1
|
Nicolas E, Kosmider B, Cukierman E, Borghaei H, Golemis EA, Borriello L. Cancer treatments as paradoxical catalysts of tumor awakening in the lung. Cancer Metastasis Rev 2024; 43:1165-1183. [PMID: 38963567 PMCID: PMC11554904 DOI: 10.1007/s10555-024-10196-5] [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: 03/16/2024] [Accepted: 06/12/2024] [Indexed: 07/05/2024]
Abstract
Much of the fatality of tumors is linked to the growth of metastases, which can emerge months to years after apparently successful treatment of primary tumors. Metastases arise from disseminated tumor cells (DTCs), which disperse through the body in a dormant state to seed distant sites. While some DTCs lodge in pre-metastatic niches (PMNs) and rapidly develop into metastases, other DTCs settle in distinct microenvironments that maintain them in a dormant state. Subsequent awakening, induced by changes in the microenvironment of the DTC, causes outgrowth of metastases. Hence, there has been extensive investigation of the factors causing survival and subsequent awakening of DTCs, with the goal of disrupting these processes to decrease cancer lethality. We here provide a detailed overview of recent developments in understanding of the factors controlling dormancy and awakening in the lung, a common site of metastasis for many solid tumors. These factors include dynamic interactions between DTCs and diverse epithelial, mesenchymal, and immune cell populations resident in the lung. Paradoxically, among key triggers for metastatic outgrowth, lung tissue remodeling arising from damage induced by the treatment of primary tumors play a significant role. In addition, growing evidence emphasizes roles for inflammation and aging in opposing the factors that maintain dormancy. Finally, we discuss strategies being developed or employed to reduce the risk of metastatic recurrence.
Collapse
Affiliation(s)
- Emmanuelle Nicolas
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA, 19111, USA
| | - Beata Kosmider
- Center for Inflammation and Lung Research, Lewis Katz School of Medicine, Temple University, 3500 N Broad St., Philadelphia, PA, 19140, USA
- Department of Microbiology, Immunology, and Inflammation, Lewis Katz School of Medicine, Temple University, 3500 N Broad St., Philadelphia, PA, 19140, USA
| | - Edna Cukierman
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA, 19111, USA
| | - Hossein Borghaei
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA, 19111, USA
| | - Erica A Golemis
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA, 19111, USA
- Department of Cancer and Cellular Biology, Lewis Katz School of Medicine, Temple University, 3500 N Broad St., Philadelphia, PA, 19140, USA
| | - Lucia Borriello
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA, 19111, USA.
- Department of Cancer and Cellular Biology, Lewis Katz School of Medicine, Temple University, 3500 N Broad St., Philadelphia, PA, 19140, USA.
| |
Collapse
|
2
|
Bakhshandeh S, Heras U, Taïeb HM, Varadarajan AR, Lissek SM, Hücker SM, Lu X, Garske DS, Young SAE, Abaurrea A, Caffarel MM, Riestra A, Bragado P, Contzen J, Gossen M, Kirsch S, Warfsmann J, Honarnejad K, Klein CA, Cipitria A. Dormancy-inducing 3D engineered matrix uncovers mechanosensitive and drug-protective FHL2-p21 signaling axis. SCIENCE ADVANCES 2024; 10:eadr3997. [PMID: 39504377 PMCID: PMC11540038 DOI: 10.1126/sciadv.adr3997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 09/26/2024] [Indexed: 11/08/2024]
Abstract
Solid cancers frequently relapse with distant metastasis, despite local and systemic treatment. Cellular dormancy has been identified as an important mechanism underlying drug resistance enabling late relapse. Therefore, relapse from invisible, minimal residual cancer of seemingly disease-free patients call for in vitro models of dormant cells suited for drug discovery. Here, we explore dormancy-inducing 3D engineered matrices, which generate mechanical confinement and induce growth arrest and survival against chemotherapy in cancer cells. We characterized the dormant phenotype of solitary cells by P-ERKlow:P-p38high dormancy signaling ratio, along with Ki67- expression. As underlying mechanism, we identified stiffness-dependent nuclear localization of the four-and-a-half LIM domain 2 (FHL2) protein, leading to p53-independent high p21Cip1/Waf1 nuclear expression, validated in murine and human tissue. Suggestive of a resistance-causing role, cells in the dormancy-inducing matrix became sensitive against chemotherapy upon FHL2 down-regulation. Thus, our biomaterial-based approach will enable systematic screens for previously unidentified compounds suited to eradicate potentially relapsing dormant cancer cells.
Collapse
Affiliation(s)
- Sadra Bakhshandeh
- Department of Biomaterials, Max Planck Institute of Colloids and Interfaces, Potsdam, Germany
| | - Unai Heras
- Group of Bioengineering in Regeneration and Cancer, Biogipuzkoa Health Research Institute, San Sebastian, Spain
- Department of Cell Biology and Histology, Faculty of Medicine and Nursing, University of the Basque Country UPV/EHU, Leioa, Spain
| | - Hubert M. Taïeb
- Department of Biomaterials, Max Planck Institute of Colloids and Interfaces, Potsdam, Germany
| | - Adithi R. Varadarajan
- Division of Personalized Tumor Therapy, Fraunhofer Institute for Toxicology and Experimental Medicine, Regensburg, Germany
| | - Susanna M. Lissek
- Experimental Medicine and Therapy Research, University of Regensburg, Regensburg, Germany
| | - Sarah M. Hücker
- Division of Personalized Tumor Therapy, Fraunhofer Institute for Toxicology and Experimental Medicine, Regensburg, Germany
| | - Xin Lu
- Division of Personalized Tumor Therapy, Fraunhofer Institute for Toxicology and Experimental Medicine, Regensburg, Germany
| | - Daniela S. Garske
- Department of Biomaterials, Max Planck Institute of Colloids and Interfaces, Potsdam, Germany
| | - Sarah A. E. Young
- Department of Biomaterials, Max Planck Institute of Colloids and Interfaces, Potsdam, Germany
| | - Andrea Abaurrea
- Group of Breast Cancer, Biogipuzkoa Health Research Institute, San Sebastian, Spain
| | - Maria M Caffarel
- Group of Breast Cancer, Biogipuzkoa Health Research Institute, San Sebastian, Spain
- IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| | - Ana Riestra
- Department of Pharmacy, Fundación Onkologikoa Fundazioa, San Sebastian, Spain
- Department of Medicine, University of Deusto, Bilbao, Spain
| | - Paloma Bragado
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University of Madrid, Madrid, Spain
- Health Research Institute of the Hospital Clínico San Carlos, Madrid, Spain
| | - Jörg Contzen
- Department of Experimental Neurology, Charité - Universitätsmedizin Berlin, Berlin, Germany
- Institute of Active Polymers, Helmholtz-Zentrum Hereon, Teltow, Germany
- Berlin-Brandenburg Center for Regenerative Therapies, Charité Campus Virchow Klinikum, Berlin, Germany
| | - Manfred Gossen
- Institute of Active Polymers, Helmholtz-Zentrum Hereon, Teltow, Germany
- Berlin-Brandenburg Center for Regenerative Therapies, Charité Campus Virchow Klinikum, Berlin, Germany
| | - Stefan Kirsch
- Division of Personalized Tumor Therapy, Fraunhofer Institute for Toxicology and Experimental Medicine, Regensburg, Germany
| | - Jens Warfsmann
- Division of Personalized Tumor Therapy, Fraunhofer Institute for Toxicology and Experimental Medicine, Regensburg, Germany
| | - Kamran Honarnejad
- Division of Personalized Tumor Therapy, Fraunhofer Institute for Toxicology and Experimental Medicine, Regensburg, Germany
| | - Christoph A. Klein
- Division of Personalized Tumor Therapy, Fraunhofer Institute for Toxicology and Experimental Medicine, Regensburg, Germany
- Experimental Medicine and Therapy Research, University of Regensburg, Regensburg, Germany
| | - Amaia Cipitria
- Department of Biomaterials, Max Planck Institute of Colloids and Interfaces, Potsdam, Germany
- Group of Bioengineering in Regeneration and Cancer, Biogipuzkoa Health Research Institute, San Sebastian, Spain
- IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| |
Collapse
|
3
|
Bushnell GG, Sharma D, Wilmot HC, Zheng M, Fashina TD, Hutchens CM, Osipov S, Burness M, Wicha MS. Natural Killer Cell Regulation of Breast Cancer Stem Cells Mediates Metastatic Dormancy. Cancer Res 2024; 84:3337-3353. [PMID: 39106452 PMCID: PMC11474167 DOI: 10.1158/0008-5472.can-24-0030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 06/04/2024] [Accepted: 07/31/2024] [Indexed: 08/09/2024]
Abstract
Patients with breast cancer with estrogen receptor-positive tumors face a constant risk of disease recurrence for the remainder of their lives. Dormant tumor cells residing in tissues such as the bone marrow may generate clinically significant metastases many years after initial diagnosis. Previous studies suggest that dormant cancer cells display "stem-like" properties (cancer stem cell, CSC), which may be regulated by the immune system. To elucidate the role of the immune system in controlling dormancy and its escape, we studied dormancy in immunocompetent, syngeneic mouse breast cancer models. Three mouse breast cancer cell lines, PyMT, Met1, and D2.0R, contained CSCs that displayed short- and long-term metastatic dormancy in vivo, which was dependent on the host immune system. Each model was regulated by different components of the immune system. Natural killer (NK) cells were key for the metastatic dormancy phenotype in D2.0R cells. Quiescent D2.0R CSCs were resistant to NK cell cytotoxicity, whereas proliferative CSCs were sensitive. Resistance to NK cell cytotoxicity was mediated, in part, by the expression of BACH1 and SOX2 transcription factors. Expression of STING and STING targets was decreased in quiescent CSCs, and the STING agonist MSA-2 enhanced NK cell killing. Collectively, these findings demonstrate the role of immune regulation of breast tumor dormancy and highlight the importance of utilizing immunocompetent models to study this phenomenon. Significance: The immune system controls disseminated breast cancer cells during disease latency, highlighting the need to utilize immunocompetent models to identify strategies for targeting dormant cancer cells and reducing metastatic recurrence. See related commentary by Cackowski and Korkaya, p. 3319.
Collapse
Affiliation(s)
- Grace G. Bushnell
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan.
| | - Deeksha Sharma
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan.
| | - Henry C. Wilmot
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan.
| | - Michelle Zheng
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan.
| | | | - Chloe M. Hutchens
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan.
| | - Samuel Osipov
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan.
| | - Monika Burness
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan.
| | - Max S. Wicha
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan.
| |
Collapse
|
4
|
Baldassarri I, Tavakol DN, Graney PL, Chramiec AG, Hibshoosh H, Vunjak-Novakovic G. An engineered model of metastatic colonization of human bone marrow reveals breast cancer cell remodeling of the hematopoietic niche. Proc Natl Acad Sci U S A 2024; 121:e2405257121. [PMID: 39374382 PMCID: PMC11494322 DOI: 10.1073/pnas.2405257121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 08/16/2024] [Indexed: 10/09/2024] Open
Abstract
Incomplete understanding of metastatic disease mechanisms continues to hinder effective treatment of cancer. Despite remarkable advancements toward the identification of druggable targets, treatment options for patients in remission following primary tumor resection remain limited. Bioengineered human tissue models of metastatic sites capable of recreating the physiologically relevant milieu of metastatic colonization may strengthen our grasp of cancer progression and contribute to the development of effective therapeutic strategies. We report the use of an engineered tissue model of human bone marrow (eBM) to identify microenvironmental cues regulating cancer cell proliferation and to investigate how triple-negative breast cancer (TNBC) cell lines influence hematopoiesis. Notably, individual stromal components of the bone marrow niche (osteoblasts, endothelial cells, and mesenchymal stem/stromal cells) were each critical for regulating tumor cell quiescence and proliferation in the three-dimensional eBM niche. We found that hematopoietic stem and progenitor cells (HSPCs) impacted TNBC cell growth and responded to cancer cell presence with a shift of HSPCs (CD34+CD38-) to downstream myeloid lineages (CD11b+CD14+). To account for tumor heterogeneity and show proof-of-concept ability for patient-specific studies, we demonstrate that patient-derived tumor organoids survive and proliferate in the eBM, resulting in distinct shifts in myelopoiesis that are similar to those observed for aggressively metastatic cell lines. We envision that this human tissue model will facilitate studies of niche-specific metastatic progression and individualized responses to treatment.
