1
|
Hulahan TS, Angel PM. From ductal carcinoma in situ to invasive breast cancer: the prognostic value of the extracellular microenvironment. J Exp Clin Cancer Res 2024; 43:329. [PMID: 39716322 DOI: 10.1186/s13046-024-03236-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] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 11/19/2024] [Indexed: 12/25/2024] Open
Abstract
Ductal carcinoma in situ (DCIS) is a noninvasive breast disease that variably progresses to invasive breast cancer (IBC). Given the unpredictability of this progression, most DCIS patients are aggressively managed similar to IBC patients. Undoubtedly, this treatment paradigm places many DCIS patients at risk of overtreatment and its significant consequences. Historically, prognostic modeling has included the assessment of clinicopathological features and genomic markers. Although these provide valuable insights into tumor biology, they remain insufficient to predict which DCIS patients will progress to IBC. Contemporary work has begun to focus on the microenvironment surrounding the ductal cells for molecular patterns that might predict progression. In this review, extracellular microenvironment alterations occurring with the malignant transformation from DCIS to IBC are detailed. Not only do changes in collagen abundance, organization, and localization mediate the transition to IBC, but also the discrete post-translational regulation of collagen fibers is understood to promote invasion. Other extracellular matrix proteins, such as matrix metalloproteases, decorin, and tenascin C, have been characterized for their role in invasive transformation and further demonstrate the prognostic value of the extracellular matrix. Importantly, these extracellular matrix proteins influence immune cells and fibroblasts toward pro-tumorigenic phenotypes. Thus, the progressive changes in the extracellular microenvironment play a key role in invasion and provide promise for prognostic development.
Collapse
Affiliation(s)
- Taylor S Hulahan
- Department of Pharmacology and Immunology, Medical University of South Carolina, Charleston, SC, USA
| | - Peggi M Angel
- Department of Pharmacology and Immunology, Medical University of South Carolina, Charleston, SC, USA.
| |
Collapse
|
2
|
Torre-Cea I, Berlana-Galán P, Guerra-Paes E, Cáceres-Calle D, Carrera-Aguado I, Marcos-Zazo L, Sánchez-Juanes F, Muñoz-Félix JM. Basement membranes in lung metastasis growth and progression. Matrix Biol 2024:S0945-053X(24)00150-1. [PMID: 39719224 DOI: 10.1016/j.matbio.2024.12.008] [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: 11/05/2024] [Revised: 12/19/2024] [Accepted: 12/20/2024] [Indexed: 12/26/2024]
Abstract
The lung is a highly vascularized tissue that often harbors metastases from various extrathoracic malignancies. Lung parenchyma consists of a complex network of alveolar epithelial cells and microvessels, structured within an architecture defined by basement membranes. Consequently, understanding the role of the extracellular matrix (ECM) in the growth of lung metastases is essential to uncover the biology of this pathology and developing targeted therapies. These basement membranes play a critical role in the progression of lung metastases, influencing multiple stages of the metastatic cascade, from the acquisition of an aggressive phenotype to intravasation, extravasation and colonization of secondary sites. This review examines the biological composition of basement membranes, focusing on their core components-collagens, fibronectin, and laminin-and their specific roles in cancer progression. Additionally, we discuss the function of integrins as primary mediators of cell adhesion and signaling between tumor cells, basement membranes and the extracellular matrix, as well as their implications for metastatic growth in the lung. We also explore vascular co-option (VCO) as a form of tumor growth resistance linked to basement membranes and tumor vasculature. Finally, the review covers current clinical therapies targeting tumor adhesion, extracellular matrix remodeling, and vascular development, aiming to improve the precision and effectiveness of treatments against lung metastases.
Collapse
Affiliation(s)
- Irene Torre-Cea
- Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, Instituto de Investigación Biomédica de Salamanca (IBSAL)
| | - Patricia Berlana-Galán
- Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, Instituto de Investigación Biomédica de Salamanca (IBSAL)
| | - Elena Guerra-Paes
- Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, Instituto de Investigación Biomédica de Salamanca (IBSAL)
| | - Daniel Cáceres-Calle
- Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, Instituto de Investigación Biomédica de Salamanca (IBSAL)
| | - Iván Carrera-Aguado
- Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, Instituto de Investigación Biomédica de Salamanca (IBSAL)
| | - Laura Marcos-Zazo
- Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, Instituto de Investigación Biomédica de Salamanca (IBSAL)
| | - Fernando Sánchez-Juanes
- Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, Instituto de Investigación Biomédica de Salamanca (IBSAL).
| | - Jose M Muñoz-Félix
- Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, Instituto de Investigación Biomédica de Salamanca (IBSAL).
| |
Collapse
|
3
|
Lv K, He T. Cancer-associated fibroblasts: heterogeneity, tumorigenicity and therapeutic targets. MOLECULAR BIOMEDICINE 2024; 5:70. [PMID: 39680287 DOI: 10.1186/s43556-024-00233-8] [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: 07/24/2024] [Revised: 11/04/2024] [Accepted: 11/19/2024] [Indexed: 12/17/2024] Open
Abstract
Cancer, characterized by its immune evasion, active metabolism, and heightened proliferation, comprises both stroma and cells. Although the research has always focused on parenchymal cells, the non-parenchymal components must not be overlooked. Targeting cancer parenchymal cells has proven to be a formidable challenge, yielding limited success on a broad scale. The tumor microenvironment(TME), a critical niche for cancer cell survival, presents a novel way for cancer treatment. Cancer-associated fibroblast (CAF), as a main component of TME, is a dynamically evolving, dual-functioning stromal cell. Furthermore, their biological activities span the entire spectrum of tumor development, metastasis, drug resistance, and prognosis. A thorough understanding of CAFs functions and therapeutic advances holds significant clinical implications. In this review, we underscore the heterogeneity of CAFs by elaborating on their origins, types and function. Most importantly, by elucidating the direct or indirect crosstalk between CAFs and immune cells, the extracellular matrix, and cancer cells, we emphasize the tumorigenicity of CAFs in cancer. Finally, we highlight the challenges encountered in the exploration of CAFs and list targeted therapies for CAF, which have implications for clinical treatment.
Collapse
Affiliation(s)
- Keke Lv
- Department of Hepatopanreatobiliary Surgery, Changhai Hospital, 168 Changhai Road, Yangpu District, Shanghai, 200433, China
| | - Tianlin He
- Department of Hepatopanreatobiliary Surgery, Changhai Hospital, 168 Changhai Road, Yangpu District, Shanghai, 200433, China.
| |
Collapse
|
4
|
Rodponthukwaji K, Thongchot S, Deureh S, Thongkleang T, Thaweesuvannasak M, Srichan K, Srisawat C, Thuwajit P, Nguyen KT, Tadpetch K, Thuwajit C, Punnakitikashem P. Development of cancer-associated fibroblasts-targeting polymeric nanoparticles loaded with 8- O-methylfusarubin for breast cancer treatment. Int J Pharm X 2024; 8:100294. [PMID: 39507587 PMCID: PMC11539325 DOI: 10.1016/j.ijpx.2024.100294] [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: 07/17/2024] [Revised: 10/05/2024] [Accepted: 10/15/2024] [Indexed: 11/08/2024] Open
Abstract
Cancer-associated fibroblasts (CAFs) are abundant stromal cells residing in a tumor microenvironment (TME) which are associated with the progression of tumor. Herein, we developed novel CAFs-targeting polymeric nanoparticles encapsulating a synthetic 8-O-methylfusarubin (OMF) compound (OMF@NPs-anti-FAP). Anti-FAP/fibroblast activation protein antibody was employed as a CAFs-targeting ligand. The physicochemical properties of the synthesized nanomaterials were firstly investigated with various techniques. The cytocompatibility of polymeric nanoparticles (NPs) was elicited through cell viability of CAFs and human breast epithelial cells, MCF-10A. Additionally, the anti-FAP-conjugated NPs displayed different degrees of cellular internalization regarding the FAP expression level on the CAFs' surface. However, CAFs exposed to NPs containing OMF demonstrated significant cell death which were associated with the apoptotic pathway as confirmed by caspase-3/7 activity. Upon OMF@NPs-anti-FAP treatment, an enhanced toxicity was clearly observed in 3D spheroid models. High FAP-expressed PC-B-132CAFs demonstrated a high percentage of cell death compared to other cells with a low level of FAP expression analyzed by flow cytometry (e.g. MCF-10A, HDFa, and PC-B-142CAFs). This result emphasized the importance of anti-FAP antibody as a targeting ligand. These findings suggest that the fabricated nanosystem of OMF-loaded polymeric NPs with CAFs' high specificity holds a potential NP-based platform for improvement in breast cancer treatment.
Collapse
Affiliation(s)
- Kamonlatth Rodponthukwaji
- Department of Biochemistry, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
- Siriraj Center of Research Excellence in Theranostic Nanomedicine, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Suyanee Thongchot
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
- Siriraj Center of Research Excellence for Cancer Immunotherapy (SiCORE-CIT), Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Suttikiat Deureh
- Department of Biochemistry, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Tanva Thongkleang
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Mattika Thaweesuvannasak
- Department of Biochemistry, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Kornrawee Srichan
- Department of Biochemistry, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Chatchawan Srisawat
- Department of Biochemistry, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
- Siriraj Center of Research Excellence in Theranostic Nanomedicine, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Peti Thuwajit
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Kytai T. Nguyen
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX 76019, USA
| | - Kwanruthai Tadpetch
- Division of Physical Science and Center of Excellence for Innovation in Chemistry, Faculty of Science, Prince of Songkla University, Hat Yai, Songkhla 90110, Thailand
| | - Chanitra Thuwajit
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Primana Punnakitikashem
- Department of Biochemistry, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
- Siriraj Center of Research Excellence in Theranostic Nanomedicine, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| |
Collapse
|
5
|
de la Jara Ortiz F, Cimmino C, Ventre M, Cambi A. Understanding and measuring mechanical signals in the tumor stroma. FEBS Open Bio 2024. [PMID: 39523476 DOI: 10.1002/2211-5463.13923] [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: 06/30/2024] [Revised: 09/30/2024] [Accepted: 10/25/2024] [Indexed: 11/16/2024] Open
Abstract
The tumor microenvironment (TME) is well known for its immune suppressive role, especially in solid tumors which are characterized by a thick, dense stroma. Apart from cell-cell interactions and biochemical signals, the tumor stroma is also characterized by its distinct mechanical properties, which are dictated by the composition and architecture of its extracellular matrix (ECM). Cancer-associated fibroblasts (CAFs) are the main producers and remodelers of the stromal ECM, and their heterogeneity has recently become a focus of intense research. This review describes recent findings highlighting CAF subtypes and their specific functions, as well as the development of 3D models to study tumor stroma mechanics in vitro. Finally, we discuss the quantitative techniques used to measure tissue mechanical properties at different scales. Given the diagnostic and prognostic value of stroma stiffness and composition, and the recent development of anti-tumor therapeutic strategies targeting the stroma, understanding and measuring tumor stroma mechanical properties has never been more timely or relevant.
Collapse
Affiliation(s)
- Fàtima de la Jara Ortiz
- Department of Medical BioSciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Chiara Cimmino
- Department of Chemical, Materials and Industrial Production Engineering, University of Naples Federico II, Naples, Italy
- Center for Advanced Biomaterials for Healthcare@CRIB, Fondazione Istituto Italiano di Tecnologia, Naples, Italy
| | - Maurizio Ventre
- Department of Chemical, Materials and Industrial Production Engineering, University of Naples Federico II, Naples, Italy
- Center for Advanced Biomaterials for Healthcare@CRIB, Fondazione Istituto Italiano di Tecnologia, Naples, Italy
- Interdisciplinary Research Centre on Biomaterials, University of Naples Federico II, Naples, Italy
| | - Alessandra Cambi
- Department of Medical BioSciences, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
6
|
Yuan T, Fu X, Hu R, Zheng X, Jiang D, Jing L, Kuang X, Guo Z, Luo X, Liu Y, Zou X, Luker GD, Mi S, Liu C, Sun W. Bioprinted, spatially defined breast tumor microenvironment models of intratumoral heterogeneity and drug resistance. Trends Biotechnol 2024; 42:1523-1550. [PMID: 39112274 DOI: 10.1016/j.tibtech.2024.06.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 06/13/2024] [Accepted: 06/17/2024] [Indexed: 11/17/2024]
Abstract
Cellular, extracellular matrix (ECM), and spatial heterogeneity of tumor microenvironments (TMEs) regulate disease progression and treatment efficacy. Developing in vitro models that recapitulate the TME promises to accelerate studies of tumor biology and identify new targets for therapy. Here, we used extrusion-based, multi-nozzle 3D bioprinting to spatially pattern triple-negative MDA-MB-231 breast cancer cells, endothelial cells (ECs), and human mammary cancer-associated fibroblasts (HMCAFs) with biomimetic ECM inks. Bioprinted models captured key features of the spatial architecture of human breast tumors, including varying-sized dense regions of cancer cells and surrounding microvessel-rich stroma. Angiogenesis and ECM stiffening occurred in the stromal area but not the cancer cell-rich (CCR) regions, mimicking pathological changes in patient samples. Transcriptomic analyses revealed upregulation of angiogenesis-related and ECM remodeling-related signatures in the stroma region and identified potential ligand-receptor (LR) mediators of these processes. Breast cancer cells in distinct parts of the bioprinted TME showed differing sensitivities to chemotherapy, highlighting environmentally mediated drug resistance. In summary, our 3D-bioprinted tumor model will act as a platform to discover integrated functions of the TME in cancer biology and therapy.
Collapse
Affiliation(s)
- Tianying Yuan
- Tsinghua-Berkeley Shenzhen Institute, Tsinghua University, 518055, Shenzhen, China; Tsinghua Shenzhen International Graduate School, Tsinghua University, 518055, Shenzhen, China
| | - Xihong Fu
- Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-sen University, 510080, Guangzhou, China; Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, 510080, Guangzhou, China
| | - Rongcheng Hu
- Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-sen University, 510080, Guangzhou, China; Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, 510080, Guangzhou, China
| | - Xiaochun Zheng
- Tsinghua Shenzhen International Graduate School, Tsinghua University, 518055, Shenzhen, China
| | - Dong Jiang
- Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-sen University, 510080, Guangzhou, China; Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, 510080, Guangzhou, China
| | - Lanyu Jing
- Department of Breast Surgery, The First Affiliated Hospital of Sun Yat-sen University, 510080, Guangzhou, China
| | - Xiaying Kuang
- Department of Breast Surgery, The First Affiliated Hospital of Sun Yat-sen University, 510080, Guangzhou, China
| | - Zhongwei Guo
- School of Mechanics and Safety Engineering, Zhengzhou University, 450001, Zhengzhou, China
| | - Xu Luo
- Tsinghua Shenzhen International Graduate School, Tsinghua University, 518055, Shenzhen, China
| | - Yixin Liu
- Tsinghua Shenzhen International Graduate School, Tsinghua University, 518055, Shenzhen, China
| | - Xuenong Zou
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, 510080, Guangzhou, China
| | - Gary D Luker
- Department of Radiology, University of Michigan, Ann Arbor, MI 48109, USA; Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA.
| | - Shengli Mi
- Tsinghua Shenzhen International Graduate School, Tsinghua University, 518055, Shenzhen, China.
| | - Chun Liu
- Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-sen University, 510080, Guangzhou, China; Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, 510080, Guangzhou, China.
| | - Wei Sun
- Tsinghua-Berkeley Shenzhen Institute, Tsinghua University, 518055, Shenzhen, China; Department of Mechanical Engineering, Biomanufacturing Center, Tsinghua University, 100084, Beijing, China; Department of Mechanical Engineering, Drexel University, Philadelphia, PA 19104, USA.
| |
Collapse
|
7
|
Zhang B, Chen X, Song H, Gao X, Ma S, Ji H, Qu H, Xia S, Shang D. Identification of basement membrane-related prognostic model associated with the immune microenvironment and synthetic therapy response in pancreatic cancer: integrated bioinformatics analysis and clinical validation. J Cancer 2024; 15:6273-6298. [PMID: 39513120 PMCID: PMC11540510 DOI: 10.7150/jca.100891] [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: 07/13/2024] [Accepted: 09/28/2024] [Indexed: 11/15/2024] Open
Abstract
Pancreatic cancer (PC) is a common and highly malignant tumor. Basement membrane (BM) is formed by the crosslinking of extracellular matrix macromolecules and acts as a barrier against tumor cell metastasis. However, the role of BM in PC prognosis, immune infiltration, and treatment remains unclear. This study collected transcriptome and clinical survival data of PC via TCGA, GEO, and ICGC databases. PC patients (PCs) from the First Affiliated Hospital of Dalian Medical University were obtained as the clinical validation cohort. BM-related genes (BMRGs) were acquired from GeneCards and basement membraneBASE databases. A total of 46 differential-expressed BMRGs were identified. Then the BM-related prognostic model (including DSG3, MET, and PLAU) was built and validated. PCs with a low BM-related score had a better outcome and were more likely to benefit from oxaliplatin, irinotecan, and KRAS(G12C) inhibitor-12, and immunotherapy. Immune analysis revealed that BM-related score was positively correlated with neutrophils, cancer-associated fibroblasts, and macrophages infiltration, but negatively correlated with CD8+ T cells, NK cells, and B cells infiltration. PCs from the clinical cohort further verified that BM-related model could accurately predict PCs' outcomes. DSG3, MET, and PLAU were notably up-regulated within PC tissues and linked to a poor prognosis. In vitro experiments showed that DSG3 knockdown markedly suppressed the proliferation, migration, and invasion of PC cells. Molecular docking indicated that epigallocatechin gallate had a strong binding activity with DSG3, MET, and PLAU and may be used as a potential therapeutic agent for PC. In conclusion, this study developed a BM-related model associated with PC prognosis, immune infiltration, and treatment, which provided new insights into PC stratification and drug intervention.
