholesterol Enhances Secretion of Extracellular Vesicles by ROS-Induced Dysregulation of Lysosomes. Endocrinology 2024; 165:bqae127. [PMID: 39298675 PMCID: PMC11448339 DOI: 10.1210/endocr/bqae127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 08/30/2024] [Accepted: 09/17/2024] [Indexed: 09/22/2024]
Abstract
Extracellular vesicles (EVs) serve as crucial mediators of cell-to-cell communication in normal physiology as well as in diseased states; they have been largely studied in regard to their role in cancer progression. However, the mechanisms by which their biogenesis and secretion are regulated by metabolic or endocrine factors remain unknown. Here, we delineate a mechanism by which EV secretion is regulated by a cholesterol metabolite, 27-hydroxycholesterol (27HC), where treatment of myeloid immune cells (RAW 264.7 and J774A.1) with 27HC impairs lysosomal homeostasis, leading to shunting of multivesicular bodies (MVBs) away from lysosomal degradation, toward secretion as EVs. This altered lysosomal function is likely caused by mitochondrial dysfunction and subsequent increase in reactive oxygen species (ROS). Interestingly, cotreatment with a mitochondria-targeted antioxidant rescued the lysosomal impairment and attenuated the 27HC-mediated increase in EV secretion. Overall, our findings establish how a cholesterol metabolite regulates EV secretion and paves the way for the development of strategies to regulate cancer progression by controlling EV secretion.
Collapse
Affiliation(s)
- Anasuya Das Gupta
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Jaena Park
- Department of Bioengineering, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Janet E Sorrells
- Department of Bioengineering, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Hannah Kim
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Natalia Krawczynska
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Dhanya Pradeep
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Yu Wang
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Hashni Epa Vidana Gamage
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Adam T Nelczyk
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Stephen A Boppart
- Department of Bioengineering, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Department of Electrical and Computer Engineering, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
- Carle Illinois College of Medicine, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
- Interdisciplinary Health Sciences Institute, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
- NIH/NIBIB Center for Label-free Imaging and Multi-scale Biophotonics (CLIMB), University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
- Cancer Center at Illinois, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| | - Marni D Boppart
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Carle Illinois College of Medicine, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
- Department of Kinesiology and Community Health, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| | - Erik R Nelson
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Carl R. Woese Institute for Genomic Biology- Anticancer Discovery from Pets to People, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Division of Nutritional Sciences, University of Illinois Urbana-Champaign, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Cancer Center at Illinois, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| |
Collapse
|
6
|
D'Antonio C, Liguori GL. Dormancy and awakening of cancer cells: the extracellular vesicle-mediated cross-talk between Dr. Jekill and Mr. Hyde. Front Immunol 2024; 15:1441914. [PMID: 39301024 PMCID: PMC11410588 DOI: 10.3389/fimmu.2024.1441914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 08/08/2024] [Indexed: 09/22/2024] Open
Abstract
Cancer cell dormancy is a reversible process whereby cancer cells enter a quiescent state characterized by cell cycle arrest, inhibition of cell migration and invasion, and increased chemoresistance. Because of its reversibility and resistance to treatment, dormancy is a key process to study, monitor, and interfere with, in order to prevent tumor recurrence and metastasis and improve the prognosis of cancer patients. However, to achieve this goal, further studies are needed to elucidate the mechanisms underlying this complex and dynamic dual process. Here, we review the contribution of extracellular vesicles (EVs) to the regulation of cancer cell dormancy/awakening, focusing on the cross-talk between tumor and non-tumor cells in both the primary tumor and the (pre-)metastatic niche. Although EVs are recognized as key players in tumor progression and metastasis, as well as in tumor diagnostics and therapeutics, their role specifically in dormancy induction/escape is still largely elusive. We report on the most recent and promising results on this topic, focusing on the EV-associated nucleic acids involved. We highlight how EV studies could greatly contribute to the identification of dormancy signaling pathways and a dormancy/early awakening signature for the development of successful diagnostic/prognostic and therapeutic approaches.
Collapse
Affiliation(s)
- Concetta D'Antonio
- Institute of Genetics and Biophysics (IGB) "Adriano Buzzati-Traverso", National Research Council (CNR) of Italy, Naples, Italy
| | - Giovanna L Liguori
- Institute of Genetics and Biophysics (IGB) "Adriano Buzzati-Traverso", National Research Council (CNR) of Italy, Naples, Italy
| |
Collapse
|
7
|
Xin Z, Qin L, Tang Y, Guo S, Li F, Fang Y, Li G, Yao Y, Zheng B, Zhang B, Wu D, Xiao J, Ni C, Wei Q, Zhang T. Immune mediated support of metastasis: Implication for bone invasion. Cancer Commun (Lond) 2024; 44:967-991. [PMID: 39003618 PMCID: PMC11492328 DOI: 10.1002/cac2.12584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 06/05/2024] [Accepted: 06/18/2024] [Indexed: 07/15/2024] Open
Abstract
Bone is a common organ affected by metastasis in various advanced cancers, including lung, breast, prostate, colorectal, and melanoma. Once a patient is diagnosed with bone metastasis, the patient's quality of life and overall survival are significantly reduced owing to a wide range of morbidities and the increasing difficulty of treatment. Many studies have shown that bone metastasis is closely related to bone microenvironment, especially bone immune microenvironment. However, the effects of various immune cells in the bone microenvironment on bone metastasis remain unclear. Here, we described the changes in various immune cells during bone metastasis and discussed their related mechanisms. Osteoblasts, adipocytes, and other non-immune cells closely related to bone metastasis were also included. This review also summarized the existing treatment methods and potential therapeutic targets, and provided insights for future studies of cancer bone metastasis.
Collapse
Affiliation(s)
- Zengfeng Xin
- Department of Orthopedic SurgerySecond Affiliated HospitalZhejiang University School of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
| | - Luying Qin
- Cancer Institute (Key Laboratory of Cancer Prevention and InterventionNational Ministry of Education)Second Affiliated HospitalZhejiang University School of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
| | - Yang Tang
- Cancer Institute (Key Laboratory of Cancer Prevention and InterventionNational Ministry of Education)Second Affiliated HospitalZhejiang University School of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
| | - Siyu Guo
- Cancer Institute (Key Laboratory of Cancer Prevention and InterventionNational Ministry of Education)Second Affiliated HospitalZhejiang University School of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
- Department of Radiation OncologySecond Affiliated HospitalZhejiang University School of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
| | - Fangfang Li
- Cancer Institute (Key Laboratory of Cancer Prevention and InterventionNational Ministry of Education)Second Affiliated HospitalZhejiang University School of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
| | - Yuan Fang
- Cancer Institute (Key Laboratory of Cancer Prevention and InterventionNational Ministry of Education)Second Affiliated HospitalZhejiang University School of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
| | - Gege Li
- Cancer Institute (Key Laboratory of Cancer Prevention and InterventionNational Ministry of Education)Second Affiliated HospitalZhejiang University School of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
| | - Yihan Yao
- Cancer Institute (Key Laboratory of Cancer Prevention and InterventionNational Ministry of Education)Second Affiliated HospitalZhejiang University School of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
| | - Binbin Zheng
- Department of Respiratory MedicineNingbo Hangzhou Bay HospitalNingboZhejiangP. R. China
| | - Bicheng Zhang
- Cancer Institute (Key Laboratory of Cancer Prevention and InterventionNational Ministry of Education)Second Affiliated HospitalZhejiang University School of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
- Department of Radiation OncologySecond Affiliated HospitalZhejiang University School of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
| | - Dang Wu
- Cancer Institute (Key Laboratory of Cancer Prevention and InterventionNational Ministry of Education)Second Affiliated HospitalZhejiang University School of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
- Department of Radiation OncologySecond Affiliated HospitalZhejiang University School of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
| | - Jie Xiao
- Department of Orthopedic SurgerySecond Affiliated Hospital (Jiande Branch)Zhejiang University School of MedicineHangzhouZhejiangP. R. China
| | - Chao Ni
- Cancer Institute (Key Laboratory of Cancer Prevention and InterventionNational Ministry of Education)Second Affiliated HospitalZhejiang University School of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
- Department of Breast SurgerySecond Affiliated HospitalZhejiang University School of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
| | - Qichun Wei
- Cancer Institute (Key Laboratory of Cancer Prevention and InterventionNational Ministry of Education)Second Affiliated HospitalZhejiang University School of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
- Department of Radiation OncologySecond Affiliated HospitalZhejiang University School of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
| | - Ting Zhang
- Cancer Institute (Key Laboratory of Cancer Prevention and InterventionNational Ministry of Education)Second Affiliated HospitalZhejiang University School of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
- Department of Radiation OncologySecond Affiliated HospitalZhejiang University School of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
| |
Collapse
|
8
|
Dong Y, Bai J, Zhou J. Developing a dormancy-associated ECM signature in TNBC that is linked to immunosuppressive tumor microenvironment and selective sensitivity to MAPK inhibitors. Heliyon 2024; 10:e32106. [PMID: 38868025 PMCID: PMC11168407 DOI: 10.1016/j.heliyon.2024.e32106] [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: 03/11/2024] [Revised: 05/12/2024] [Accepted: 05/28/2024] [Indexed: 06/14/2024] Open
Abstract
Aims Cellular dormancy is a state of quiescence subpopulation of tumor cells, characterized by low differentiation and lack of mitotic activity. They could evade chemotherapy and targeted therapy, leading to drug resistance and disease recurrence. Recent studies have shown a correlation between dormant cancer cells and unique extracellular matrix (ECM) composition, which is critical in regulating cell behavior. However, their interacting roles in TNBC patients remains to be characterized. Main methods Dormant cancer cells in MDA-MB-231 cell line with highest PKH26 dye-retaining were FACS-sorted and gene expression was then analyzed. Dormant associated ECM (DA-ECM) signature was characterized by pathway analysis. Unsupervised hierarchical clustering was used to define distinct ECM features for TNBC patients. ECM-specific tumor biology was defined by integration of bulk RNA-seq with single-cell RNA-seq data, analysis of ligand-receptor interactions and enriched biological pathways, and in silico drug screening. We validated the sensitivity of dormant cancer cells to MAPK inhibitors by flow cytometry in vitro. Key findings We observed that dormant TNBC cells preferentially expressed ∼10 % DA-ECM genes. The DA-ECM High subtype defined by unsupervised hierarchical clustering analysis was associated with immunosuppressive tumor microenvironment. Moreover, ligand-receptor interaction and pathway analysis revealed that the DA-ECM High subtype may likely help maintain tumor cell dormancy through MAPK, Hedgehog and Notch signaling pathways. Finally, in silico drug screening against the DA-ECM signature and in vitro assay showed dormant cancer cells were relatively sensitive to the MAPK pathway inhibitors, which may represent a potential therapeutic strategy for treating TNBC. Significance Collectively, our research revealed that dormancy-associated ECM characterized tumor cells possess significant ECM remodeling capacity, and treatment strategies towards these cells could improve TNBC patient outcome.
Collapse
Affiliation(s)
- Yang Dong
- Research Center for Translational Medicine, Cancer Stem Cell Institute, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200120, China
| | - Jin Bai
- Department of Oncology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200120, China
| | - Jianjun Zhou
- Research Center for Translational Medicine, Cancer Stem Cell Institute, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200120, China
| |
Collapse
|
9
|
Das Gupta A, Park J, Sorrells JE, Kim H, Krawczynska N, Gamage HEV, Nelczyk AT, Boppart SA, Boppart MD, Nelson ER. Cholesterol Metabolite 27-Hydroxycholesterol Enhances the Secretion of Cancer Promoting Extracellular Vesicles by a Mitochondrial ROS-Induced Impairment of Lysosomal Function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.01.591500. [PMID: 38746134 PMCID: PMC11092642 DOI: 10.1101/2024.05.01.591500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Extracellular vesicles (EVs) serve as crucial mediators of cell-to-cell communication in normal physiology as well as in diseased states, and have been largely studied in regard to their role in cancer progression. However, the mechanisms by which their biogenesis and secretion are regulated by metabolic or endocrine factors remain unknown. Here, we delineate a mechanism by which EV secretion is regulated by a cholesterol metabolite, 27-Hydroxycholesterol (27HC), where treatment of myeloid immune cells (RAW 264.7 and J774A.1) with 27HC impairs lysosomal homeostasis, leading to shunting of multivesicular bodies (MVBs) away from lysosomal degradation, towards secretion as EVs. This impairment of lysosomal function is caused by mitochondrial dysfunction and subsequent increase in reactive oxygen species (ROS). Interestingly, cotreatment with a mitochondria-targeted antioxidant rescued the lysosomal impairment and attenuated the 27HC-mediated increase in EV secretion. Overall, our findings establish how a cholesterol metabolite regulates EV secretion and paves the way for the development of strategies to regulate cancer progression by controlling EV secretion.