Collapse
Affiliation(s)
- Ilaria Baldassarri
- Department of Biomedical Engineering, Columbia University, New York, NY10025
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY10032
| | - Daniel Naveed Tavakol
- Department of Biomedical Engineering, Columbia University, New York, NY10025
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY10032
| | - Pamela L. Graney
- Department of Biomedical Engineering, Columbia University, New York, NY10025
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY10032
| | - Alan G. Chramiec
- Department of Biomedical Engineering, Columbia University, New York, NY10025
| | - Hanina Hibshoosh
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY10032
- Department of Pathology and Cell Biology, Columbia University, New York, NY10032
| | - Gordana Vunjak-Novakovic
- Department of Biomedical Engineering, Columbia University, New York, NY10025
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY10032
- Department of Medicine, Columbia University, New York, NY10032
- College of Dental Medicine, Columbia University, New York, NY10032
| |
Collapse
|
5
|
Reinecke JB, Jimenez Garcia L, Gross AC, Cam M, Cannon MV, Gust MJ, Sheridan JP, Gryder BE, Dries R, Roberts RD. Aberrant activation of wound healing programs within the metastatic niche facilitates lung colonization by osteosarcoma cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.10.575008. [PMID: 38260361 PMCID: PMC10802507 DOI: 10.1101/2024.01.10.575008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
PURPOSE Lung metastasis is responsible for nearly all deaths caused by osteosarcoma, the most common pediatric bone tumor. How malignant bone cells coerce the lung microenvironment to support metastatic growth is unclear. The purpose of this study is to identify metastasis-specific therapeutic vulnerabilities by delineating the cellular and molecular mechanisms underlying osteosarcoma lung metastatic niche formation. EXPERIMENTAL DESIGN Using single-cell transcriptomics (scRNA-seq), we characterized genome- and tissue-wide molecular changes induced within lung tissues by disseminated osteosarcoma cells in both immunocompetent murine models of metastasis and patient samples. We confirmed transcriptomic findings at the protein level and determined spatial relationships with multi-parameter immunofluorescence and spatial transcriptomics. Based on these findings, we evaluated the ability of nintedanib, a kinase inhibitor used to treat patients with pulmonary fibrosis, to impair metastasis progression in both immunocompetent murine osteosarcoma and immunodeficient human xenograft models. Single-nucleus and spatial transcriptomics was used to perform molecular pharmacodynamic studies that define the effects of nintedanib on tumor and non-tumor cells within the metastatic microenvironment. RESULTS Osteosarcoma cells induced acute alveolar epithelial injury upon lung dissemination. scRNA-seq demonstrated that the surrounding lung stroma adopts a chronic, non-resolving wound-healing phenotype similar to that seen in other models of lung injury. Accordingly, metastasis-associated lung demonstrated marked fibrosis, likely due to the accumulation of pathogenic, pro-fibrotic, partially differentiated epithelial intermediates and macrophages. Our data demonstrated that nintedanib prevented metastatic progression in multiple murine and human xenograft models by inhibiting osteosarcoma-induced fibrosis. CONCLUSIONS Fibrosis represents a targetable vulnerability to block the progression of osteosarcoma lung metastasis. Our data support a model wherein interactions between osteosarcoma cells and epithelial cells create a pro-metastatic niche by inducing tumor deposition of extracellular matrix proteins such as fibronectin that is disrupted by the anti-fibrotic TKI nintedanib. Our data shed light on the non-cell autonomous effects of TKIs on metastasis and provide a roadmap for using single-cell and spatial transcriptomics to define the mechanism of action of TKI on metastases in animal models.
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
|
Libby JR, Royce H, Walker SR, Li L. The role of extracellular matrix in angiogenesis: Beyond adhesion and structure. BIOMATERIALS AND BIOSYSTEMS 2024; 15:100097. [PMID: 39129826 PMCID: PMC11315062 DOI: 10.1016/j.bbiosy.2024.100097] [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: 02/09/2024] [Revised: 06/19/2024] [Accepted: 07/06/2024] [Indexed: 08/13/2024] Open
Abstract
While the extracellular matrix (ECM) has long been recognized for its structural contributions, anchoring cells for adhesion, providing mechanical support, and maintaining tissue integrity, recent efforts have elucidated its dynamic, reciprocal, and diverse properties on angiogenesis. The ECM modulates angiogenic signaling and mechanical transduction, influences the extent and degree of receptor activation, controls cellular behaviors, and serves as a reservoir for bioactive macromolecules. Collectively, these factors guide the formation, maturation, and stabilization of a functional vascular network. This review aims to shed light on the versatile roles of the ECM in angiogenesis, transcending its traditional functions as a mere structural material. We will explore its engagement and synergy in signaling modulation, interactions with various angiogenic factors, and highlight its importance in both health and disease. By capturing the essence of the ECM's diverse functionalities, we highlight the significance in the broader context of vascular biology, enabling the design of novel biomaterials to engineer vascularized tissues and their potential therapeutic implications.
Collapse
Affiliation(s)
- Jaxson R. Libby
- Department of Molecular, Cellular, and Biomedical Sciences, University of New Hampshire, Durham, NH, USA
| | - Haley Royce
- Department of Chemical Engineering and Bioengineering, University of New Hampshire, Durham, NH, USA
| | - Sarah R. Walker
- Department of Molecular, Cellular, and Biomedical Sciences, University of New Hampshire, Durham, NH, USA
| | - Linqing Li
- Department of Chemical Engineering and Bioengineering, University of New Hampshire, Durham, NH, USA
- Department of Chemistry, University of New Hampshire, Durham, NH, USA
| |
Collapse
|
8
|
Giles C, Lee J. Inflammation drives tumor growth in an immunocompetent implantable metastasis model. RESEARCH SQUARE 2024:rs.3.rs-4719290. [PMID: 39149496 PMCID: PMC11326373 DOI: 10.21203/rs.3.rs-4719290/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Nearly 90% of cancer deaths are due to metastasis. Conventional cancer therapeutics including chemotherapy, surgery, and radiotherapy, are effective in treating primary tumors, but may aggravate disseminated tumor cells (DTCs) into regaining a proliferative state. Models isolating the post dissemination environment are needed to address the potential risks of these therapies, however modeling post dissemination environments is challenging. Often, host organisms become moribund due to primary tumor mass before native metastatic niches can evolve. Implantable tissue engineered niches have been used to attract circulating tumor cells independent of the primary tumor. Here, we serially transplant such tissue engineered niches with recruited DTCs in order to isolate the post dissemination environment. After transplantaion, 69% of scaffolds developed overt post-dissemination cancer growth, however 100% of scaffolds did not grow to a life-threatening critical size within twelve weeks. Adjuvant chemotherapy, while initially effective, did not prevent long-term DTC growth in scaffolds. Subjecting these transplanted niches to surgical resection via biopsy punch enhanced CD31, MMP9, Ly6G, and tumor burden compared to control scaffolds. Biopsy punching was able to rescue tumor incidence from prior chemotherapy. This model of serial transplantation of engineered DTC niches is a highly controllable and flexible method of establishing and systematically investigating the post-dissemination niche.
Collapse
|
9
|
Ferrer CM, Cho HM, Boon R, Bernasocchi T, Wong LP, Cetinbas M, Haggerty ER, Mitsiades I, Wojtkiewicz GR, McLoughlin DE, Aboushousha R, Abdelhamid H, Kugel S, Rheinbay E, Sadreyev R, Juric D, Janssen-Heininger YMW, Mostoslavsky R. The glutathione S-transferase Gstt1 drives survival and dissemination in metastases. Nat Cell Biol 2024; 26:975-990. [PMID: 38862786 DOI: 10.1038/s41556-024-01426-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 04/18/2024] [Indexed: 06/13/2024]
Abstract
Identifying the adaptive mechanisms of metastatic cancer cells remains an elusive question in the treatment of metastatic disease, particularly in pancreatic cancer (pancreatic adenocarcinoma, PDA). A loss-of-function shRNA targeted screen in metastatic-derived cells identified Gstt1, a member of the glutathione S-transferase superfamily, as uniquely required for dissemination and metastasis, but dispensable for primary tumour growth. Gstt1 is expressed in latent disseminated tumour cells (DTCs), is retained within a subpopulation of slow-cycling cells within existing metastases, and its inhibition leads to complete regression of macrometastatic tumours. This distinct Gstt1high population is highly metastatic and retains slow-cycling phenotypes, epithelial-mesenchymal transition features and DTC characteristics compared to the Gstt1low population. Mechanistic studies indicate that in this subset of cancer cells, Gstt1 maintains metastases by binding and glutathione-modifying intracellular fibronectin, in turn promoting its secretion and deposition into the metastatic microenvironment. We identified Gstt1 as a mediator of metastasis, highlighting the importance of heterogeneity and its influence on the metastatic tumour microenvironment.
Collapse
Affiliation(s)
- Christina M Ferrer
- The Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA.
- The Broad Institute of Harvard and MIT, Cambridge, MA, USA.
- University of Maryland School of Medicine and the Greenebaum Comprehensive Cancer Center, Baltimore, MD, USA.
| | - Hyo Min Cho
- The Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
- The Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Ruben Boon
- The Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
- Galapagos NV, 2800 Mechelen, Belgium
| | - Tiziano Bernasocchi
- The Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
- The Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Lai Ping Wong
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, USA
| | - Murat Cetinbas
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, USA
| | - Elizabeth R Haggerty
- The Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Irene Mitsiades
- The Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | | | - Daniel E McLoughlin
- The Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
- Termeer Center for Targeted Therapies, Massachusetts General Hospital, Boston, MA, USA
| | - Reem Aboushousha
- University of Vermont Larner College of Medicine, Burlington, VT, USA
| | - Hend Abdelhamid
- University of Vermont Larner College of Medicine, Burlington, VT, USA
| | - Sita Kugel
- Fred Hutchison Cancer Research Center, Seattle, WA, USA
| | - Esther Rheinbay
- The Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Ruslan Sadreyev
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, USA
| | - Dejan Juric
- The Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
- Termeer Center for Targeted Therapies, Massachusetts General Hospital, Boston, MA, USA
| | | | - Raul Mostoslavsky
- The Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA.
- The Broad Institute of Harvard and MIT, Cambridge, MA, USA.
| |
Collapse
|
10
|
Wu C, Weis SM, Cheresh DA. Tumor-initiating cells establish a niche to overcome isolation stress. Trends Cell Biol 2024; 34:380-387. [PMID: 37640611 DOI: 10.1016/j.tcb.2023.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 07/26/2023] [Accepted: 08/02/2023] [Indexed: 08/31/2023]
Abstract
While the tumor microenvironment is a critical contributor to cancer progression, early steps of tumor initiation and metastasis also rely on the ability of individual tumor cells to survive and thrive at locations where tumor stroma or immune infiltration has yet to be established. In this opinion article, we use the term 'isolation stress' to broadly describe the challenges that individual tumor cells must overcome during the initiation and expansion of the primary tumor beyond permissive boundaries and metastatic spread into distant sites, including a lack of cell-cell contact, adhesion to protumor extracellular matrix proteins, and access to nutrients, oxygen, and soluble factors that support growth. In particular, we highlight the ability of solitary tumor cells to autonomously generate a specialized fibronectin-enriched extracellular matrix to create their own pericellular niche that supports tumor initiation. Cancer cells that can creatively evade the effects of isolation stress not only become more broadly stress tolerant, they also tend to show enhanced stemness, drug resistance, tumor initiation, and metastasis.
Collapse
Affiliation(s)
- Chengsheng Wu
- Department of Pathology, Moores Cancer Center, San Diego, La Jolla, CA, USA; Sanford Consortium for Regenerative Medicine at the University of California, San Diego, La Jolla, CA, USA
| | - Sara M Weis
- Department of Pathology, Moores Cancer Center, San Diego, La Jolla, CA, USA; Sanford Consortium for Regenerative Medicine at the University of California, San Diego, La Jolla, CA, USA
| | - David A Cheresh
- Department of Pathology, Moores Cancer Center, San Diego, La Jolla, CA, USA; Sanford Consortium for Regenerative Medicine at the University of California, San Diego, La Jolla, CA, USA.
| |
Collapse
|
11
|
Guerrero-Barberà G, Burday N, Costell M. Shaping Oncogenic Microenvironments: Contribution of Fibronectin. Front Cell Dev Biol 2024; 12:1363004. [PMID: 38660622 PMCID: PMC11039881 DOI: 10.3389/fcell.2024.1363004] [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: 12/29/2023] [Accepted: 03/25/2024] [Indexed: 04/26/2024] Open
Abstract
The extracellular matrix (ECM) is a complex network of proteins and glycans, dynamically remodeled and specifically tailored to the structure/function of each organ. The malignant transformation of cancer cells is determined by both cell intrinsic properties, such as mutations, and extrinsic variables, such as the mixture of surrounding cells in the tumor microenvironment and the biophysics of the ECM. During cancer progression, the ECM undergoes extensive remodeling, characterized by disruption of the basal lamina, vascular endothelial cell invasion, and development of fibrosis in and around the tumor cells resulting in increased tissue stiffness. This enhanced rigidity leads to aberrant mechanotransduction and further malignant transformation potentiating the de-differentiation, proliferation and invasion of tumor cells. Interestingly, this fibrotic microenvironment is primarily secreted and assembled by non-cancerous cells. Among them, the cancer-associated fibroblasts (CAFs) play a central role. CAFs massively produce fibronectin together with type I collagen. This review delves into the primary interactions and signaling pathways through which fibronectin can support tumorigenesis and metastasis, aiming to provide critical molecular insights for better therapy response prediction.