Collapse
Affiliation(s)
- Biao Zhang
- Pancreas & Biliary Center, Department of General Surgery, Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Xu Chen
- Pancreas & Biliary Center, Department of General Surgery, Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Huiyi Song
- Pancreas & Biliary Center, Department of General Surgery, Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, China
| | - Xue Gao
- Department of Pathology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Shurong Ma
- Pancreas & Biliary Center, Department of General Surgery, Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, China
| | - Hongying Ji
- Department of Pathology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Huixian Qu
- Department of Pathology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Shilin Xia
- Pancreas & Biliary Center, Department of General Surgery, Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, China
| | - Dong Shang
- Pancreas & Biliary Center, Department of General Surgery, Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, China
| |
Collapse
|
8
|
Puebla-Osorio N, Fowlkes NW, Barsoumian HB, Xega K, Srivastava G, Kettlun-Leyton C, Nizzero S, Voss T, Riad TS, Wong C, Huang A, Hu Y, Mitchell J, Kim M, Rafiq Z, He K, Sezen D, Hsu E, Masrorpour F, Maleki A, Leuschner C, Cortez MA, Oertle P, Loparic M, Plodinec M, Markman JL, Welsh JW. Enhanced tumor control and survival in preclinical models with adoptive cell therapy preceded by low-dose radiotherapy. Front Oncol 2024; 14:1407143. [PMID: 39445067 PMCID: PMC11496962 DOI: 10.3389/fonc.2024.1407143] [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: 03/26/2024] [Accepted: 09/04/2024] [Indexed: 10/25/2024] Open
Abstract
Introduction Effective infiltration of chimeric antigen receptor T (CAR-T) cells into solid tumors is critical for achieving a robust antitumor response and improving therapeutic outcomes. While CAR-T cell therapies have succeeded in hematologic malignancies, their efficacy in solid tumors remains limited due to poor tumor penetration and an immunosuppressive tumor microenvironment. This study aimed to evaluate the potential of low-dose radiotherapy (LDRT) administered before T-cell therapy to enhance the antitumor effect by promoting CAR-T cell infiltration. We hypothesized that combining LDRT with T-cell therapy would improve tumor control and survival compared to either treatment alone. Methods We investigated this hypothesis using two NSG mouse models bearing GSU or CAPAN-2 solid tumors. The mice were treated with engineered CAR-T cells targeting guanyl cyclase-C (GCC) or mesothelin as monotherapy or in combination with LDRT. Additionally, we extended this approach to a C57BL/6 mouse model implanted with MC38-gp100+ cells, followed by adoptive transfer of pmel+ T cells before and after LDRT. Tumor growth and survival outcomes were monitored in all models. Furthermore, we employed atomic force microscopy (AFM) in a small cohort to assess the effects of radiotherapy on tumor stiffness and plasticity, exploring the role of tumor nanomechanics as a potential biomarker for treatment efficacy. Results Our results demonstrated enhanced tumor control and prolonged survival in mice treated with LDRT followed by T-cell therapy across all models. The combination of LDRT with CAR-T or pmel+ T-cell therapy led to superior tumor suppression and survival compared to monotherapy, highlighting the synergistic impact of the combined approach. Additionally, AFM analysis revealed significant changes in tumor stiffness and plasticity in response to LDRT, suggesting that the nanomechanical properties of the tumor may be predictive of therapeutic response. Discussion The findings of this study highlight the transformative potential of incorporating LDRT as a precursor to adoptive T-cell therapy in solid tumors. By promoting CAR-T and pmel+ T-cell infiltration into the tumor microenvironment, LDRT enhanced tumor control and improved survival outcomes, offering a promising strategy to overcome the challenges associated with CAR-T therapy in solid tumors. Additionally, the changes in tumor nanomechanics observed through AFM suggest that tumor stiffness and plasticity could be biomarkers for predicting treatment outcomes. These results support further investigation into the clinical application of this combined approach to improve the efficacy of cell-based therapies in patients with solid tumors.
Collapse
Affiliation(s)
- Nahum Puebla-Osorio
- Department of Radiation Oncology—Research, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Natalie Wall Fowlkes
- Department of Veterinary Medicine and Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Hampartsoum B. Barsoumian
- Department of Radiation Oncology—Research, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Kristina Xega
- Takeda Development Centers Americas, Inc, Lexington, MA, United States
| | | | - Claudia Kettlun-Leyton
- Department of Radiation Oncology—Research, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | | | - Tiffany Voss
- Department of Radiation Oncology—Research, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Thomas S. Riad
- Department of Radiation Oncology—Research, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Christina Wong
- Takeda Development Centers Americas, Inc, Lexington, MA, United States
| | - Ailing Huang
- Department of Radiation Oncology—Research, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Yun Hu
- Department of Radiation Oncology—Research, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Joylise Mitchell
- Department of Radiation Oncology—Research, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Mingee Kim
- Medical College of Wisconsin, Milwaukee, WI, United States
| | - Zahid Rafiq
- Department of Radiation Oncology—Research, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Kewen He
- Department of Radiation Oncology—Research, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Duygu Sezen
- Department of Radiation Oncology—Research, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Ethan Hsu
- Department of Radiation Oncology—Research, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Fatemeh Masrorpour
- Department of Radiation Oncology—Research, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Aurian Maleki
- Department of Radiation Oncology—Research, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Carola Leuschner
- Department of Radiation Oncology—Research, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Maria Angelica Cortez
- Department of Radiation Oncology—Research, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | | | | | | | - Janet L. Markman
- Takeda Development Centers Americas, Inc, Lexington, MA, United States
| | - James W. Welsh
- Department of Radiation Oncology—Research, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
9
|
Alekseenko I, Zhukova L, Kondratyeva L, Buzdin A, Chernov I, Sverdlov E. Tumor Cell Communications as Promising Supramolecular Targets for Cancer Chemotherapy: A Possible Strategy. Int J Mol Sci 2024; 25:10454. [PMID: 39408784 PMCID: PMC11476449 DOI: 10.3390/ijms251910454] [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: 09/05/2024] [Revised: 09/25/2024] [Accepted: 09/26/2024] [Indexed: 10/20/2024] Open
Abstract
Fifty-two years have passed since President Nixon launched the "War on Cancer". Despite unparalleled efforts and funds allocated worldwide, the outlined goals were not achieved because cancer treatment approaches such as chemotherapy, radiation therapy, hormonal and targeted therapies have not fully met the expectations. Based on the recent literature, a new direction in cancer therapy can be proposed which targets connections between cancer cells and their microenvironment by chemical means. Cancer-stromal synapses such as immunological synapses between cancer and immune cells provide an attractive target for this approach. Such synapses form ligand-receptor clusters on the interface of the interacting cells. They share a common property of involving intercellular clusters of spatially proximate and cooperatively acting proteins. Synapses provide the space for the focused intercellular signaling molecules exchange. Thus, the disassembly of cancer-stromal synapses may potentially cause the collapse of various tumors. Additionally, the clustered arrangement of synapse components offers opportunities to enhance treatment safety and precision by using targeted crosslinking chemical agents which may inactivate cancer synapses even in reduced concentrations. Furthermore, attaching a cleavable cell-permeable toxic agent(s) to a crosslinker may further enhance the anti-cancer effect of such therapeutics. The highlighted approach promises to be universal, relatively simple and cost-efficient. We also hope that, unlike chemotherapeutic and immune drugs that interact with a single target, by using supramolecular large clusters that include many different components as a target, the emergence of a resistance characteristic of chemo- and immunotherapy is extremely unlikely.
Collapse
Affiliation(s)
- Irina Alekseenko
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 117997 Moscow, Russia; (I.A.); (A.B.); (I.C.)
- National Research Center “Kurchatov Institute”, 123182 Moscow, Russia
| | - Lyudmila Zhukova
- Department of Oncology, SBIH “Moscow Clinical Scientific and Practical Center Named After A.S. Loginov” DHM, 111123 Moscow, Russia;
| | - Liya Kondratyeva
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 117997 Moscow, Russia; (I.A.); (A.B.); (I.C.)
- National Research Center “Kurchatov Institute”, 123182 Moscow, Russia
| | - Anton Buzdin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 117997 Moscow, Russia; (I.A.); (A.B.); (I.C.)
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University, 119992 Moscow, Russia
- Oncobox LLC, 121205 Moscow, Russia
| | - Igor Chernov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 117997 Moscow, Russia; (I.A.); (A.B.); (I.C.)
| | - Eugene Sverdlov
- National Research Center “Kurchatov Institute”, 123182 Moscow, Russia
| |
Collapse
|
10
|
Xu N, Wu J, Wang W, Sun S, Sun M, Bian Y, Zhang H, Liu S, Yu G. Anti-tumor therapy of glycyrrhetinic acid targeted liposome co-delivery of doxorubicin and berberine for hepatocellular carcinoma. Drug Deliv Transl Res 2024; 14:2386-2402. [PMID: 38236508 DOI: 10.1007/s13346-023-01512-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/26/2023] [Indexed: 01/19/2024]
Abstract
During the development of hepatocellular carcinoma (HCC), hepatic stellate cells undergo activation and transform into cancer-associated fibroblasts (CAFs) due to the influence of tumor cells. The interaction between CAFs and tumor cells can compromise the effectiveness of chemotherapy drugs and promote tumor proliferation, invasion, and metastasis. This study explores the potential of glycyrrhetinic acid (GA)-modified liposomes (lip-GA) as a strategy for co-delivery of berberine (Ber) and doxorubicin (Dox) to treat HCC. The characterizations of liposomes, including particle size, zeta potential, polydispersity index, stability and in vitro drug release, were investigated. The study evaluated the anti-proliferation and anti-migration effects of Dox&Ber@lip-GA on the Huh-7 + LX-2 cell model were through MTT and wound-healing assays. Additionally, the in vivo drug distribution and anti-tumor efficacy were investigated using the H22 + NIH-3T3-bearing mouse model. The results indicated that Dox&Ber@lip-GA exhibited a nanoscale particle size, accumulated specifically in the tumor region, and was efficiently taken up by tumor cells. Compared to other groups, Dox&Ber@lip-GA demonstrated higher cytotoxicity and lower migration rates. Additionally, it significantly reduced the deposition of extracellular matrix (ECM) and inhibited tumor angiogenesis, thereby suppressing tumor growth. In conclusion, Dox&Ber@lip-GA exhibited superior anti-tumor effects both in vitro and in vivo, highlighting its potential as an effective therapeutic strategy for combating HCC.
Collapse
Affiliation(s)
- Na Xu
- School of Clinical Medicine, Weifang Medicine University, Weifang, China
- Department of Oncology, The First Affiliated Hospital of Weifang Medical College: Weifang People's Hospital, Weifang, China
| | - Jingliang Wu
- School of Nursing, Weifang University of Science and Technology, Weifang, China.
| | - Weihao Wang
- School of Clinical Medicine, Weifang Medicine University, Weifang, China
| | - Shujie Sun
- School of Nursing, Weifang University of Science and Technology, Weifang, China
| | - Mengmeng Sun
- School of Clinical Medicine, Weifang Medicine University, Weifang, China
- Department of Oncology, The First Affiliated Hospital of Weifang Medical College: Weifang People's Hospital, Weifang, China
| | - Yandong Bian
- School of Clinical Medicine, Weifang Medicine University, Weifang, China
| | - Huien Zhang
- School of Clinical Medicine, Weifang Medicine University, Weifang, China
| | - Shuzhen Liu
- School of Clinical Medicine, Weifang Medicine University, Weifang, China
- Department of Oncology, The First Affiliated Hospital of Weifang Medical College: Weifang People's Hospital, Weifang, China
| | - Guohua Yu
- School of Clinical Medicine, Weifang Medicine University, Weifang, China.
- Department of Oncology, The First Affiliated Hospital of Weifang Medical College: Weifang People's Hospital, Weifang, China.
| |
Collapse
|
11
|
Yang Y, Huang G, Lian J, Long C, Zhao B, Liu X, Zhang B, Ye W, Chen J, Du L, Jiang Z, Liu J, Zhang J, Hu C, Chen Q, Hong X. Circulating tumour cell clusters: isolation, biological significance and therapeutic implications. BMJ ONCOLOGY 2024; 3:e000437. [DOI: 10.1136/bmjonc-2024-000437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
Abstract
Circulating tumour cells (CTCs) and CTC clusters are considered metastatic precursors due to their ability to seed distant metastasis. However, navigating the bloodstream presents a significant challenge for CTCs, as they must endure fluid shear forces and resist detachment-induced anoikis. Consequently, while a large number of cells from the primary tumour may enter the circulation, only a tiny fraction will result in metastasis. Nevertheless, the metastatic potency dramatically increases when CTCs travel in conjunction with other cell types to form CTC clusters, including neutrophils, myeloid-derived suppressor cells, macrophages, platelets, cancer-associated fibroblasts and red blood cells found in circulation. Such heterotypic CTC clustering events have been identified in a variety of cancer types and may serve as intriguing therapeutic targets and novel biomarkers for liquid biopsy. This review summarises recent advances in microfluidic technologies designed for the isolation of CTC clusters and explores the biological properties of distinct types of CTC clusters within the circulatory system. Investigation of the mechanisms of CTC cluster–blood microenvironment interactions may offer a promising avenue for gaining fresh insights into CTC cluster-mediated metastatic progression and reveal potential opportunities for devising personalised antimetastasis treatments.
Collapse
|
12
|
Suh YJ, Li AT, Pandey M, Nordmann CS, Huang YL, Wu M. Decoding physical principles of cell migration under controlled environment using microfluidics. BIOPHYSICS REVIEWS 2024; 5:031302. [PMID: 39091432 PMCID: PMC11290890 DOI: 10.1063/5.0199161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Accepted: 06/26/2024] [Indexed: 08/04/2024]
Abstract
Living cells can perform incredible tasks that man-made micro/nano-sized robots have not yet been able to accomplish. One example is that white blood cells can sense and move to the site of pathogen attack within minutes. The robustness and precision of cellular functions have been perfected through billions of years of evolution. In this context, we ask the question whether cells follow a set of physical principles to sense, adapt, and migrate. Microfluidics has emerged as an enabling technology for recreating well-defined cellular environment for cell migration studies, and its ability to follow single cell dynamics allows for the results to be amenable for theoretical modeling. In this review, we focus on the development of microfluidic platforms for recreating cellular biophysical (e.g., mechanical stress) and biochemical (e.g., nutrients and cytokines) environments for cell migration studies in 3D. We summarize the basic principles that cells (including bacteria, algal, and mammalian cells) use to respond to chemical gradients learned from microfluidic systems. We also discuss about novel biological insights gained from studies of cell migration under biophysical cues and the need for further quantitative studies of cell function under well-controlled biophysical environments in the future.
Collapse
Affiliation(s)
- Young Joon Suh
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, New York 14853, USA
| | - Alan T. Li
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, New York 14853, USA
| | - Mrinal Pandey
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, New York 14853, USA
| | - Cassidy S. Nordmann
- Department of Biomedical Engineering, Cornell University, Ithaca, New York 14853, USA
| | - Yu Ling Huang
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, New York 14853, USA
| | - Mingming Wu
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, New York 14853, USA
| |
Collapse
|
13
|
Cai S, Zhao M, Yang G, Li C, Hu M, Yang L, Xing L, Sun X. Modified spatial architecture of regulatory T cells after neoadjuvant chemotherapy in non-small cell lung cancer patients. Int Immunopharmacol 2024; 137:112434. [PMID: 38889507 DOI: 10.1016/j.intimp.2024.112434] [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: 02/04/2024] [Revised: 05/17/2024] [Accepted: 06/05/2024] [Indexed: 06/20/2024]
Abstract
It is crucial to decipher the modulation of regulatory T cells (Tregs) in tumor microenvironment (TME) induced by chemotherapy, which may contribute to improving the efficacy of neoadjuvant chemoimmunotherapy in resectable non-small cell lung cancer (NSCLC). We retrospectively collected specimens from patients with II-III NSCLC, constituting two cohorts: a neoadjuvant chemotherapy (NAC) cohort (N = 141) with biopsy (N = 58) and postoperative specimens (N = 141), and a surgery-only cohort (N = 122) as the control group. Then, the cell density (Dens), infiltration score (InS), and Treg-cell proximity score (TrPS) were conducted using a panel of multiplex fluorescence staining (Foxp3, CD4, CD8, CK, CD31, ɑSMA). Subsequently, the association of Tregs with cancer microvessels (CMVs) and cancer-associated fibroblasts (CAFs) was analyzed. Patients with NAC treatment have a higher density of Tregs in both paired (P < 0.001) and unpaired analysis (P = 0.022). Additionally, patients with NAC treatment showed higher infiltration score (paired, P < 0.001; unpaired, P = 0.014) and more CD8+T cells around Tregs (paired/unpaired, both P < 0.001). Subgroup analysis indicated that tumors with a diameter of ≤ 5 cm exhibited increase in both Dens(Treg) and InS(Treg), and gemcitabine, pemetrexed and taxel enhanced Dens(Treg) and TrPS(CD8) following NAC. Multivariate analysis identified that the Dens(Tregs), InS(Tregs) and TrPS(CD8) were significantly associated with better chemotherapy response [OR = 8.54, 95%CI (1.69, 43.14), P = 0.009; OR = 7.14, 95%CI (1.70, 30.08), P = 0.024; OR = 5.50, 95%CI (1.09, 27.75), P = 0.039, respectively] and positive recurrence-free survival [HR = 3.23, 95%CI (1.47, 7.10), P = 0.004; HR = 2.70; 95%CI (1.27, 5.72); P = 0.010; HR = 2.55, 95%CI (1.21, 5.39), P = 0.014, respectively]. Moreover, TrPS(CD8) and TrPS(CD4) were negatively correlated with the CMVs and CAFs. These discoveries have deepened our comprehension of the immune-modulating impact of chemotherapy and underscored that the modified spatial landscape of Tregs after chemotherapy should be taken into account for personalized immunotherapy, aiming to ultimately improve clinical outcomes in patients with NSCLC.