Collapse
Affiliation(s)
- Anasuya Das Gupta
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana Illinois, 61801 USA
| | - Jaena Park
- Department of Bioengineering, University of Illinois Urbana-Champaign, Urbana, Illinois, 61801, USA
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana Illinois, 61801 USA
| | - Janet E. Sorrells
- Department of Bioengineering, University of Illinois Urbana-Champaign, Urbana, Illinois, 61801, USA
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana Illinois, 61801 USA
| | - Hannah Kim
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana Illinois, 61801 USA
| | - Natalia Krawczynska
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana Illinois, 61801 USA
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana Illinois, 61801 USA
| | - Hashni Epa Vidana Gamage
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana Illinois, 61801 USA
| | - Adam T. Nelczyk
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana Illinois, 61801 USA
| | - Stephen A. Boppart
- Department of Bioengineering, University of Illinois Urbana-Champaign, Urbana, Illinois, 61801, USA
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana Illinois, 61801 USA
- Department of Electrical and Computer Engineering, University of Illinois Urbana-Champaign, Urbana Illinois, 61801 USA
- Carle Illinois College of Medicine, University of Illinois Urbana-Champaign, Urbana Illinois, 61801 USA
- Interdisciplinary Health Sciences Institute, University of Illinois Urbana-Champaign, Urbana Illinois, 61801 USA
- NIH/NIBIB Center for Label-free Imaging and Multi-scale Biophotonics (CLIMB), University of Illinois Urbana-Champaign, Urbana, Illinois, 61801 USA
- Cancer Center at Illinois, University of Illinois Urbana-Champaign, Urbana Illinois, 61801 USA
| | - Marni D. Boppart
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana Illinois, 61801 USA
- Carle Illinois College of Medicine, University of Illinois Urbana-Champaign, Urbana Illinois, 61801 USA
- Department of Kinesiology and Community Health, University of Illinois Urbana-Champaign, Urbana Illinois, 61801 USA
| | - Erik R. Nelson
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana Illinois, 61801 USA
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana Illinois, 61801 USA
- Carl R. Woese Institute for Genomic Biology-Anticancer Discovery from Pets to People, University of Illinois at Urbana-Champaign, Urbana Illinois, 61801 USA
- Division of Nutritional Sciences, University of Illinois Urbana-Champaign, University of Illinois at Urbana-Champaign, Urbana Illinois, 61801 USA
- Cancer Center at Illinois, University of Illinois Urbana-Champaign, Urbana Illinois, 61801 USA
| |
Collapse
|
10
|
Marrone L, Romano S, Malasomma C, Di Giacomo V, Cerullo A, Abate R, Vecchione MA, Fratantonio D, Romano MF. Metabolic vulnerability of cancer stem cells and their niche. Front Pharmacol 2024; 15:1375993. [PMID: 38659591 PMCID: PMC11039812 DOI: 10.3389/fphar.2024.1375993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 03/25/2024] [Indexed: 04/26/2024] Open
Abstract
Cancer stem cells (CSC) are the leading cause of the failure of anti-tumor treatments. These aggressive cancer cells are preserved and sustained by adjacent cells forming a specialized microenvironment, termed niche, among which tumor-associated macrophages (TAMs) are critical players. The cycle of tricarboxylic acids, fatty acid oxidation path, and electron transport chain have been proven to play central roles in the development and maintenance of CSCs and TAMs. By improving their oxidative metabolism, cancer cells are able to extract more energy from nutrients, which allows them to survive in nutritionally defective environments. Because mitochondria are crucial bioenergetic hubs and sites of these metabolic pathways, major hopes are posed for drugs targeting mitochondria. A wide range of medications targeting mitochondria, electron transport chain complexes, or oxidative enzymes are currently investigated in phase 1 and phase 2 clinical trials against hard-to-treat tumors. This review article aims to highlight recent literature on the metabolic adaptations of CSCs and their supporting macrophages. A focus is provided on the resistance and dormancy behaviors that give CSCs a selection advantage and quiescence capacity in particularly hostile microenvironments and the role of TAMs in supporting these attitudes. The article also describes medicaments that have demonstrated a robust ability to disrupt core oxidative metabolism in preclinical cancer studies and are currently being tested in clinical trials.
Collapse
Affiliation(s)
- Laura Marrone
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| | - Simona Romano
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| | - Chiara Malasomma
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| | - Valeria Di Giacomo
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| | - Andrea Cerullo
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| | - Rosetta Abate
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| | | | - Deborah Fratantonio
- Department of Medicine and Surgery, LUM University Giuseppe Degennaro, Bari, Italy
| | - Maria Fiammetta Romano
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| |
Collapse
|
11
|
Guo J, Ma RY, Qian BZ. Macrophage heterogeneity in bone metastasis. J Bone Oncol 2024; 45:100598. [PMID: 38585688 PMCID: PMC10997910 DOI: 10.1016/j.jbo.2024.100598] [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: 02/06/2024] [Revised: 03/16/2024] [Accepted: 03/20/2024] [Indexed: 04/09/2024] Open
Abstract
Previous studies illustrated that macrophage, a type of innate immune cell, plays critical roles in tumour progression and metastasis. Bone is the most frequent site of metastasis for several cancer types including breast, prostate, and lung. In bone metastasis, osteoclast, a macrophage subset specialized in bone resorption, was heavily investigated in the past. Recent studies illustrated that other macrophage subsets, e.g. monocyte-derived macrophages, and bone resident macrophages, promoted bone metastasis independent of osteoclast function. These novel mechanisms further improved our understanding of macrophage heterogeneity in the context of bone metastasis and illustrated new opportunities for future studies.
Collapse
Affiliation(s)
| | | | - Bin-Zhi Qian
- Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, The Human Phenome Institute, Zhangjiang-Fudan International Innovation Center, Fudan University, Shanghai 200438, China
| |
Collapse
|
12
|
Dou T, Li J, Zhang Y, Pei W, Zhang B, Wang B, Wang Y, Jia H. The cellular composition of the tumor microenvironment is an important marker for predicting therapeutic efficacy in breast cancer. Front Immunol 2024; 15:1368687. [PMID: 38487526 PMCID: PMC10937353 DOI: 10.3389/fimmu.2024.1368687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 02/19/2024] [Indexed: 03/17/2024] Open
Abstract
At present, the incidence rate of breast cancer ranks first among new-onset malignant tumors in women. The tumor microenvironment is a hot topic in tumor research. There are abundant cells in the tumor microenvironment that play a protumor or antitumor role in breast cancer. During the treatment of breast cancer, different cells have different influences on the therapeutic response. And after treatment, the cellular composition in the tumor microenvironment will change too. In this review, we summarize the interactions between different cell compositions (such as immune cells, fibroblasts, endothelial cells, and adipocytes) in the tumor microenvironment and the treatment mechanism of breast cancer. We believe that detecting the cellular composition of the tumor microenvironment is able to predict the therapeutic efficacy of treatments for breast cancer and benefit to combination administration of breast cancer.
Collapse
Affiliation(s)
- Tingyao Dou
- Department of First Clinical Medicine, Shanxi Medical University, Taiyuan, China
| | - Jing Li
- Department of Breast Surgery, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yaochen Zhang
- Department of First Clinical Medicine, Shanxi Medical University, Taiyuan, China
| | - Wanru Pei
- Department of First Clinical Medicine, Shanxi Medical University, Taiyuan, China
| | - Binyue Zhang
- Department of Breast Surgery, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Bin Wang
- Department of Breast Surgery, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yanhong Wang
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, Shanxi, China
- Key Laboratory of Cellular Physiology (Shanxi Medical University), Ministry of Education, Taiyuan, Shanxi, China
| | - Hongyan Jia
- Department of Breast Surgery, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Key Laboratory of Cellular Physiology (Shanxi Medical University), Ministry of Education, Taiyuan, Shanxi, China
| |
Collapse
|
13
|
Ferrer-Diaz AI, Sinha G, Petryna A, Gonzalez-Bermejo R, Kenfack Y, Adetayo O, Patel SA, Hooda-Nehra A, Rameshwar P. Revealing role of epigenetic modifiers and DNA oxidation in cell-autonomous regulation of Cancer stem cells. Cell Commun Signal 2024; 22:119. [PMID: 38347590 PMCID: PMC10863086 DOI: 10.1186/s12964-024-01512-1] [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: 12/30/2023] [Accepted: 02/01/2024] [Indexed: 02/15/2024] Open
Abstract
BACKGROUND Breast cancer cells (BCCs) can remain undetected for decades in dormancy. These quiescent cells are similar to cancer stem cells (CSCs); hence their ability to initiate tertiary metastasis. Dormancy can be regulated by components of the tissue microenvironment such as bone marrow mesenchymal stem cells (MSCs) that release exosomes to dedifferentiate BCCs into CSCs. The exosomes cargo includes histone 3, lysine 4 (H3K4) methyltransferases - KMT2B and KMT2D. A less studied mechanism of CSC maintenance is the process of cell-autonomous regulation, leading us to examine the roles for KMT2B and KMT2D in sustaining CSCs, and their potential as drug targets. METHODS Use of pharmacological inhibitor of H3K4 (WDR5-0103), knockdown (KD) of KMT2B or KMT2D in BCCs, real time PCR, western blot, response to chemotherapy, RNA-seq, and flow cytometry for circulating markers of CSCs and DNA hydroxylases in BC patients. In vivo studies using a dormancy model studied the effects of KMT2B/D to chemotherapy. RESULTS H3K4 methyltransferases sustain cell autonomous regulation of CSCs, impart chemoresistance, maintain cycling quiescence, and reduce migration and proliferation of BCCs. In vivo studies validated KMT2's role in dormancy and identified these genes as potential drug targets. DNA methylase (DNMT), predicted within a network with KMT2 to regulate CSCs, was determined to sustain circulating CSC-like in the blood of patients. CONCLUSION H3K4 methyltransferases and DNA methylation mediate cell autonomous regulation to sustain CSC. The findings provide crucial insights into epigenetic regulatory mechanisms underlying BC dormancy with KMT2B and KMT2D as potential therapeutic targets, along with standard care. Stem cell and epigenetic markers in circulating BCCs could monitor treatment response and this could be significant for long BC remission to partly address health disparity.
Collapse
Affiliation(s)
- Alejandra I Ferrer-Diaz
- Department of Medicine - Division of Hematology/Oncology, Rutgers, New Jersey Medical School, Newark, NJ, 07103, USA
- Rutgers School of Graduate Studies at New Jersey Medical School, Newark, NJ, USA
| | - Garima Sinha
- Department of Medicine - Division of Hematology/Oncology, Rutgers, New Jersey Medical School, Newark, NJ, 07103, USA
- Rutgers School of Graduate Studies at New Jersey Medical School, Newark, NJ, USA
| | - Andrew Petryna
- Department of Medicine - Division of Hematology/Oncology, Rutgers, New Jersey Medical School, Newark, NJ, 07103, USA
- Rutgers School of Graduate Studies at New Jersey Medical School, Newark, NJ, USA
| | | | - Yannick Kenfack
- Department of Medicine - Division of Hematology/Oncology, Rutgers, New Jersey Medical School, Newark, NJ, 07103, USA
- Rutgers School of Graduate Studies at New Jersey Medical School, Newark, NJ, USA
| | | | - Shyam A Patel
- Division of Hematology and Oncology, Department of Medicine, UMass Memorial Medical Center, UMass Chan Medical School, Worcester, MA, USA
| | - Anupama Hooda-Nehra
- Department of Medicine - Division of Hematology/Oncology, Rutgers, New Jersey Medical School, Newark, NJ, 07103, USA
- Rutgers Cancer Institute of New Jersey, Newark, NJ, USA
| | - Pranela Rameshwar
- Department of Medicine - Division of Hematology/Oncology, Rutgers, New Jersey Medical School, Newark, NJ, 07103, USA.
| |
Collapse
|
14
|
Khan SU, Fatima K, Aisha S, Malik F. Unveiling the mechanisms and challenges of cancer drug resistance. Cell Commun Signal 2024; 22:109. [PMID: 38347575 PMCID: PMC10860306 DOI: 10.1186/s12964-023-01302-1] [Citation(s) in RCA: 64] [Impact Index Per Article: 64.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Accepted: 08/30/2023] [Indexed: 02/15/2024] Open
Abstract
Cancer treatment faces many hurdles and resistance is one among them. Anti-cancer treatment strategies are evolving due to innate and acquired resistance capacity, governed by genetic, epigenetic, proteomic, metabolic, or microenvironmental cues that ultimately enable selected cancer cells to survive and progress under unfavorable conditions. Although the mechanism of drug resistance is being widely studied to generate new target-based drugs with better potency than existing ones. However, due to the broader flexibility in acquired drug resistance, advanced therapeutic options with better efficacy need to be explored. Combination therapy is an alternative with a better success rate though the risk of amplified side effects is commonplace. Moreover, recent groundbreaking precision immune therapy is one of the ways to overcome drug resistance and has revolutionized anticancer therapy to a greater extent with the only limitation of being individual-specific and needs further attention. This review will focus on the challenges and strategies opted by cancer cells to withstand the current therapies at the molecular level and also highlights the emerging therapeutic options -like immunological, and stem cell-based options that may prove to have better potential to challenge the existing problem of therapy resistance. Video Abstract.
Collapse
Affiliation(s)
- Sameer Ullah Khan
- Division of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Holcombe Blvd, Houston, TX, 77030, USA.