Collapse
Affiliation(s)
| | | | - Mercedes Costell
- Departament of Biochemistry and Molecular Biology, Institut Universitari de Biotecnologia i Biomedicina, Universitat de València, Valencia, Spain
| |
Collapse
|
12
|
Richbourg NR, Irakoze N, Kim H, Peyton SR. Outlook and opportunities for engineered environments of breast cancer dormancy. SCIENCE ADVANCES 2024; 10:eadl0165. [PMID: 38457510 PMCID: PMC10923521 DOI: 10.1126/sciadv.adl0165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Accepted: 02/01/2024] [Indexed: 03/10/2024]
Abstract
Dormant, disseminated breast cancer cells resist treatment and may relapse into malignant metastases after decades of quiescence. Identifying how and why these dormant breast cancer cells are triggered into outgrowth is a key unsolved step in treating latent, metastatic breast cancer. However, our understanding of breast cancer dormancy in vivo is limited by technical challenges and ethical concerns with triggering the activation of dormant breast cancer. In vitro models avoid many of these challenges by simulating breast cancer dormancy and activation in well-controlled, bench-top conditions, creating opportunities for fundamental insights into breast cancer biology that complement what can be achieved through animal and clinical studies. In this review, we address clinical and preclinical approaches to treating breast cancer dormancy, how precisely controlled artificial environments reveal key interactions that regulate breast cancer dormancy, and how future generations of biomaterials could answer further questions about breast cancer dormancy.
Collapse
Affiliation(s)
- Nathan R. Richbourg
- Department of Chemical Engineering, University of Massachusetts Amherst, MA 01003, USA
| | - Ninette Irakoze
- Department of Chemical Engineering, University of Massachusetts Amherst, MA 01003, USA
| | - Hyuna Kim
- Molecular and Cellular Biology Graduate Program, University of Massachusetts Amherst, MA 01003, USA
| | - Shelly R. Peyton
- Department of Chemical Engineering, University of Massachusetts Amherst, MA 01003, USA
- Molecular and Cellular Biology Graduate Program, University of Massachusetts Amherst, MA 01003, USA
- Department of Biomedical Engineering, University of Massachusetts Amherst Amherst, MA 01003, USA
| |
Collapse
|
13
|
Jiao Y, Yu Y, Zheng M, Yan M, Wang J, Zhang Y, Zhang S. Dormant cancer cells and polyploid giant cancer cells: The roots of cancer recurrence and metastasis. Clin Transl Med 2024; 14:e1567. [PMID: 38362620 PMCID: PMC10870057 DOI: 10.1002/ctm2.1567] [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: 10/26/2023] [Revised: 01/11/2024] [Accepted: 01/16/2024] [Indexed: 02/17/2024] Open
Abstract
Tumour cell dormancy is critical for metastasis and resistance to chemoradiotherapy. Polyploid giant cancer cells (PGCCs) with giant or multiple nuclei and high DNA content have the properties of cancer stem cell and single PGCCs can individually generate tumours in immunodeficient mice. PGCCs represent a dormant form of cancer cells that survive harsh tumour conditions and contribute to tumour recurrence. Hypoxic mimics, chemotherapeutics, radiation and cytotoxic traditional Chinese medicines can induce PGCCs formation through endoreduplication and/or cell fusion. After incubation, dormant PGCCs can recover from the treatment and produce daughter cells with strong proliferative, migratory and invasive abilities via asymmetric cell division. Additionally, PGCCs can resist hypoxia or chemical stress and have a distinct protein signature that involves chromatin remodelling and cell cycle regulation. Dormant PGCCs form the cellular basis for therapeutic resistance, metastatic cascade and disease recurrence. This review summarises regulatory mechanisms governing dormant cancer cells entry and exit of dormancy, which may be used by PGCCs, and potential therapeutic strategies for targeting PGCCs.
Collapse
Affiliation(s)
- Yuqi Jiao
- School of Integrative MedicineTianjin University of Traditional Chinese MedicineTianjinChina
| | - Yongjun Yu
- Department of PathologyTianjin Union Medical CenterTianjinChina
| | - Minying Zheng
- Department of PathologyTianjin Union Medical CenterNankai UniversityTianjinChina
| | - Man Yan
- School of Integrative MedicineTianjin University of Traditional Chinese MedicineTianjinChina
| | - Jiangping Wang
- School of Integrative MedicineTianjin University of Traditional Chinese MedicineTianjinChina
| | - Yue Zhang
- School of Integrative MedicineTianjin University of Traditional Chinese MedicineTianjinChina
| | - Shiwu Zhang
- Department of PathologyTianjin Union Medical CenterTianjinChina
| |
Collapse
|
14
|
Bhattacharya A, Alam K, Roy NS, Kaur K, Kaity S, Ravichandiran V, Roy S. Exploring the interaction between extracellular matrix components in a 3D organoid disease model to replicate the pathophysiology of breast cancer. J Exp Clin Cancer Res 2023; 42:343. [PMID: 38102637 PMCID: PMC10724947 DOI: 10.1186/s13046-023-02926-4] [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: 10/20/2023] [Accepted: 12/06/2023] [Indexed: 12/17/2023] Open
Abstract
In vitro models are necessary to study the pathophysiology of the disease and the development of effective, tailored treatment methods owing to the complexity and heterogeneity of breast cancer and the large population affected by it. The cellular connections and tumor microenvironments observed in vivo are often not recapitulated in conventional two-dimensional (2D) cell cultures. Therefore, developing 3D in vitro models that mimic the complex architecture and physiological circumstances of breast tumors is crucial for advancing our understanding of the illness. A 3D scaffold-free in vitro disease model mimics breast cancer pathophysiology by allowing cells to self-assemble/pattern into 3D structures, in contrast with other 3D models that rely on artificial scaffolds. It is possible that this model, whether applied to breast tumors using patient-derived primary cells (fibroblasts, endothelial cells, and cancer cells), can accurately replicate the observed heterogeneity. The complicated interactions between different cell types are modelled by integrating critical components of the tumor microenvironment, such as the extracellular matrix, vascular endothelial cells, and tumor growth factors. Tissue interactions, immune cell infiltration, and the effects of the milieu on drug resistance can be studied using this scaffold-free 3D model. The scaffold-free 3D in vitro disease model for mimicking tumor pathophysiology in breast cancer is a useful tool for studying the molecular basis of the disease, identifying new therapeutic targets, and evaluating treatment modalities. It provides a more physiologically appropriate high-throughput platform for screening large compound library in a 96-384 well format. We critically discussed the rapid development of personalized treatment strategies and accelerated drug screening platforms to close the gap between traditional 2D cell culture and in vivo investigations.
Collapse
Affiliation(s)
- Anamitra Bhattacharya
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal, 700054, India
| | - Kamare Alam
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal, 700054, India
| | - Nakka Sharmila Roy
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal, 700054, India
| | - Kulwinder Kaur
- School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine a Health Sciences, Dublin, Ireland
- Tissue Engineering Research Group, Department of Anatomy & Regenerative Medicine, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Santanu Kaity
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal, 700054, India
| | - Velayutham Ravichandiran
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal, 700054, India
| | - Subhadeep Roy
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal, 700054, India.
| |
Collapse
|
15
|
Nasr MM, Lynch CC. How circulating tumor cluster biology contributes to the metastatic cascade: from invasion to dissemination and dormancy. Cancer Metastasis Rev 2023; 42:1133-1146. [PMID: 37442876 PMCID: PMC10713810 DOI: 10.1007/s10555-023-10124-z] [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: 05/26/2023] [Accepted: 07/05/2023] [Indexed: 07/15/2023]
Abstract
Circulating tumor cells (CTCs) are known to be prognostic for metastatic relapse and are detected in patients as solitary cells or cell clusters. Circulating tumor cell clusters (CTC clusters) have been observed clinically for decades and are of significantly higher metastatic potential compared to solitary CTCs. Recent studies suggest distinct differences in CTC cluster biology regarding invasion and survival in circulation. However, differences regarding dissemination, dormancy, and reawakening require more investigations compared to solitary CTCs. Here, we review the current state of CTC cluster research and consider their clinical significance. In addition, we discuss the concept of collective invasion by CTC clusters and molecular evidence as to how cluster survival in circulation compares to that of solitary CTCs. Molecular differences between solitary and clustered CTCs during dormancy and reawakening programs will also be discussed. We also highlight future directions to advance our current understanding of CTC cluster biology.
Collapse
Affiliation(s)
- Mostafa M Nasr
- Tumor Biology Department, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA
- Cancer Biology Ph.D. Program, University of South Florida, Tampa, FL, USA
| | - Conor C Lynch
- Tumor Biology Department, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA.
| |
Collapse
|
16
|
Zhao Y, Sheldon M, Sun Y, Ma L. New Insights into YAP/TAZ-TEAD-Mediated Gene Regulation and Biological Processes in Cancer. Cancers (Basel) 2023; 15:5497. [PMID: 38067201 PMCID: PMC10705714 DOI: 10.3390/cancers15235497] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 11/07/2023] [Accepted: 11/17/2023] [Indexed: 02/12/2024] Open
Abstract
The Hippo pathway is conserved across species. Key mammalian Hippo pathway kinases, including MST1/2 and LATS1/2, inhibit cellular growth by inactivating the TEAD coactivators, YAP, and TAZ. Extensive research has illuminated the roles of Hippo signaling in cancer, development, and regeneration. Notably, dysregulation of Hippo pathway components not only contributes to tumor growth and metastasis, but also renders tumors resistant to therapies. This review delves into recent research on YAP/TAZ-TEAD-mediated gene regulation and biological processes in cancer. We focus on several key areas: newly identified molecular patterns of YAP/TAZ activation, emerging mechanisms that contribute to metastasis and cancer therapy resistance, unexpected roles in tumor suppression, and advances in therapeutic strategies targeting this pathway. Moreover, we provide an updated view of YAP/TAZ's biological functions, discuss ongoing controversies, and offer perspectives on specific debated topics in this rapidly evolving field.
Collapse
Affiliation(s)
- Yang Zhao
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (Y.Z.); (M.S.)
| | - Marisela Sheldon
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (Y.Z.); (M.S.)
| | - Yutong Sun
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - Li Ma
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (Y.Z.); (M.S.)
- The University of Texas MD Anderson Cancer Center UTHealth Houston Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| |
Collapse
|
17
|
Weston WA, Barr AR. A cell cycle centric view of tumour dormancy. Br J Cancer 2023; 129:1535-1545. [PMID: 37608096 PMCID: PMC10645753 DOI: 10.1038/s41416-023-02401-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 07/31/2023] [Accepted: 08/10/2023] [Indexed: 08/24/2023] Open
Abstract
Tumour dormancy and recurrent metastatic cancer remain the greatest clinical challenge for cancer patients. Dormant tumour cells can evade treatment and detection, while retaining proliferative potential, often for years, before relapsing to tumour outgrowth. Cellular quiescence is one mechanism that promotes and maintains tumour dormancy due to its central role in reducing proliferation, elevating cyto-protective mechanisms, and retaining proliferative potential. Quiescence/proliferation decisions are dictated by intrinsic and extrinsic signals, which regulate the activity of cyclin-dependent kinases (CDKs) to modulate cell cycle gene expression. By clarifying the pathways regulating CDK activity and the signals which activate them, we can better understand how cancer cells enter, maintain, and escape from quiescence throughout the progression of dormancy and metastatic disease. Here we review how CDK activity is regulated to modulate cellular quiescence in the context of tumour dormancy and highlight the therapeutic challenges and opportunities it presents.
Collapse
Affiliation(s)
- William A Weston
- MRC London Institute of Medical Sciences, Du Cane Road, London, W12 0NN, UK
| | - Alexis R Barr
- MRC London Institute of Medical Sciences, Du Cane Road, London, W12 0NN, UK.