Collapse
Affiliation(s)
- Siqi Cai
- Shandong University Cancer Center, Shandong University, Jinan, Shandong, China; Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Miaoqing Zhao
- Department of Pathology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Guanqun Yang
- Shandong University Cancer Center, Shandong University, Jinan, Shandong, China; Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Chaozhuo Li
- School of Clinical Medicine, Shandong Second Medical University, Weifang, China; Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Mengyu Hu
- Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China; Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Liying Yang
- Shandong University Cancer Center, Shandong University, Jinan, Shandong, China; Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Ligang Xing
- Shandong University Cancer Center, Shandong University, Jinan, Shandong, China; Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Xiaorong Sun
- Shandong University Cancer Center, Shandong University, Jinan, Shandong, China; Department of Nuclear Medicine, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China.
| |
Collapse
|
14
|
Rodríguez-Cruz D, Boquet-Pujadas A, López-Muñoz E, Rincón-Heredia R, Paredes-Díaz R, Flores-Fortis M, Olivo-Marin JC, Guillén N, Aguilar-Rojas A. Three-dimensional cell culture conditions promoted the Mesenchymal-Amoeboid Transition in the Triple-Negative Breast Cancer cell line MDA-MB-231. Front Cell Dev Biol 2024; 12:1435708. [PMID: 39156975 PMCID: PMC11327030 DOI: 10.3389/fcell.2024.1435708] [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: 05/20/2024] [Accepted: 07/17/2024] [Indexed: 08/20/2024] Open
Abstract
Introduction Breast cancer (BC) is the leading cause of death among women, primarily due to its potential for metastasis. As BC progresses, the extracellular matrix (ECM) produces more type-I collagen, resulting in increased stiffness. This alteration influences cellular behaviors such as migration, invasion, and metastasis. Specifically, cancer cells undergo changes in gene expression that initially promote an epithelial-to-mesenchymal transition (EMT) and subsequently, a transition from a mesenchymal to an amoeboid (MAT) migration mode. In this way, cancer cells can migrate more easily through the stiffer microenvironment. Despite their importance, understanding MATs remains challenging due to the difficulty of replicating in vitro the conditions for cell migration that are observed in vivo. Methods To address this challenge, we developed a three-dimensional (3D) growth system that replicates the different matrix properties observed during the progression of a breast tumor. We used this model to study the migration and invasion of the Triple-Negative BC (TNBC) cell line MDA-MB-231, which is particularly subject to metastasis. Results Our results indicate that denser collagen matrices present a reduction in porosity, collagen fiber size, and collagen fiber orientation, which are associated with the transition of cells to a rounder morphology with bleb-like protrusions. We quantified how this transition is associated with a more persistent migration, an enhanced invasion capacity, and a reduced secretion of matrix metalloproteinases. Discussion Our findings suggest that the proposed 3D growth conditions (especially those with high collagen concentrations) mimic key features of MATs, providing a new platform to study the physiology of migratory transitions and their role in BC progression.
Collapse
Affiliation(s)
- Daniela Rodríguez-Cruz
- Medical Research Unit in Reproductive Medicine, High Specialty Medical Unit in Gynecology and Obstetrics No. 4 “Luis Castelazo Ayala”, Mexican Social Security Institute, Mexico City, Mexico
| | - Aleix Boquet-Pujadas
- École Polytechnique Fédérale de Lausanne, Biomedical Imaging Group, Lausanne, Switzerland
- Bioimage Analysis Unit, Pasteur Institute, Paris, France
- National Center for Scientific Research, CNRS UMR3691, Paris, France
| | - Eunice López-Muñoz
- Medical Research Unit in Reproductive Medicine, High Specialty Medical Unit in Gynecology and Obstetrics No. 4 “Luis Castelazo Ayala”, Mexican Social Security Institute, Mexico City, Mexico
| | - Ruth Rincón-Heredia
- Microscopy Core Unit, Institute of Cellular Physiology, National Autonomous University of Mexico, Mexico City, Mexico
| | - Rodolfo Paredes-Díaz
- Microscopy Core Unit, Institute of Cellular Physiology, National Autonomous University of Mexico, Mexico City, Mexico
| | - Mauricio Flores-Fortis
- Cuajimalpa Unit, Engineering and Natural Science Doctoral Program, Metropolitan Autonomous University, Mexico City, Mexico
- Cuajimalpa Unit, Department of Natural Science, Metropolitan Autonomous University, Mexico City, Mexico
| | - Jean-Christophe Olivo-Marin
- Bioimage Analysis Unit, Pasteur Institute, Paris, France
- National Center for Scientific Research, CNRS UMR3691, Paris, France
| | - Nancy Guillén
- Bioimage Analysis Unit, Pasteur Institute, Paris, France
- National Center for Scientific Research, CNRS ERL9195, Paris, France
| | - Arturo Aguilar-Rojas
- Medical Research Unit in Reproductive Medicine, High Specialty Medical Unit in Gynecology and Obstetrics No. 4 “Luis Castelazo Ayala”, Mexican Social Security Institute, Mexico City, Mexico
- Bioimage Analysis Unit, Pasteur Institute, Paris, France
| |
Collapse
|
15
|
Shi X, Wang X, Yao W, Shi D, Shao X, Lu Z, Chai Y, Song J, Tang W, Wang X. Mechanism insights and therapeutic intervention of tumor metastasis: latest developments and perspectives. Signal Transduct Target Ther 2024; 9:192. [PMID: 39090094 PMCID: PMC11294630 DOI: 10.1038/s41392-024-01885-2] [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: 07/29/2023] [Revised: 05/29/2024] [Accepted: 06/10/2024] [Indexed: 08/04/2024] Open
Abstract
Metastasis remains a pivotal characteristic of cancer and is the primary contributor to cancer-associated mortality. Despite its significance, the mechanisms governing metastasis are not fully elucidated. Contemporary findings in the domain of cancer biology have shed light on the molecular aspects of this intricate process. Tumor cells undergoing invasion engage with other cellular entities and proteins en route to their destination. Insights into these engagements have enhanced our comprehension of the principles directing the movement and adaptability of metastatic cells. The tumor microenvironment plays a pivotal role in facilitating the invasion and proliferation of cancer cells by enabling tumor cells to navigate through stromal barriers. Such attributes are influenced by genetic and epigenetic changes occurring in the tumor cells and their surrounding milieu. A profound understanding of the metastatic process's biological mechanisms is indispensable for devising efficacious therapeutic strategies. This review delves into recent developments concerning metastasis-associated genes, important signaling pathways, tumor microenvironment, metabolic processes, peripheral immunity, and mechanical forces and cancer metastasis. In addition, we combine recent advances with a particular emphasis on the prospect of developing effective interventions including the most popular cancer immunotherapies and nanotechnology to combat metastasis. We have also identified the limitations of current research on tumor metastasis, encompassing drug resistance, restricted animal models, inadequate biomarkers and early detection methods, as well as heterogeneity among others. It is anticipated that this comprehensive review will significantly contribute to the advancement of cancer metastasis research.
Collapse
Affiliation(s)
- Xiaoli Shi
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences; NHC Key Laboratory of Hepatobiliary Cancers, Nanjing, Jiangsu, China
- School of Medicine, Southeast University, Nanjing, Jiangsu, China
| | - Xinyi Wang
- The First Clinical Medical College, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Wentao Yao
- Department of Urology, Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou, Jiangsu, China
| | - Dongmin Shi
- Department of Medical Oncology, Shanghai Changzheng Hospital, Shanghai, China
| | - Xihuan Shao
- The Fourth Clinical Medical College, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Zhengqing Lu
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences; NHC Key Laboratory of Hepatobiliary Cancers, Nanjing, Jiangsu, China
| | - Yue Chai
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences; NHC Key Laboratory of Hepatobiliary Cancers, Nanjing, Jiangsu, China
| | - Jinhua Song
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences; NHC Key Laboratory of Hepatobiliary Cancers, Nanjing, Jiangsu, China.
| | - Weiwei Tang
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences; NHC Key Laboratory of Hepatobiliary Cancers, Nanjing, Jiangsu, China.
| | - Xuehao Wang
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences; NHC Key Laboratory of Hepatobiliary Cancers, Nanjing, Jiangsu, China.
- School of Medicine, Southeast University, Nanjing, Jiangsu, China.
| |
Collapse
|
16
|
Vitale S, Calapà F, Colonna F, Luongo F, Biffoni M, De Maria R, Fiori ME. Advancements in 3D In Vitro Models for Colorectal Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2405084. [PMID: 38962943 PMCID: PMC11348154 DOI: 10.1002/advs.202405084] [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] [Received: 05/14/2024] [Indexed: 07/05/2024]
Abstract
The process of drug discovery and pre-clinical testing is currently inefficient, expensive, and time-consuming. Most importantly, the success rate is unsatisfactory, as only a small percentage of tested drugs are made available to oncological patients. This is largely due to the lack of reliable models that accurately predict drug efficacy and safety. Even animal models often fail to replicate human-specific pathologies and human body's complexity. These factors, along with ethical concerns regarding animal use, urge the development of suitable human-relevant, translational in vitro models.
Collapse
Affiliation(s)
- Sara Vitale
- Department of Oncology and Molecular Medicine (OMM)Istituto Superiore di SanitàViale Regina Elena 299Rome00161Italy
| | - Federica Calapà
- Dipartimento di Medicina e Chirurgia traslazionaleUniversità Cattolica del Sacro CuoreLargo F. Vito 1RomeItaly
| | - Francesca Colonna
- Department of Oncology and Molecular Medicine (OMM)Istituto Superiore di SanitàViale Regina Elena 299Rome00161Italy
| | - Francesca Luongo
- Dipartimento di Medicina e Chirurgia traslazionaleUniversità Cattolica del Sacro CuoreLargo F. Vito 1RomeItaly
| | - Mauro Biffoni
- Department of Oncology and Molecular Medicine (OMM)Istituto Superiore di SanitàViale Regina Elena 299Rome00161Italy
| | - Ruggero De Maria
- Dipartimento di Medicina e Chirurgia traslazionaleUniversità Cattolica del Sacro CuoreLargo F. Vito 1RomeItaly
- Fondazione Policlinico Universitario “A. Gemelli” – IRCCSLargo F. Vito 1RomeItaly
| | - Micol E. Fiori
- Department of Oncology and Molecular Medicine (OMM)Istituto Superiore di SanitàViale Regina Elena 299Rome00161Italy
| |
Collapse
|
17
|
Wang G, Mao X, Wang W, Wang X, Li S, Wang Z. Bioprinted research models of urological malignancy. EXPLORATION (BEIJING, CHINA) 2024; 4:20230126. [PMID: 39175884 PMCID: PMC11335473 DOI: 10.1002/exp.20230126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 01/08/2024] [Indexed: 08/24/2024]
Abstract
Urological malignancy (UM) is among the leading threats to health care worldwide. Recent years have seen much investment in fundamental UM research, including mechanistic investigation, early diagnosis, immunotherapy, and nanomedicine. However, the results are not fully satisfactory. Bioprinted research models (BRMs) with programmed spatial structures and functions can serve as powerful research tools and are likely to disrupt traditional UM research paradigms. Herein, a comprehensive review of BRMs of UM is presented. It begins with a brief introduction and comparison of existing UM research models, emphasizing the advantages of BRMs, such as modeling real tissues and organs. Six kinds of mainstream bioprinting techniques used to fabricate such BRMs are summarized with examples. Thereafter, research advances in the applications of UM BRMs, such as culturing tumor spheroids and organoids, modeling cancer metastasis, mimicking the tumor microenvironment, constructing organ chips for drug screening, and isolating circulating tumor cells, are comprehensively discussed. At the end of this review, current challenges and future development directions of BRMs and UM are highlighted from the perspective of interdisciplinary science.
Collapse
Affiliation(s)
- Guanyi Wang
- Department of UrologyCancer Precision Diagnosis and Treatment and Translational Medicine Hubei Engineering Research CenterZhongnan Hospital of Wuhan UniversityWuhanChina
- Department of Biomedical Engineering and Hubei Province Key Laboratory of Allergy and Immune Related DiseaseTaiKang Medical School (School of Basic Medical Sciences)Wuhan UniversityWuhanChina
| | - Xiongmin Mao
- Department of UrologyCancer Precision Diagnosis and Treatment and Translational Medicine Hubei Engineering Research CenterZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Wang Wang
- Department of UrologyCancer Precision Diagnosis and Treatment and Translational Medicine Hubei Engineering Research CenterZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Xiaolong Wang
- Lewis Katz School of MedicineTemple UniversityPhiladelphiaPennsylvaniaUSA
| | - Sheng Li
- Department of UrologyCancer Precision Diagnosis and Treatment and Translational Medicine Hubei Engineering Research CenterZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Zijian Wang
- Department of UrologyCancer Precision Diagnosis and Treatment and Translational Medicine Hubei Engineering Research CenterZhongnan Hospital of Wuhan UniversityWuhanChina
- Department of Biomedical Engineering and Hubei Province Key Laboratory of Allergy and Immune Related DiseaseTaiKang Medical School (School of Basic Medical Sciences)Wuhan UniversityWuhanChina
| |
Collapse
|
18
|
Peng AY, Lee BE. Microphysiological Systems for Cancer Immunotherapy Research and Development. Adv Biol (Weinh) 2024; 8:e2300077. [PMID: 37409385 PMCID: PMC10770294 DOI: 10.1002/adbi.202300077] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 06/13/2023] [Indexed: 07/07/2023]
Abstract
Cancer immunotherapy focuses on the use of patients' adaptive immune systems to combat cancer. In the past decade, FDA has approved many immunotherapy products for cancer patients who suffer from primary tumors, tumor relapse, and metastases. However, these immunotherapies still show resistance in many patients and often lead to inconsistent responses in patients due to variations in tumor genetic mutations and tumor immune microenvironment. Microfluidics-based organ-on-a-chip technologies or microphysiological systems have opened new ways that can provide relatively fast screening for personalized immunotherapy and help researchers and clinicians understand tumor-immune interactions in a patient-specific manner. They also have the potential to overcome the limitations of traditional drug screening and testing, given the models provide a more realistic 3D microenvironment with better controllability, reproducibility, and physiological relevance. This review focuses on the cutting-edge microphysiological organ-on-a-chip devices developed in recent years for studying cancer immunity and testing cancer immunotherapeutic agents, as well as some of the largest challenges of translating this technology to clinical applications in immunotherapy and personalized medicine.
Collapse
Affiliation(s)
- A. Yansong Peng
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | - B. Esak Lee
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| |
Collapse
|
19
|
Mao Y, Wickström SA. Mechanical state transitions in the regulation of tissue form and function. Nat Rev Mol Cell Biol 2024; 25:654-670. [PMID: 38600372 DOI: 10.1038/s41580-024-00719-x] [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] [Accepted: 02/26/2024] [Indexed: 04/12/2024]
Abstract
From embryonic development, postnatal growth and adult homeostasis to reparative and disease states, cells and tissues undergo constant changes in genome activity, cell fate, proliferation, movement, metabolism and growth. Importantly, these biological state transitions are coupled to changes in the mechanical and material properties of cells and tissues, termed mechanical state transitions. These mechanical states share features with physical states of matter, liquids and solids. Tissues can switch between mechanical states by changing behavioural dynamics or connectivity between cells. Conversely, these changes in tissue mechanical properties are known to control cell and tissue function, most importantly the ability of cells to move or tissues to deform. Thus, tissue mechanical state transitions are implicated in transmitting information across biological length and time scales, especially during processes of early development, wound healing and diseases such as cancer. This Review will focus on the biological basis of tissue-scale mechanical state transitions, how they emerge from molecular and cellular interactions, and their roles in organismal development, homeostasis, regeneration and disease.
Collapse
Affiliation(s)
- Yanlan Mao
- Laboratory for Molecular Cell Biology, University College London, London, UK.
- Institute for the Physics of Living Systems, University College London, London, UK.
| | - Sara A Wickström
- Department of Cell and Tissue Dynamics, Max Planck Institute for Molecular Biomedicine, Münster, Germany.