- Division of Cancer Pharmacology, CSIR-Indian Institute of Integrative Medicine, Srinagar-190005, Jammu and Kashmir, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad-201002, India.
| | - Kaneez Fatima
- Division of Cancer Pharmacology, CSIR-Indian Institute of Integrative Medicine, Srinagar-190005, Jammu and Kashmir, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad-201002, India
| | - Shariqa Aisha
- Division of Cancer Pharmacology, CSIR-Indian Institute of Integrative Medicine, Srinagar-190005, Jammu and Kashmir, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad-201002, India
| | - Fayaz Malik
- Division of Cancer Pharmacology, CSIR-Indian Institute of Integrative Medicine, Srinagar-190005, Jammu and Kashmir, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad-201002, India.
| |
Collapse
|
15
|
Naaldijk Y, Sherman LS, Turrini N, Kenfack Y, Ratajczak MZ, Souayah N, Rameshwar P, Ulrich H. Mesenchymal Stem Cell-Macrophage Crosstalk Provides Specific Exosomal Cargo to Direct Immune Response Licensing of Macrophages during Inflammatory Responses. Stem Cell Rev Rep 2024; 20:218-236. [PMID: 37851277 DOI: 10.1007/s12015-023-10612-3] [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] [Accepted: 08/19/2023] [Indexed: 10/19/2023]
Abstract
Neurodegenerative diseases (NDDs) continue to be a significant healthcare problem. The economic and social implications of NDDs increase with longevity. NDDs are linked to neuroinflammation and activated microglia and astrocytes play a central role. There is a growing interest for stem cell-based therapy to deliver genes, and for tissue regeneration. The promise of mesenchymal stem cells (MSC) is based on their availability as off-the-shelf source, and ease of expanding from discarded tissues. We tested the hypothesis that MSC have a major role of resetting activated microglial cells. We modeled microglial cell lines by using U937 cell-derived M1 and M2 macrophages. We studied macrophage types, alone, or in a non-contact culture with MSCs. MSCs induced significant release of exosomes from both types of macrophages, but significantly more of the M1 type. RNA sequencing showed enhanced gene expression within the exosomes with the major changes linked to the inflammatory response, including cytokines and the purinergic receptors. Computational analyses of the transcripts supported the expected effect of MSCs in suppressing the inflammatory response of M1 macrophages. The inflammatory cargo of M1 macrophage-derived exosomes revealed involvement of cytokines and purinergic receptors. At the same time, the exosomes from MSC-M2 macrophages were able to reset the classical M2 macrophages to more balanced inflammation. Interestingly, we excluded transfer of purinergic receptor transcripts from the co-cultured MSCs by analyzing these cells for the identified purinergic receptors. Since exosomes are intercellular communicators, these findings provide insights into how MSCs may modulate tissue regeneration and neuroinflammation.
Collapse
Affiliation(s)
- Yahaira Naaldijk
- Department of Medicine, Rutgers New Jersey Medical School (NJMS), Newark, NJ, USA
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, SP, 05508-000, Brazil
| | - Lauren S Sherman
- Department of Medicine, Rutgers New Jersey Medical School (NJMS), Newark, NJ, USA
- Rutgers School of Graduate Studies at NHMS, Newark, NJ, USA
| | - Natalia Turrini
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, SP, 05508-000, Brazil
| | | | - Mariusz Z Ratajczak
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA
- Laboratory of Regenerative Medicine at Medical University of Warsaw, Warsaw, Poland
| | - Nizar Souayah
- Department of Neurology, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Pranela Rameshwar
- Department of Medicine, Rutgers New Jersey Medical School (NJMS), Newark, NJ, USA.
| | - Henning Ulrich
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, SP, 05508-000, Brazil.
- Department of Neuroscience and Physiology, Rutgers New Jersey Medical School, Newark, NJ, USA.
| |
Collapse
|
16
|
Cooper TT, Postovit LM. Wounding the stroma: Docetaxel's role in dormant breast cancer escape. PLoS Biol 2023; 21:e3002297. [PMID: 37703292 PMCID: PMC10499231 DOI: 10.1371/journal.pbio.3002297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/15/2023] Open
Abstract
The mechanistic underpinnings of breast cancer recurrence following periods of dormancy are largely undetermined. A new study in PLOS Biology reveals that docetaxel-induced injury of tumour stromal cells stimulates the release of cytokines that support dormancy escape of breast cancer cells.
Collapse
Affiliation(s)
- Tyler T. Cooper
- Biomedical and Molecular Sciences, Queen’s University, Kingston, Canada
| | | |
Collapse
|
17
|
Zhang C, Huang G, Yang J, Jiang Y, Huang R, Ye Z, Huang Y, Hu H, Xi X. Overexpression of DBT suppresses the aggressiveness of renal clear cell carcinoma and correlates with immune infiltration. Front Immunol 2023; 14:1197011. [PMID: 37383233 PMCID: PMC10293648 DOI: 10.3389/fimmu.2023.1197011] [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: 03/30/2023] [Accepted: 05/22/2023] [Indexed: 06/30/2023] Open
Abstract
Conventional therapy for kidney renal clear cell carcinoma (KIRC) is unpromising. The tumor microenvironment (TME) is intimately linked to the invasiveness of a variety of tumor forms, including KIRC. The purpose of this research is to establish the prognostic and immune-related significance of dihydrolipoamide branched chain transacylase E2 (DBT) in individuals with KIRC. In this investigation, we discovered that DBT expression was down-regulated in a range of human malignancies, and low DBT expression in KIRC was linked to higher-level clinicopathological characteristics as well as a poor prognosis for KIRC patients. Based on the findings of univariate and multivariate Cox regression analyses, DBT might be employed as an independent prognostic factor in KIRC patients. Furthermore, we developed a nomogram to better investigate DBT's predictive usefulness. To confirm DBT expression, we examined KIRC cell lines using RT-qPCR and Western blotting. We also examined the role of DBT in KIRC using colony formation, CCK-8, EdU, transwell, and wound healing assays. We discovered that plasmid-mediated overexpression of DBT in KIRC cells slowed cell proliferation and decreased migration and invasion. Multiple enrichment analyses revealed that DBT may be involved in processes and pathways related to immunotherapy and drug metabolism. We computed the immune infiltration score and discovered that the immunological score and the ESTIMATE score were both greater in the DBT low expression group. According to the CIBERSORT algorithm, DBT seems to promote anti-cancer immune responses in KIRC by activating M1 macrophages, mast cells, and dendritic cells while inhibiting regulatory T cells. Finally, in KIRC, DBT expression was found to be highly linked to immunological checkpoints, targeted medicines, and immunotherapeutic agents. Our findings suggest that DBT is a distinct predictive biomarker for KIRC patients, playing a significant role in the TME of KIRC and serving as a reference for the selection of targeted treatment and immunotherapy.
Collapse
Affiliation(s)
- Chiyu Zhang
- Department of Urology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Gaomin Huang
- Department of Urology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jiale Yang
- Hepato-Biliary-Pancreatic Surgery Division, Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yi Jiang
- Department of Urology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Ruizhen Huang
- Department of Urology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Zhenfeng Ye
- Department of Urology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yawei Huang
- Department of Urology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Honglin Hu
- Department of Urology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xiaoqing Xi
- Department of Urology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
18
|
Bhatia R, Chang J, Munoz JL, Walker ND. Forging New Therapeutic Targets: Efforts of Tumor Derived Exosomes to Prepare the Pre-Metastatic Niche for Cancer Cell Dissemination and Dormancy. Biomedicines 2023; 11:1614. [PMID: 37371709 PMCID: PMC10295689 DOI: 10.3390/biomedicines11061614] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 05/23/2023] [Accepted: 05/24/2023] [Indexed: 06/29/2023] Open
Abstract
Tumor-derived exosomes play a multifaceted role in preparing the pre-metastatic niche, promoting cancer dissemination, and regulating cancer cell dormancy. A brief review of three types of cells implicated in metastasis and an overview of other types of extracellular vesicles related to metastasis are described. A central focus of this review is on how exosomes influence cancer progression throughout metastatic disease. Exosomes are crucial mediators of intercellular communication by transferring their cargo to recipient cells, modulating their behavior, and promoting tumor pro-gression. First, their functional role in cancer cell dissemination in the peripheral blood by facilitating the establishment of a pro-angiogenic and pro-inflammatory niche is described during organotro-pism and in lymphatic-mediated metastasis. Second, tumor-derived exosomes can transfer molecular signals that induce cell cycle arrest, dormancy, and survival pathways in disseminated cells, promoting a dormant state are reviewed. Third, several studies highlight exosome involvement in maintaining cellular dormancy in the bone marrow endosteum. Finally, the clinical implications of exosomes as biomarkers or diagnostic tools for cancer progression are also outlined. Understanding the complex interplay between tumor-derived exosomes and the pre-metastatic niche is crucial for developing novel therapeutic strategies to target metastasis and prevent cancer recurrence. To that end, several examples of how exosomes or other nanocarriers are used as a drug delivery system to inhibit cancer metastasis are discussed. Strategies are discussed to alter exosome cargo content for better loading capacity or direct cell targeting by integrins. Further, pre-clinical models or Phase I clinical trials implementing exosomes or other nanocarriers to attack metastatic cancer cells are highlighted.
Collapse
Affiliation(s)
- Ranvir Bhatia
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Joanna Chang
- Department of Biological Sciences, University of Maryland, Baltimore, MD 21250, USA
| | - Jessian L Munoz
- Division of Perinatal Surgery, Texas Children's Hospital, Houston, TX 77030, USA
- Division of Maternal Fetal Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Nykia D Walker
- Department of Biological Sciences, University of Maryland, Baltimore, MD 21250, USA
| |
Collapse
|
19
|
Wieder R. Awakening of Dormant Breast Cancer Cells in the Bone Marrow. Cancers (Basel) 2023; 15:cancers15113021. [PMID: 37296983 DOI: 10.3390/cancers15113021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 05/23/2023] [Accepted: 05/26/2023] [Indexed: 06/12/2023] Open
Abstract
Up to 40% of patients with breast cancer (BC) have metastatic cells in the bone marrow (BM) at the initial diagnosis of localized disease. Despite definitive systemic adjuvant therapy, these cells survive in the BM microenvironment, enter a dormant state and recur stochastically for more than 20 years. Once they begin to proliferate, recurrent macrometastases are not curable, and patients generally succumb to their disease. Many potential mechanisms for initiating recurrence have been proposed, but no definitive predictive data have been generated. This manuscript reviews the proposed mechanisms that maintain BC cell dormancy in the BM microenvironment and discusses the data supporting specific mechanisms for recurrence. It addresses the well-described mechanisms of secretory senescence, inflammation, aging, adipogenic BM conversion, autophagy, systemic effects of trauma and surgery, sympathetic signaling, transient angiogenic bursts, hypercoagulable states, osteoclast activation, and epigenetic modifications of dormant cells. This review addresses proposed approaches for either eliminating micrometastases or maintaining a dormant state.
Collapse
Affiliation(s)
- Robert Wieder
- Rutgers New Jersey Medical School and the Cancer Institute of New Jersey, 185 South Orange Avenue, MSB F671, Newark, NJ 07103, USA
| |
Collapse
|
20
|
Salemme V, Centonze G, Avalle L, Natalini D, Piccolantonio A, Arina P, Morellato A, Ala U, Taverna D, Turco E, Defilippi P. The role of tumor microenvironment in drug resistance: emerging technologies to unravel breast cancer heterogeneity. Front Oncol 2023; 13:1170264. [PMID: 37265795 PMCID: PMC10229846 DOI: 10.3389/fonc.2023.1170264] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 04/28/2023] [Indexed: 06/03/2023] Open
Abstract
Breast cancer is a highly heterogeneous disease, at both inter- and intra-tumor levels, and this heterogeneity is a crucial determinant of malignant progression and response to treatments. In addition to genetic diversity and plasticity of cancer cells, the tumor microenvironment contributes to tumor heterogeneity shaping the physical and biological surroundings of the tumor. The activity of certain types of immune, endothelial or mesenchymal cells in the microenvironment can change the effectiveness of cancer therapies via a plethora of different mechanisms. Therefore, deciphering the interactions between the distinct cell types, their spatial organization and their specific contribution to tumor growth and drug sensitivity is still a major challenge. Dissecting intra-tumor heterogeneity is currently an urgent need to better define breast cancer biology and to develop therapeutic strategies targeting the microenvironment as helpful tools for combined and personalized treatment. In this review, we analyze the mechanisms by which the tumor microenvironment affects the characteristics of tumor heterogeneity that ultimately result in drug resistance, and we outline state of the art preclinical models and emerging technologies that will be instrumental in unraveling the impact of the tumor microenvironment on resistance to therapies.
Collapse
Affiliation(s)
- Vincenzo Salemme
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
- Molecular Biotechnology Center (MBC) “Guido Tarone”, Turin, Italy
| | - Giorgia Centonze
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
- Molecular Biotechnology Center (MBC) “Guido Tarone”, Turin, Italy
| | - Lidia Avalle
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
- Molecular Biotechnology Center (MBC) “Guido Tarone”, Turin, Italy
| | - Dora Natalini
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
- Molecular Biotechnology Center (MBC) “Guido Tarone”, Turin, Italy
| | - Alessio Piccolantonio
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
- Molecular Biotechnology Center (MBC) “Guido Tarone”, Turin, Italy
| | - Pietro Arina
- UCL, Bloomsbury Institute of Intensive Care Medicine, Division of Medicine, University College London, London, United Kingdom
| | - Alessandro Morellato
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
- Molecular Biotechnology Center (MBC) “Guido Tarone”, Turin, Italy
| | - Ugo Ala
- Department of Veterinary Sciences, University of Turin, Grugliasco, TO, Italy
| | - Daniela Taverna
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
- Molecular Biotechnology Center (MBC) “Guido Tarone”, Turin, Italy
| | - Emilia Turco
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Paola Defilippi
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
- Molecular Biotechnology Center (MBC) “Guido Tarone”, Turin, Italy
| |
Collapse
|
21
|
Loric S, Denis JA, Desbene C, Sabbah M, Conti M. Extracellular Vesicles in Breast Cancer: From Biology and Function to Clinical Diagnosis and Therapeutic Management. Int J Mol Sci 2023; 24:7208. [PMID: 37108371 PMCID: PMC10139222 DOI: 10.3390/ijms24087208] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 04/03/2023] [Accepted: 04/09/2023] [Indexed: 04/29/2023] Open
Abstract
Breast cancer (BC) is the first worldwide most frequent cancer in both sexes and the most commonly diagnosed in females. Although BC mortality has been thoroughly declining over the past decades, there are still considerable differences between women diagnosed with early BC and when metastatic BC is diagnosed. BC treatment choice is widely dependent on precise histological and molecular characterization. However, recurrence or distant metastasis still occurs even with the most recent efficient therapies. Thus, a better understanding of the different factors underlying tumor escape is mainly mandatory. Among the leading candidates is the continuous interplay between tumor cells and their microenvironment, where extracellular vesicles play a significant role. Among extracellular vesicles, smaller ones, also called exosomes, can carry biomolecules, such as lipids, proteins, and nucleic acids, and generate signal transmission through an intercellular transfer of their content. This mechanism allows tumor cells to recruit and modify the adjacent and systemic microenvironment to support further invasion and dissemination. By reciprocity, stromal cells can also use exosomes to profoundly modify tumor cell behavior. This review intends to cover the most recent literature on the role of extracellular vesicle production in normal and cancerous breast tissues. Specific attention is paid to the use of extracellular vesicles for early BC diagnosis, follow-up, and prognosis because exosomes are actually under the spotlight of researchers as a high-potential source of liquid biopsies. Extracellular vesicles in BC treatment as new targets for therapy or efficient nanovectors to drive drug delivery are also summarized.