- Institute of Clinical Sciences, Imperial College London, Du Cane Rd, London, W12 0NN, UK.
| |
Collapse
|
18
|
Yu TY, Zhang G, Chai XX, Ren L, Yin DC, Zhang CY. Recent progress on the effect of extracellular matrix on occurrence and progression of breast cancer. Life Sci 2023; 332:122084. [PMID: 37716504 DOI: 10.1016/j.lfs.2023.122084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 09/07/2023] [Accepted: 09/11/2023] [Indexed: 09/18/2023]
Abstract
Breast cancer (BC) metastasis is an enormous challenge targeting BC therapy. The extracellular matrix (ECM), the principal component of the BC metastasis niche, is the pivotal driver of breast tumor development, whose biochemical and biophysical characteristics have attracted widespread attention. Here, we review the biological effects of ECM constituents and the influence of ECM stiffness on BC metastasis and drug resistance. We provide an overview of the relative signal transduction mechanisms, existing metastasis models, and targeted drug strategies centered around ECM stiffness. It will shed light on exploring more underlying targets and developing specific drugs aimed at ECM utilizing biomimetic platforms, which are promising for breast cancer treatment.
Collapse
Affiliation(s)
- Tong-Yao Yu
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, Shanxi, PR China
| | - Ge Zhang
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, Shanxi, PR China
| | - Xiao-Xia Chai
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, Shanxi, PR China
| | - Li Ren
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, Shanxi, PR China; Key Laboratory of Flexible Electronics of Zhejiang Province, Ningbo Institute of Northwestern Polytechnical University, Ningbo 315103, Zhejiang, PR China
| | - Da-Chuan Yin
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, Shanxi, PR China.
| | - Chen-Yan Zhang
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, Shanxi, PR China.
| |
Collapse
|
19
|
Choi S, Whitman MA, Shimpi AA, Sempertegui ND, Chiou AE, Druso JE, Verma A, Lux SC, Cheng Z, Paszek M, Elemento O, Estroff LA, Fischbach C. Bone-matrix mineralization dampens integrin-mediated mechanosignalling and metastatic progression in breast cancer. Nat Biomed Eng 2023; 7:1455-1472. [PMID: 37550422 DOI: 10.1038/s41551-023-01077-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 06/30/2023] [Indexed: 08/09/2023]
Abstract
In patients with breast cancer, lower bone mineral density increases the risk of bone metastasis. Although the relationship between bone-matrix mineralization and tumour-cell phenotype in breast cancer is not well understood, mineralization-induced rigidity is thought to drive metastatic progression via increased cell-adhesion forces. Here, by using collagen-based matrices with adjustable intrafibrillar mineralization, we show that, unexpectedly, matrix mineralization dampens integrin-mediated mechanosignalling and induces a less proliferative stem-cell-like phenotype in breast cancer cells. In mice with xenografted decellularized physiological bone matrices seeded with human breast tumour cells, the presence of bone mineral reduced tumour growth and upregulated a gene-expression signature that is associated with longer metastasis-free survival in patients with breast cancer. Our findings suggest that bone-matrix changes in osteogenic niches regulate metastatic progression in breast cancer and that in vitro models of bone metastasis should integrate organic and inorganic matrix components to mimic physiological and pathologic mineralization.
Collapse
Affiliation(s)
- Siyoung Choi
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Matthew A Whitman
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Adrian A Shimpi
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Nicole D Sempertegui
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Aaron E Chiou
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Joseph E Druso
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Akanksha Verma
- Caryl and Israel Englander Institute for Precision Medicine, Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA
| | - Stephanie C Lux
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Zhu Cheng
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, NY, USA
| | - Matthew Paszek
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, NY, USA
| | - Olivier Elemento
- Caryl and Israel Englander Institute for Precision Medicine, Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA
| | - Lara A Estroff
- Department of Materials Science and Engineering, Cornell University, Ithaca, NY, USA.
- Kavli Institute at Cornell for Nanoscale Science, Cornell University, Ithaca, NY, USA.
| | - Claudia Fischbach
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA.
- Kavli Institute at Cornell for Nanoscale Science, Cornell University, Ithaca, NY, USA.
| |
Collapse
|
20
|
Wu C, Weis SM, Cheresh DA. Upregulation of fibronectin and its integrin receptors - an adaptation to isolation stress that facilitates tumor initiation. J Cell Sci 2023; 136:jcs261483. [PMID: 37870164 PMCID: PMC10652044 DOI: 10.1242/jcs.261483] [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] [Indexed: 10/24/2023] Open
Abstract
Tumor initiation at either primary or metastatic sites is an inefficient process in which tumor cells must fulfill a series of conditions. One critical condition involves the ability of individual tumor-initiating cells to overcome 'isolation stress', enabling them to survive within harsh isolating microenvironments that can feature nutrient stress, hypoxia, oxidative stress and the absence of a proper extracellular matrix (ECM). In response to isolation stress, tumor cells can exploit various adaptive strategies to develop stress tolerance and gain stemness features. In this Opinion, we discuss how strategies such as the induction of certain cell surface receptors and deposition of ECM proteins enable tumor cells to endure isolation stress, thereby gaining tumor-initiating potential. As examples, we highlight recent findings from our group demonstrating how exposure of tumor cells to isolation stress upregulates the G-protein-coupled receptor lysophosphatidic acid receptor 4 (LPAR4), its downstream target fibronectin and two fibronectin-binding integrins, α5β1 and αvβ3. These responses create a fibronectin-rich niche for tumor cells, ultimately driving stress tolerance, cancer stemness and tumor initiation. We suggest that approaches to prevent cancer cells from adapting to stress by suppressing LPAR4 induction, blocking its downstream signaling or disrupting fibronectin-integrin interactions hold promise as potential strategies for cancer treatment.
Collapse
Affiliation(s)
- Chengsheng Wu
- Department of Pathology, Moores Cancer Center, University of California San Diego, La Jolla, CA 92037, USA
- Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, CA 92037, USA
| | - Sara M. Weis
- Department of Pathology, Moores Cancer Center, University of California San Diego, La Jolla, CA 92037, USA
- Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, CA 92037, USA
| | - David A. Cheresh
- Department of Pathology, Moores Cancer Center, University of California San Diego, La Jolla, CA 92037, USA
- Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, CA 92037, USA
| |
Collapse
|
21
|
Bushnell GG, Sharma D, Wilmot HC, Zheng M, Fashina TD, Hutchens CM, Osipov S, Wicha MS. Natural killer cell regulation of breast cancer stem cells mediates metastatic dormancy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.02.560493. [PMID: 37873211 PMCID: PMC10592904 DOI: 10.1101/2023.10.02.560493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Breast cancer patients with estrogen receptor positive tumors face a constant risk of disease recurrence for the remainder of their lives. Dormant tumor cells residing in tissues such as the bone marrow may generate clinically significant metastases many years after initial diagnosis. Previous studies suggest that dormant cells display "stem like" properties (CSCs), which may be regulated by the immune system. Although many studies have examined tumor cell intrinsic characteristics of dormancy, the role of the immune system in controlling dormancy and its escape is not well understood. This scientific gap is due, in part, to a lack of immunocompetent mouse models of breast cancer dormancy with many studies involving human xenografts in immunodeficient mice. To overcome this limitation, we studied dormancy in immunocompetent, syngeneic mouse breast cancer models. We find that PyMT, Met-1 and D2.0R cell lines contain CSCs that display both short- and long-term metastatic dormancy in vivo, which is dependent on the host immune system. Natural killer cells were key for the metastatic dormancy phenotype observed for D2.0R and the role of NK cells in regulating CSCs was further investigated.Quiescent D2.0R CSC are resistant to NK cytotoxicity, while proliferative D2.0R CSC were sensitive to NK cytotoxicity both in vitro and in vivo. This resistance was mediated, in part, by the expression of Bach1 and Sox2 transcription factors. NK killing was enhanced by the STING agonist MSA-2. Collectively, our findings demonstrate the important role of immune regulation of breast tumor dormancy and highlight the importance of utilizing immunocompetent models to study this phenomenon.
Collapse
|
22
|
Richbourg NR, Peppas NA. Structurally decoupled stiffness and solute transport in multi-arm poly(ethylene glycol) hydrogels. Biomaterials 2023; 301:122272. [PMID: 37573839 PMCID: PMC10785603 DOI: 10.1016/j.biomaterials.2023.122272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 08/01/2023] [Accepted: 08/07/2023] [Indexed: 08/15/2023]
Abstract
Synthetic hydrogels are widely used as artificial 3D environments for cell culture, facilitating the controlled study of cell-environment interactions. However, most hydrogels are limited in their ability to represent the physical properties of biological tissues because stiffness and solute transport properties in hydrogels are closely correlated. Resultingly, experimental investigations of cell-environment interactions in hydrogels are confounded by simultaneous changes in multiple physical properties. Here, we overcame this limitation by simultaneously manipulating four structural parameters to synthesize a library of multi-arm poly (ethylene glycol) (PEG) hydrogel formulations with robustly decoupled stiffness and solute transport. This structural design approach avoids chemical alterations or additions to the network that might have unanticipated effects on encapsulated cells. An algorithm created to statistically evaluate stiffness-transport decoupling within the dataset identified 46 of the 73 synthesized formulations as robustly decoupled. We show that the swollen polymer network model accurately predicts 11 out of 12 structure-property relationships, suggesting that this approach to decoupling stiffness and solute transport in hydrogels is fundamentally validated and potentially broadly applicable. Furthermore, the unprecedented control of hydrogel network structure provided by multi-arm PEG hydrogels confirmed several fundamental modeling assumptions. This study enables nuanced hydrogel design for uncompromised investigation of cell-environment interactions.
Collapse
Affiliation(s)
- Nathan R Richbourg
- Department of Biomedical Engineering, University of Texas, Austin, TX, 78712, USA.
| | - Nicholas A Peppas
- Department of Biomedical Engineering, University of Texas, Austin, TX, 78712, USA; McKetta Department of Chemical Engineering, University of Texas, Austin, TX, 78712, USA; Division of Molecular Therapeutics and Drug Delivery, College of Pharmacy, University of Texas, Austin, TX, 78712, USA; Departments of Surgery and Pediatrics, Dell Medical School, University of Texas, Austin, TX, 78712, USA.
| |
Collapse
|
23
|
Khan SU, Fatima K, Malik F, Kalkavan H, Wani A. Cancer metastasis: Molecular mechanisms and clinical perspectives. Pharmacol Ther 2023; 250:108522. [PMID: 37661054 DOI: 10.1016/j.pharmthera.2023.108522] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 08/22/2023] [Accepted: 08/29/2023] [Indexed: 09/05/2023]
Abstract
Metastatic progression combined with non-responsiveness towards systemic therapy often shapes the course of disease for cancer patients and commonly determines its lethal outcome. The complex molecular events that promote metastasis are a combination of both, the acquired pro-metastatic properties of cancer cells and a metastasis-permissive or -supportive tumor micro-environment (TME). Yet, dissemination is a challenging process for cancer cells that requires a series of events to enable cancer cell survival and growth. Metastatic cancer cells have to initially detach themselves from primary tumors, overcome the challenges of their intravasal journey and colonize distant sites that are suited for their metastases. The implicated obstacles including anoikis and immune surveillance, can be overcome by intricate intra- and extracellular signaling pathways, which we will summarize and discuss in this review. Further, emerging modulators of metastasis, like the immune-microenvironment, microbiome, sublethal cell death engagement, or the nervous system will be integrated into the existing working model of metastasis.
Collapse
Affiliation(s)
- Sameer Ullah Khan
- The University of Texas MD Anderson Cancer Center, Division of Genitourinary Medical Oncology, Holcombe Blvd, Houston, TX 77030, USA; Division of Cancer Pharmacology, CSIR-Indian Institute of Integrative Medicine, Jammu and Kashmir, India
| | - Kaneez Fatima
- Division of Cancer Pharmacology, CSIR-Indian Institute of Integrative Medicine, Jammu and Kashmir, India; Academy of Scientific and Innovative Research (ASIR), Ghaziabad 201002, India
| | - Fayaz Malik
- Division of Cancer Pharmacology, CSIR-Indian Institute of Integrative Medicine, Jammu and Kashmir, India; Academy of Scientific and Innovative Research (ASIR), Ghaziabad 201002, India.
| | - Halime Kalkavan
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen, Essen, Germany; German Cancer Consortium (DKTK), Partner Site University Hospital Essen, Essen, Germany.
| | - Abubakar Wani
- St. Jude Children's Research Hospital, 262 Danny Thomas Pl, Memphis, TN 38105, United States.
| |
Collapse
|
24
|
Closset L, Gultekin O, Salehi S, Sarhan D, Lehti K, Gonzalez-Molina J. The extracellular matrix - immune microenvironment crosstalk in cancer therapy: Challenges and opportunities. Matrix Biol 2023; 121:217-228. [PMID: 37524251 DOI: 10.1016/j.matbio.2023.07.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 07/28/2023] [Accepted: 07/28/2023] [Indexed: 08/02/2023]
Abstract
Targeting the tumour immune microenvironment (TIME) by cancer immunotherapy has led to improved patient outcomes. However, response to these treatments is heterogeneous and cancer-type dependant. The therapeutic activity of classical cancer therapies such as chemotherapy, radiotherapy, and surgical oncology is modulated by alterations of the TIME. A major regulator of immune cell function and resistance to both immune and classical therapies is the extracellular matrix (ECM). Concurrently, cancer therapies reshape the TIME as well as the ECM, causing both pro- and anti-tumour responses. Accordingly, the TIME-ECM crosstalk presents attractive opportunities to improve therapy outcomes. Here, we review the molecular crosstalk between the TIME and the ECM in cancer and its implications in cancer progression and clinical intervention. Additionally, we discuss examples and future directions of ECM and TIME co-targeting in combination with oncological therapies including surgery, chemotherapy, and radiotherapy.