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
- Helsinki Institute of Life Science, Biomedicum Helsinki, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
20
|
Zhang X, Zhang X, Yong T, Gan L, Yang X. Boosting antitumor efficacy of nanoparticles by modulating tumor mechanical microenvironment. EBioMedicine 2024; 105:105200. [PMID: 38876044 PMCID: PMC11225208 DOI: 10.1016/j.ebiom.2024.105200] [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: 01/16/2024] [Revised: 05/30/2024] [Accepted: 05/30/2024] [Indexed: 06/16/2024] Open
Abstract
Nanoparticles have shown great potential for tumor targeting delivery via enhanced permeability and retention effect. However, the tumor mechanical microenvironment, characterized by dense extracellular matrix (ECM), high tumor stiffness and solid stress, leads to only 0.7% of administered dose accumulating in solid tumors and even fewer (∼0.0014%) reaching tumor cells, limiting the therapeutic efficacy of nanoparticles. Furthermore, the tumor mechanical microenvironment can regulate tumor cell stemness, promote tumor invasion, metastasis and reduce treatment efficacy. In this review, methods detecting the mechanical are introduced. Strategies for modulating the mechanical microenvironment including elimination of dense ECM by physical, chemical and biological methods, disruption of ECM formation, depletion or inhibition of cancer-associated fibroblasts, are then summarized. Finally, prospects and challenges for further clinical applications of mechano-modulating strategies to enhance the therapeutic efficacy of nanomedicines are discussed. This review may provide guidance for the rational design and application of nanoparticles in clinical settings.
Collapse
Affiliation(s)
- Xiaoqiong Zhang
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Xiaojuan Zhang
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Tuying Yong
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China; Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China; Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, Huazhong University of Science and Technology, Wuhan, 430074, China.
| | - Lu Gan
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China; Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China; Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, Huazhong University of Science and Technology, Wuhan, 430074, China.
| | - Xiangliang Yang
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China; Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China; Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, Huazhong University of Science and Technology, Wuhan, 430074, China.
| |
Collapse
|
21
|
Ruocco MR, Gisonna A, Acampora V, D’Agostino A, Carrese B, Santoro J, Venuta A, Nasso R, Rocco N, Russo D, Cavaliere A, Altobelli GG, Masone S, Avagliano A, Arcucci A, Fiume G. Guardians and Mediators of Metastasis: Exploring T Lymphocytes, Myeloid-Derived Suppressor Cells, and Tumor-Associated Macrophages in the Breast Cancer Microenvironment. Int J Mol Sci 2024; 25:6224. [PMID: 38892411 PMCID: PMC11172575 DOI: 10.3390/ijms25116224] [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: 02/20/2024] [Revised: 05/30/2024] [Accepted: 06/01/2024] [Indexed: 06/21/2024] Open
Abstract
Breast cancers (BCs) are solid tumors composed of heterogeneous tissues consisting of cancer cells and an ever-changing tumor microenvironment (TME). The TME includes, among other non-cancer cell types, immune cells influencing the immune context of cancer tissues. In particular, the cross talk of immune cells and their interactions with cancer cells dramatically influence BC dissemination, immunoediting, and the outcomes of cancer therapies. Tumor-infiltrating lymphocytes (TILs), tumor-associated macrophages (TAMs), and myeloid-derived suppressor cells (MDSCs) represent prominent immune cell populations of breast TMEs, and they have important roles in cancer immunoescape and dissemination. Therefore, in this article we review the features of TILs, TAMs, and MDSCs in BCs. Moreover, we highlight the mechanisms by which these immune cells remodel the immune TME and lead to breast cancer metastasis.
Collapse
Affiliation(s)
- Maria Rosaria Ruocco
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy; (M.R.R.); (A.G.)
| | - Armando Gisonna
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy; (M.R.R.); (A.G.)
| | - Vittoria Acampora
- Department of Public Health, University of Naples Federico II, 80131 Naples, Italy; (V.A.); (A.V.); (A.A.)
| | - Anna D’Agostino
- IRCCS SYNLAB SDN, Via Emanuele Gianturco 113, 80143 Naples, Italy; (A.D.); (B.C.); (J.S.)
| | - Barbara Carrese
- IRCCS SYNLAB SDN, Via Emanuele Gianturco 113, 80143 Naples, Italy; (A.D.); (B.C.); (J.S.)
| | - Jessie Santoro
- IRCCS SYNLAB SDN, Via Emanuele Gianturco 113, 80143 Naples, Italy; (A.D.); (B.C.); (J.S.)
| | - Alessandro Venuta
- Department of Public Health, University of Naples Federico II, 80131 Naples, Italy; (V.A.); (A.V.); (A.A.)
| | - Rosarita Nasso
- Department of Movement Sciences and Wellness, University of Naples “Parthenope”, 80133 Naples, Italy;
| | - Nicola Rocco
- Department of Advanced Biomedical Science, University of Naples Federico II, 80131 Naples, Italy; (N.R.); (D.R.); (G.G.A.)
| | - Daniela Russo
- Department of Advanced Biomedical Science, University of Naples Federico II, 80131 Naples, Italy; (N.R.); (D.R.); (G.G.A.)
| | | | - Giovanna Giuseppina Altobelli
- Department of Advanced Biomedical Science, University of Naples Federico II, 80131 Naples, Italy; (N.R.); (D.R.); (G.G.A.)
| | - Stefania Masone
- Department of Clinical Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy;
| | - Angelica Avagliano
- Department of Public Health, University of Naples Federico II, 80131 Naples, Italy; (V.A.); (A.V.); (A.A.)
| | - Alessandro Arcucci
- Department of Public Health, University of Naples Federico II, 80131 Naples, Italy; (V.A.); (A.V.); (A.A.)
| | - Giuseppe Fiume
- Department of Experimental and Clinical Medicine, University of Catanzaro “Magna Graecia”, 88100 Catanzaro, Italy;
| |
Collapse
|
22
|
Haake SM, Rios BL, Pozzi A, Zent R. Integrating integrins with the hallmarks of cancer. Matrix Biol 2024; 130:20-35. [PMID: 38677444 DOI: 10.1016/j.matbio.2024.04.003] [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: 01/09/2024] [Revised: 04/02/2024] [Accepted: 04/23/2024] [Indexed: 04/29/2024]
Abstract
Epithelial cells adhere to a specialized extracellular matrix called the basement membrane which allows them to polarize and form epithelial tissues. The extracellular matrix provides essential physical scaffolding and biochemical and biophysical cues required for tissue morphogenesis, differentiation, function, and homeostasis. Epithelial cell adhesion to the extracellular matrix (i.e., basement membrane) plays a critical role in organizing epithelial tissues, separating the epithelial cells from the stroma. Epithelial cell detachment from the basement membrane classically results in death, though detachment or invasion through the basement membrane represents a critical step in carcinogenesis. Epithelial cells bind to the extracellular matrix via specialized matrix receptors, including integrins. Integrins are transmembrane receptors that form a mechanical linkage between the extracellular matrix and the intracellular cytoskeleton and are required for anchorage-dependent cellular functions such as proliferation, migration, and invasion. The role of integrins in the development, growth, and dissemination of multiple types of carcinomas has been investigated by numerous methodologies, which has led to great complexity. To organize this vast array of information, we have utilized the "Hallmarks of Cancer" from Hanahan and Weinberg as a convenient framework to discuss the role of integrins in the pathogenesis of cancers. This review explores this biology and how its complexity has impacted the development of integrin-targeted anti-cancer therapeutics.
Collapse
Affiliation(s)
- Scott M Haake
- Division of Hematology, Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Veterans Affairs, Nashville, TN, USA; Vanderbilt-Ingram Cancer Center, Nashville, TN, USA; Cancer Biology Program, Vanderbilt University, Nashville, TN, USA.
| | - Brenda L Rios
- Vanderbilt-Ingram Cancer Center, Nashville, TN, USA; Cancer Biology Program, Vanderbilt University, Nashville, TN, USA
| | - Ambra Pozzi
- Department of Veterans Affairs, Nashville, TN, USA; Vanderbilt-Ingram Cancer Center, Nashville, TN, USA; Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Roy Zent
- Department of Veterans Affairs, Nashville, TN, USA; Vanderbilt-Ingram Cancer Center, Nashville, TN, USA; Cancer Biology Program, Vanderbilt University, Nashville, TN, USA; Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA
| |
Collapse
|
23
|
Wu Y, Chen R, Ni S, Hu K. Biomimetic "nano-spears" for CAFs-targeting: splintered three "shields" with enhanced cisplatin anti-TNBC efficiency. J Control Release 2024; 370:556-569. [PMID: 38697316 DOI: 10.1016/j.jconrel.2024.04.053] [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: 01/26/2024] [Revised: 04/12/2024] [Accepted: 04/29/2024] [Indexed: 05/04/2024]
Abstract
The treatment dilemma of triple-negative breast cancer (TNBC) revolves around drug resistance and metastasis. Cancer-associated fibroblasts (CAFs) contribute to cisplatin (Cis) resistance and further metastasis in TNBC, making TNBC a difficult-to-treat disease. The dense stromal barrier which restricts drug delivery, invasive phenotype of tumor cells, and immunosuppressive tumor microenvironment (TME) induced by CAFs serve as three "shields" for TNBC against Cis therapy. Here, we designed a silybin-loaded biomimetic nanoparticle coated with anisamide-modified red blood cell membrane (ARm@SNP) as a "nanospear" for CAFs-targeting, which could shatter the "shields" and significantly exhibit inhibitory effect on 4T1 cells in combination with Cis both in vitro and in vivo. The ARm@SNP/Cis elicited 4T1 tumor growth arrest and destroyed three "shields" as follows: disintegrating the stromal barrier by inhibiting blood vessels growth and the expression of fibronectin; decreasing 4T1 cell invasion and metastasis by affecting the TGF-β/Twist/EMT pathway which impeded EMT activation; reversing the immunosuppressive microenvironment by increasing the activity and infiltration of immunocompetent cells. Based on CAFs-targeting, ARm@SNP reversed the resistance of Cis, remodeled the TME and inhibited invasion and metastasis while significantly improving the therapeutic effect of Cis on 4T1 tumor-bearing mice, providing a promising approach for treating intractable TNBC.
Collapse
Affiliation(s)
- Yufan Wu
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Rujing Chen
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Department of Pharmacy, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, China
| | - Shuting Ni
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Kaili Hu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
24
|
Wilkie IC. Basement Membranes, Brittlestar Tendons, and Their Mechanical Adaptability. BIOLOGY 2024; 13:375. [PMID: 38927255 PMCID: PMC11200632 DOI: 10.3390/biology13060375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 05/20/2024] [Accepted: 05/21/2024] [Indexed: 06/28/2024]
Abstract
Basement membranes (BMs) are thin layers of extracellular matrix that separate epithelia, endothelia, muscle cells, and nerve cells from adjacent interstitial connective tissue. BMs are ubiquitous in almost all multicellular animals, and their composition is highly conserved across the Metazoa. There is increasing interest in the mechanical functioning of BMs, including the involvement of altered BM stiffness in development and pathology, particularly cancer metastasis, which can be facilitated by BM destabilization. Such BM weakening has been assumed to occur primarily through enzymatic degradation by matrix metalloproteinases. However, emerging evidence indicates that non-enzymatic mechanisms may also contribute. In brittlestars (Echinodermata, Ophiuroidea), the tendons linking the musculature to the endoskeleton consist of extensions of muscle cell BMs. During the process of brittlestar autotomy, in which arms are detached for the purpose of self-defense, muscles break away from the endoskeleton as a consequence of the rapid destabilization and rupture of their BM-derived tendons. This contribution provides a broad overview of current knowledge of the structural organization and biomechanics of non-echinoderm BMs, compares this with the equivalent information on brittlestar tendons, and discusses the possible relationship between the weakening phenomena exhibited by BMs and brittlestar tendons, and the potential translational value of the latter as a model system of BM destabilization.
Collapse
Affiliation(s)
- Iain C Wilkie
- School of Biodiversity, One Health and Veterinary Medicine, University of Glasgow, Glasgow G12 8QQ, UK
| |
Collapse
|
25
|
Hartmann B, Fleischhauer L, Nicolau M, Jensen THL, Taran FA, Clausen-Schaumann H, Reuten R. Profiling native pulmonary basement membrane stiffness using atomic force microscopy. Nat Protoc 2024; 19:1498-1528. [PMID: 38429517 DOI: 10.1038/s41596-024-00955-7] [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: 02/20/2023] [Accepted: 11/27/2023] [Indexed: 03/03/2024]
Abstract
Mammalian cells sense and react to the mechanics of their immediate microenvironment. Therefore, the characterization of the biomechanical properties of tissues with high spatial resolution provides valuable insights into a broad variety of developmental, homeostatic and pathological processes within living organisms. The biomechanical properties of the basement membrane (BM), an extracellular matrix (ECM) substructure measuring only ∼100-400 nm across, are, among other things, pivotal to tumor progression and metastasis formation. Although the precise assignment of the Young's modulus E of such a thin ECM substructure especially in between two cell layers is still challenging, biomechanical data of the BM can provide information of eminent diagnostic potential. Here we present a detailed protocol to quantify the elastic modulus of the BM in murine and human lung tissue, which is one of the major organs prone to metastasis. This protocol describes a streamlined workflow to determine the Young's modulus E of the BM between the endothelial and epithelial cell layers shaping the alveolar wall in lung tissues using atomic force microscopy (AFM). Our step-by-step protocol provides instructions for murine and human lung tissue extraction, inflation of these tissues with cryogenic cutting medium, freezing and cryosectioning of the tissue samples, and AFM force-map recording. In addition, it guides the reader through a semi-automatic data analysis procedure to identify the pulmonary BM and extract its Young's modulus E using an in-house tailored user-friendly AFM data analysis software, the Center for Applied Tissue Engineering and Regenerative Medicine processing toolbox, which enables automatic loading of the recorded force maps, conversion of the force versus piezo-extension curves to force versus indentation curves, calculation of Young's moduli and generation of Young's modulus maps, where the pulmonary BM can be identified using a semi-automatic spatial filtering tool. The entire protocol takes 1-2 d.
Collapse
Affiliation(s)
- Bastian Hartmann
- Munich University of Applied Sciences, Center for Applied Tissue Engineering and Regenerative Medicine - CANTER, Munich, Germany
- Center for Nanoscience, Munich, Germany
| | - Lutz Fleischhauer
- Munich University of Applied Sciences, Center for Applied Tissue Engineering and Regenerative Medicine - CANTER, Munich, Germany
- Center for Nanoscience, Munich, Germany
| | - Monica Nicolau
- Experimental and Clinical Pharmacology and Toxicology, Medical Faculty, University of Freiburg, Freiburg, Germany
- Department of Obstetrics and Gynecology, Medical Center, University of Freiburg, Freiburg, Germany
| | - Thomas Hartvig Lindkær Jensen
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Pathology, Rigshospitalet, Copenhagen, Denmark
| | - Florin-Andrei Taran
- Department of Obstetrics and Gynecology, Medical Center, University of Freiburg, Freiburg, Germany
| | - Hauke Clausen-Schaumann
- Munich University of Applied Sciences, Center for Applied Tissue Engineering and Regenerative Medicine - CANTER, Munich, Germany.
- Center for Nanoscience, Munich, Germany.
| | - Raphael Reuten
- Experimental and Clinical Pharmacology and Toxicology, Medical Faculty, University of Freiburg, Freiburg, Germany.
- Department of Obstetrics and Gynecology, Medical Center, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
26
|
Chang J, Saraswathibhatla A, Song Z, Varma S, Sanchez C, Alyafei NHK, Indana D, Slyman R, Srivastava S, Liu K, Bassik MC, Marinkovich MP, Hodgson L, Shenoy V, West RB, Chaudhuri O. Cell volume expansion and local contractility drive collective invasion of the basement membrane in breast cancer. NATURE MATERIALS 2024; 23:711-722. [PMID: 37957268 PMCID: PMC11185842 DOI: 10.1038/s41563-023-01716-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Accepted: 10/05/2023] [Indexed: 11/15/2023]
Abstract
Breast cancer becomes invasive when carcinoma cells invade through the basement membrane (BM)-a nanoporous layer of matrix that physically separates the primary tumour from the stroma. Single cells can invade through nanoporous three-dimensional matrices due to protease-mediated degradation or force-mediated widening of pores via invadopodial protrusions. However, how multiple cells collectively invade through the physiological BM, as they do during breast cancer progression, remains unclear. Here we developed a three-dimensional in vitro model of collective invasion of the BM during breast cancer. We show that cells utilize both proteases and forces-but not invadopodia-to breach the BM. Forces are generated from a combination of global cell volume expansion, which stretches the BM, and local contractile forces that act in the plane of the BM to breach it, allowing invasion. These results uncover a mechanism by which cells collectively interact to overcome a critical barrier to metastasis.
Collapse
Affiliation(s)
- Julie Chang
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | | | - Zhaoqiang Song
- Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, PA, USA
- Department of Materials Science and Engineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Sushama Varma
- Department of Pathology, Stanford University Medical Center, Palo Alto, CA, USA
| | - Colline Sanchez
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | | | - Dhiraj Indana
- Department of Mechanical Engineering, Stanford University, Stanford, CA, USA
| | - Raleigh Slyman
- Department of Mechanical Engineering, Stanford University, Stanford, CA, USA
| | - Sucheta Srivastava
- Department of Pathology, Stanford University Medical Center, Palo Alto, CA, USA
| | - Katherine Liu
- Department of Genetics, Stanford University Medical Center, Palo Alto, CA, USA
| | - Michael C Bassik
- Department of Genetics, Stanford University Medical Center, Palo Alto, CA, USA
| | - M Peter Marinkovich
- Department of Dermatology, Stanford University School of Medicine, Stanford, CA, USA
- Dermatology Service, VA Medical Center, Palo Alto, CA, USA
| | - Louis Hodgson
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Vivek Shenoy
- Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, PA, USA
- Department of Materials Science and Engineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Robert B West
- Department of Pathology, Stanford University Medical Center, Palo Alto, CA, USA
| | - Ovijit Chaudhuri
- Department of Mechanical Engineering, Stanford University, Stanford, CA, USA.