Collapse
Affiliation(s)
- Sylvain Loric
- INSERM U538, CRSA, Saint-Antoine University Hospital, 75012 Paris, France; (J.A.D.)
| | | | - Cédric Desbene
- INSERM U538, CRSA, Saint-Antoine University Hospital, 75012 Paris, France; (J.A.D.)
| | - Michèle Sabbah
- INSERM U538, CRSA, Saint-Antoine University Hospital, 75012 Paris, France; (J.A.D.)
| | - Marc Conti
- INSERM U538, CRSA, Saint-Antoine University Hospital, 75012 Paris, France; (J.A.D.)
- INTEGRACELL SAS, 91160 Longjumeau, France
| |
Collapse
|
22
|
Lamouline A, Bersini S, Moretti M. In vitro models of breast cancer bone metastasis: analyzing drug resistance through the lens of the microenvironment. Front Oncol 2023; 13:1135401. [PMID: 37182144 PMCID: PMC10168004 DOI: 10.3389/fonc.2023.1135401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Accepted: 03/27/2023] [Indexed: 05/16/2023] Open
Abstract
Even though breast cancers usually have a good outcome compared to other tumors, the cancer can progress and create metastases in different parts of the organism, the bone being a predilection locus. These metastases are usually the cause of death, as they are mostly resistant to treatments. This resistance can be caused by intrinsic properties of the tumor, such as its heterogeneity, but it can also be due to the protective role of the microenvironment. By activating signaling pathways protecting cancer cells when exposed to chemotherapy, contributing to their ability to reach dormancy, or even reducing the amount of drug able to reach the metastases, among other mechanisms, the specificities of the bone tissue are being investigated as important players of drug resistance. To this date, most mechanisms of this resistance are yet to be discovered, and many researchers are implementing in vitro models to study the interaction between the tumor cells and their microenvironment. Here, we will review what is known about breast cancer drug resistance in bone metastasis due to the microenvironment and we will use those observations to highlight which features in vitro models should include to properly recapitulate these biological aspects in vitro. We will also detail which elements advanced in vitro models should implement in order to better recapitulate in vivo physiopathology and drug resistance.
Collapse
Affiliation(s)
- Anaïs Lamouline
- Regenerative Medicine Technologies Laboratory, Laboratories for Translational Research (LRT), Ente Ospedaliero Cantonale (EOC), Bellinzona, Switzerland
- Service of Orthopaedics and Traumatology, Department of Surgery, EOC, Lugano, Switzerland
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milano, Italy
| | - Simone Bersini
- Regenerative Medicine Technologies Laboratory, Laboratories for Translational Research (LRT), Ente Ospedaliero Cantonale (EOC), Bellinzona, Switzerland
- Service of Orthopaedics and Traumatology, Department of Surgery, EOC, Lugano, Switzerland
- Euler Institute, Faculty of Biomedical Sciences, Università della Svizzera italiana (USI), Lugano, Switzerland
| | - Matteo Moretti
- Regenerative Medicine Technologies Laboratory, Laboratories for Translational Research (LRT), Ente Ospedaliero Cantonale (EOC), Bellinzona, Switzerland
- Service of Orthopaedics and Traumatology, Department of Surgery, EOC, Lugano, Switzerland
- Euler Institute, Faculty of Biomedical Sciences, Università della Svizzera italiana (USI), Lugano, Switzerland
- Cell and Tissue Engineering Laboratory, IRCCS Istituto Ortopedico Galeazzi, Milano, Italy
| |
Collapse
|
23
|
Nahas GR, Sherman LS, Sinha G, El Far MH, Petryna A, Munoz SM, Silverio KA, Shaker M, Neopane P, Mariotti V, Rameshwar P. Increased expression of musashi 1 on breast cancer cells has implication to understand dormancy and survival in bone marrow. Aging (Albany NY) 2023; 15:3230-3248. [PMID: 36996499 PMCID: PMC10449290 DOI: 10.18632/aging.204620] [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: 03/01/2023] [Accepted: 03/13/2023] [Indexed: 03/31/2023]
Abstract
Breast cancer (BC) stem cells (CSCs) resist treatment and can exist as dormant cells in tissues such as the bone marrow (BM). Years before clinical diagnosis, BC cells (BCCs) could migrate from the primary site where the BM niche cells facilitate dedifferentiation into CSCs. Additionally, dedifferentiation could occur by cell autonomous methods. Here we studied the role of Msi 1, a RNA-binding protein, Musashi I (Msi 1). We also analyzed its relationship with the T-cell inhibitory molecule programmed death-ligand 1 (PD-L1) in CSCs. PD-L1 is an immune checkpoint that is a target in immune therapy for cancers. Msi 1 can support BCC growth through stabilization of oncogenic transcripts and modulation of stem cell-related gene expression. We reported on a role for Msi 1 to maintain CSCs. This seemed to occur by the differentiation of CSCs to more matured BCCs. This correlated with increased transition from cycling quiescence and reduced expression of stem cell-linked genes. CSCs co-expressed Msi 1 and PD-L1. Msi 1 knockdown led to a significant decrease in CSCs with undetectable PD-L1. This study has implications for Msi 1 as a therapeutic target, in combination with immune checkpoint inhibitor. Such treatment could also prevent dedifferentiation of breast cancer to CSCs, and to reverse tumor dormancy. The proposed combined treatment might be appropriate for other solid tumors.
Collapse
Affiliation(s)
- George R. Nahas
- Department of Medicine, Hematology-Oncology, Rutgers New Jersey Medical School, Newark, NJ 07103, USA
| | - Lauren S. Sherman
- Department of Medicine, Hematology-Oncology, Rutgers New Jersey Medical School, Newark, NJ 07103, USA
- Rutgers School of Graduate Studies at New Jersey Medical School, Newark, NJ 07103, USA
| | - Garima Sinha
- Department of Medicine, Hematology-Oncology, Rutgers New Jersey Medical School, Newark, NJ 07103, USA
- Rutgers School of Graduate Studies at New Jersey Medical School, Newark, NJ 07103, USA
| | - Markos H. El Far
- Department of Medicine, Hematology-Oncology, Rutgers New Jersey Medical School, Newark, NJ 07103, USA
| | - Andrew Petryna
- Department of Medicine, Hematology-Oncology, Rutgers New Jersey Medical School, Newark, NJ 07103, USA
- Rutgers School of Graduate Studies at New Jersey Medical School, Newark, NJ 07103, USA
| | - Steven M. Munoz
- Department of Medicine, Hematology-Oncology, Rutgers New Jersey Medical School, Newark, NJ 07103, USA
| | - Kimberly A. Silverio
- Department of Medicine, Hematology-Oncology, Rutgers New Jersey Medical School, Newark, NJ 07103, USA
- Rutgers School of Graduate Studies at New Jersey Medical School, Newark, NJ 07103, USA
| | - Maran Shaker
- Department of Medicine, Hematology-Oncology, Rutgers New Jersey Medical School, Newark, NJ 07103, USA
- Rutgers School of Graduate Studies at New Jersey Medical School, Newark, NJ 07103, USA
| | - Pujan Neopane
- Department of Medicine, Hematology-Oncology, Rutgers New Jersey Medical School, Newark, NJ 07103, USA
| | - Veronica Mariotti
- Department of Medicine, Hematology-Oncology, Rutgers New Jersey Medical School, Newark, NJ 07103, USA
| | - Pranela Rameshwar
- Department of Medicine, Hematology-Oncology, Rutgers New Jersey Medical School, Newark, NJ 07103, USA
| |
Collapse
|
24
|
Ruggieri L, Moretti A, Berardi R, Cona MS, Dalu D, Villa C, Chizzoniti D, Piva S, Gambaro A, La Verde N. Host-Related Factors in the Interplay among Inflammation, Immunity and Dormancy in Breast Cancer Recurrence and Prognosis: An Overview for Clinicians. Int J Mol Sci 2023; 24:4974. [PMID: 36902406 PMCID: PMC10002538 DOI: 10.3390/ijms24054974] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 02/28/2023] [Accepted: 03/02/2023] [Indexed: 03/08/2023] Open
Abstract
A significant proportion of patients treated for early breast cancer develop medium-term and late distant recurrence. The delayed manifestation of metastatic disease is defined as "dormancy". This model describes the aspects of the clinical latency of isolated metastatic cancer cells. Dormancy is regulated by extremely complex interactions between disseminated cancer cells and the microenvironment where they reside, the latter in turn influenced directly by the host. Among these entangled mechanisms, inflammation and immunity may play leading roles. This review is divided into two parts: the first describes the biological underpinnings of cancer dormancy and the role of the immune response, in particular, for breast cancer; the second provides an overview of the host-related factors that may influence systemic inflammation and immune response, subsequently impacting the dynamics of breast cancer dormancy. The aim of this review is to provide physicians and medical oncologists a useful tool to understand the clinical implications of this relevant topic.
Collapse
Affiliation(s)
- Lorenzo Ruggieri
- Medical Oncology Unit, Luigi Sacco University Hospital, ASST Fatebenefratelli-Sacco, Via G.B. Grassi, n° 74, 20157 Milan, Italy
| | - Anna Moretti
- Medical Oncology Unit, S. Carlo Hospital, ASST Santi Paolo e Carlo, 20153 Milan, Italy
| | - Rossana Berardi
- Department of Oncology, Università Politecnica delle Marche—AOU delle Marche, 60121 Ancona, Italy
| | - Maria Silvia Cona
- Medical Oncology Unit, Luigi Sacco University Hospital, ASST Fatebenefratelli-Sacco, Via G.B. Grassi, n° 74, 20157 Milan, Italy
| | - Davide Dalu
- Medical Oncology Unit, Luigi Sacco University Hospital, ASST Fatebenefratelli-Sacco, Via G.B. Grassi, n° 74, 20157 Milan, Italy
| | - Cecilia Villa
- Medical Oncology Unit, Luigi Sacco University Hospital, ASST Fatebenefratelli-Sacco, Via G.B. Grassi, n° 74, 20157 Milan, Italy
| | - Davide Chizzoniti
- Medical Oncology Unit, Luigi Sacco University Hospital, ASST Fatebenefratelli-Sacco, Via G.B. Grassi, n° 74, 20157 Milan, Italy
| | - Sheila Piva
- Medical Oncology Unit, Fatebenefratelli Hospital, ASST Fatebenefratelli-Sacco, 20157 Milan, Italy
| | - Anna Gambaro
- Medical Oncology Unit, Luigi Sacco University Hospital, ASST Fatebenefratelli-Sacco, Via G.B. Grassi, n° 74, 20157 Milan, Italy
| | - Nicla La Verde
- Medical Oncology Unit, Luigi Sacco University Hospital, ASST Fatebenefratelli-Sacco, Via G.B. Grassi, n° 74, 20157 Milan, Italy
| |
Collapse
|
25
|
Li D, Zhang X, Liu R, Long M, Zhou S, Lin J, Zhang L. Kainic acid induced hyperexcitability in thalamic reticular nucleus that initiates an inflammatory response through the HMGB1/TLR4 pathway. Neurotoxicology 2023; 95:94-106. [PMID: 36669621 DOI: 10.1016/j.neuro.2023.01.007] [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: 10/24/2022] [Revised: 01/11/2023] [Accepted: 01/12/2023] [Indexed: 01/20/2023]
Abstract
OBJECTIVE To explore the relationship between the proinflammatory factor high-mobility group box 1 (HMGB1) and glutamatergic alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors in the development of epilepsy. METHODS Thalamic reticular nucleus (TRN) slices were treated with kainic acid (KA) to simulate seizures. Action potentials and spontaneous inhibitory postsynaptic currents (sIPSCs) were recorded within TRN slices using whole-cell patch clamp techniques. The translocation of HMGB1 was detected by immunofluorescence. The HMGB1/TLR4 signaling pathway and its downstream inflammatory factors (IL-1β and NF-κB) were detected by RTPCR, Western blot and ELISA. RESULTS KA-evoked spikings were observed in TRN slices and blocked by perampanel. sIPSCs in the TRN were enhanced by KA and reduced by perampanel. The translocation of HMGB1 in the TRN was promoted by KA and inhibited by perampanel. The expression of the HMGB1/TLR4 signaling pathway was promoted by KA and suppressed by perampanel. CONCLUSION KA induced hyperexcitability activates the HMGB1/TLR4 pathway, which potentially leading to neuroinflammation in epilepsy.