Collapse
Affiliation(s)
- Lara Closset
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Solnavägen 9, Stockholm 171 65, Sweden; Saint-Antoine Research center (CRSA), UMR_S 938, INSERM, Sorbonne Université, Paris F-75012, France
| | - Okan Gultekin
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Solnavägen 9, Stockholm 171 65, Sweden
| | - Sahar Salehi
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Solnavägen 9, Stockholm 171 65, Sweden; Department of Women's and Children's Health, Division of Obstetrics and Gynecology, Karolinska Institutet, Stockholm, Sweden; Department of Pelvic Cancer, Theme Cancer, Karolinska University Hospital, Stockholm, Sweden
| | - Dhifaf Sarhan
- Department of Laboratory Medicine, Division of Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Kaisa Lehti
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Solnavägen 9, Stockholm 171 65, Sweden; Department of Biomedical Laboratory Science, Norwegian University of Science and Technology, Trondheim, Norway
| | - Jordi Gonzalez-Molina
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Solnavägen 9, Stockholm 171 65, Sweden.
| |
Collapse
|
25
|
Limam I, Abdelkarim M, El Ayeb M, Crepin M, Marrakchi N, Di Benedetto M. Disintegrin-like Protein Strategy to Inhibit Aggressive Triple-Negative Breast Cancer. Int J Mol Sci 2023; 24:12219. [PMID: 37569595 PMCID: PMC10418936 DOI: 10.3390/ijms241512219] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 07/14/2023] [Accepted: 07/28/2023] [Indexed: 08/13/2023] Open
Abstract
Venoms are a rich source of bioactive compounds, and among them is leberagin-C (Leb-C), a disintegrin-like protein derived from the venom of Macrovipera lebetina transmediterrannea snakes. Leb-C has shown promising inhibitory effects on platelet aggregation. Previous studies have demonstrated that this SECD protein specifically targets α5β1, αvβ3, and αvβ6 integrins through a mimic mechanism of RGD disintegrins. In our current study, we focused on exploring the potential effects of Leb-C on metastatic breast cancer. Our findings revealed that Leb-C disrupted the adhesion, migration, and invasion capabilities of MDA-MB-231 breast cancer cells and its highly metastatic D3H2LN sub-population. Additionally, we observed significant suppression of adhesion, migration, and invasion of human umbilical vein endothelial cells (HUVECs). Furthermore, Leb-C demonstrated a strong inhibitory effect on fibroblast-growth-factor-2-induced proliferation of HUVEC. We conducted in vivo experiments using nude mice and found that treatment with 2 µM of Leb-C resulted in a remarkable 73% reduction in D3H2LN xenograft tumor size. Additionally, quantification of intratumor microvessels revealed a 50% reduction in tumor angiogenesis in xenograft after 21 days of twice-weekly treatment with 2 µM of Leb-C. Collectively, these findings suggest the potential utility of this disintegrin-like protein for inhibiting aggressive and resistant metastatic breast cancer.
Collapse
Affiliation(s)
- Inès Limam
- Laboratory of Biomolecules, Venoms and Theranostic Applications, LR20IPT01, Institut Pasteur of Tunis, Tunis El Manar University, Tunis 1068, Tunisia
| | - Mohamed Abdelkarim
- INSERM Unité 553, Laboratoire d’Hémostase, Endothélium et Angiogenèse, Hôpital Saint-Louis, 75010 Paris, France; (M.A.)
- LR99ES10, Faculty of Medicine of Tunis, Tunis El Manar University, 1 Rue Djebal Lakhdar, Tunis 1006, Tunisia
| | - Mohamed El Ayeb
- Laboratory of Biomolecules, Venoms and Theranostic Applications, LR20IPT01, Institut Pasteur of Tunis, Tunis El Manar University, Tunis 1068, Tunisia
| | - Michel Crepin
- INSERM Unité 553, Laboratoire d’Hémostase, Endothélium et Angiogenèse, Hôpital Saint-Louis, 75010 Paris, France; (M.A.)
| | - Naziha Marrakchi
- Laboratory of Biomolecules, Venoms and Theranostic Applications, LR20IPT01, Institut Pasteur of Tunis, Tunis El Manar University, Tunis 1068, Tunisia
| | - Mélanie Di Benedetto
- IUT of Saint-Denis, Department HSE, Université Paris 13, UMRS941 SMBH, 1 Rue de Chablis, 93000 Bobigny, France
| |
Collapse
|
26
|
Li C, Qiu S, Liu X, Guo F, Zhai J, Li Z, Deng L, Ge L, Qian H, Yang L, Xu B. Extracellular matrix-derived mechanical force governs breast cancer cell stemness and quiescence transition through integrin-DDR signaling. Signal Transduct Target Ther 2023; 8:247. [PMID: 37369642 DOI: 10.1038/s41392-023-01453-0] [Citation(s) in RCA: 30] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 04/13/2023] [Accepted: 04/23/2023] [Indexed: 06/29/2023] Open
Abstract
The extracellular matrix (ECM) serves as signals that regulate specific cell states in tumor tissues. Increasing evidence suggests that extracellular biomechanical force signals are critical in tumor progression. In this study, we aimed to explore the influence of ECM-derived biomechanical force on breast cancer cell status. Experiments were conducted using 3D collagen, fibrinogen, and Matrigel matrices to investigate the role of mechanical force in tumor development. Integrin-cytoskeleton-AIRE and DDR-STAT signals were examined using RNA sequencing and western blotting. Data from 1358 patients and 86 clinical specimens were used for ECM signature-prognosis analysis. Our findings revealed that ECM-derived mechanical force regulated tumor stemness and cell quiescence in breast cancer cells. A mechanical force of ~45 Pa derived from the extracellular substrate activated integrin β1/3 receptors, stimulating stem cell signaling pathways through the cytoskeleton/AIRE axis and promoting tumorigenic potential and stem-like phenotypes. However, excessive mechanical force (450 Pa) could drive stem-like cancer cells into a quiescent state, with the removal of mechanical forces leading to vigorous proliferation in quiescent cancer stem cells. Mechanical force facilitated cell cycle arrest to induce quiescence, dependent on DDR2/STAT1/P27 signaling. Therefore, ECM-derived mechanical force governs breast cancer cell status and proliferative characteristics through stiffness alterations. We further established an ECM signature based on the fibrinogen/fibronectin/vitronectin/elastin axis, which efficiently predicts patient prognosis in breast cancer. Our findings highlight the vital role of ECM-derived mechanical force in governing breast cancer cell stemness/quiescence transition and suggest the novel use of ECM signature in predicting the clinical prognosis of breast cancer.
Collapse
Affiliation(s)
- Cong Li
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Shi Qiu
- Department of Urology, Institute of Urology and National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, 610041, China
- National Clinical Research Center of Geriatrics, The Center of Gerontology and Geriatrics, West China Hospital, Sichuan University, Chengdu, 610065, China
- Institute of Oncology Research (IOR), Oncology Institute of Southern Switzerland (IOSI), Bellinzona, 6500, Switzerland
| | - Xiaohan Liu
- Department of Histology and Embryology, Basic Medical College, China Medical University, Shenyang, Liaoning, 110122, China
| | - Fengzhu Guo
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Jingtong Zhai
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Zhijun Li
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Linghui Deng
- Institute of Oncology Research (IOR), Oncology Institute of Southern Switzerland (IOSI), Bellinzona, 6500, Switzerland
| | - Liming Ge
- Department of Pharmaceutical and Bioengineering, School of Chemical Engineering, Sichuan University, Chengdu, 610065, China
| | - Haili Qian
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Lu Yang
- Department of Urology, Institute of Urology and National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, 610041, China.
| | - Binghe Xu
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
27
|
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
|
28
|
Kim H, Wirasaputra A, Mohammadi F, Kundu AN, Esteves JAE, Heiser LM, Meyer AS, Peyton SR. Live Cell Lineage Tracing of Dormant Cancer Cells. Adv Healthc Mater 2023; 12:e2202275. [PMID: 36625629 PMCID: PMC10238615 DOI: 10.1002/adhm.202202275] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 12/02/2022] [Indexed: 01/11/2023]
Abstract
Breast cancer is a leading cause of global cancer-related deaths, and metastasis is the overwhelming culprit of poor patient prognosis. The most nefarious aspect of metastasis is dormancy, a prolonged period between primary tumor resection and relapse. Current therapies are insufficient at killing dormant cells; thus, they can remain quiescent in the body for decades until eventually undergoing a phenotypic switch, resulting in metastases that are more adaptable and drug resistant. Unfortunately, dormancy has few in vitro models, largely because lab-derived cell lines are highly proliferative. Existing models address tumor dormancy, not cellular dormancy, because tracking individual cells is technically challenging. To combat this problem, a live cell lineage approach to find and track individual dormant cells, distinguishing them from proliferative and dying cells over multiple days, is adapted. This approach is applied across a range of different in vitro microenvironments. This approach reveals that the proportion of cells that exhibit long-term quiescence is regulated by both cell intrinsic and extrinsic factors, with the most dormant cells found in 3D collagen gels. This paper envisions that this approach will prove useful to biologists and bioengineers in the dormancy community to identify, quantify, and study dormant tumor cells.
Collapse
Affiliation(s)
- Hyuna Kim
- Molecular and Cell Biology Graduate Program, University of Massachusetts, Amherst, MA, 01002, USA
| | - Anna Wirasaputra
- Department of Chemical Engineering, University of Massachusetts, Amherst, MA, 01002, USA
| | - Farnaz Mohammadi
- Department of Bioengineering, University of California, Los Angeles, CA, 90095, USA
| | - Aritra Nath Kundu
- Department of Chemical Engineering, University of Massachusetts, Amherst, MA, 01002, USA
| | - Jennifer A E Esteves
- Department of Biomedical Engineering, University of Massachusetts, Amherst, MA, 01002, USA
| | - Laura M Heiser
- Department of Biomedical Engineering, Knight Cancer Institute, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Aaron S Meyer
- Department of Bioengineering, University of California, Los Angeles, CA, 90095, USA
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA, 90095, USA
| | - Shelly R Peyton
- Molecular and Cell Biology Graduate Program, University of Massachusetts, Amherst, MA, 01002, USA
- Department of Chemical Engineering, University of Massachusetts, Amherst, MA, 01002, USA
- Department of Biomedical Engineering, University of Massachusetts, Amherst, MA, 01002, USA
| |
Collapse
|
29
|
Brummer AB, Xella A, Woodall R, Adhikarla V, Cho H, Gutova M, Brown CE, Rockne RC. Data driven model discovery and interpretation for CAR T-cell killing using sparse identification and latent variables. Front Immunol 2023; 14:1115536. [PMID: 37256133 PMCID: PMC10226275 DOI: 10.3389/fimmu.2023.1115536] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Accepted: 03/27/2023] [Indexed: 06/01/2023] Open
Abstract
In the development of cell-based cancer therapies, quantitative mathematical models of cellular interactions are instrumental in understanding treatment efficacy. Efforts to validate and interpret mathematical models of cancer cell growth and death hinge first on proposing a precise mathematical model, then analyzing experimental data in the context of the chosen model. In this work, we present the first application of the sparse identification of non-linear dynamics (SINDy) algorithm to a real biological system in order discover cell-cell interaction dynamics in in vitro experimental data, using chimeric antigen receptor (CAR) T-cells and patient-derived glioblastoma cells. By combining the techniques of latent variable analysis and SINDy, we infer key aspects of the interaction dynamics of CAR T-cell populations and cancer. Importantly, we show how the model terms can be interpreted biologically in relation to different CAR T-cell functional responses, single or double CAR T-cell-cancer cell binding models, and density-dependent growth dynamics in either of the CAR T-cell or cancer cell populations. We show how this data-driven model-discovery based approach provides unique insight into CAR T-cell dynamics when compared to an established model-first approach. These results demonstrate the potential for SINDy to improve the implementation and efficacy of CAR T-cell therapy in the clinic through an improved understanding of CAR T-cell dynamics.