- Chemistry, Engineering, and Medicine for Human Health (ChEM-H), Stanford University, Stanford, CA, USA.
| |
Collapse
|
27
|
Yamaguchi H, Miyazaki M. Heterocellular Adhesion in Cancer Invasion and Metastasis: Interactions between Cancer Cells and Cancer-Associated Fibroblasts. Cancers (Basel) 2024; 16:1636. [PMID: 38730588 PMCID: PMC11082996 DOI: 10.3390/cancers16091636] [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: 03/06/2024] [Revised: 04/19/2024] [Accepted: 04/22/2024] [Indexed: 05/13/2024] Open
Abstract
Cancer invasion is a requisite for the most malignant progression of cancer, that is, metastasis. The mechanisms of cancer invasion were originally studied using in vitro cell culture systems, in which cancer cells were cultured using artificial extracellular matrices (ECMs). However, conventional culture systems do not precisely recapitulate in vivo cancer invasion because the phenotypes of cancer cells in tumor tissues are strongly affected by the tumor microenvironment (TME). Cancer-associated fibroblasts (CAFs) are the most abundant cell type in the TME and accelerate cancer progression through invasion, metastasis, therapy resistance, and immune suppression. Thus, the reciprocal interactions between CAFs and cancer cells have been extensively studied, leading to the identification of factors that mediate cellular interactions, such as growth factors, cytokines, and extracellular vesicles. In addition, the importance of direct heterocellular adhesion between cancer cells and CAFs in cancer progression has recently been elucidated. In particular, CAFs are directly associated with cancer cells, allowing them to invade the ECM and metastasize to distant organs. In this review, we summarize the recent progress in understanding the molecular and cellular mechanisms of the direct heterocellular interaction in CAF-led cancer invasion and metastasis, with an emphasis on gastric cancer.
Collapse
Affiliation(s)
- Hideki Yamaguchi
- Department of Cancer Cell Research, Sasaki Institute, Sasaki Foundation, 2-2 Kandasurugadai, Chiyoda-ku, Tokyo 101-0062, Japan;
| | | |
Collapse
|
28
|
Huang Q, Ge Y, He Y, Wu J, Tong Y, Shang H, Liu X, Ba X, Xia D, Peng E, Chen Z, Tang K. The Application of Nanoparticles Targeting Cancer-Associated Fibroblasts. Int J Nanomedicine 2024; 19:3333-3365. [PMID: 38617796 PMCID: PMC11012801 DOI: 10.2147/ijn.s447350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Accepted: 03/23/2024] [Indexed: 04/16/2024] Open
Abstract
Cancer-associated fibroblasts (CAF) are the most abundant stromal cells in the tumor microenvironment (TME), especially in solid tumors. It has been confirmed that it can not only interact with tumor cells to promote cancer progression and metastasis, but also affect the infiltration and function of immune cells to induce chemotherapy and immunotherapy resistance. So, targeting CAF has been considered an important method in cancer treatment. The rapid development of nanotechnology provides a good perspective to improve the efficiency of targeting CAF. At present, more and more researches have focused on the application of nanoparticles (NPs) in targeting CAF. These studies explored the effects of different types of NPs on CAF and the multifunctional nanomedicines that can eliminate CAF are able to enhance the EPR effect which facilitate the anti-tumor effect of themselves. There also exist amounts of studies focusing on using NPs to inhibit the activation and function of CAF to improve the therapeutic efficacy. The application of NPs targeting CAF needs to be based on an understanding of CAF biology. Therefore, in this review, we first summarized the latest progress of CAF biology, then discussed the types of CAF-targeting NPs and the main strategies in the current. The aim is to elucidate the application of NPs in targeting CAF and provide new insights for engineering nanomedicine to enhance immune response in cancer treatment.
Collapse
Affiliation(s)
- Qiu Huang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, People’s Republic of China
| | - Yue Ge
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, People’s Republic of China
| | - Yu He
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, People’s Republic of China
| | - Jian Wu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, People’s Republic of China
| | - Yonghua Tong
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, People’s Republic of China
| | - Haojie Shang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, People’s Republic of China
| | - Xiao Liu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, People’s Republic of China
| | - Xiaozhuo Ba
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, People’s Republic of China
| | - Ding Xia
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, People’s Republic of China
| | - Ejun Peng
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, People’s Republic of China
| | - Zhiqiang Chen
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, People’s Republic of China
| | - Kun Tang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, People’s Republic of China
| |
Collapse
|
29
|
Jouybar M, de Winde CM, Wolf K, Friedl P, Mebius RE, den Toonder JMJ. Cancer-on-chip models for metastasis: importance of the tumor microenvironment. Trends Biotechnol 2024; 42:431-448. [PMID: 37914546 DOI: 10.1016/j.tibtech.2023.10.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 10/05/2023] [Accepted: 10/06/2023] [Indexed: 11/03/2023]
Abstract
Cancer-on-chip (CoC) models, based on microfluidic chips harboring chambers for 3D tumor-cell culture, enable us to create a controlled tumor microenvironment (TME). CoC models are therefore increasingly used to systematically study effects of the TME on the various steps in cancer metastasis. Moreover, CoC models have great potential for developing novel cancer therapies and for predicting patient-specific response to cancer treatments. We review recent developments in CoC models, focusing on three main TME components: (i) the anisotropic extracellular matrix (ECM) architectures, (ii) the vasculature, and (iii) the immune system. We aim to provide guidance to biologists to choose the best CoC approach for addressing questions about the role of the TME in metastasis, and to inspire engineers to develop novel CoC technologies.
Collapse
Affiliation(s)
- Mohammad Jouybar
- Microsystems, Eindhoven University of Technology, Eindhoven, The Netherlands; Institute for Complex Molecular Systems, Eindhoven, The Netherlands
| | - Charlotte M de Winde
- Amsterdam UMC location Vrije Universiteit Amsterdam, Molecular Cell Biology & Immunology, Amsterdam, The Netherlands; Amsterdam Institute for Infection and Immunity, Cancer Immunology, Amsterdam, The Netherlands; Cancer Center Amsterdam, Cancer Biology & Immunology, Amsterdam, The Netherlands
| | - Katarina Wolf
- Department of Medical BioSciences, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Peter Friedl
- Department of Medical BioSciences, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands; Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Cancer Genomics Center, Utrecht, The Netherlands
| | - Reina E Mebius
- Amsterdam UMC location Vrije Universiteit Amsterdam, Molecular Cell Biology & Immunology, Amsterdam, The Netherlands; Amsterdam Institute for Infection and Immunity, Cancer Immunology, Amsterdam, The Netherlands; Cancer Center Amsterdam, Cancer Biology & Immunology, Amsterdam, The Netherlands; Amsterdam Institute for Infection and Immunity, Inflammatory diseases, Amsterdam, The Netherlands
| | - Jaap M J den Toonder
- Microsystems, Eindhoven University of Technology, Eindhoven, The Netherlands; Institute for Complex Molecular Systems, Eindhoven, The Netherlands.
| |
Collapse
|
30
|
Prieto-Díaz R, Fojo-Carballo H, Majellaro M, Tandarić T, Azuaje J, Brea J, Loza MI, Barbazán J, Salort G, Chotalia M, Rodríguez-Pampín I, Mallo-Abreu A, Rita Paleo M, García-Mera X, Ciruela F, Gutiérrez-de-Terán H, Sotelo E. Exploring Biginelli-based scaffolds as A 2B adenosine receptor antagonists: Unveiling novel structure-activity relationship trends, lead compounds, and potent colorectal anticancer agents. Biomed Pharmacother 2024; 173:116345. [PMID: 38442670 DOI: 10.1016/j.biopha.2024.116345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 02/19/2024] [Accepted: 02/23/2024] [Indexed: 03/07/2024] Open
Abstract
Antagonists of the A2B adenosine receptor have recently emerged as targeted anticancer agents and immune checkpoint inhibitors within the realm of cancer immunotherapy. This study presents a comprehensive evaluation of novel Biginelli-assembled pyrimidine chemotypes, including mono-, bi-, and tricyclic derivatives, as A2BAR antagonists. We conducted a comprehensive examination of the adenosinergic profile (both binding and functional) of a large compound library consisting of 168 compounds. This approach unveiled original lead compounds and enabled the identification of novel structure-activity relationship (SAR) trends, which were supported by extensive computational studies, including quantum mechanical calculations and free energy perturbation (FEP) analysis. In total, 25 molecules showed attractive affinity (Ki < 100 nM) and outstanding selectivity for A2BAR. From these, five molecules corresponding to the new benzothiazole scaffold were below the Ki < 10 nM threshold, in addition to a novel dual A2A/A2B antagonist. The most potent compounds, and the dual antagonist, showed enantiospecific recognition in the A2BAR. Two A2BAR selective antagonists and the dual A2AAR/A2BAR antagonist reported in this study were assessed for their impact on colorectal cancer cell lines. The results revealed a significant and dose-dependent reduction in cell proliferation. Notably, the A2BAR antagonists exhibited remarkable specificity, as they did not impede the proliferation of non-tumoral cell lines. These findings support the efficacy and potential that A2BAR antagonists as valuable candidates for cancer therapy, but also that they can effectively complement strategies involving A2AAR antagonism in the context of immune checkpoint inhibition.
Collapse
Affiliation(s)
- Rubén Prieto-Díaz
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Universidade de Santiago de Compostela, Santiago de Compostela 15782, Spain; Departamento de Química Orgánica, Facultade de Farmacia, Universidade de Santiago de Compostela, Santiago de Compostela 15782, Spain
| | - Hugo Fojo-Carballo
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Universidade de Santiago de Compostela, Santiago de Compostela 15782, Spain; Departamento de Química Orgánica, Facultade de Farmacia, Universidade de Santiago de Compostela, Santiago de Compostela 15782, Spain
| | - Maria Majellaro
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Universidade de Santiago de Compostela, Santiago de Compostela 15782, Spain; Departamento de Química Orgánica, Facultade de Farmacia, Universidade de Santiago de Compostela, Santiago de Compostela 15782, Spain.
| | - Tana Tandarić
- Science for Life Laboratory, Department of Cell and Molecular Biology, Uppsala University, Biomedical Centre, Uppsala 75124, Sweden
| | - Jhonny Azuaje
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Universidade de Santiago de Compostela, Santiago de Compostela 15782, Spain; Departamento de Química Orgánica, Facultade de Farmacia, Universidade de Santiago de Compostela, Santiago de Compostela 15782, Spain
| | - José Brea
- Centro Singular de Investigación en Medicina Molecular y Enfermedades Crónicas (CiMUS), Universidade de Santiago de Compostela, Santiago de Compostela 15782, Spain; Departamento de Farmacoloxía, Farmacia e Tecnoloxía Farmacéutica, Facultade de Farmacia, Universidade de Santiago de Compostela, Santiago de Compostela 15782, Spain.
| | - María I Loza
- Centro Singular de Investigación en Medicina Molecular y Enfermedades Crónicas (CiMUS), Universidade de Santiago de Compostela, Santiago de Compostela 15782, Spain; Departamento de Farmacoloxía, Farmacia e Tecnoloxía Farmacéutica, Facultade de Farmacia, Universidade de Santiago de Compostela, Santiago de Compostela 15782, Spain
| | - Jorge Barbazán
- Grupo de Oncología Médica Traslacional (ONCOMET), Instituto de Investigación Sanitaria, Santiago de Compostela (IDIS), Hospital Universitario de Santiago de Compostela (SERGAS), Santiago de Compostela 15706, Spain.
| | - Glòria Salort
- Unidad de Farmacología, Departamento de Patología y Terapéutica Experimental, Facultad de Medicina y Ciencias de la Salud, Instituto de Neurociencia, Universitat de Barcelona, L'Hospitalet de Llobregat 08907, Spain; Neuropharmacology and Pain Group, Neuroscience Program, Institut d'Investigació Biomèdica de Bellvitge, IDIBELL, L'Hospitalet de Llobregat 08907, Spain; Laboratorio de Neurofarmacología, Institut Universitari d'Investigació en Ciències de la Salut (IUNICS), Institut d'investigació Sanitària Illes Balears (IdISBa), University of the Balearic Islands, Palma de Mallorca 07122, Spain
| | - Meera Chotalia
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Universidade de Santiago de Compostela, Santiago de Compostela 15782, Spain; Departamento de Química Orgánica, Facultade de Farmacia, Universidade de Santiago de Compostela, Santiago de Compostela 15782, Spain
| | - Iván Rodríguez-Pampín
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Universidade de Santiago de Compostela, Santiago de Compostela 15782, Spain; Departamento de Química Orgánica, Facultade de Farmacia, Universidade de Santiago de Compostela, Santiago de Compostela 15782, Spain
| | - Ana Mallo-Abreu
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Universidade de Santiago de Compostela, Santiago de Compostela 15782, Spain; Departamento de Química Orgánica, Facultade de Farmacia, Universidade de Santiago de Compostela, Santiago de Compostela 15782, Spain
| | - M Rita Paleo
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Universidade de Santiago de Compostela, Santiago de Compostela 15782, Spain; Departamento de Química Orgánica, Facultade de Farmacia, Universidade de Santiago de Compostela, Santiago de Compostela 15782, Spain
| | - Xerardo García-Mera
- Departamento de Química Orgánica, Facultade de Farmacia, Universidade de Santiago de Compostela, Santiago de Compostela 15782, Spain
| | - Francisco Ciruela
- Unidad de Farmacología, Departamento de Patología y Terapéutica Experimental, Facultad de Medicina y Ciencias de la Salud, Instituto de Neurociencia, Universitat de Barcelona, L'Hospitalet de Llobregat 08907, Spain; Neuropharmacology and Pain Group, Neuroscience Program, Institut d'Investigació Biomèdica de Bellvitge, IDIBELL, L'Hospitalet de Llobregat 08907, Spain
| | - Hugo Gutiérrez-de-Terán
- Science for Life Laboratory, Department of Cell and Molecular Biology, Uppsala University, Biomedical Centre, Uppsala 75124, Sweden.
| | - Eddy Sotelo
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Universidade de Santiago de Compostela, Santiago de Compostela 15782, Spain; Departamento de Química Orgánica, Facultade de Farmacia, Universidade de Santiago de Compostela, Santiago de Compostela 15782, Spain.
| |
Collapse
|
31
|
Ozmen E, Demir TD, Ozcan G. Cancer-associated fibroblasts: protagonists of the tumor microenvironment in gastric cancer. Front Mol Biosci 2024; 11:1340124. [PMID: 38562556 PMCID: PMC10982390 DOI: 10.3389/fmolb.2024.1340124] [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: 11/17/2023] [Accepted: 01/31/2024] [Indexed: 04/04/2024] Open
Abstract
Enhanced knowledge of the interaction of cancer cells with their environment elucidated the critical role of tumor microenvironment in tumor progression and chemoresistance. Cancer-associated fibroblasts act as the protagonists of the tumor microenvironment, fostering the metastasis, stemness, and chemoresistance of cancer cells and attenuating the anti-cancer immune responses. Gastric cancer is one of the most aggressive cancers in the clinic, refractory to anti-cancer therapies. Growing evidence indicates that cancer-associated fibroblasts are the most prominent risk factors for a poor tumor immune microenvironment and dismal prognosis in gastric cancer. Therefore, targeting cancer-associated fibroblasts may be central to surpassing resistance to conventional chemotherapeutics, molecular-targeted agents, and immunotherapies, improving survival in gastric cancer. However, the heterogeneity in cancer-associated fibroblasts may complicate the development of cancer-associated fibroblast targeting approaches. Although single-cell sequencing studies started dissecting the heterogeneity of cancer-associated fibroblasts, the research community should still answer these questions: "What makes a cancer-associated fibroblast protumorigenic?"; "How do the intracellular signaling and the secretome of different cancer-associated fibroblast subpopulations differ from each other?"; and "Which cancer-associated fibroblast subtypes predominate specific cancer types?". Unveiling these questions can pave the way for discovering efficient cancer-associated fibroblast targeting strategies. Here, we review current knowledge and perspectives on these questions, focusing on how CAFs induce aggressiveness and therapy resistance in gastric cancer. We also review potential therapeutic approaches to prevent the development and activation of cancer-associated fibroblasts via inhibition of CAF inducers and CAF markers in cancer.