Collapse
Affiliation(s)
- Dongbin Li
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, China; First Department of Neurology and Neuroscience Center, Heilongjiang Provincial Hospital, Harbin, China
| | - Xiaosi Zhang
- Metro-Medic Clinic, 1538 sherbrooke Ouest, suite 100, Montreal, QC H3G 1L5, Canada
| | - Ruoshi Liu
- Department of Neurology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Meixin Long
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Shanshan Zhou
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jinghan Lin
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Liming Zhang
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, China.
| |
Collapse
|
26
|
Dormancy, stemness, and therapy resistance: interconnected players in cancer evolution. Cancer Metastasis Rev 2023; 42:197-215. [PMID: 36757577 PMCID: PMC10014678 DOI: 10.1007/s10555-023-10092-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 01/26/2023] [Indexed: 02/10/2023]
Abstract
The biological complexity of cancer represents a tremendous clinical challenge, resulting in the frequent failure of current treatment protocols. In the rapidly evolving scenario of a growing tumor, anticancer treatments impose a drastic perturbation not only to cancer cells but also to the tumor microenvironment, killing a portion of the cells and inducing a massive stress response in the survivors. Consequently, treatments can act as a double-edged sword by inducing a temporary response while laying the ground for therapy resistance and subsequent disease progression. Cancer cell dormancy (or quiescence) is a central theme in tumor evolution, being tightly linked to the tumor's ability to survive cytotoxic challenges, metastasize, and resist immune-mediated attack. Accordingly, quiescent cancer cells (QCCs) have been detected in virtually all the stages of tumor development. In recent years, an increasing number of studies have focused on the characterization of quiescent/therapy resistant cancer cells, unveiling QCCs core transcriptional programs, metabolic plasticity, and mechanisms of immune escape. At the same time, our partial understanding of tumor quiescence reflects the difficulty to identify stable QCCs biomarkers/therapeutic targets and to control cancer dormancy in clinical settings. This review focuses on recent discoveries in the interrelated fields of dormancy, stemness, and therapy resistance, discussing experimental evidences in the frame of a nonlinear dynamics approach, and exploring the possibility that tumor quiescence may represent not only a peril but also a potential therapeutic resource.
Collapse
|
27
|
Zhou M, Zheng M, Zhou X, Tian S, Yang X, Ning Y, Li Y, Zhang S. The roles of connexins and gap junctions in the progression of cancer. Cell Commun Signal 2023; 21:8. [PMID: 36639804 PMCID: PMC9837928 DOI: 10.1186/s12964-022-01009-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 12/03/2022] [Indexed: 01/15/2023] Open
Abstract
Gap junctions (GJs), which are composed of connexins (Cxs), provide channels for direct information exchange between cells. Cx expression has a strong spatial specificity; however, its influence on cell behavior and information exchange between cells cannot be ignored. A variety of factors in organisms can modulate Cxs and subsequently trigger a series of responses that have important effects on cellular behavior. The expression and function of Cxs and the number and function of GJs are in dynamic change. Cxs have been characterized as tumor suppressors in the past, but recent studies have highlighted the critical roles of Cxs and GJs in cancer pathogenesis. The complex mechanism underlying Cx and GJ involvement in cancer development is a major obstacle to the evolution of therapy targeting Cxs. In this paper, we review the post-translational modifications of Cxs, the interactions of Cxs with several chaperone proteins, and the effects of Cxs and GJs on cancer. Video Abstract.
Collapse
Affiliation(s)
- Mingming Zhou
- Graduate School, Tianjin Medical University, Tianjin, 300070 People’s Republic of China
| | - Minying Zheng
- Department of Pathology, Tianjin Union Medical Center, Nankai University, Tianjin, 300121 People’s Republic of China
| | - Xinyue Zhou
- Graduate School, Tianjin Medical University, Tianjin, 300070 People’s Republic of China
| | - Shifeng Tian
- Graduate School, Tianjin Medical University, Tianjin, 300070 People’s Republic of China
| | - Xiaohui Yang
- Nankai University School of Medicine, Nankai University, Tianjin, 300071 People’s Republic of China
| | - Yidi Ning
- Nankai University School of Medicine, Nankai University, Tianjin, 300071 People’s Republic of China
| | - Yuwei Li
- Department of Colorectal Surgery, Tianjin Union Medical Center, Tianjin, 300121 People’s Republic of China
| | - Shiwu Zhang
- Department of Pathology, Tianjin Union Medical Center, Nankai University, Tianjin, 300121 People’s Republic of China
| |
Collapse
|
28
|
Kim R, Kawai A, Wakisaka M, Shimoyama M, Yasuda N, Kin T, Arihiro K. Breast cancer recurrence and survival rates in patients who underwent breast-conserving surgery under non-mechanically ventilated anesthesia. Cancer Rep (Hoboken) 2023; 6:e1643. [PMID: 35655440 PMCID: PMC9875645 DOI: 10.1002/cnr2.1643] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 02/25/2022] [Accepted: 05/12/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Recurrence after primary treatment is an important obstacle to the curing of primary breast cancer. Less-immunosuppressive anesthetic techniques, such as local anesthesia with lidocaine, intravenous anesthesia (IVA) with propofol, and/or sedation with midazolam under spontaneous breathing may reduce breast cancer recurrence compared with standard general anesthesia techniques such as IVA and inhalation anesthesia with opioids under mechanical ventilation. AIM The aim of this study was to analyze the factors involved in breast cancer recurrence in patients who underwent breast-conserving surgery (BCS) under non-mechanically ventilated anesthesia. METHODS The study included 491 consecutive patients with stages 0-III breast cancer who underwent BCS/axillary lymph-node management with local anesthesia and IVA and/or sedation under non-mechanical ventilation between May 2008 and September 2021. Survival and recurrence were assessed by retrospective cohort analysis. RESULTS The median follow-up period was 2565 days (range, 28-4834 days). The overall and breast cancer-specific survival rates were 92.9% and 95.6%, respectively. Twenty-one deaths, of which 11 were breast cancer-related, occurred. Disease recurred in 29 (5.9%) patients, of whom 15 patients received neoadjuvant chemotherapy (NAC) and 14 patients received adjuvant therapy (chemotherapy in 12 cases). The surgical procedure performed, but not other clinicopathological factors [recurrence site, P stage, tumor subtype, and disease-free interval (DFI)], differed between the NAC and adjuvant therapy groups. The DFI tended to be shorter in the NAC group than in the adjuvant therapy group. The pathological therapeutic effect grade after NAC was 1 in 12 patients and ≥2 in 3 patients. CONCLUSION More than 50% (15/29) of patients with recurrence who underwent BCS were given NAC, but most patients did not respond to it. Similarly, adjuvant chemotherapy may not have contributed to the eradication of residual tumor cells after BCS. To reduce breast cancer recurrence in patients undergoing BCS, treatment strategies, especially for patients who do not respond to NAC or adjuvant chemotherapy, need to be developed. Non-mechanical ventilation anesthesia may also affect the incidence of breast cancer recurrence.
Collapse
Affiliation(s)
- Ryungsa Kim
- Department of Breast SurgeryHiroshima Mark ClinicHiroshimaJapan
| | - Ami Kawai
- Department of Breast SurgeryHiroshima Mark ClinicHiroshimaJapan
| | - Megumi Wakisaka
- Department of Breast SurgeryHiroshima Mark ClinicHiroshimaJapan
| | - Mika Shimoyama
- Department of Breast SurgeryHiroshima Mark ClinicHiroshimaJapan
| | - Naomi Yasuda
- Department of Breast SurgeryHiroshima Mark ClinicHiroshimaJapan
| | - Takanori Kin
- Department of Breast SurgeryHiroshima City HospitalHiroshimaJapan
| | - Koji Arihiro
- Department of Anatomical PathologyHiroshima University HospitalHiroshimaJapan
| |
Collapse
|
29
|
Ollodart J, Contino KF, Deep G, Shiozawa Y. The impacts of exosomes on bone metastatic progression and their potential clinical utility. Bone Rep 2022; 17:101606. [PMID: 35910404 PMCID: PMC9335387 DOI: 10.1016/j.bonr.2022.101606] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 07/05/2022] [Accepted: 07/18/2022] [Indexed: 11/10/2022] Open
Abstract
Bone is one of the most common sites of cancer metastasis. Once cancer metastasizes to the bone, the mortality rate of cancer patients dramatically increases. Although the exact mechanisms for this observation remain elusive, recent studies have revealed that the complex crosstalk between bone marrow microenvironment and bone metastatic cancer cells is responsible for the induction of treatment resistance. Consequently, bone metastasis is currently considered incurable. Bone metastasis not only impairs the patients' survival, but also negatively affects their quality of life by causing painful complications. It has recently been implicated the regulatory role of exosomes in cancer development and/or progression as a delivery biomaterial between cancer cells and tumor microenvironment. However, little is known as to how exosomes contribute to the progression of bone metastasis by impaction on the crosstalk between bone metastatic cancer cells and bone marrow microenvironment. Here, we highlighted the emerging roles of cancer-derived exosomes in (i) the process of dissemination and bone colonization of bone metastatic cancer cells, (ii) the enhancement of crosstalk between bone marrow microenvironment and bone metastatic cancer cells, (iii) the development of its resultant painful complications, and (iv) the clinical applications of exosomes in the bone metastatic setting. Cancer-derived exosomes facilitate cancer dissemination and colonization to bone. Cancer-derived exosomes are crucial for controlling bone metastatic phenotype. Cancer-derived exosomes prime bone marrow microenvironment for further metastasis. Cancer-derived exosomes are involved in development of cancer-induced bone pain. Exosomes can be used as therapies and/or diagnostic tools for bone metastasis.
Collapse
Affiliation(s)
- Jenna Ollodart
- Department of Cancer Biology and Comprehensive Cancer Center, Wake Forest University Health Sciences, Winston-Salem, NC 27157, USA
| | - Kelly F Contino
- Department of Cancer Biology and Comprehensive Cancer Center, Wake Forest University Health Sciences, Winston-Salem, NC 27157, USA
| | - Gagan Deep
- Department of Cancer Biology and Comprehensive Cancer Center, Wake Forest University Health Sciences, Winston-Salem, NC 27157, USA
| | - Yusuke Shiozawa
- Department of Cancer Biology and Comprehensive Cancer Center, Wake Forest University Health Sciences, Winston-Salem, NC 27157, USA
| |
Collapse
|
30
|
Alptekin A, Parvin M, Chowdhury HI, Rashid MH, Arbab AS. Engineered exosomes for studies in tumor immunology. Immunol Rev 2022; 312:76-102. [PMID: 35808839 DOI: 10.1111/imr.13107] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 05/27/2022] [Indexed: 12/14/2022]
Abstract
Exosomes are a type of extracellular vesicle (EV) with diameters of 30-150 nm secreted by most of the cells into the extracellular spaces and can alter the microenvironment through cell-to-cell interactions by fusion with the plasma membrane and subsequent endocytosis and release of the cargo. Because of their biocompatibility, low toxicity and immunogenicity, permeability (even through the blood-brain barrier (BBB)), stability in biological fluids, and ability to accumulate in the lesions with higher specificity, investigators have started making designer's exosomes or engineered exosomes to carry biologically active protein on the surface or inside the exosomes as well as using exosomes to carry drugs, micro RNA, and other products to the site of interest. In this review, we have discussed biogenesis, markers, and contents of various exosomes including exosomes of immune cells. We have also discussed the current methods of making engineered and designer's exosomes as well as the use of engineered exosomes targeting different immune cells in the tumors, stroke, as well as at peripheral blood. Genetic engineering and customizing exosomes create an unlimited opportunity to use in diagnosis and treatment. Very little use has been discovered, and we are far away to reach its limits.
Collapse
Affiliation(s)
- Ahmet Alptekin
- Georgia Cancer Center, Augusta University, Augusta, Georgia, USA
| | - Mahrima Parvin
- Georgia Cancer Center, Augusta University, Augusta, Georgia, USA
| | | | | | - Ali S Arbab
- Georgia Cancer Center, Augusta University, Augusta, Georgia, USA
| |
Collapse
|
31
|
Lucotti S, Kenific CM, Zhang H, Lyden D. Extracellular vesicles and particles impact the systemic landscape of cancer. EMBO J 2022; 41:e109288. [PMID: 36052513 PMCID: PMC9475536 DOI: 10.15252/embj.2021109288] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Revised: 02/16/2022] [Accepted: 03/23/2022] [Indexed: 11/09/2022] Open
Abstract
Intercellular cross talk between cancer cells and stromal and immune cells is essential for tumor progression and metastasis. Extracellular vesicles and particles (EVPs) are a heterogeneous class of secreted messengers that carry bioactive molecules and that have been shown to be crucial for this cell-cell communication. Here, we highlight the multifaceted roles of EVPs in cancer. Functionally, transfer of EVP cargo between cells influences tumor cell growth and invasion, alters immune cell composition and function, and contributes to stromal cell activation. These EVP-mediated changes impact local tumor progression, foster cultivation of pre-metastatic niches at distant organ-specific sites, and mediate systemic effects of cancer. Furthermore, we discuss how exploiting the highly selective enrichment of molecules within EVPs has profound implications for advancing diagnostic and prognostic biomarker development and for improving therapy delivery in cancer patients. Altogether, these investigations into the role of EVPs in cancer have led to discoveries that hold great promise for improving cancer patient care and outcome.