Collapse
Affiliation(s)
- Alexander B. Brummer
- Division of Mathematical Oncology, Department of Computational and Quantitative Medicine, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, United States
- Department of Physics and Astronomy, College of Charleston, Charleston, SC, United States
| | - Agata Xella
- Department of Hemtaology and Hematopoietic Cell Translation and Immuno-Oncology, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, United States
| | - Ryan Woodall
- Division of Mathematical Oncology, Department of Computational and Quantitative Medicine, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, United States
| | - Vikram Adhikarla
- Division of Mathematical Oncology, Department of Computational and Quantitative Medicine, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, United States
| | - Heyrim Cho
- Department of Mathematics, University of California, Riverside, Riverside, CA, United States
| | - Margarita Gutova
- Department of Stem Cell Biology and Regenerative Medicine, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, United States
| | - Christine E. Brown
- Department of Hemtaology and Hematopoietic Cell Translation and Immuno-Oncology, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, United States
| | - Russell C. Rockne
- Division of Mathematical Oncology, Department of Computational and Quantitative Medicine, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, United States
| |
Collapse
|
30
|
Ceyhan Y, Garcia NMG, Alvarez JV. Immune cells in residual disease and recurrence. Trends Cancer 2023:S2405-8033(23)00057-2. [PMID: 37150627 DOI: 10.1016/j.trecan.2023.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 04/03/2023] [Accepted: 04/05/2023] [Indexed: 05/09/2023]
Abstract
Tumor recurrence following potentially curative therapy constitutes a major obstacle to achieving cures in patients with cancer. Recurrent tumors frequently arise from a population of residual cancer cells - also referred to as minimal residual disease (RD) or persister cells - that survive therapy and persist for prolonged periods prior to tumor relapse. While there has been significant recent progress in deciphering tumor-cell-intrinsic pathways that regulate residual cancer cell survival and recurrence, much less is known about how the tumor microenvironment (TME) of residual tumors impacts persister cancer cells or tumor recurrence. In this review, we highlight recent studies exploring the regulation and function of immune cells in RD and discuss therapeutic opportunities to target immune cells in residual tumors.
Collapse
Affiliation(s)
- Yasemin Ceyhan
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - Nina Marie G Garcia
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - James V Alvarez
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA.
| |
Collapse
|
31
|
Yuan Z, Li Y, Zhang S, Wang X, Dou H, Yu X, Zhang Z, Yang S, Xiao M. Extracellular matrix remodeling in tumor progression and immune escape: from mechanisms to treatments. Mol Cancer 2023; 22:48. [PMID: 36906534 PMCID: PMC10007858 DOI: 10.1186/s12943-023-01744-8] [Citation(s) in RCA: 154] [Impact Index Per Article: 154.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 02/11/2023] [Indexed: 03/13/2023] Open
Abstract
The malignant tumor is a multi-etiological, systemic and complex disease characterized by uncontrolled cell proliferation and distant metastasis. Anticancer treatments including adjuvant therapies and targeted therapies are effective in eliminating cancer cells but in a limited number of patients. Increasing evidence suggests that the extracellular matrix (ECM) plays an important role in tumor development through changes in macromolecule components, degradation enzymes and stiffness. These variations are under the control of cellular components in tumor tissue via the aberrant activation of signaling pathways, the interaction of the ECM components to multiple surface receptors, and mechanical impact. Additionally, the ECM shaped by cancer regulates immune cells which results in an immune suppressive microenvironment and hinders the efficacy of immunotherapies. Thus, the ECM acts as a barrier to protect cancer from treatments and supports tumor progression. Nevertheless, the profound regulatory network of the ECM remodeling hampers the design of individualized antitumor treatment. Here, we elaborate on the composition of the malignant ECM, and discuss the specific mechanisms of the ECM remodeling. Precisely, we highlight the impact of the ECM remodeling on tumor development, including proliferation, anoikis, metastasis, angiogenesis, lymphangiogenesis, and immune escape. Finally, we emphasize ECM "normalization" as a potential strategy for anti-malignant treatment.
Collapse
Affiliation(s)
- Zhennan Yuan
- Department of Oncological Surgery, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Yingpu Li
- Department of Oncological Surgery, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Sifan Zhang
- Department of Neurobiology, Harbin Medical University, Harbin, 150081, China
| | - Xueying Wang
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - He Dou
- Department of Oncological Surgery, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Xi Yu
- Department of Gynecological Oncology, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Zhiren Zhang
- NHC Key Laboratory of Cell Transplantation, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China.,Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Heilongjiang Key Laboratory for Metabolic Disorder and Cancer Related Cardiovascular Diseases, Harbin, 150001, China
| | - Shanshan Yang
- Department of Gynecological Radiotherapy, Harbin Medical University Cancer Hospital, Harbin, 150000, China.
| | - Min Xiao
- Department of Oncological Surgery, Harbin Medical University Cancer Hospital, Harbin, 150081, China.
| |
Collapse
|
32
|
Pradhan L, Moore D, Ovadia EM, Swedzinski SL, Cossette T, Sikes RA, van Golen K, Kloxin AM. Dynamic bioinspired coculture model for probing ER + breast cancer dormancy in the bone marrow niche. SCIENCE ADVANCES 2023; 9:eade3186. [PMID: 36888709 PMCID: PMC9995072 DOI: 10.1126/sciadv.ade3186] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 02/03/2023] [Indexed: 05/28/2023]
Abstract
Late recurrences of breast cancer are hypothesized to arise from disseminated tumor cells (DTCs) that reactivate after dormancy and occur most frequently with estrogen receptor-positive (ER+) breast cancer cells (BCCs) in bone marrow (BM). Interactions between the BM niche and BCCs are thought to play a pivotal role in recurrence, and relevant model systems are needed for mechanistic insights and improved treatments. We examined dormant DTCs in vivo and observed DTCs near bone lining cells and exhibiting autophagy. To study underlying cell-cell interactions, we established a well-defined, bioinspired dynamic indirect coculture model of ER+ BCCs with BM niche cells, human mesenchymal stem cells (hMSCs) and fetal osteoblasts (hFOBs). hMSCs promoted BCC growth, whereas hFOBs promoted dormancy and autophagy, regulated in part by tumor necrosis factor-α and monocyte chemoattractant protein 1 receptor signaling. This dormancy was reversible by dynamically changing the microenvironment or inhibiting autophagy, presenting further opportunities for mechanistic and targeting studies to prevent late recurrence.
Collapse
Affiliation(s)
- Lina Pradhan
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19716, USA
| | - DeVonte Moore
- Department of Chemistry and Biochemistry, University of Delaware, Newark, DE 19716, USA
| | - Elisa M. Ovadia
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19716, USA
| | - Samantha L. Swedzinski
- Department of Materials Science and Engineering, University of Delaware, Newark, DE 19716, USA
| | - Travis Cossette
- Office of Laboratory Animal Medicine, University of Delaware, Newark, DE 19716, USA
| | - Robert A. Sikes
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
| | - Kenneth van Golen
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
| | - April M. Kloxin
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19716, USA
- Department of Materials Science and Engineering, University of Delaware, Newark, DE 19716, USA
| |
Collapse
|
33
|
Mukherjee A, Bravo-Cordero JJ. Regulation of dormancy during tumor dissemination: the role of the ECM. Cancer Metastasis Rev 2023; 42:99-112. [PMID: 36802311 PMCID: PMC10027413 DOI: 10.1007/s10555-023-10094-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 02/07/2023] [Indexed: 02/23/2023]
Abstract
The study of the metastatic cascade has revealed the complexity of the process and the multiple cellular states that disseminated cancer cells must go through. The tumor microenvironment and in particular the extracellular matrix (ECM) plays an important role in regulating the transition from invasion, dormancy to ultimately proliferation during the metastatic cascade. The time delay from primary tumor detection to metastatic growth is regulated by a molecular program that maintains disseminated tumor cells in a non-proliferative, quiescence state known as tumor cell dormancy. Identifying dormant cells and their niches in vivo and how they transition to the proliferative state is an active area of investigation, and novel approaches have been developed to track dormant cells during dissemination. In this review, we highlight the latest research on the invasive nature of disseminated tumor cells and their link to dormancy programs. We also discuss the role of the ECM in sustaining dormant niches at distant sites.
Collapse
Affiliation(s)
- Ananya Mukherjee
- Division of Hematology and Medical Oncology, Department of Medicine, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jose Javier Bravo-Cordero
- Division of Hematology and Medical Oncology, Department of Medicine, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
34
|
The Journey of Cancer Cells to the Brain: Challenges and Opportunities. Int J Mol Sci 2023; 24:ijms24043854. [PMID: 36835266 PMCID: PMC9967224 DOI: 10.3390/ijms24043854] [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: 12/22/2022] [Revised: 02/09/2023] [Accepted: 02/10/2023] [Indexed: 02/17/2023] Open
Abstract
Cancer metastases into the brain constitute one of the most severe, but not uncommon, manifestations of cancer progression. Several factors control how cancer cells interact with the brain to establish metastasis. These factors include mediators of signaling pathways participating in migration, infiltration of the blood-brain barrier, interaction with host cells (e.g., neurons, astrocytes), and the immune system. Development of novel therapies offers a glimpse of hope for increasing the diminutive life expectancy currently forecasted for patients suffering from brain metastasis. However, applying these treatment strategies has not been sufficiently effective. Therefore, there is a need for a better understanding of the metastasis process to uncover novel therapeutic targets. In this review, we follow the journey of various cancer cells from their primary location through the diverse processes that they undergo to colonize the brain. These processes include EMT, intravasation, extravasation, and infiltration of the blood-brain barrier, ending up with colonization and angiogenesis. In each phase, we focus on the pathways engaging molecules that potentially could be drug target candidates.
Collapse
|
35
|
Liu H, Yin H, Wang Z, Yuan Q, Xu F, Chen Y, Li C. Rho A/ROCK1 signaling-mediated metabolic reprogramming of valvular interstitial cells toward Warburg effect accelerates aortic valve calcification via AMPK/RUNX2 axis. Cell Death Dis 2023; 14:108. [PMID: 36774349 PMCID: PMC9922265 DOI: 10.1038/s41419-023-05642-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 01/29/2023] [Accepted: 02/01/2023] [Indexed: 02/13/2023]
Abstract
The aberrant differentiation of valvular interstitial cells (VICs) to osteogenic lineages promotes calcified aortic valves disease (CAVD), partly activated by potentially destructive hemodynamic forces. These involve Rho A/ROCK1 signaling, a mechano-sensing pathway. However, how Rho A/ROCK1 signaling transduces mechanical signals into cellular responses and disrupts normal VIC homeostasis remain unclear. We examined Rho A/ROCK1 signaling in human aortic valves, and further detected how Rho A/ROCK1 signaling regulates mineralization in human VICs. Aortic valves (CAVD n = 22, normal control (NC) n = 12) from patients undergoing valve replacement were investigated. Immunostaining and western blotting analysis indicated that Rho A/ROCK1 signaling, as well as key transporters and enzymes involved in the Warburg effect, were markedly upregulated in human calcified aortic valves compared with those in the controls. In vitro, Rho A/ROCK1-induced calcification was confirmed as AMPK-dependent, via a mechanism involving metabolic reprogramming of human VICs to Warburg effect. Y-27632, a selective ROCK1 inhibitor, suppressed the Warburg effect, rescued AMPK activity and subsequently increased RUNX2 ubiquitin-proteasome degradation, leading to decreased RUNX2 protein accumulation in human VICs under pathological osteogenic stimulus. Rho A/ROCK1 signaling, which is elevated in human calcified aortic valves, plays a positive role in valvular calcification, partially through its ability to drive metabolic switching of VICs to the Warburg effect, leading to altered AMPK activity and RUNX2 protein accumulation. Thus, Rho A/ROCK1 signaling could be an important and unrecognized hub of destructive hemodynamics and cellular aerobic glycolysis that is essential to promote the CAVD process.