Collapse
Affiliation(s)
- Ece Ozmen
- Koç University Graduate School of Health Sciences, Istanbul, Türkiye
| | - Tevriz Dilan Demir
- Koç University Research Center for Translational Medicine (KUTTAM), Istanbul, Türkiye
| | - Gulnihal Ozcan
- Koç University Research Center for Translational Medicine (KUTTAM), Istanbul, Türkiye
- Department of Medical Pharmacology, Koç University School of Medicine, Istanbul, Türkiye
| |
Collapse
|
32
|
Sarkar M, Burkel BM, Ponik SM, Notbohm J. Unexpected softening of a fibrous matrix by contracting inclusions. Acta Biomater 2024; 177:253-264. [PMID: 38272198 PMCID: PMC10948310 DOI: 10.1016/j.actbio.2024.01.025] [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: 08/15/2023] [Revised: 01/16/2024] [Accepted: 01/18/2024] [Indexed: 01/27/2024]
Abstract
Cells respond to the stiffness of their surrounding environment, but quantifying the stiffness of a fibrous matrix at the scale of a cell is complicated, due to the effects of nonlinearity and complex force transmission pathways resulting from randomness in fiber density and connections. While it is known that forces produced by individual contractile cells can stiffen the matrix, it remains unclear how simultaneous contraction of multiple cells in a fibrous matrix alters the stiffness at the scale of a cell. Here, we used computational modeling and experiments to quantify the stiffness of a random fibrous matrix embedded with multiple contracting inclusions, which mimicked the contractile forces of a cell. The results showed that when the matrix was free to contract as a result of the forces produced by the inclusions, the matrix softened rather than stiffened, which was surprising given that the contracting inclusions applied tensile forces to the matrix. Using the computational model, we identified that the underlying cause of the softening was that the majority of the fibers were under a local state of axial compression, causing buckling. We verified that this buckling-induced matrix softening was sufficient for cells to sense and respond by altering their morphology and force generation. Our findings reveal that the localized forces induced by cells do not always stiffen the matrix; rather, softening can occur in instances wherein the matrix can contract in response to the cell-generated forces. This study opens up new possibilities to investigate whether cell-induced softening contributes to maintenance of homeostatic conditions or progression of disease. STATEMENT OF SIGNIFICANCE: Mechanical interactions between cells and the surrounding matrix strongly influence cellular functions. Cell-induced forces can alter matrix properties, and much prior literature in this area focused on the influence of individual contracting cells. Cells in tissues are rarely solitary; rather, they are interspersed with neighboring cells throughout the matrix. As a result, the mechanics are complicated, leaving it unclear how the multiple contracting cells affect matrix stiffness. Here, we show that multiple contracting inclusions within a fibrous matrix can cause softening that in turn affects cell sensing and response. Our findings provide new directions to determine impacts of cell-induced softening on maintenance of tissue or progression of disease.
Collapse
Affiliation(s)
- Mainak Sarkar
- Department of Mechanical Engineering, University of Wisconsin-Madison, Madison, WI, USA
| | - Brian M Burkel
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI, USA; University of Wisconsin Carbone Cancer Center, Madison, WI, USA
| | - Suzanne M Ponik
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI, USA; University of Wisconsin Carbone Cancer Center, Madison, WI, USA
| | - Jacob Notbohm
- Department of Mechanical Engineering, University of Wisconsin-Madison, Madison, WI, USA; University of Wisconsin Carbone Cancer Center, Madison, WI, USA.
| |
Collapse
|
33
|
Chandler T, Guo M, Su Y, Chen J, Wu Y, Liu J, Agashe A, Fischer RS, Mehta SB, Kumar A, Baskin TI, Jamouillé V, Liu H, Swaminathan V, Nain A, Oldenbourg R, Riviére PL, Shroff H. Three-dimensional spatio-angular fluorescence microscopy with a polarized dual-view inverted selective-plane illumination microscope (pol-diSPIM). BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.09.584243. [PMID: 38712306 PMCID: PMC11071302 DOI: 10.1101/2024.03.09.584243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Polarized fluorescence microscopy is a valuable tool for measuring molecular orientations, but techniques for recovering three-dimensional orientations and positions of fluorescent ensembles are limited. We report a polarized dual-view light-sheet system for determining the three-dimensional orientations and diffraction-limited positions of ensembles of fluorescent dipoles that label biological structures, and we share a set of visualization, histogram, and profiling tools for interpreting these positions and orientations. We model our samples, their excitation, and their detection using coarse-grained representations we call orientation distribution functions (ODFs). We apply ODFs to create physics-informed models of image formation with spatio-angular point-spread and transfer functions. We use theory and experiment to conclude that light-sheet tilting is a necessary part of our design for recovering all three-dimensional orientations. We use our system to extend known two-dimensional results to three dimensions in FM1-43-labelled giant unilamellar vesicles, fast-scarlet-labelled cellulose in xylem cells, and phalloidin-labelled actin in U2OS cells. Additionally, we observe phalloidin-labelled actin in mouse fibroblasts grown on grids of labelled nanowires and identify correlations between local actin alignment and global cell-scale orientation, indicating cellular coordination across length scales.
Collapse
Affiliation(s)
- Talon Chandler
- CZ Biohub SF, San Francisco, 94158, California, USA
- Department of Radiology, University of Chicago, Chicago, 60637, Illinois, USA
| | - Min Guo
- State Key Laboratory of Extreme Photonics and Instrumentation, College of Optical Science and Engineering, Zhejiang University, Hangzhou, 310027, Zhejiang, China
- Laboratory of High Resolution Optical Imaging, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, 20892, Maryland, USA
| | - Yijun Su
- Laboratory of High Resolution Optical Imaging, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, 20892, Maryland, USA
- Advanced Imaging and Microscopy Resource, National Institutes of Health, Bethesda, 20892, Maryland, USA
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, 20147, Virginia, USA
| | - Jiji Chen
- Advanced Imaging and Microscopy Resource, National Institutes of Health, Bethesda, 20892, Maryland, USA
| | - Yicong Wu
- Laboratory of High Resolution Optical Imaging, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, 20892, Maryland, USA
| | - Junyu Liu
- State Key Laboratory of Extreme Photonics and Instrumentation, College of Optical Science and Engineering, Zhejiang University, Hangzhou, 310027, Zhejiang, China
| | - Atharva Agashe
- Department of Mechanical Engineering, Virginia Tech, Blacksburg, 24061, Virginia, USA
| | - Robert S. Fischer
- Cell Biology and Physiology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, 20892, Maryland, USA
| | - Shalin B. Mehta
- CZ Biohub SF, San Francisco, 94158, California, USA
- Department of Radiology, University of Chicago, Chicago, 60637, Illinois, USA
- Bell Center, Marine Biological Laboratory, Woods Hole, 02543, Massachusetts, USA
| | - Abhishek Kumar
- Bell Center, Marine Biological Laboratory, Woods Hole, 02543, Massachusetts, USA
| | - Tobias I. Baskin
- Biology Department, University of Massachusetts, Amherst, 01003, Maryland, USA
- Whitman Center, Marine Biological Laboratory, Woods Hole, 02543, Massachusetts, USA
| | - Valentin Jamouillé
- Cell Biology and Physiology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, 20892, Maryland, USA
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, V5A 1S6, British Columbia, Canada
| | - Huafeng Liu
- State Key Laboratory of Extreme Photonics and Instrumentation, College of Optical Science and Engineering, Zhejiang University, Hangzhou, 310027, Zhejiang, China
| | - Vinay Swaminathan
- Department of Clinical Sciences, Lund University, Lund, SE-221 00, Scania, Sweden
- Wallenberg Centre for Molecular Medicine, Lund University, Lund, SE-221 00, Scania, Sweden
| | - Amrinder Nain
- Department of Mechanical Engineering, Virginia Tech, Blacksburg, 24061, Virginia, USA
- Department of Biomedical Engineering and Mechanics, Virginia Tech, Blacksburg, 24061, Virginia, USA
| | - Rudolf Oldenbourg
- Bell Center, Marine Biological Laboratory, Woods Hole, 02543, Massachusetts, USA
| | - Patrick La Riviére
- Department of Radiology, University of Chicago, Chicago, 60637, Illinois, USA
- Whitman Center, Marine Biological Laboratory, Woods Hole, 02543, Massachusetts, USA
| | - Hari Shroff
- Laboratory of High Resolution Optical Imaging, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, 20892, Maryland, USA
- Advanced Imaging and Microscopy Resource, National Institutes of Health, Bethesda, 20892, Maryland, USA
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, 20147, Virginia, USA
- Whitman Center, Marine Biological Laboratory, Woods Hole, 02543, Massachusetts, USA
| |
Collapse
|
34
|
Gorczynski R. Translation of Data from Animal Models of Cancer to Immunotherapy of Breast Cancer and Chronic Lymphocytic Leukemia. Genes (Basel) 2024; 15:292. [PMID: 38540350 PMCID: PMC10970502 DOI: 10.3390/genes15030292] [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: 01/23/2024] [Revised: 02/18/2024] [Accepted: 02/23/2024] [Indexed: 06/14/2024] Open
Abstract
The field of clinical oncology has been revolutionized over the past decade with the introduction of many new immunotherapies the existence of which have depended to a large extent on experimentation with both in vitro analysis and the use of various animal models, including gene-modified mice. The discussion below will review my own laboratory's studies, along with those of others in the field, on cancer immunotherapy. Our own studies have predominantly dwelt on two models of malignancy, namely a solid tumor model (breast cancer) and lymphoma. The data from our own laboratory, and that of other scientists, highlights the novel information so obtained, and the evidence that application of such information has already had an impact on immunotherapy of human oncologic diseases.
Collapse
Affiliation(s)
- Reginald Gorczynski
- Institute of Medical Science, Department of Immunology and Surgery, University of Toronto, C/O 429 Drewry Avenue, Toronto, ON M2R 2K6, Canada
| |
Collapse
|
35
|
Kenny-Ganzert IW, Sherwood DR. The C. elegans anchor cell: A model to elucidate mechanisms underlying invasion through basement membrane. Semin Cell Dev Biol 2024; 154:23-34. [PMID: 37422376 PMCID: PMC10592375 DOI: 10.1016/j.semcdb.2023.07.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Revised: 06/30/2023] [Accepted: 07/01/2023] [Indexed: 07/10/2023]
Abstract
Cell invasion through basement membrane barriers is crucial during many developmental processes and in immune surveillance. Dysregulation of invasion also drives the pathology of numerous human diseases, such as metastasis and inflammatory disorders. Cell invasion involves dynamic interactions between the invading cell, basement membrane, and neighboring tissues. Owing to this complexity, cell invasion is challenging to study in vivo, which has hampered the understanding of mechanisms controlling invasion. Caenorhabditis elegans anchor cell invasion is a powerful in vivo model where subcellular imaging of cell-basement membrane interactions can be combined with genetic, genomic, and single-cell molecular perturbation studies. In this review, we outline insights gained by studying anchor cell invasion, which span transcriptional networks, translational regulation, secretory apparatus expansion, dynamic and adaptable protrusions that breach and clear basement membrane, and a complex, localized metabolic network that fuels invasion. Together, investigation of anchor cell invasion is building a comprehensive understanding of the mechanisms that underlie invasion, which we expect will ultimately facilitate better therapeutic strategies to control cell invasive activity in human disease.
Collapse
Affiliation(s)
| | - David R Sherwood
- Department of Biology, Duke University, Box 90338, Durham, NC 27708, USA.
| |
Collapse
|
36
|
Bertillot F, Andrique L, Ureña Martin C, Zajac O, de Plater L, Norton MM, Richard A, Alessandri K, Gurchenkov BG, Fage F, Asnacios A, Lamaze C, Das M, Maître JL, Nassoy P, Matic Vignjevic D. Compressive stress triggers fibroblasts spreading over cancer cells to generate carcinoma in situ organization. Commun Biol 2024; 7:184. [PMID: 38360973 PMCID: PMC10869726 DOI: 10.1038/s42003-024-05883-6] [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: 03/09/2022] [Accepted: 02/02/2024] [Indexed: 02/17/2024] Open
Abstract
At the early stage of tumor progression, fibroblasts are located at the outer edges of the tumor, forming an encasing layer around it. In this work, we have developed a 3D in vitro model where fibroblasts' layout resembles the structure seen in carcinoma in situ. We use a microfluidic encapsulation technology to co-culture fibroblasts and cancer cells within hollow, permeable, and elastic alginate shells. We find that in the absence of spatial constraint, fibroblasts and cancer cells do not mix but segregate into distinct aggregates composed of individual cell types. However, upon confinement, fibroblasts enwrap cancer cell spheroid. Using a combination of biophysical methods and live imaging, we find that buildup of compressive stress is required to induce fibroblasts spreading over the aggregates of tumor cells. We propose that compressive stress generated by the tumor growth might be a mechanism that prompts fibroblasts to form a capsule around the tumor.
Collapse
Affiliation(s)
- Fabien Bertillot
- Institut Curie, PSL Research University, CNRS UMR 144, F-75005, Paris, France
- Department of Cell and Tissue Dynamics, Max Planck Institute for Molecular Biomedicine, 48149, Münster, Germany
| | - Laetitia Andrique
- LP2N, Laboratoire Photonique Numérique et Nanosciences, Univ. Bordeaux, F-33400, Talence, France
- Institut d'Optique Graduate School & CNRS UMR 5298, F-33400, Talence, France
- VoxCell, TBM-Core, CNRS UMS 3427 & INSERM US 005, Univ. Bordeaux, F-33000, Bordeaux, France
| | - Carlos Ureña Martin
- Institut Curie, PSL Research University, CNRS UMR3666-INSERM U1143, F-75005, Paris, France
| | - Olivier Zajac
- Institut Curie, PSL Research University, CNRS UMR 144, F-75005, Paris, France
| | - Ludmilla de Plater
- Institut Curie, PSL Research University, U934/UMR3215, F-75005, Paris, France
| | - Michael M Norton
- VoxCell, TBM-Core, CNRS UMS 3427 & INSERM US 005, Univ. Bordeaux, F-33000, Bordeaux, France
| | - Aurélien Richard
- LP2N, Laboratoire Photonique Numérique et Nanosciences, Univ. Bordeaux, F-33400, Talence, France
- Institut d'Optique Graduate School & CNRS UMR 5298, F-33400, Talence, France
- VoxCell, TBM-Core, CNRS UMS 3427 & INSERM US 005, Univ. Bordeaux, F-33000, Bordeaux, France
| | - Kevin Alessandri
- Institut Curie, PSL Research University, CNRS UMR 144, F-75005, Paris, France
| | - Basile G Gurchenkov
- Institut Curie, PSL Research University, CNRS UMR 144, F-75005, Paris, France
| | - Florian Fage
- Laboratoire Matière et Systèmes Complexes, Université Paris Cité, CNRS UMR7057, F-75013, Paris, France
| | - Atef Asnacios
- Laboratoire Matière et Systèmes Complexes, Université Paris Cité, CNRS UMR7057, F-75013, Paris, France
| | - Christophe Lamaze
- Institut Curie, PSL Research University, CNRS UMR3666-INSERM U1143, F-75005, Paris, France
| | - Moumita Das
- Rochester Institute of Technology, Rochester, NY, USA
| | - Jean- Léon Maître
- Institut Curie, PSL Research University, U934/UMR3215, F-75005, Paris, France
| | - Pierre Nassoy
- LP2N, Laboratoire Photonique Numérique et Nanosciences, Univ. Bordeaux, F-33400, Talence, France.
- Institut d'Optique Graduate School & CNRS UMR 5298, F-33400, Talence, France.
| | | |
Collapse
|
37
|
Jones RA, Trejo B, Sil P, Little KA, Pasolli HA, Joyce B, Posfai E, Devenport D. An mTurq2-Col4a1 mouse model allows for live visualization of mammalian basement membrane development. J Cell Biol 2024; 223:e202309074. [PMID: 38051393 PMCID: PMC10697824 DOI: 10.1083/jcb.202309074] [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: 09/25/2023] [Revised: 11/03/2023] [Accepted: 11/15/2023] [Indexed: 12/07/2023] Open
Abstract
Basement membranes (BMs) are specialized sheets of extracellular matrix that underlie epithelial and endothelial tissues. BMs regulate the traffic of cells and molecules between compartments, and participate in signaling, cell migration, and organogenesis. The dynamics of mammalian BMs, however, are poorly understood, largely due to a lack of models in which core BM components are endogenously labeled. Here, we describe the mTurquoise2-Col4a1 mouse in which we fluorescently tag collagen IV, the main component of BMs. Using an innovative planar-sagittal live imaging technique to visualize the BM of developing skin, we directly observe BM deformation during hair follicle budding and basal progenitor cell divisions. The BM's inherent pliability enables dividing cells to remain attached to and deform the BM, rather than lose adhesion as generally thought. Using FRAP, we show BM collagen IV is extremely stable, even during periods of rapid epidermal growth. These findings demonstrate the utility of the mTurq2-Col4a1 mouse to shed new light on mammalian BM developmental dynamics.
Collapse
Affiliation(s)
- Rebecca A. Jones
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Brandon Trejo
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Parijat Sil
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | | | - H. Amalia Pasolli
- Electron Microscopy Resource Center, The Rockefeller University, New York, NY, USA
| | - Bradley Joyce
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Eszter Posfai
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Danelle Devenport
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| |
Collapse
|
38
|
Jiang L, Khawaja H, Tahsin S, Clarkson TA, Miranti CK, Zohar Y. Microfluidic-based human prostate-cancer-on-chip. Front Bioeng Biotechnol 2024; 12:1302223. [PMID: 38322789 PMCID: PMC10844564 DOI: 10.3389/fbioe.2024.1302223] [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: 09/26/2023] [Accepted: 01/09/2024] [Indexed: 02/08/2024] Open
Abstract
Lack of adequate models significantly hinders advances in prostate cancer treatment, where resistance to androgen-deprivation therapies and bone metastasis remain as major challenges. Current in vitro models fail to faithfully mimic the complex prostate physiology. In vivo animal models can shed light on the oncogenes involved in prostate cancer development and progression; however, the animal prostate gland is fundamentally different from that of human, and the underlying genetic mechanisms are different. To address this problem, we developed the first in vitro microfluidic human Prostate-Cancer-on-Chip (PCoC) model, where human prostate cancer and stromal fibroblast cells were co-cultivated in two channels separated by a porous membrane under culture medium flow. The established microenvironment enables soluble signaling factors secreted by each culture to locally diffuse through the membrane pores affecting the neighboring culture. We particularly explored the conversion of the stromal fibroblasts into cancer-associated fibroblasts (CAFs) due to the interaction between the 2 cell types. Immunofluorescence microscopy revealed that tumor cells induced CAF biomarkers, αSMA and COL1A1, in stromal fibroblasts. The stromal CAF conversion level was observed to increase along the flow direction in response to diffusion agents, consistent with simulations of solute concentration gradients. The tumor cells also downregulated androgen receptor (AR) expression in stromal fibroblasts, while an adequate level of stromal AR expression is maintained in normal prostate homeostasis. We further investigated tumor invasion into the stroma, an early step in the metastatic cascade, in devices featuring a serpentine channel with orthogonal channel segments overlaying a straight channel and separated by an 8 µm-pore membrane. Both tumor cells and stromal CAFs were observed to cross over into their neighboring channel, and the stroma's role seemed to be proactive in promoting cell invasion. As control, normal epithelial cells neither induced CAF conversion nor promoted cell invasion. In summary, the developed PCoC model allows spatiotemporal analysis of the tumor-stroma dynamic interactions, due to bi-directional signaling and physical contact, recapitulating tissue-level multicellular responses associated with prostate cancer in vivo. Hence, it can serve as an in vitro model to dissect mechanisms in human prostate cancer development and seek advanced therapeutic strategies.