Collapse
Affiliation(s)
- Serena Lucotti
- Children’s Cancer and Blood Foundation Laboratories, Departments of Pediatrics, and Cell and Developmental Biology, Drukier Institute for Children’s Health, Meyer Cancer CenterWeill Cornell MedicineNew YorkNYUSA
| | - Candia M Kenific
- Children’s Cancer and Blood Foundation Laboratories, Departments of Pediatrics, and Cell and Developmental Biology, Drukier Institute for Children’s Health, Meyer Cancer CenterWeill Cornell MedicineNew YorkNYUSA
| | - Haiying Zhang
- Children’s Cancer and Blood Foundation Laboratories, Departments of Pediatrics, and Cell and Developmental Biology, Drukier Institute for Children’s Health, Meyer Cancer CenterWeill Cornell MedicineNew YorkNYUSA
| | - David Lyden
- Children’s Cancer and Blood Foundation Laboratories, Departments of Pediatrics, and Cell and Developmental Biology, Drukier Institute for Children’s Health, Meyer Cancer CenterWeill Cornell MedicineNew YorkNYUSA
| |
Collapse
|
32
|
Tamura T, Yoshioka Y, Sakamoto S, Ichikawa T, Ochiya T. Extracellular vesicles in bone homeostasis: key roles of physiological and pathological conditions. J Bone Miner Metab 2022; 41:345-357. [PMID: 35943593 DOI: 10.1007/s00774-022-01362-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 07/08/2022] [Indexed: 11/25/2022]
Abstract
Extracellular vesicles (EVs) are small particles with lipid bilayer membranes that are secreted by all cell types and are widely known as crucial intercellular communication mediators, shuttling biologically active molecules. The bone is a typically preferred site of cancer metastasis due to its unique cellular compositions and dynamics. Bone cell-derived EVs serve as regulators that orchestrate harmonious bone homeostasis. Cancer cells secrete specific EVs in a series of the bone metastatic process to dominate the bone microenvironment. Additionally, cancer cell-related EVs contribute to pre-metastatic niche formation, bone homeostasis disruption, and tumor bone progression and survival. Here, we investigated recent studies on EV-mediated crosstalk in the bone tumor microenvironment. Furthermore, this review aimed to elucidate the EV-based therapeutic perspectives for bone metastasis.
Collapse
Affiliation(s)
- Takaaki Tamura
- Department of Molecular and Cellular Medicine, Institute of Medical Science, Tokyo Medical University, Tokyo, 160-0023, Japan
- Department of Urology, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan
| | - Yusuke Yoshioka
- Department of Molecular and Cellular Medicine, Institute of Medical Science, Tokyo Medical University, Tokyo, 160-0023, Japan
| | - Shinichi Sakamoto
- Department of Urology, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan
| | - Tomohiko Ichikawa
- Department of Urology, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan
| | - Takahiro Ochiya
- Department of Molecular and Cellular Medicine, Institute of Medical Science, Tokyo Medical University, Tokyo, 160-0023, Japan.
| |
Collapse
|
33
|
Stromal Co-Cultivation for Modeling Breast Cancer Dormancy in the Bone Marrow. Cancers (Basel) 2022; 14:cancers14143344. [PMID: 35884405 PMCID: PMC9320268 DOI: 10.3390/cancers14143344] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 07/05/2022] [Accepted: 07/05/2022] [Indexed: 11/16/2022] Open
Abstract
Cancers metastasize to the bone marrow before primary tumors can be detected. Bone marrow micrometastases are resistant to therapy, and while they are able to remain dormant for decades, they recur steadily and result in incurable metastatic disease. The bone marrow microenvironment maintains the dormancy and chemoresistance of micrometastases through interactions with multiple cell types and through structural and soluble factors. Modeling dormancy in vitro can identify the mechanisms of these interactions. Modeling also identifies mechanisms able to disrupt these interactions or define novel interactions that promote the reawakening of dormant cells. The in vitro modeling of the interactions of cancer cells with various bone marrow elements can generate hypotheses on the mechanisms that control dormancy, treatment resistance and reawakening in vivo. These hypotheses can guide in vivo murine experiments that have high probabilities of succeeding in order to verify in vitro findings while minimizing the use of animals in experiments. This review outlines the existing data on predominant stromal cell types and their use in 2D co-cultures with cancer cells.
Collapse
|
34
|
Xavier CPR, Belisario DC, Rebelo R, Assaraf YG, Giovannetti E, Kopecka J, Vasconcelos MH. The role of extracellular vesicles in the transfer of drug resistance competences to cancer cells. Drug Resist Updat 2022; 62:100833. [PMID: 35429792 DOI: 10.1016/j.drup.2022.100833] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Revised: 02/20/2022] [Accepted: 03/13/2022] [Indexed: 02/07/2023]
Abstract
Drug resistance remains a major hurdle to successful cancer treatment, being accountable for approximately 90% of cancer-related deaths. In the past years, increasing attention has been given to the role of extracellular vesicles (EVs) in the horizontal transfer of drug resistance in cancer. Indeed, many studies have described the dissemination of therapy resistance traits mediated by EVs, which may be transferred from drug resistant tumor cells to their drug sensitive counterparts. Importantly, different key players of drug resistance have been identified in the cargo of those EVs, such as drug efflux pumps, oncoproteins, antiapoptotic proteins, or microRNAs, among others. Interestingly, the EVs-mediated crosstalk between cells from the tumor microenvironment (TME) and tumor cells has emerged as another important mechanism that leads to cancer cells drug resistance. Recently, the cargo of the TME-derived EVs responsible for the transfer of drug resistance traits has also become a focus of attention. In addition, the possible mechanisms involved in drug sequestration by EVs, likely to contribute to cancer drug resistance, are also described and discussed herein. Despite the latest scientific advances in the field of EVs, this is still a challenging area of research, particularly in the clinical setting. Therefore, further investigation is needed to assess the relevance of EVs to the failure of cancer patients to drug treatment, to identify biomarkers of drug resistance in the EV's cargo, and to develop effective therapeutic strategies to surmount drug resistance. This up-to-date review summarizes relevant literature on the role of EVs in the transfer of drug resistance competences to cancer cells, and the relevance of tumor cells and of TME cells in this process. Finally, this knowledge is integrated with a discussion of possible future clinical applications of EVs as biomarkers of drug resistance.
Collapse
Affiliation(s)
- Cristina P R Xavier
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; Cancer Drug Resistance Group, IPATIMUP - Institute of Molecular Pathology and Immunology, University of Porto, 4200-135 Porto, Portugal.
| | | | - Rita Rebelo
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; Cancer Drug Resistance Group, IPATIMUP - Institute of Molecular Pathology and Immunology, University of Porto, 4200-135 Porto, Portugal.
| | - Yehuda G Assaraf
- The Fred Wyszkowski Cancer Research Laboratory, Department of Biology, Technion-Israel Institute of Technology, Haifa 3200000, Israel.
| | - Elisa Giovannetti
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands; Fondazione Pisana per La Scienza, Pisa, Italy.
| | | | - M Helena Vasconcelos
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; Cancer Drug Resistance Group, IPATIMUP - Institute of Molecular Pathology and Immunology, University of Porto, 4200-135 Porto, Portugal; Department of Biological Sciences, FFUP - Faculty of Pharmacy of the University of Porto, Porto, Portugal.
| |
Collapse
|
35
|
Altei WF, Pachane BC, Souza C, Marques MMC, Selistre-de-Araújo H. New insights into the discovery of drugs for triple-negative breast cancer metastasis. Expert Opin Drug Discov 2022; 17:365-376. [PMID: 35179448 DOI: 10.1080/17460441.2022.2039619] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
INTRODUCTION Triple-negative breast cancer (TNBC) is of great concern due to its aggressiveness and lack of targeted therapy. For these reasons, TNBC is one of the main causes of death in women, mainly due to metastases. Tumor dissemination has highlighted a set of possible targets, with extensive research into new single-target drugs, in addition to drug repurposing strategies, being undertaken to discover new classes of potential inhibitors of metastasis. AREAS COVERED The authors here describe the main proposed targets and the bases of their pharmacological inhibition with different chemical compounds. The authors also discuss the state-of-the-art from the latest clinical trials and highlight other potential targets for metastatic TNBC. EXPERT OPINION In the last decade, oncology research has changed its focus from primary tumors to moving tumor cells, their products, and to the secondary tumor and its surroundings, for the purpose of finding targets to treat metastasis. Consequently, our comprehension of the complexity of the metastatic process has increased drastically, with, furthermore, the discovery of new potential targets. Although promising, the wide range of strategies is still not effective to suppress TNBC metastasis in terms of increasing patient survival or decreasing the number of metastases. Treating or preventing metastasis continues to be a great challenge.
Collapse
Affiliation(s)
- Wanessa Fernanda Altei
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, Brazil.,Radiotherapy Department, Barretos Cancer Hospital, Barretos, Brazil
| | - Bianca Cruz Pachane
- Graduate Program of Evolutionary Genetics and Molecular Biology, Federal University of São Carlos, São Carlos, Brazil
| | - Cristiano Souza
- Department of Clinical Oncology, Barretos Cancer Hospital, Barretos, Brazil
| | - Márcia Maria Chiquitelli Marques
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, Brazil.,Barretos School of Health Sciences, Dr. Paulo Prata-FACISB, Barretos, Brazil
| | | |
Collapse
|
36
|
Xiao M, He J, Yin L, Chen X, Zu X, Shen Y. Tumor-Associated Macrophages: Critical Players in Drug Resistance of Breast Cancer. Front Immunol 2022; 12:799428. [PMID: 34992609 PMCID: PMC8724912 DOI: 10.3389/fimmu.2021.799428] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 12/01/2021] [Indexed: 12/11/2022] Open
Abstract
Drug resistance is one of the most critical challenges in breast cancer (BC) treatment. The occurrence and development of drug resistance are closely related to the tumor immune microenvironment (TIME). Tumor-associated macrophages (TAMs), the most important immune cells in TIME, are essential for drug resistance in BC treatment. In this article, we summarize the effects of TAMs on the resistance of various drugs in endocrine therapy, chemotherapy, targeted therapy, and immunotherapy, and their underlying mechanisms. Based on the current overview of the key role of TAMs in drug resistance, we discuss the potential possibility for targeting TAMs to reduce drug resistance in BC treatment, By inhibiting the recruitment of TAMs, depleting the number of TAMs, regulating the polarization of TAMs and enhancing the phagocytosis of TAMs. Evidences in our review support it is important to develop novel therapeutic strategies to target TAMs in BC to overcome the treatment of resistance.
Collapse
Affiliation(s)
- Maoyu Xiao
- Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Jun He
- Department of Spine Surgery, The Nanhua Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Liyang Yin
- Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Xiguan Chen
- Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Xuyu Zu
- Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Yingying Shen
- Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| |
Collapse
|
37
|
Metabolic Features of Tumor Dormancy: Possible Therapeutic Strategies. Cancers (Basel) 2022; 14:cancers14030547. [PMID: 35158815 PMCID: PMC8833651 DOI: 10.3390/cancers14030547] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/17/2022] [Accepted: 01/19/2022] [Indexed: 02/01/2023] Open
Abstract
Simple Summary Tumor recurrence still represents a major clinical challenge for cancer patients. Cancer cells may undergo a dormant state for long times before re-emerging. Both intracellular- and extracellular-driven pathways are involved in maintaining the dormant state and the subsequent awakening, with a mechanism that is still mostly unknown. In this scenario, cancer metabolism is emerging as a critical driver of tumor progression and dissemination and have gained increasing attention in cancer research. This review focuses on the metabolic adaptations characterizing the dormant phenotype and supporting tumor re-growth. Deciphering the metabolic adaptation sustaining tumor dormancy may pave the way for novel therapeutic approaches to prevent tumor recurrence based on combined metabolic drugs. Abstract Tumor relapse represents one of the main obstacles to cancer treatment. Many patients experience cancer relapse even decades from the primary tumor eradication, developing more aggressive and metastatic disease. This phenomenon is associated with the emergence of dormant cancer cells, characterized by cell cycle arrest and largely insensitive to conventional anti-cancer therapies. These rare and elusive cells may regain proliferative abilities upon the induction of cell-intrinsic and extrinsic factors, thus fueling tumor re-growth and metastasis formation. The molecular mechanisms underlying the maintenance of resistant dormant cells and their awakening are intriguing but, currently, still largely unknown. However, increasing evidence recently underlined a strong dependency of cell cycle progression to metabolic adaptations of cancer cells. Even if dormant cells are frequently characterized by a general metabolic slowdown and an increased ability to cope with oxidative stress, different factors, such as extracellular matrix composition, stromal cells influence, and nutrient availability, may dictate specific changes in dormant cells, finally resulting in tumor relapse. The main topic of this review is deciphering the role of the metabolic pathways involved in tumor cells dormancy to provide new strategies for selectively targeting these cells to prevent fatal recurrence and maximize therapeutic benefit.
Collapse
|
38
|
Extracellular Vesicles as Mediators of Therapy Resistance in the Breast Cancer Microenvironment. Biomolecules 2022; 12:biom12010132. [PMID: 35053279 PMCID: PMC8773878 DOI: 10.3390/biom12010132] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/07/2022] [Accepted: 01/11/2022] [Indexed: 12/17/2022] Open
Abstract
Resistance to various therapies, including novel immunotherapies, poses a major challenge in the management of breast cancer and is the leading cause of treatment failure. Bidirectional communication between breast cancer cells and the tumour microenvironment is now known to be an important contributor to therapy resistance. Several studies have demonstrated that crosstalk with the tumour microenvironment through extracellular vesicles is an important mechanism employed by cancer cells that leads to drug resistance via changes in protein, lipid and nucleic acid cargoes. Moreover, the cargo content enables extracellular vesicles to be used as effective biomarkers for predicting response to treatments and as potential therapeutic targets. This review summarises the literature to date regarding the role of extracellular vesicles in promoting therapy resistance in breast cancer through communication with the tumour microenvironment.