Collapse
Affiliation(s)
- Huiruo Liu
- Department of Emergency Medicine and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, Shandong, China
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital of Shandong University, Jinan, Shandong, China
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Hang Yin
- Department of Emergency Medicine and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, Shandong, China
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital of Shandong University, Jinan, Shandong, China
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Zhen Wang
- Department of Emergency Medicine and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, Shandong, China
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital of Shandong University, Jinan, Shandong, China
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Qiuhuan Yuan
- Department of Emergency Medicine and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, Shandong, China
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital of Shandong University, Jinan, Shandong, China
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Feng Xu
- Department of Emergency Medicine and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, Shandong, China
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital of Shandong University, Jinan, Shandong, China
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Yuguo Chen
- Department of Emergency Medicine and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, Shandong, China.
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital of Shandong University, Jinan, Shandong, China.
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Qilu Hospital of Shandong University, Jinan, Shandong, China.
| | - Chuanbao Li
- Department of Emergency Medicine and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, Shandong, China.
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital of Shandong University, Jinan, Shandong, China.
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Qilu Hospital of Shandong University, Jinan, Shandong, China.
| |
Collapse
|
36
|
Santiago-Gómez A, Barkan D, Chambers AF. Editorial: Revisiting seed and soil: A new approach to target hibernating dormant tumor cells. Front Oncol 2023; 13:1126924. [PMID: 36798827 PMCID: PMC9927640 DOI: 10.3389/fonc.2023.1126924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Accepted: 01/19/2023] [Indexed: 02/04/2023] Open
Affiliation(s)
- Angélica Santiago-Gómez
- Manchester Breast Centre, Division of Cancer Sciences, University of Manchester, Manchester, United Kingdom
- Molecular Oncology Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Dalit Barkan
- Department of Human Biology and Medical Sciences, University of Haifa, Haifa, Israel
| | - Ann F. Chambers
- Departments of Oncology, Medical Biophysics, and Pathology & Laboratory Medicine, University of Western Ontario, London, ON, Canada
| |
Collapse
|
37
|
Peyton SR, Platt MO, Cukierman E. Challenges and Opportunities Modeling the Dynamic Tumor Matrisome. BME FRONTIERS 2023; 4:0006. [PMID: 37849664 PMCID: PMC10521682 DOI: 10.34133/bmef.0006] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Accepted: 11/28/2022] [Indexed: 10/19/2023] Open
Abstract
We need novel strategies to target the complexity of cancer and, particularly, of metastatic disease. As an example of this complexity, certain tissues are particularly hospitable environments for metastases, whereas others do not contain fertile microenvironments to support cancer cell growth. Continuing evidence that the extracellular matrix (ECM) of tissues is one of a host of factors necessary to support cancer cell growth at both primary and secondary tissue sites is emerging. Research on cancer metastasis has largely been focused on the molecular adaptations of tumor cells in various cytokine and growth factor environments on 2-dimensional tissue culture polystyrene plates. Intravital imaging, conversely, has transformed our ability to watch, in real time, tumor cell invasion, intravasation, extravasation, and growth. Because the interstitial ECM that supports all cells in the tumor microenvironment changes over time scales outside the possible window of typical intravital imaging, bioengineers are continuously developing both simple and sophisticated in vitro controlled environments to study tumor (and other) cell interactions with this matrix. In this perspective, we focus on the cellular unit responsible for upholding the pathologic homeostasis of tumor-bearing organs, cancer-associated fibroblasts (CAFs), and their self-generated ECM. The latter, together with tumoral and other cell secreted factors, constitute the "tumor matrisome". We share the challenges and opportunities for modeling this dynamic CAF/ECM unit, the tools and techniques available, and how the tumor matrisome is remodeled (e.g., via ECM proteases). We posit that increasing information on tumor matrisome dynamics may lead the field to alternative strategies for personalized medicine outside genomics.
Collapse
Affiliation(s)
- Shelly R. Peyton
- Department of Chemical Engineering, University of Massachusetts Amherst, Amherst, MA, USA
| | - Manu O. Platt
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Edna Cukierman
- Cancer Signaling & Microenvironment Program, Marvin and Concetta Greenberg Pancreatic Cancer Institute, Fox Chase Cancer Center, Temple Health, Philadelphia, PA, USA
| |
Collapse
|
38
|
Regulation of Metastatic Tumor Dormancy and Emerging Opportunities for Therapeutic Intervention. Int J Mol Sci 2022; 23:ijms232213931. [PMID: 36430404 PMCID: PMC9698240 DOI: 10.3390/ijms232213931] [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: 09/30/2022] [Revised: 10/26/2022] [Accepted: 11/02/2022] [Indexed: 11/16/2022] Open
Abstract
Cancer recurrence and metastasis, following successful treatment, constitutes a critical threat in clinical oncology and are the leading causes of death amongst cancer patients. This phenomenon is largely attributed to metastatic tumor dormancy, a rate-limiting stage during cancer progression, in which disseminated cancer cells remain in a viable, yet not proliferating state for a prolonged period. Dormant cancer cells are characterized by their entry into cell cycle arrest and survival in a quiescence state to adapt to their new microenvironment through the acquisition of mutations and epigenetic modifications, rendering them resistant to anti-cancer treatment and immune surveillance. Under favorable conditions, disseminated dormant tumor cells 're-awake', resume their proliferation and thus colonize distant sites. Due to their rarity, detection of dormant cells using current diagnostic tools is challenging and, thus, therapeutic targets are hard to be identified. Therefore, unraveling the underlying mechanisms required for keeping disseminating tumor cells dormant, along with signals that stimulate their "re-awakening" are crucial for the discovery of novel pharmacological treatments. In this review, we shed light into the main mechanisms that control dormancy induction and escape as well as emerging therapeutic strategies for the eradication of metastatic dormant cells, including dormancy maintenance, direct targeting of dormant cells and re-awakening dormant cells. Studies on the ability of the metastatic cancer cells to cease proliferation and survive in a quiescent state before re-initiating proliferation and colonization years after successful treatment, will pave the way toward developing innovative therapeutic strategies against dormancy-mediated metastatic outgrowth.
Collapse
|
39
|
Peng Z, Lv X, Huang S. Recent Progress on the Role of Fibronectin in Tumor Stromal Immunity and Immunotherapy. Curr Top Med Chem 2022; 22:2494-2505. [PMID: 35708087 DOI: 10.2174/1568026622666220615152647] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 04/18/2022] [Accepted: 04/20/2022] [Indexed: 01/20/2023]
Abstract
As a major component of the stromal microenvironment of various solid tumors, the extracellular matrix (ECM) has attracted increasing attention in cancer-related studies. ECM in the tumor stroma not only provides an external barrier and framework for tumor cell adhesion and movement, but also acts as an active regulator that modulates the tumor microenvironment, including stromal immunity. Fibronectin (Fn), as a core component of the ECM, plays a key role in the assembly and remodeling of the ECM. Hence, understanding the role of Fn in the modulation of tumor stromal immunity is of great importance for cancer immunotherapy. Hence, in-depth studies on the underlying mechanisms of Fn in tumors are urgently needed to clarify the current understanding and issues and to identify new and specific targets for effective diagnosis and treatment purposes. In this review, we summarize the structure and role of Fn, its potent derivatives in tumor stromal immunity, and their biological effects and mechanisms in tumor development. In addition, we discuss the novel applications of Fn in tumor treatment. Therefore, this review can provide prospective insight into Fn immunotherapeutic applications in tumor treatment.
Collapse
Affiliation(s)
- Zheng Peng
- Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Xiaolan Lv
- Department of Laboratory Medicine, Liuzhou Maternity and Child Healthcare Hospital, Liuzhou, Guangxi, China
| | - Shigao Huang
- Department of Radiation Oncology, The First Affiliated Hospital, Air Force Medical University, Xi an, Shaan Xi, China
| |
Collapse
|
40
|
Gonzalez-Molina J, Moyano-Galceran L, Single A, Gultekin O, Alsalhi S, Lehti K. Chemotherapy as a regulator of extracellular matrix-cell communication: Implications in therapy resistance. Semin Cancer Biol 2022; 86:224-236. [PMID: 35331851 DOI: 10.1016/j.semcancer.2022.03.012] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 03/16/2022] [Accepted: 03/17/2022] [Indexed: 02/08/2023]
Abstract
The development of most solid cancers, including pancreatic, breast, lung, liver, and ovarian cancer, involves a desmoplastic reaction: a process of major remodeling of the extracellular matrix (ECM) affecting the ECM composition, mechanics, and microarchitecture. These properties of the ECM influence key cancer cell functions, including treatment resistance. Furthermore, emerging data show that various chemotherapeutic treatments lead to alterations in ECM features and ECM-cell communication. Here, we summarize the current knowledge around the effects of chemotherapy on both the ECM remodeling and ECM-cell signaling and discuss the implications of these alterations on distinct mechanisms of chemoresistance. Additionally, we provide an overview of current therapeutic strategies and ongoing clinical trials utilizing anti-cancer drugs to target the ECM-cell communication and explore the future challenges of these strategies.
Collapse
Affiliation(s)
- Jordi Gonzalez-Molina
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden.
| | - Lidia Moyano-Galceran
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Andrew Single
- Department of Biomedical Laboratory Science, Norwegian University of Science and Technology, Trondheim, Norway
| | - Okan Gultekin
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Shno Alsalhi
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Kaisa Lehti
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden; Department of Biomedical Laboratory Science, Norwegian University of Science and Technology, Trondheim, Norway.
| |
Collapse
|
41
|
A pan-cancer analysis of matrisome proteins reveals CTHRC1 and a related network as major ECM regulators across cancers. PLoS One 2022; 17:e0270063. [PMID: 36190948 PMCID: PMC9529084 DOI: 10.1371/journal.pone.0270063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 06/02/2022] [Indexed: 11/07/2022] Open
Abstract
The extracellular matrix in the tumour microenvironment can regulate cancer cell growth and progression. A pan-cancer analysis of TCGA data from 30 cancer types, identified the top 5% of matrisome genes with amplifications or deletions in their copy number, that affect their expression and cancer survival. A similar analysis of matrisome genes in individual cancers identified CTHRC1 to be significantly altered. CTHRC1, a regulator of collagen synthesis, was identified as the most prominently upregulated matrisome gene of interest across cancers. Differential gene expression analysis identified 19 genes whose expression is increased with CTHRC1. STRING analysis of these genes classified them as ‘extracellular’, involved most prominently in ECM organization and cell adhesion. KEGG analysis showed their involvement in ECM-receptor and growth factor signalling. Cytohubba analysis of these genes revealed 13 hub genes, of which MMP13, POSTN, SFRP4, ADAMTS16 and FNDC1 were significantly altered in their expression with CTHRC1 and seen to affect survival across cancers. This could in part be mediated by their overlapping roles in regulating ECM (collagen or fibronectin) expression and organisation. In breast cancer tumour samples CTHRC1 protein levels are significantly upregulated with POSTN and MMP13, further supporting the need to evaluate their crosstalk in cancers.
Collapse
|
42
|
Si H, Zhao N, Pedroza A, Zaske AM, Rosen JM, Creighton CJ, Roarty K. Noncanonical Wnt/Ror2 signaling regulates cell-matrix adhesion to prompt directional tumor cell invasion in breast cancer. Mol Biol Cell 2022; 33:ar103. [PMID: 36001375 PMCID: PMC9582800 DOI: 10.1091/mbc.e22-02-0055] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 07/25/2022] [Accepted: 08/18/2022] [Indexed: 12/03/2022] Open
Abstract
Cell-extracellular matrix (ECM) interactions represent fundamental exchanges during tumor progression, yet how particular signal-transduction factors prompt the conversion of tumor cells into migratory populations capable of systemic spread during metastasis remains elusive. We demonstrate that the noncanonical Wnt receptor, Ror2, regulates tumor cell-driven matrix remodeling and invasion in breast cancer. Ror2 loss-of-function (LOF) triggers the disruption of E-cadherin within tumor cells, accompanied by an increase in tumor cell invasion and collagen realignment in three-dimensional cultures. RNA sequencing of Ror2-deficient organoids further uncovered alterations in actin cytoskeleton, cell adhesion, and collagen cross-linking gene expression programs. Spatially, we pinpoint the up-regulation and redistribution of α5 and β3 integrins together with the production of fibronectin in areas of invasion downstream of Ror2 loss. Wnt/β-catenin-dependent and Wnt/Ror2 alternative Wnt signaling appear to regulate distinct functions for tumor cells regarding their ability to modify cell-ECM exchanges during invasion. Furthermore, blocking either integrin or focal adhesion kinase (FAK), a downstream mediator of integrin-mediated signal transduction, abrogates the enhanced migration observed upon Ror2 loss. These results reveal a critical function for the alternative Wnt receptor, Ror2, as a determinant of tumor cell-driven ECM exchanges during cancer invasion and metastasis.