Collapse
Affiliation(s)
- Linan Jiang
- Department of Aerospace and Mechanical Engineering, Tucson, AZ, United States
| | - Hunain Khawaja
- Cancer Biology Graduate Interdisciplinary Program, Tucson, AZ, United States
| | - Shekha Tahsin
- Cancer Biology Graduate Interdisciplinary Program, Tucson, AZ, United States
| | | | - Cindy K. Miranti
- Department of Molecular and Cellular Biology, Tucson, AZ, United States
- University of Arizona Cancer Center, University of Arizona, Tucson, AZ, United States
| | - Yitshak Zohar
- Department of Aerospace and Mechanical Engineering, Tucson, AZ, United States
- University of Arizona Cancer Center, University of Arizona, Tucson, AZ, United States
| |
Collapse
|
39
|
Mitha M, Aden D, Zaheer S, Alvi Y, Chintamani. Role of tumor budding and fibrotic cancer stroma in head and neck squamous cell carcinoma. Pathol Res Pract 2024; 253:155052. [PMID: 38176309 DOI: 10.1016/j.prp.2023.155052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 12/14/2023] [Accepted: 12/19/2023] [Indexed: 01/06/2024]
Abstract
BACKGROUND Head and neck squamous cell carcinoma (HNSCC) is an aggressive cancer with an increased frequency of lymph node metastasis at the time of presentation. Tumour budding, characterised by the presence of a single cell or a small grouping of tumour cells (a cluster containing fewer than five malignant cells) at the invasive front and composition of the fibrotic cancer stroma has been demonstrated to have a growing impact on the behaviour of the solid tumour. However exact role played by them is yet to be defined and a standardized scoring system needs to be incorporated. MATERIAL AND METHODS A total of 45 histopathologically confirmed cases of HNSCC were included in the study. Hematoxylin and Eosin staining (H&E staining), and immunohistochemistry for CK and alpha-SMA were applied to study the tumour budding and fibrotic cancer stroma in all HNSCC cases. The tumour budding was graded as, Grade 1: 0-4 tumour buds, Grade 2: 5-9 buds and Grade 3: ≥ 10 buds and the nature of fibrotic cancer stroma was categorized as mature, intermediate or immature. RESULTS Among 45 cases analyzed, well differentiated squamous cell carcinoma (WDSCC; Grade 1) accounted for 42.22% (19 cases), whereas moderately differentiated squamous cell carcinoma (MDSCC; Grade 2) and poorly differentiated squamous cell carcinoma (PDSCC; Grade 3) comprised 48.89% (22 cases) and 8.89% (4 cases) respectively. Tumour budding showed instances of 0-4 buds in 33.3% (Grade 1), 5-9 buds in 48.9% (Grade 2), and ≥ 10 buds in 17.8% of cases. Evaluating tumour stroma, Intermediate stroma led at 51.1%, Mature at 37.8%, and 11.1% displayed Immature stroma. Histologically, < 5 buds were seen in 47.4% of Grade 1 cases, while ≥ 10 buds were in 75.0% of Grade 3 cases, proven statistically significant (p = 0.021). However, an association between T&N Stage and tumour budding lacked significance. WDSCC notably had more mature stroma than MDSCC and PDSCC, whereas MDSCC showed higher rates of intermediate and immature stroma (p < 0.001). Comparatively, no significant correlation existed between fibrotic stroma and tumour budding (p = 0.076). Also, fibrotic stroma was compared with tumour budding, however, no significant correlation was found (p = 0.076) CONCLUSION: This study reveals a significant link between tumour budding, cancer stroma, and WHO tumour grade. Thus, evaluating these factors in HNSCC cases can serve as valuable histological prognostic indicators, aiding in treatment planning and prognosis assessment.
Collapse
Affiliation(s)
- Madhu Mitha
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India
| | - Durre Aden
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India
| | - Sufian Zaheer
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India.
| | - Yasir Alvi
- Department of Community Medicine, Hamdard Institute of Medical Sciences and Research New Delhi, India
| | - Chintamani
- Department of Surgery, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India
| |
Collapse
|
40
|
Zhou F, Liu Y, Liu D, Xie Y, Zhou X. Identification of basement membrane-related signatures for estimating prognosis, immune infiltration landscape and drug candidates in pancreatic adenocarcinoma. J Cancer 2024; 15:401-417. [PMID: 38169540 PMCID: PMC10758037 DOI: 10.7150/jca.89665] [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: 08/30/2023] [Accepted: 11/10/2023] [Indexed: 01/05/2024] Open
Abstract
Background: Pancreatic adenocarcinoma (PAAD) is a frequent digestive system cancer, which has high mortality and bad outcome. However, the role of basement membrane (BM)-related gene in assessing patient's outcome, microenvironment and treatment response remain unclear. Methods: Basement membrane (BM)-associated genes were detected by univariate and least absolute shrinkage and selection operator (LASSO) Cox regression analyses using data from the TCGA databases. A risk score system was constructed to distinguish patients in the high- and low-risk groups. Prognostic gene distribution in various immune cell forms was explored through scRNA-seq. Immune cell infiltration was assessed using CIBERSORT and ESTIMATE. The IC50 of common chemotherapeutic drugs and useful molecule compounds were evaluated. The mRNA and protein expression of important signatures were validated utilizing GEPIA and HPA databases. Results: Compared to low risk PAAD patients, PAAD patients with high risk showed a significant much worse overall survival (OS). Risk score of BM-associated genes could estimate patient outcome well, and areas under the curve (AUC) of receiver operating characteristic (ROC) survival curve were 0.76, 0.85, and 0.85 at 1-, 3-, and 5-year. Clinical impact curve (CIC) curve demonstrated the clinical importance of risk score. scRNA-seq revealed that BM-related genes were mainly distributed in malignant cells. Significant variations existed in the immune microenvironment, immune checkpoint expression and chemotherapy response between the studied groups. Furthermore, the mRNA expression levels of FAM83A, LY6D, MET, MUC16, MYEOV, and WNT7A were elevated in PAAD tissues, while the protein expression patterns of LY6D, MET, MUC16, and WNT7A were higher in tumor sample. RO-90-7501, Scriptaid, TG-101348, XMD-892, and XMD-1150 may be valuable small molecule drugs to treat PAAD. Conclusions: In conclusion, we develop a novel BM-related gene signature provide new insights and targets for the diagnosis, outcome estimation, candidate drugs and therapy management of PAAD patients.
Collapse
Affiliation(s)
- Feng Zhou
- Department of Gastroenterology, Digestive disease Hospital, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
- Gastroenterology Institute of Jiangxi Province, Nanchang, Jiangxi Province, China
- Key Laboratory of Digestive Diseases of Jiangxi Province, Nanchang, Jiangxi Province, China
- Jiangxi Clinical Research Center for Gastroenterology, Nanchang, Jiangxi Province, China
| | - Yang Liu
- Department of Gastroenterology, Digestive disease Hospital, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
- Gastroenterology Institute of Jiangxi Province, Nanchang, Jiangxi Province, China
- Key Laboratory of Digestive Diseases of Jiangxi Province, Nanchang, Jiangxi Province, China
- Jiangxi Clinical Research Center for Gastroenterology, Nanchang, Jiangxi Province, China
| | - Dingwei Liu
- Department of Gastroenterology, Digestive disease Hospital, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
- Gastroenterology Institute of Jiangxi Province, Nanchang, Jiangxi Province, China
- Key Laboratory of Digestive Diseases of Jiangxi Province, Nanchang, Jiangxi Province, China
- Jiangxi Clinical Research Center for Gastroenterology, Nanchang, Jiangxi Province, China
| | - Yong Xie
- Department of Gastroenterology, Digestive disease Hospital, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
- Gastroenterology Institute of Jiangxi Province, Nanchang, Jiangxi Province, China
- Key Laboratory of Digestive Diseases of Jiangxi Province, Nanchang, Jiangxi Province, China
- Jiangxi Clinical Research Center for Gastroenterology, Nanchang, Jiangxi Province, China
| | - Xiaojiang Zhou
- Department of Gastroenterology, Digestive disease Hospital, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
- Gastroenterology Institute of Jiangxi Province, Nanchang, Jiangxi Province, China
- Key Laboratory of Digestive Diseases of Jiangxi Province, Nanchang, Jiangxi Province, China
- Jiangxi Clinical Research Center for Gastroenterology, Nanchang, Jiangxi Province, China
| |
Collapse
|
41
|
Thorlacius-Ussing J, Jensen C, Nissen NI, Cox TR, Kalluri R, Karsdal M, Willumsen N. The collagen landscape in cancer: profiling collagens in tumors and in circulation reveals novel markers of cancer-associated fibroblast subtypes. J Pathol 2024; 262:22-36. [PMID: 37728068 DOI: 10.1002/path.6207] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 07/07/2023] [Accepted: 08/18/2023] [Indexed: 09/21/2023]
Abstract
Cancer-associated fibroblasts (CAFs) deposit and remodel collagens in the tumor stroma, impacting cancer progression and efficacy of interventions. CAFs are the focus of new therapeutics with the aim of normalizing the tumor microenvironment. To do this, a better understanding of CAF heterogeneity and collagen composition in cancer is needed. In this study, we sought to profile the expression of collagens at multiple levels with the goal of identifying cancer biomarkers. We investigated the collagen expression pattern in various cell types and CAF subtypes in a publicly available single-cell RNA sequencing (RNA-seq) dataset of pancreatic ductal adenocarcinoma. Next, we investigated the collagen expression profile in tumor samples across cancer types from The Cancer Genome Atlas (TCGA) database and evaluated if specific patterns of collagen expression were associated with prognosis. Finally, we profiled circulating collagen peptides using a panel of immunoassays to measure collagen fragments in the serum of cancer patients. We found that pancreatic stellate cells and fibroblasts were the primary producers of collagens in the pancreas. COL1A1, COL3A1, COL5A1, COL6A1 were expressed in all CAF subtypes, whereas COL8A1, COL10A1, COL11A1, COL12A1 were specific to myofibroblast CAFs (myCAF) and COL14A1 specific to inflammatory CAFs (iCAF). In TCGA database, myCAF collagens COL10A1 and COL11A1 were elevated across solid tumor types, and multiple associations between high expression and worse survival were found. Finally, circulating collagen biomarkers were elevated in the serum of patients with cancer relative to healthy controls with COL11A1 (myCAF) having the best diagnostic accuracy of the markers measured. In conclusion, CAFs express a noncanonical collagen profile with specific collagen subtypes associated with iCAFs and myCAFs in PDAC. These collagens are deregulated at the cellular, tumor, and systemic levels across different solid tumors and associate with survival. These findings could lead to new discoveries such as novel biomarkers and therapeutic targets. © 2023 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Jeppe Thorlacius-Ussing
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, København N, Denmark
- Biomarkers & Research, Nordic Bioscience A/S, Herlev, Denmark
| | | | - Neel I Nissen
- Biomarkers & Research, Nordic Bioscience A/S, Herlev, Denmark
| | - Thomas R Cox
- Matrix and Metastasis Lab, Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Darlinghurst, NSW, Australia
- School of Clinical Medicine, UNSW Medicine & Health, UNSW Sydney, Sydney, NSW, Australia
| | - Raghu Kalluri
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Morten Karsdal
- Biomarkers & Research, Nordic Bioscience A/S, Herlev, Denmark
| | | |
Collapse
|
42
|
Perez-Gonzalez C, Barbazán J. Addressing the Mechanical Interaction Between Cancer-Associated Fibroblasts and Cancer Cells by Laser Ablation. Methods Mol Biol 2024; 2764:179-203. [PMID: 38393596 DOI: 10.1007/978-1-0716-3674-9_13] [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] [Indexed: 02/25/2024]
Abstract
Cells within a tumor interact by generating, transmitting, and sensing mechanical forces. Among all the cells of the tumor microenvironment, cancer-associated fibroblasts (CAFs) are a paradigmatic example of mechanical communication. In different steps of tumor progression, CAFs pull and push on cancer cells, regulating cancer cell migration, invasion, compartmentalization, and signaling. There is thus an increasing need to experimentally address mechanical interactions within a tumor. A common technique to measure these interactions is laser ablation. Cutting a tissue region with a high-power laser triggers a sudden tissue displacement whose direction and magnitude reveal the local mechanical stresses. In this chapter, we provide a detailed protocol to perform laser ablations in vitro and ex vivo. First, we describe how to prepare cocultures of primary CAFs and cancer cells and tumor explants. Then, we explain how to perform laser ablations in these two systems and how to analyze the induced tissue displacements using particle image velocimetry (PIV). Overall, we provide a workflow to perform, analyze, and interpret laser ablations to explore tumor mechanical interactions.
Collapse
Affiliation(s)
- Carlos Perez-Gonzalez
- Cell Biology and Cancer Unit, Institut Curie, PSL Research University, CNRS, Paris, France.
| | - Jorge Barbazán
- Translational Medical Oncology Group (ONCOMET), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain.
| |
Collapse
|
43
|
Matsuoka T, Yashiro M. Molecular Insight into Gastric Cancer Invasion-Current Status and Future Directions. Cancers (Basel) 2023; 16:54. [PMID: 38201481 PMCID: PMC10778111 DOI: 10.3390/cancers16010054] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 12/15/2023] [Accepted: 12/20/2023] [Indexed: 01/12/2024] Open
Abstract
Gastric cancer (GC) is one of the most common malignancies worldwide. There has been no efficient therapy for stage IV GC patients due to this disease's heterogeneity and dissemination ability. Despite the rapid advancement of molecular targeted therapies, such as HER2 and immune checkpoint inhibitors, survival of GC patients is still unsatisfactory because the understanding of the mechanism of GC progression is still incomplete. Invasion is the most important feature of GC metastasis, which causes poor mortality in patients. Recently, genomic research has critically deepened our knowledge of which gene products are dysregulated in invasive GC. Furthermore, the study of the interaction of GC cells with the tumor microenvironment has emerged as a principal subject in driving invasion and metastasis. These results are expected to provide a profound knowledge of how biological molecules are implicated in GC development. This review summarizes the advances in our current understanding of the molecular mechanism of GC invasion. We also highlight the future directions of the invasion therapeutics of GC. Compared to conventional therapy using protease or molecular inhibitors alone, multi-therapy targeting invasion plasticity may seem to be an assuring direction for the progression of novel strategies.
Collapse
Affiliation(s)
| | - Masakazu Yashiro
- Molecular Oncology and Therapeutics, Osaka Metropolitan University Graduate School of Medicine, Osaka 5458585, Japan;
| |
Collapse
|
44
|
Agrawal A, Lasli S, Javanmardi Y, Coursier D, Micalet A, Watson S, Shahreza S, Serwinski B, Djordjevic B, Szita N, Cheema U, Bertazzo S, Calvo F, Moeendarbary E. Stromal cells regulate mechanics of tumour spheroid. Mater Today Bio 2023; 23:100821. [PMID: 37868949 PMCID: PMC10585335 DOI: 10.1016/j.mtbio.2023.100821] [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: 05/04/2023] [Revised: 09/15/2023] [Accepted: 09/24/2023] [Indexed: 10/24/2023] Open
Abstract
The remarkable contractility and force generation ability exhibited by cancer cells empower them to overcome the resistance and steric hindrance presented by a three-dimensional, interconnected matrix. Cancer cells disseminate by actively remodelling and deforming their extracellular matrix (ECM). The process of tumour growth and its ECM remodelling have been extensively studied, but the effect of the cellular tumour microenvironment (TME) has been ignored in most studies that investigated tumour-cell-mediated ECM deformations and realignment. This study reports the integration of stromal cells in spheroid contractility assays that impacts the ECM remodelling and invasion abilities of cancer spheroids. To investigate this, we developed a novel multilayer in vitro assay that incorporates stromal cells and quantifies the contractile deformations that tumour spheroids exert on the ECM. We observed a negative correlation between the spheroid invasion potential and the levels of collagen deformation. The presence of stromal cells significantly increased cancer cell invasiveness and altered the cancer cells' ability to deform and realign collagen gel, due to upregulation of proinflammatory cytokines. Interestingly, this was observed consistently in both metastatic and non-metastatic cancer cells. Our findings contribute to a better understanding of the vital role played by the cellular TME in regulating the invasive outgrowth of cancer cells and underscore the potential of utilising matrix deformation measurements as a biophysical marker for evaluating invasiveness and informing targeted therapeutic opportunities.