Collapse
|
39
|
Hourani T, Holden JA, Li W, Lenzo JC, Hadjigol S, O’Brien-Simpson NM. Tumor Associated Macrophages: Origin, Recruitment, Phenotypic Diversity, and Targeting. Front Oncol 2021; 11:788365. [PMID: 34988021 PMCID: PMC8722774 DOI: 10.3389/fonc.2021.788365] [Citation(s) in RCA: 86] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Accepted: 11/30/2021] [Indexed: 12/20/2022] Open
Abstract
The tumor microenvironment (TME) is known to have a strong influence on tumorigenesis, with various components being involved in tumor suppression and tumor growth. A protumorigenic TME is characterized by an increased infiltration of tumor associated macrophages (TAMs), where their presence is strongly associated with tumor progression, therapy resistance, and poor survival rates. This association between the increased TAMs and poor therapeutic outcomes are stemming an increasing interest in investigating TAMs as a potential therapeutic target in cancer treatment. Prominent mechanisms in targeting TAMs include: blocking recruitment, stimulating repolarization, and depletion methods. For enhancing targeting specificity multiple nanomaterials are currently being explored for the precise delivery of chemotherapeutic cargo, including the conjugation with TAM-targeting peptides. In this paper, we provide a focused literature review of macrophage biology in relation to their role in tumorigenesis. First, we discuss the origin, recruitment mechanisms, and phenotypic diversity of TAMs based on recent investigations in the literature. Then the paper provides a detailed review on the current methods of targeting TAMs, including the use of nanomaterials as novel cancer therapeutics.
Collapse
Affiliation(s)
| | | | | | | | | | - Neil M. O’Brien-Simpson
- Antimicrobial, Cancer Therapeutics and Vaccines (ACTV) Research Group, Melbourne Dental School, Centre for Oral Health Research, Royal Dental Hospital, The University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
40
|
Moore CA, Ferrer AI, Alonso S, Pamarthi SH, Sandiford OA, Rameshwar P. Exosomes in the Healthy and Malignant Bone Marrow Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1350:67-89. [PMID: 34888844 DOI: 10.1007/978-3-030-83282-7_3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The bone marrow (BM) is a complex organ that sustains hematopoiesis via mechanisms involving the microenvironment. The microenvironment includes several cell types, neurotransmitters from innervated fibers, growth factors, extracellular matrix proteins, and extracellular vesicles. The main function of the BM is to regulate hematopoietic function to sustain the production of blood and immune cells. However, the BM microenvironment can also accommodate the survival of malignant cells. A major mechanism by which the cancer cells communicate with cells of the BM microenvironment is through the exchange of exosomes, a subset of extracellular vesicles that deliver molecular signals bidirectionally between malignant and healthy cells. The field of exosomes is an active area of investigation since an understanding of how the exosomal packaging, cargo, and production can be leveraged therapeutically to deter cancer progression and sensitize malignant cells to other therapies. Altogether, this chapter discusses the crucial role of exosomes in the development and progression of BM-associated cancers, such as hematologic malignancies and marrow-metastatic breast cancer. Exosome-based therapeutic strategies and their limitations are also considered.
Collapse
Affiliation(s)
- Caitlyn A Moore
- Rutgers New Jersey Medical School, Rutgers University, Newark, NJ, United States
- Rutgers School of Graduate Studies at New Jersey Medical School, Rutgers University, Newark, NJ, United States
| | - Alejandra I Ferrer
- Rutgers New Jersey Medical School, Rutgers University, Newark, NJ, United States
- Rutgers School of Graduate Studies at New Jersey Medical School, Rutgers University, Newark, NJ, United States
| | - Sara Alonso
- Rutgers School of Graduate Studies at New Jersey Medical School, Rutgers University, Newark, NJ, United States
| | - Sri Harika Pamarthi
- Rutgers New Jersey Medical School, Rutgers University, Newark, NJ, United States
| | - Oleta A Sandiford
- Rutgers New Jersey Medical School, Rutgers University, Newark, NJ, United States
- Rutgers School of Graduate Studies at New Jersey Medical School, Rutgers University, Newark, NJ, United States
| | - Pranela Rameshwar
- Rutgers New Jersey Medical School, Rutgers University, Newark, NJ, United States.
- Rutgers School of Graduate Studies at New Jersey Medical School, Rutgers University, Newark, NJ, United States.
| |
Collapse
|
41
|
Wang X, Sun C, Huang X, Li J, Fu Z, Li W, Yin Y. The Advancing Roles of Exosomes in Breast Cancer. Front Cell Dev Biol 2021; 9:731062. [PMID: 34790660 PMCID: PMC8591197 DOI: 10.3389/fcell.2021.731062] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Accepted: 09/27/2021] [Indexed: 12/12/2022] Open
Abstract
Breast cancer (BC) develops from breast tissue and is the most common aggressive malignant tumor in women worldwide. Although advanced treatment strategies have been applied and reduced current mortality rates, BC control remains unsatisfactory. It is essential to elucidate the underlying molecular mechanisms to assist clinical options. Exosomes are a type of extracellular vesicles and mediate cellular communications by delivering various biomolecules (oncogenes, oncomiRs, proteins, and even pharmacological compounds). These bioactive molecules can be transferred to change the transcriptome of target cells and influence tumor-related signaling pathways. Extensive studies have implicated exosomes in BC biology, including therapeutic resistance and the surrounding microenvironment. This review focuses on discussing the functions of exosomes in tumor treatment resistance, invasion and metastasis of BC. Moreover, we will also summarize multiple interactions between exosomes and the BC tumor microenvironment. Finally, we propose promising clinical applications of exosomes in BC.
Collapse
Affiliation(s)
- Xi Wang
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,The First School of Clinical Medicine, Nanjing Medical University, Nanjing, China
| | - Chunxiao Sun
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xiang Huang
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jun Li
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Ziyi Fu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Nanjing Maternity and Child Medical Institute, Obstetrics and Gynecology Hospital, Nanjing Medical University, Nanjing, China
| | - Wei Li
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yongmei Yin
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
42
|
Batoon L, McCauley LK. Cross Talk Between Macrophages and Cancer Cells in the Bone Metastatic Environment. Front Endocrinol (Lausanne) 2021; 12:763846. [PMID: 34803925 PMCID: PMC8597897 DOI: 10.3389/fendo.2021.763846] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 10/11/2021] [Indexed: 12/12/2022] Open
Abstract
The skeleton is a common site for cancer metastases with the bone microenvironment providing the appropriate conditions for cancer cell colonization. Once in bone, cancer cells effectively manipulate their microenvironment to support their growth and survival. Despite previous efforts to improve treatment modalities, skeletal metastases remain with poor prognoses. This warrants an improved understanding of the mechanisms leading to bone metastasis that will aid development of effective treatments. Macrophages in the tumor microenvironment are termed tumor associated macrophages (TAMs) and their crosstalk with cancer cells is critical in regulating tumorigenicity in multiple cancers. In bone metastases, this crosstalk is also being increasingly implicated but the specific signaling pathways remain incompletely understood. Here, we summarize the reported functions, interactions, and signaling of macrophages with cancer cells during the metastatic cascade to bone. Specifically, we review and discuss how these specific interactions impact macrophages and their profiles to promote tumor development. We also discuss the potential of targeting this crosstalk to inhibit disease progression. Finally, we identify the remaining knowledge gaps that will need to be addressed in order to fully consider therapeutic targeting to improve clinical outcomes in cancer patients.
Collapse
Affiliation(s)
- Lena Batoon
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI, United States
- Bones and Immunology Group, Mater Research Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Laurie K. McCauley
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI, United States
| |
Collapse
|
43
|
Ghoroghi S, Mary B, Asokan N, Goetz JG, Hyenne V. Tumor extracellular vesicles drive metastasis (it's a long way from home). FASEB Bioadv 2021; 3:930-943. [PMID: 34761175 PMCID: PMC8565230 DOI: 10.1096/fba.2021-00079] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Accepted: 07/27/2021] [Indexed: 12/13/2022] Open
Abstract
Among a plethora of functions, extracellular vesicles released by primary tumors spread in the organism and reach distant organs where they can induce the formation of a premetastatic niche. This constitutes a favorable microenvironment for circulating tumor cells which facilitates their seeding and colonization. In this review, we describe the journey of extracellular vesicles (EVs) from the primary tumor to the future metastatic organ, with a focus on the mechanisms used by EVs to target organs with a specific tropism (i.e., organotropism). We then highlight important tumor EV cargos in the context of premetastatic niche formation and summarize their known effects on extracellular matrix remodeling, angiogenesis, vessel permeabilization, resident cell activation, recruitment of foreign cells, and ultimately the formation of a pro-inflammatory and immuno-tolerant microenvironment. Finally, we discuss current experimental limitations and remaining opened questions in light of metastatic diagnosis and potential therapies targeting PMN formation.
Collapse
Affiliation(s)
- Shima Ghoroghi
- Tumor Biomechanics INSERM UMR_S1109 Strasbourg France
- Université de Strasbourg Strasbourg France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS) Strasbourg France
- Equipe Labellisée Ligue Contre le Cancer Strasbourg France
| | - Benjamin Mary
- Tumor Biomechanics INSERM UMR_S1109 Strasbourg France
- Université de Strasbourg Strasbourg France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS) Strasbourg France
- Equipe Labellisée Ligue Contre le Cancer Strasbourg France
| | - Nandini Asokan
- Tumor Biomechanics INSERM UMR_S1109 Strasbourg France
- Université de Strasbourg Strasbourg France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS) Strasbourg France
- Equipe Labellisée Ligue Contre le Cancer Strasbourg France
| | - Jacky G Goetz
- Tumor Biomechanics INSERM UMR_S1109 Strasbourg France
- Université de Strasbourg Strasbourg France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS) Strasbourg France
- Equipe Labellisée Ligue Contre le Cancer Strasbourg France
| | - Vincent Hyenne
- Tumor Biomechanics INSERM UMR_S1109 Strasbourg France
- Université de Strasbourg Strasbourg France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS) Strasbourg France
- Equipe Labellisée Ligue Contre le Cancer Strasbourg France
- CNRS SNC5055 Strasbourg France
| |
Collapse
|
44
|
Cappariello A, Rucci N. Extracellular Vesicles in Bone Tumors: How to Seed in the Surroundings Molecular Information for Malignant Transformation and Progression. Front Oncol 2021; 11:722922. [PMID: 34616676 PMCID: PMC8488258 DOI: 10.3389/fonc.2021.722922] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 08/26/2021] [Indexed: 12/19/2022] Open
Abstract
Bone is a very dynamic tissue hosting different cell types whose functions are regulated by a plethora of membrane-bound and soluble molecules. Intercellular communication was recently demonstrated to be also sustained by the exchange of extracellular vesicles (EVs). These are cell-derived nanosized structures shuttling biologically active molecules, such as nucleic acids and proteins. The bone microenvironment is a preferential site of primary and metastatic tumors, in which cancer cells find a fertile soil to “seed and blossom”. Nowadays, many oncogenic processes are recognized to be sustained by EVs. For example, EVs can directly fuel the vicious cycle in the bone/bone marrow microenvironment. EVs create a favourable environment for tumor growth by affecting osteoblasts, osteoclasts, osteocytes, adipocytes, leukocytes, and endothelial cells. At the same time other crucial tumor-mediated events, such as the premetastatic niche formation, tumor cell dormancy, as well as drug resistance, have been described to be fostered by tumor-derived EVs. In this review, we will discuss the main body of literature describing how the cancer cells use the EVs for their growth into the bone and for educating the bone microenvironment to host metastases.
Collapse
Affiliation(s)
- Alfredo Cappariello
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Nadia Rucci
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| |
Collapse
|
45
|
da Costa VR, Araldi RP, Vigerelli H, D’Ámelio F, Mendes TB, Gonzaga V, Policíquio B, Colozza-Gama GA, Valverde CW, Kerkis I. Exosomes in the Tumor Microenvironment: From Biology to Clinical Applications. Cells 2021; 10:2617. [PMID: 34685596 PMCID: PMC8533895 DOI: 10.3390/cells10102617] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 08/25/2021] [Accepted: 08/26/2021] [Indexed: 12/12/2022] Open
Abstract
Cancer is one of the most important health problems and the second leading cause of death worldwide. Despite the advances in oncology, cancer heterogeneity remains challenging to therapeutics. This is because the exosome-mediated crosstalk between cancer and non-cancer cells within the tumor microenvironment (TME) contributes to the acquisition of all hallmarks of cancer and leads to the formation of cancer stem cells (CSCs), which exhibit resistance to a range of anticancer drugs. Thus, this review aims to summarize the role of TME-derived exosomes in cancer biology and explore the clinical potential of mesenchymal stem-cell-derived exosomes as a cancer treatment, discussing future prospects of cell-free therapy for cancer treatment and challenges to be overcome.
Collapse
Affiliation(s)
- Vitor Rodrigues da Costa
- Programa de Pós-Graduação em Biologia Estrutural e Funcional, Escola Paulista de Medicina (EPM), Federal University of São Paulo (UNIFES), São Paulo 04039-032, Brazil; (V.R.d.C.); (T.B.M.); (G.A.C.-G.)
- Genetics Laboratory, Instituto Butantan, São Paulo 05508-010, Brazil; (H.V.); (F.D.); (V.G.); (B.P.)
| | - Rodrigo Pinheiro Araldi
- Programa de Pós-Graduação em Biologia Estrutural e Funcional, Escola Paulista de Medicina (EPM), Federal University of São Paulo (UNIFES), São Paulo 04039-032, Brazil; (V.R.d.C.); (T.B.M.); (G.A.C.-G.)
- Genetics Laboratory, Instituto Butantan, São Paulo 05508-010, Brazil; (H.V.); (F.D.); (V.G.); (B.P.)
- Cellavita Pesquisas Científicas Ltd.a., Valinhos 13271-650, Brazil;
| | - Hugo Vigerelli
- Genetics Laboratory, Instituto Butantan, São Paulo 05508-010, Brazil; (H.V.); (F.D.); (V.G.); (B.P.)
| | - Fernanda D’Ámelio
- Genetics Laboratory, Instituto Butantan, São Paulo 05508-010, Brazil; (H.V.); (F.D.); (V.G.); (B.P.)
| | - Thais Biude Mendes
- Programa de Pós-Graduação em Biologia Estrutural e Funcional, Escola Paulista de Medicina (EPM), Federal University of São Paulo (UNIFES), São Paulo 04039-032, Brazil; (V.R.d.C.); (T.B.M.); (G.A.C.-G.)