Collapse
Affiliation(s)
- Hongjiang Si
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030
| | - Na Zhao
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030
| | - Andrea Pedroza
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030
| | - Ana-Maria Zaske
- University of Texas Health Science Center at Houston, Houston, TX 77054
| | - Jeffrey M. Rosen
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030
- Breast Cancer Program, Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030
| | - Chad J. Creighton
- Breast Cancer Program, Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030
| | - Kevin Roarty
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030
- Breast Cancer Program, Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030
| |
Collapse
|
43
|
Cuccu A, Francescangeli F, De Angelis ML, Bruselles A, Giuliani A, Zeuner A. Analysis of Dormancy-Associated Transcriptional Networks Reveals a Shared Quiescence Signature in Lung and Colorectal Cancer. Int J Mol Sci 2022; 23:9869. [PMID: 36077264 PMCID: PMC9456317 DOI: 10.3390/ijms23179869] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 08/23/2022] [Accepted: 08/25/2022] [Indexed: 11/17/2022] Open
Abstract
Quiescent cancer cells (QCCs) are a common feature of solid tumors, representing a major obstacle to the long-term success of cancer therapies. We isolated QCCs ex vivo from non-small cell lung cancer (NSCLC) and colorectal cancer (CRC) xenografts with a label-retaining strategy and compared QCCs gene expression profiles to identify a shared "quiescence signature". Principal Component Analysis (PCA) revealed a specific component neatly discriminating quiescent and replicative phenotypes in NSCLC and CRC. The discriminating component showed significant overlapping, with 688 genes in common including ZEB2, a master regulator of stem cell plasticity and epithelial-to-mesenchymal transition (EMT). Gene set enrichment analysis showed that QCCs of both NSCLC and CRC had an increased expression of factors related to stemness/self renewal, EMT, TGF-β, morphogenesis, cell adhesion and chemotaxis, whereas proliferating cells overexpressed Myc targets and factors involved in RNA metabolism. Eventually, we analyzed in depth by means of a complex network approach, both the 'morphogenesis module' and the subset of differentially expressed genes shared by NCSLC and CRC. This allowed us to recognize different gene regulation network wiring for quiescent and proliferating cells and to underpin few genes central for network integration that may represent new therapeutic vulnerabilities. Altogether, our results highlight common regulatory pathways in QCCs of lung and colorectal tumors that may be the target of future therapeutic interventions.
Collapse
Affiliation(s)
- Adriano Cuccu
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | - Federica Francescangeli
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | - Maria Laura De Angelis
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | - Alessandro Bruselles
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | - Alessandro Giuliani
- Environment and Health Department, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | - Ann Zeuner
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| |
Collapse
|
44
|
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: 3.0] [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
|
45
|
Kadonosono T, Miyamoto K, Sakai S, Matsuo Y, Kitajima S, Wang Q, Endo M, Niibori M, Kuchimaru T, Soga T, Hirota K, Kizaka-Kondoh S. AGE/RAGE axis regulates reversible transition to quiescent states of ALK-rearranged NSCLC and pancreatic cancer cells in monolayer cultures. Sci Rep 2022; 12:9886. [PMID: 35701529 PMCID: PMC9198021 DOI: 10.1038/s41598-022-14272-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 06/03/2022] [Indexed: 12/17/2022] Open
Abstract
Cancer recurrence due to tumor cell quiescence after therapy and long-term remission is associated with cancer-related death. Previous studies have used cell models that are unable to return to a proliferative state; thus, the transition between quiescent and proliferative states is not well understood. Here, we report monolayer cancer cell models wherein the human non-small cell lung carcinoma cell line H2228 and pancreatic cancer cell line AsPC-1 can be reversibly induced to a quiescent state under hypoxic and serum-starved (HSS) conditions. Transcriptome and metabolome dual-omics profiles of these cells were compared with those of the human lung adenocarcinoma cell line A549, which was unable to enter a quiescent state under HSS conditions. The quiescence-inducible cells had substantially lower intracellular pyruvate and ATP levels in the quiescent state than in the proliferative state, and their response to sudden demand for energy was dramatically reduced. Furthermore, in quiescence-inducible cells, the transition between quiescent and proliferative states of these cells was regulated by the balance between the proliferation-promoting Ras and Rap1 signaling and the suppressive AGE/RAGE signaling. These cell models elucidate the transition between quiescent and proliferative states, allowing the development of drug-screening systems for quiescent tumor cells.
Collapse
Affiliation(s)
- Tetsuya Kadonosono
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, 226-8501, Japan
| | - Kotaro Miyamoto
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, 226-8501, Japan
| | - Shiori Sakai
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, 226-8501, Japan
| | - Yoshiyuki Matsuo
- Department of Human Stress Response Science, Institute of Biomedical Science, Kansai Medical University, Hirakata, 573-1010, Japan
| | - Shojiro Kitajima
- Institute for Advanced Biosciences, Keio University, Tsuruoka, 997-0052, Japan
| | - Qiannan Wang
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, 226-8501, Japan
| | - Minori Endo
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, 226-8501, Japan
| | - Mizuho Niibori
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, 226-8501, Japan
| | - Takahiro Kuchimaru
- Center for Molecular Medicine, Jichi Medical University, Shimotsuke, 329-0498, Japan
| | - Tomoyoshi Soga
- Institute for Advanced Biosciences, Keio University, Tsuruoka, 997-0052, Japan
| | - Kiichi Hirota
- Department of Human Stress Response Science, Institute of Biomedical Science, Kansai Medical University, Hirakata, 573-1010, Japan
| | - Shinae Kizaka-Kondoh
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, 226-8501, Japan.
| |
Collapse
|
46
|
Chowdhury F, Huang B, Wang N. Forces in stem cells and cancer stem cells. Cells Dev 2022; 170:203776. [DOI: 10.1016/j.cdev.2022.203776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 02/26/2022] [Accepted: 03/22/2022] [Indexed: 10/18/2022]
|
47
|
Pritchard JR, Lee MJ, Peyton SR. Materials-driven approaches to understand extrinsic drug resistance in cancer. SOFT MATTER 2022; 18:3465-3472. [PMID: 35445686 PMCID: PMC9380814 DOI: 10.1039/d2sm00071g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2023]
Abstract
Metastatic cancer has a poor prognosis, because it is broadly disseminated and associated with both intrinsic and acquired drug resistance. Critical unmet needs in effectively killing drug resistant cancer cells include overcoming the drug desensitization characteristics of some metastatic cancers/lesions, and tailoring therapeutic regimens to both the tumor microenvironment and the genetic profiles of the resident cancer cells. Bioengineers and materials scientists are developing technologies to determine how metastatic sites exclude therapies, and how extracellular factors (including cells, proteins, metabolites, extracellular matrix, and abiotic factors) at metastatic sites significantly affect drug pharmacodynamics. Two looming challenges are determining which feature, or combination of features, from the tumor microenvironment drive drug resistance, and what the relative impact is of extracellular signals vs. intrinsic cell genetics in determining drug response. Sophisticated systems biology tools that can de-convolve a crowded network of signals and responses, as well as controllable microenvironments capable of providing discrete and tunable extracellular cues can help us begin to interrogate the high dimensional interactions governing drug resistance in patients.
Collapse
Affiliation(s)
- Justin R Pritchard
- Department of Biomedical Engineering, Pennsylvania State University, State College PA, USA
| | - Michael J Lee
- Department of Systems Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Shelly R Peyton
- Department of Chemical Engineering, University of Massachusetts Amherst, 240 Thatcher Way, Life Sciences Laboratory N531, Amherst, MA 01003, USA.
| |
Collapse
|
48
|
Ronaldson-Bouchard K, Baldassarri I, Tavakol DN, Graney PL, Samaritano M, Cimetta E, Vunjak-Novakovic G. Engineering complexity in human tissue models of cancer. Adv Drug Deliv Rev 2022; 184:114181. [PMID: 35278521 PMCID: PMC9035134 DOI: 10.1016/j.addr.2022.114181] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 02/15/2022] [Accepted: 03/04/2022] [Indexed: 02/06/2023]
Abstract
Major progress in the understanding and treatment of cancer have tremendously improved our knowledge of this complex disease and improved the length and quality of patients' lives. Still, major challenges remain, in particular with respect to cancer metastasis which still escapes effective treatment and remains responsible for 90% of cancer related deaths. In recent years, the advances in cancer cell biology, oncology and tissue engineering converged into the engineered human tissue models of cancer that are increasingly recapitulating many aspects of cancer progression and response to drugs, in a patient-specific context. The complexity and biological fidelity of these models, as well as the specific questions they aim to investigate, vary in a very broad range. When selecting and designing these experimental models, the fundamental question is "how simple is complex enough" to accomplish a specific goal of cancer research. Here we review the state of the art in developing and using the human tissue models in cancer research and developmental drug screening. We describe the main classes of models providing different levels of biological fidelity and complexity, discuss their advantages and limitations, and propose a framework for designing an appropriate model for a given study. We close by outlining some of the current needs, opportunities and challenges in this rapidly evolving field.
Collapse
Affiliation(s)
- Kacey Ronaldson-Bouchard
- Department of Biomedical Engineering, Columbia University, 622 West 168th Street, VC12-234, New York, NY 10032, USA
| | - Ilaria Baldassarri
- Department of Biomedical Engineering, Columbia University, 622 West 168th Street, VC12-234, New York, NY 10032, USA
| | - Daniel Naveed Tavakol
- Department of Biomedical Engineering, Columbia University, 622 West 168th Street, VC12-234, New York, NY 10032, USA
| | - Pamela L Graney
- Department of Biomedical Engineering, Columbia University, 622 West 168th Street, VC12-234, New York, NY 10032, USA
| | - Maria Samaritano
- Department of Biomedical Engineering, Columbia University, 622 West 168th Street, VC12-234, New York, NY 10032, USA
| | - Elisa Cimetta
- Department of Industrial Engineering, University of Padua, Via Marzolo 9, 35131 Padova, Italy; Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Corso Stati Uniti 4, 35127 Padova, Italy
| | - Gordana Vunjak-Novakovic
- Department of Biomedical Engineering, Columbia University, 622 West 168th Street, VC12-234, New York, NY 10032, USA; Department of Medicine, Columbia University, 622 West 168th Street, VC12-234, New York, NY 10032, USA; College of Dental Medicine, Columbia University, 622 West 168th Street, VC12-234, New York, NY 10032, USA.
| |
Collapse
|
49
|
Mikuła-Pietrasik J, Rutecki S, Książek K. The functional multipotency of transforming growth factor β signaling at the intersection of senescence and cancer. Cell Mol Life Sci 2022; 79:196. [PMID: 35305149 PMCID: PMC11073081 DOI: 10.1007/s00018-022-04236-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 02/22/2022] [Accepted: 03/08/2022] [Indexed: 12/12/2022]
Abstract
The transforming growth factor β (TGF-β) family of cytokines comprises a group of proteins, their receptors, and effector molecules that, in a coordinated manner, modulate a plethora of physiological and pathophysiological processes. TGF-β1 is the best known and plausibly most active representative of this group. It acts as an immunosuppressant, contributes to extracellular matrix remodeling, and stimulates tissue fibrosis, differentiation, angiogenesis, and epithelial-mesenchymal transition. In recent years, this cytokine has been established as a vital regulator of organismal aging and cellular senescence. Finally, the role of TGF-β1 in cancer progression is no longer in question. Because this protein is involved in so many, often overlapping phenomena, the question arises whether it can be considered a molecular bridge linking some of these phenomena together and governing their reciprocal interactions. In this study, we reviewed the literature from the perspective of the role of various TGF-β family members as regulators of a complex mutual interplay between senescence and cancer. These aspects are then considered in a broader context of remaining TGF-β-related functions and coexisting processes. The main narrative axis in this work is centered around the interaction between the senescence of normal peritoneal cells and ovarian cancer cells. The discussion also includes examples of TGF-β activity at the interface of other normal and cancer cell types.
Collapse
Affiliation(s)
- Justyna Mikuła-Pietrasik
- Department of Pathophysiology of Ageing and Civilization Diseases, Długa ½ Str, Poznań University of Medical Sciences, 61-848, Poznań, Poland
| | - Szymon Rutecki
- Department of Pathophysiology of Ageing and Civilization Diseases, Długa ½ Str, Poznań University of Medical Sciences, 61-848, Poznań, Poland
| | - Krzysztof Książek
- Department of Pathophysiology of Ageing and Civilization Diseases, Długa ½ Str, Poznań University of Medical Sciences, 61-848, Poznań, Poland.
| |
Collapse
|
50
|
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: 9.0] [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
|