- Genetics Laboratory, Instituto Butantan, São Paulo 05508-010, Brazil; (H.V.); (F.D.); (V.G.); (B.P.)
- Cellavita Pesquisas Científicas Ltd.a., Valinhos 13271-650, Brazil;
| | - Vivian Gonzaga
- Genetics Laboratory, Instituto Butantan, São Paulo 05508-010, Brazil; (H.V.); (F.D.); (V.G.); (B.P.)
- Cellavita Pesquisas Científicas Ltd.a., Valinhos 13271-650, Brazil;
| | - Bruna Policíquio
- Genetics Laboratory, Instituto Butantan, São Paulo 05508-010, Brazil; (H.V.); (F.D.); (V.G.); (B.P.)
- Cellavita Pesquisas Científicas Ltd.a., Valinhos 13271-650, Brazil;
| | - Gabriel Avelar Colozza-Gama
- Programa de Pós-Graduação em Biologia Estrutural e Funcional, Escola Paulista de Medicina (EPM), Federal University of São Paulo (UNIFES), São Paulo 04039-032, Brazil; (V.R.d.C.); (T.B.M.); (G.A.C.-G.)
- Genetic Bases of Thyroid Tumors Laboratory, Division of Genetics, Department of Morphology and Genetics, Federal University of São Paulo (UNIFESP), São Paulo 04039-032, Brazil
| | | | - Irina Kerkis
- Programa de Pós-Graduação em Biologia Estrutural e Funcional, Escola Paulista de Medicina (EPM), Federal University of São Paulo (UNIFES), São Paulo 04039-032, Brazil; (V.R.d.C.); (T.B.M.); (G.A.C.-G.)
- Genetics Laboratory, Instituto Butantan, São Paulo 05508-010, Brazil; (H.V.); (F.D.); (V.G.); (B.P.)
- Cellavita Pesquisas Científicas Ltd.a., Valinhos 13271-650, Brazil;
| |
Collapse
|
46
|
Bone marrow/bone pre-metastatic niche for breast cancer cells colonization: The role of mesenchymal stromal cells. Crit Rev Oncol Hematol 2021; 164:103416. [PMID: 34237436 DOI: 10.1016/j.critrevonc.2021.103416] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 06/17/2021] [Accepted: 07/04/2021] [Indexed: 02/07/2023] Open
Abstract
Breast cancer is one of the most common oncological pathologies in women worldwide. While its early diagnosis has considerably improved, about 70 % of advanced patients develop bone metastases with a high mortality rate. Several authors demonstrated that primary breast cancer cells prepare their future metastatic niche -known as the pre-metastatic niche- to turn it into an "optimal soil" for colonization. The role of the different cellular components of the bone marrow/bone niche in bone metastasis has been well described. However, studying the changes that occur in this microenvironment before tumor cells arrival has become a novel research field. Therefore, the purpose of this review is to describe the current knowledge about the modulation of the normal bone marrow/bone niche by the primary breast tumor, in particular, highlighting the role of mesenchymal stem/stromal cells in transforming this soil into a pre-metastatic niche for breast cancer cells colonization.
Collapse
|
47
|
Sinha G, Ferrer AI, Ayer S, El-Far MH, Pamarthi SH, Naaldijk Y, Barak P, Sandiford OA, Bibber BM, Yehia G, Greco SJ, Jiang JG, Bryan M, Kumar R, Ponzio NM, Etchegaray JP, Rameshwar P. Specific N-cadherin-dependent pathways drive human breast cancer dormancy in bone marrow. Life Sci Alliance 2021; 4:4/7/e202000969. [PMID: 34078741 PMCID: PMC8200294 DOI: 10.26508/lsa.202000969] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 04/19/2021] [Accepted: 05/20/2021] [Indexed: 12/12/2022] Open
Abstract
The challenge for treating breast cancer (BC) is partly due to long-term dormancy driven by cancer stem cells (CSCs) capable of evading immune response and resist chemotherapy. BC cells show preference for the BM, resulting in poor prognosis. CSCs use connexin 43 (Cx43) to form gap junctional intercellular communication with BM niche cells, fibroblasts, and mesenchymal stem cells (MSCs). However, Cx43 is an unlikely target to reverse BC dormancy because of its role as a hematopoietic regulator. We found N-cadherin (CDH2) and its associated pathways as potential drug targets. CDH2, highly expressed in CSCs, interacts intracellularly with Cx43, colocalizes with Cx43 in BC cells within BM biopsies of patients, and is required for Cx43-mediated gap junctional intercellular communication with BM niche cells. Notably, CDH2 and anti-apoptotic pathways maintained BC dormancy. We thereby propose these pathways as potential pharmacological targets to prevent dormancy and chemosensitize resistant CSCs.
Collapse
Affiliation(s)
- Garima Sinha
- Rutgers School of Graduate Studies at New Jersey Medical School, Newark, NJ, USA.,Department of Medicine, Hematology/Oncology, Rutgers New Jersey Medicine School, Newark, NJ, USA
| | - Alejandra I Ferrer
- Rutgers School of Graduate Studies at New Jersey Medical School, Newark, NJ, USA.,Department of Medicine, Hematology/Oncology, Rutgers New Jersey Medicine School, Newark, NJ, USA
| | - Seda Ayer
- Department of Medicine, Hematology/Oncology, Rutgers New Jersey Medicine School, Newark, NJ, USA
| | - Markos H El-Far
- Rutgers School of Graduate Studies at New Jersey Medical School, Newark, NJ, USA.,Department of Medicine, Hematology/Oncology, Rutgers New Jersey Medicine School, Newark, NJ, USA
| | - Sri Harika Pamarthi
- Department of Medicine, Hematology/Oncology, Rutgers New Jersey Medicine School, Newark, NJ, USA
| | - Yahaira Naaldijk
- Department of Medicine, Hematology/Oncology, Rutgers New Jersey Medicine School, Newark, NJ, USA
| | - Pradeep Barak
- Department of Pathology, Immunology and Laboratory Medicine, Rutgers New Jersey Medical School, Newark, NJ, USA.,ONI, Linacre House, Oxford, UK
| | - Oleta A Sandiford
- Department of Medicine, Hematology/Oncology, Rutgers New Jersey Medicine School, Newark, NJ, USA
| | - Bernadette M Bibber
- Rutgers School of Graduate Studies at New Jersey Medical School, Newark, NJ, USA.,Department of Medicine, Hematology/Oncology, Rutgers New Jersey Medicine School, Newark, NJ, USA
| | - Ghassan Yehia
- Genome Editing Shared Resource, Office of Research and Economic Development, Rutgers University, New Brunswick, NJ, USA
| | - Steven J Greco
- Department of Medicine, Hematology/Oncology, Rutgers New Jersey Medicine School, Newark, NJ, USA
| | - Jie-Gen Jiang
- Department of Pathology, Immunology and Laboratory Medicine, Rutgers New Jersey Medical School, Newark, NJ, USA.,ONI, Linacre House, Oxford, UK
| | - Margarette Bryan
- Department of Medicine, Hematology/Oncology, Rutgers New Jersey Medicine School, Newark, NJ, USA
| | - Rakesh Kumar
- Department of Biotechnology, Rajiv Gandhi Centre for Biotechnology, Kerala, India
| | - Nicholas M Ponzio
- Department of Pathology, Immunology and Laboratory Medicine, Rutgers New Jersey Medical School, Newark, NJ, USA.,ONI, Linacre House, Oxford, UK
| | | | - Pranela Rameshwar
- Rutgers School of Graduate Studies at New Jersey Medical School, Newark, NJ, USA .,Department of Medicine, Hematology/Oncology, Rutgers New Jersey Medicine School, Newark, NJ, USA
| |
Collapse
|
48
|
Liu T, Hooda J, Atkinson JM, Whiteside TL, Oesterreich S, Lee AV. Exosomes in Breast Cancer - Mechanisms of Action and Clinical Potential. Mol Cancer Res 2021; 19:935-945. [PMID: 33627501 PMCID: PMC8178200 DOI: 10.1158/1541-7786.mcr-20-0952] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 01/04/2021] [Accepted: 02/19/2021] [Indexed: 12/24/2022]
Abstract
Extracellular vesicles (EV) are a heterogeneous group of cell-derived membrane vesicles comprising apoptotic bodies, microvesicles, and small EVs also called as exosomes. Exosomes when initially identified were considered as a waste product but the advancement in research techniques have provided insight into the important roles of exosomes in cell-cell communication, various biological processes and diseases, including cancer. As an important component of EVs, exosomes contain various biomolecules such as miRNAs, lipids, and proteins that largely reflect the characteristics of their parent cells. Notably, cancer cells generate and secrete many more exosomes than normal cells. A growing body of evidence suggests that exosomes, as mediators of intercellular cross-talk, play a role in tumorigenesis, cancer cell invasion, angiogenesis, tumor microenvironment (TME) formation, and cancer metastasis. As we gain more insights into the association between exosomes and cancer, the potential of exosomes for clinical use is becoming more intriguing. This review is focused on the role of exosomes in breast cancer, in terms of breast cancer biology, mechanism of action, potential as biomarkers, and therapeutic opportunities.
Collapse
Affiliation(s)
- Tiantong Liu
- Women's Cancer Research Center, UPMC Hillman Cancer Center, Magee-Womens Research Institute, Pittsburgh, Pennsylvania
- School of Medicine, Tsinghua University, Beijing, China
| | - Jagmohan Hooda
- Women's Cancer Research Center, UPMC Hillman Cancer Center, Magee-Womens Research Institute, Pittsburgh, Pennsylvania
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Jennifer M Atkinson
- Women's Cancer Research Center, UPMC Hillman Cancer Center, Magee-Womens Research Institute, Pittsburgh, Pennsylvania
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Theresa L Whiteside
- Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Steffi Oesterreich
- Women's Cancer Research Center, UPMC Hillman Cancer Center, Magee-Womens Research Institute, Pittsburgh, Pennsylvania
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Adrian V Lee
- Women's Cancer Research Center, UPMC Hillman Cancer Center, Magee-Womens Research Institute, Pittsburgh, Pennsylvania.
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
49
|
Abstract
PURPOSE OF REVIEW In this review, we describe the biology of extracellular vesicles (EV) and how they contribute to bone-associated cancers. RECENT FINDINGS Crosstalk between tumor and bone has been demonstrated to promote tumor and metastatic progression. In addition to direct cell-to-cell contact and soluble factors, such as cytokines, EVs mediate crosstalk between tumor and bone. EVs are composed of a heterogenous group of membrane-delineated vesicles of varying size range, mechanisms of formation, and content. These include apoptotic bodies, microvesicles, large oncosomes, and exosomes. EVs derived from primary tumors have been shown to alter bone remodeling and create formation of a pre-metastatic niche that favors development of bone metastasis. Similarly, EVs from marrow stromal cells have been shown to promote tumor progression. Additionally, EVs can act as therapeutic delivery vehicles due to their low immunogenicity and targeting specificity. EVs play critical roles in intercellular communication. Multiple classes of EVs exist based on size on mechanism of formation. In addition to a role in pathophysiology, EVs can be exploited as therapeutic delivery vehicles.
Collapse
Affiliation(s)
- Jinlu Dai
- Department of Urology, University of Michigan, NCRC B14 RM116, 2800 Plymouth Road, Ann Arbor, MI, 48109, USA
| | - Alison B Shupp
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Karen M Bussard
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Evan T Keller
- Department of Urology, University of Michigan, NCRC B14 RM116, 2800 Plymouth Road, Ann Arbor, MI, 48109, USA.
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
50
|
Liu Z, Hu S, Yun Z, Hu W, Zhang S, Luo D. Using dynamic cell communication improves treatment strategies of breast cancer. Cancer Cell Int 2021; 21:275. [PMID: 34034721 PMCID: PMC8145794 DOI: 10.1186/s12935-021-01979-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Accepted: 05/13/2021] [Indexed: 12/28/2022] Open
Abstract
Several insights from the clinical treatment of breast cancer patients have revealed that only a portion of patients achieve the expected curative effect after traditional targeted therapy, that surgical treatment may promote the development of cancer metastasis, and that the optimal combination of neoadjuvant chemotherapy and traditional treatment is not clear. Therefore, a more precise classification of breast cancer and selection of treatment methods should be undertaken to improve the efficacy of clinical treatment. In the clinical treatment of breast cancer, cell communication molecules are often selected as therapeutic targets. However, various cell communications are not static. Their dynamic changes are related to communicating cells, communicating molecules, and various intertwined internal and external environmental factors. Understanding the dynamic microenvironment can help us improve therapeutic efficacy and provide new ways to more accurately determine the cancer status. Therefore, this review describes multiple types of cellular communication in the breast cancer microenvironment and incorporates internal and external environmental factors as variable signaling factors in cell communication. Using dynamic and developmental concepts, we summarize the functional changes in signaling molecules and cells to aid in the diagnosis and treatment of breast cancer.
Collapse
Affiliation(s)
- Zhibo Liu
- Second Clinic Medical College, The Second Affiliated Hospital of Chongqing Medical University, 76 Linjiang Road, Yuzhong District, Chongqing, People's Republic of China
| | - Song Hu
- Thrombosis Center, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Zehui Yun
- Queen Mary School, School of Medicine, Nanchang University, Nanchang, People's Republic of China
| | - Wanshan Hu
- School of Medicine, Forth Clinic Medical College, Nanchang University, Nanchang, People's Republic of China
| | - Shuhua Zhang
- Jiangxi Cardiovascular Research Institute, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Aiguo Road, No. 152, Nanchang, 330006, Jiangxi, People's Republic of China.
| | - Daya Luo
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanchang University, Bayi Road, No. 461, Nanchang, 330006, People's Republic of China.
| |
Collapse
|