Collapse
Affiliation(s)
- Ayushi Agrawal
- Department of Mechanical Engineering, University College London, London, WC1E 7JE, UK
| | - Soufian Lasli
- Department of Mechanical Engineering, University College London, London, WC1E 7JE, UK
| | - Yousef Javanmardi
- Department of Mechanical Engineering, University College London, London, WC1E 7JE, UK
| | - Diane Coursier
- 199 Biotechnologies Ltd, Gloucester Road, London, W2 6LD, UK
| | - Auxtine Micalet
- Department of Mechanical Engineering, University College London, London, WC1E 7JE, UK
- Department of Targeted Intervention, Division of Surgery and Interventional Science, University College London, London WC1E 7JE, UK
| | - Sara Watson
- Department of Mechanical Engineering, University College London, London, WC1E 7JE, UK
| | - Somayeh Shahreza
- Department of Mechanical Engineering, University College London, London, WC1E 7JE, UK
| | - Bianca Serwinski
- Department of Mechanical Engineering, University College London, London, WC1E 7JE, UK
- 199 Biotechnologies Ltd, Gloucester Road, London, W2 6LD, UK
- Faculty of Social Sciences, Northeastern University London, London, E1W 1LP, UK
| | - Boris Djordjevic
- Department of Mechanical Engineering, University College London, London, WC1E 7JE, UK
- 199 Biotechnologies Ltd, Gloucester Road, London, W2 6LD, UK
| | - Nicolas Szita
- Department of Biochemical Engineering, University College London, London WC1E 7JE, UK
| | - Umber Cheema
- Department of Targeted Intervention, Division of Surgery and Interventional Science, University College London, London WC1E 7JE, UK
| | - Sergio Bertazzo
- Department of Medical Physics and Biomedical Engineering, University College London, London, WC1E 6BT, UK
| | - Fernando Calvo
- Instituto de Biomedicina y Biotecnología de Cantabria (Consejo Superior de Investigaciones Científicas, Universidad de Cantabria), Santander, Spain
| | - Emad Moeendarbary
- Department of Mechanical Engineering, University College London, London, WC1E 7JE, UK
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| |
Collapse
|
45
|
Barbazan J, Pérez-González C, Gómez-González M, Dedenon M, Richon S, Latorre E, Serra M, Mariani P, Descroix S, Sens P, Trepat X, Vignjevic DM. Cancer-associated fibroblasts actively compress cancer cells and modulate mechanotransduction. Nat Commun 2023; 14:6966. [PMID: 37907483 PMCID: PMC10618488 DOI: 10.1038/s41467-023-42382-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 10/09/2023] [Indexed: 11/02/2023] Open
Abstract
During tumor progression, cancer-associated fibroblasts (CAFs) accumulate in tumors and produce an excessive extracellular matrix (ECM), forming a capsule that enwraps cancer cells. This capsule acts as a barrier that restricts tumor growth leading to the buildup of intratumoral pressure. Combining genetic and physical manipulations in vivo with microfabrication and force measurements in vitro, we found that the CAFs capsule is not a passive barrier but instead actively compresses cancer cells using actomyosin contractility. Abrogation of CAFs contractility in vivo leads to the dissipation of compressive forces and impairment of capsule formation. By mapping CAF force patterns in 3D, we show that compression is a CAF-intrinsic property independent of cancer cell growth. Supracellular coordination of CAFs is achieved through fibronectin cables that serve as scaffolds allowing force transmission. Cancer cells mechanosense CAF compression, resulting in an altered localization of the transcriptional regulator YAP and a decrease in proliferation. Our study unveils that the contractile capsule actively compresses cancer cells, modulates their mechanical signaling, and reorganizes tumor morphology.
Collapse
Affiliation(s)
- Jorge Barbazan
- Institut Curie, PSL Research University, CNRS UMR 144, F-75005, Paris, France
- Translational Medical Oncology Group (ONCOMET), Health Research Institute of Santiago de Compostela (IDIS), University Hospital of Santiago de Compostela (SERGAS), 15706, Santiago de Compostela, Spain
| | | | - Manuel Gómez-González
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute for Science and Technology (BIST), 08028, Barcelona, Spain
| | - Mathieu Dedenon
- Institut Curie, PSL Research University, CNRS UMR 168, F-75005, Paris, France
| | - Sophie Richon
- Institut Curie, PSL Research University, CNRS UMR 144, F-75005, Paris, France
| | - Ernest Latorre
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute for Science and Technology (BIST), 08028, Barcelona, Spain
| | - Marco Serra
- Institut Curie, PSL Research University, CNRS UMR 168, F-75005, Paris, France
| | - Pascale Mariani
- Institut Curie, Department of surgical oncology, Curie Institute, F-75005, Paris, France
| | - Stéphanie Descroix
- Institut Curie, PSL Research University, CNRS UMR 168, F-75005, Paris, France
| | - Pierre Sens
- Institut Curie, PSL Research University, CNRS UMR 168, F-75005, Paris, France
| | - Xavier Trepat
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute for Science and Technology (BIST), 08028, Barcelona, Spain.
- Facutltat de Medicina, Universitat de Barcelona, 08036, Barcelona, Spain.
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain.
- Centro de Investigación Biomédica en Red en Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 08028, Barcelona, Spain.
| | | |
Collapse
|
46
|
Xin Y, Li K, Huang M, Liang C, Siemann D, Wu L, Tan Y, Tang X. Biophysics in tumor growth and progression: from single mechano-sensitive molecules to mechanomedicine. Oncogene 2023; 42:3457-3490. [PMID: 37864030 PMCID: PMC10656290 DOI: 10.1038/s41388-023-02844-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 09/08/2023] [Accepted: 09/15/2023] [Indexed: 10/22/2023]
Abstract
Evidence from physical sciences in oncology increasingly suggests that the interplay between the biophysical tumor microenvironment and genetic regulation has significant impact on tumor progression. Especially, tumor cells and the associated stromal cells not only alter their own cytoskeleton and physical properties but also remodel the microenvironment with anomalous physical properties. Together, these altered mechano-omics of tumor tissues and their constituents fundamentally shift the mechanotransduction paradigms in tumorous and stromal cells and activate oncogenic signaling within the neoplastic niche to facilitate tumor progression. However, current findings on tumor biophysics are limited, scattered, and often contradictory in multiple contexts. Systematic understanding of how biophysical cues influence tumor pathophysiology is still lacking. This review discusses recent different schools of findings in tumor biophysics that have arisen from multi-scale mechanobiology and the cutting-edge technologies. These findings range from the molecular and cellular to the whole tissue level and feature functional crosstalk between mechanotransduction and oncogenic signaling. We highlight the potential of these anomalous physical alterations as new therapeutic targets for cancer mechanomedicine. This framework reconciles opposing opinions in the field, proposes new directions for future cancer research, and conceptualizes novel mechanomedicine landscape to overcome the inherent shortcomings of conventional cancer diagnosis and therapies.
Collapse
Grants
- R35 GM150812 NIGMS NIH HHS
- This work was financially supported by National Natural Science Foundation of China (Project no. 11972316, Y.T.), Shenzhen Science and Technology Innovation Commission (Project no. JCYJ20200109142001798, SGDX2020110309520303, and JCYJ20220531091002006, Y.T.), General Research Fund of Hong Kong Research Grant Council (PolyU 15214320, Y. T.), Health and Medical Research Fund (HMRF18191421, Y.T.), Hong Kong Polytechnic University (1-CD75, 1-ZE2M, and 1-ZVY1, Y.T.), the Cancer Pilot Research Award from UF Health Cancer Center (X. T.), the National Institute of General Medical Sciences of the National Institutes of Health under award number R35GM150812 (X. T.), the National Science Foundation under grant number 2308574 (X. T.), the Air Force Office of Scientific Research under award number FA9550-23-1-0393 (X. T.), the University Scholar Program (X. T.), UF Research Opportunity Seed Fund (X. T.), the Gatorade Award (X. T.), and the National Science Foundation REU Site at UF: Engineering for Healthcare (Douglas Spearot and Malisa Sarntinoranont). We are deeply grateful for the insightful discussions with and generous support from all members of Tang (UF)’s and Tan (PolyU)’s laboratories and all staff members of the MAE/BME/ECE/Health Cancer Center at UF and BME at PolyU.
- National Natural Science Foundation of China (National Science Foundation of China)
- Shenzhen Science and Technology Innovation Commission
Collapse
Affiliation(s)
- Ying Xin
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, China
| | - Keming Li
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, China
| | - Miao Huang
- Department of Mechanical and Aerospace Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL, USA
| | - Chenyu Liang
- Department of Mechanical and Aerospace Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL, USA
| | - Dietmar Siemann
- UF Health Cancer Center, University of Florida, Gainesville, FL, USA
| | - Lizi Wu
- UF Health Cancer Center, University of Florida, Gainesville, FL, USA
| | - Youhua Tan
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, China.
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong, China.
- Research Institute of Smart Ageing, The Hong Kong Polytechnic University, Hong Kong, China.
| | - Xin Tang
- Department of Mechanical and Aerospace Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL, USA.
- UF Health Cancer Center, University of Florida, Gainesville, FL, USA.
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA.
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
47
|
Samain R, Maiques O, Monger J, Lam H, Candido J, George S, Ferrari N, KohIhammer L, Lunetto S, Varela A, Orgaz JL, Vilardell F, Olsina JJ, Matias-Guiu X, Sarker D, Biddle A, Balkwill FR, Eyles J, Wilkinson RW, Kocher HM, Calvo F, Wells CM, Sanz-Moreno V. CD73 controls Myosin II-driven invasion, metastasis, and immunosuppression in amoeboid pancreatic cancer cells. SCIENCE ADVANCES 2023; 9:eadi0244. [PMID: 37851808 PMCID: PMC10584351 DOI: 10.1126/sciadv.adi0244] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 09/06/2023] [Indexed: 10/20/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) has a very poor prognosis because of its high propensity to metastasize and its immunosuppressive microenvironment. Using a panel of pancreatic cancer cell lines, three-dimensional (3D) invasion systems, microarray gene signatures, microfluidic devices, mouse models, and intravital imaging, we demonstrate that ROCK-Myosin II activity in PDAC cells supports a transcriptional program conferring amoeboid invasive and immunosuppressive traits and in vivo metastatic abilities. Moreover, we find that immune checkpoint CD73 is highly expressed in amoeboid PDAC cells and drives their invasive, metastatic, and immunomodulatory traits. Mechanistically, CD73 activates RhoA-ROCK-Myosin II downstream of PI3K. Tissue microarrays of human PDAC biopsies combined with bioinformatic analysis reveal that rounded-amoeboid invasive cells with high CD73-ROCK-Myosin II activity and their immunosuppressive microenvironment confer poor prognosis to patients. We propose targeting amoeboid PDAC cells as a therapeutic strategy.
Collapse
Affiliation(s)
- Remi Samain
- Centre for Tumour Microenvironment, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Oscar Maiques
- Centre for Tumour Microenvironment, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Joanne Monger
- Centre for Tumour Microenvironment, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Hoyin Lam
- School of Cancer and Pharmaceutical Sciences, Kings College London, London SE1 1UL, UK
- GSK, R&D Portfolio, Strategy and Business Insights, GSK House, 980 Great West Road, Brentford, TW8 9GS, UK
| | - Juliana Candido
- Centre for Tumour Microenvironment, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
- Oncology R&D, AstraZeneca, Cambridge CB21 6GH, UK
| | - Samantha George
- Centre for Tumour Microenvironment, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Nicola Ferrari
- Tumour Microenvironment Team, The Institute of Cancer Research, 237 Fulham Road, London SW3 6JB, UK
- Translational Science and Experimental Medicine, Early Respiratory and Immunology, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Leonie KohIhammer
- Centre for Tumour Microenvironment, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Sophia Lunetto
- Centre for Cell Biology and Cutaneous Research, Blizard Institute, Queen Mary University of London, London E1 2AT, UK
| | - Adrian Varela
- Centre for Tumour Microenvironment, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Jose L. Orgaz
- Centre for Tumour Microenvironment, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
- Instituto de Investigaciones Biomédicas Sols-Morreale CSIC-UAM, 28029 Madrid, Spain
| | - Felip Vilardell
- Department of Pathology, University Hospital Arnau de Vilanova, University of Lleida, Lleida, Spain
| | - Jorge Juan Olsina
- Department of Surgery, University Hospital Arnau de Vilanova, University of Lleida, Lleida, Spain
| | - Xavier Matias-Guiu
- Department of Pathology, University Hospital Arnau de Vilanova, University of Lleida, Lleida, Spain
- IRBLLEIDA, IDIBELL, University Hospita of Bellvitge, CIBERONC, Lleida, Spain
| | - Debashis Sarker
- School of Cancer and Pharmaceutical Sciences, Kings College London, London SE1 1UL, UK
| | - Adrian Biddle
- Centre for Cell Biology and Cutaneous Research, Blizard Institute, Queen Mary University of London, London E1 2AT, UK
| | - Frances R. Balkwill
- Centre for Tumour Microenvironment, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Jim Eyles
- Oncology R&D, AstraZeneca, Cambridge CB21 6GH, UK
| | | | - Hemant M. Kocher
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
- Barts and the London HPB Centre, The Royal London Hospital, Barts Health NHS Trust, London, UK
| | - Fernando Calvo
- Tumour Microenvironment Team, The Institute of Cancer Research, 237 Fulham Road, London SW3 6JB, UK
- Instituto de Biomedicina y Biotecnologia de Cantabria, c/ Albert Einstein 22, E39011 Santander, Spain
| | - Claire M. Wells
- School of Cancer and Pharmaceutical Sciences, Kings College London, London SE1 1UL, UK
| | - Victoria Sanz-Moreno
- Centre for Tumour Microenvironment, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| |
Collapse
|
48
|
Nazari SS, Doyle AD, Bleck CKE, Yamada KM. Long Prehensile Protrusions Can Facilitate Cancer Cell Invasion through the Basement Membrane. Cells 2023; 12:2474. [PMID: 37887318 PMCID: PMC10605924 DOI: 10.3390/cells12202474] [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: 09/26/2023] [Revised: 10/13/2023] [Accepted: 10/16/2023] [Indexed: 10/28/2023] Open
Abstract
A basic process in cancer is the breaching of basement-membrane barriers to permit tissue invasion. Cancer cells can use proteases and physical mechanisms to produce initial holes in basement membranes, but how cells squeeze through this barrier into matrix environments is not well understood. We used a 3D invasion model consisting of cancer-cell spheroids encapsulated by a basement membrane and embedded in collagen to characterize the dynamic early steps in cancer-cell invasion across this barrier. We demonstrate that certain cancer cells extend exceptionally long (~30-100 μm) protrusions through basement membranes via actin and microtubule cytoskeletal function. These long protrusions use integrin adhesion and myosin II-based contractility to pull cells through the basement membrane for initial invasion. Concurrently, these long, organelle-rich protrusions pull surrounding collagen inward while propelling cancer cells outward through perforations in the basement-membrane barrier. These exceptionally long, contractile cellular protrusions can facilitate the breaching of the basement-membrane barrier as a first step in cancer metastasis.
Collapse
Affiliation(s)
- Shayan S. Nazari
- Cell Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Andrew D. Doyle
- Cell Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Christopher K. E. Bleck
- Electron Microscopy Core, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Kenneth M. Yamada
- Cell Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
49
|
Tajaldini M, Poorkhani A, Amiriani T, Amiriani A, Javid H, Aref P, Ahmadi F, Sadani S, Khori V. Strategy of targeting the tumor microenvironment via inhibition of fibroblast/fibrosis remodeling new era to cancer chemo-immunotherapy resistance. Eur J Pharmacol 2023; 957:175991. [PMID: 37619785 DOI: 10.1016/j.ejphar.2023.175991] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Revised: 08/02/2023] [Accepted: 08/10/2023] [Indexed: 08/26/2023]
Abstract
The use of repurposing drugs that may have neoplastic and anticancer effects increases the efficiency and decrease resistance to chemotherapy drugs through a biochemical and mechanical transduction mechanisms through modulation of fibroblast/fibrosis remodeling in tumor microenvironment (TME). Interestingly, fibroblast/fibrosis remodeling plays a vital role in mediating cancer metastasis and drug resistance after immune chemotherapy. The most essential hypothesis for induction of chemo-immunotherapy resistance is via activation of fibroblast/fibrosis remodeling and preventing the infiltration of T cells after is mainly due to the interference between cytoskeleton, mechanical, biochemical, metabolic, vascular, and remodeling signaling pathways in TME. The structural components of the tumor that can be targeted in the fibroblast/fibrosis remodeling include the depletion of the TME components, targeting the cancer-associated fibroblasts and tumor associated macrophages, alleviating the mechanical stress within the ECM, and normalizing the blood vessels. It has also been found that during immune-chemotherapy, TME injury and fibroblast/fibrosis remodeling causes the up-regulation of inhibitory signals and down-regulation of activated signals, which results in immune escape or chemo-resistance of the tumor. In this regard, repurposing or neo-adjuvant drugs with various transduction signaling mechanisms, including anti-fibrotic effects, are used to target the TME and fibroblast/fibrosis signaling pathway such as angiotensin 2, transforming growth factor-beta, physical barriers of the TME, cytokines and metabolic factors which finally led to the reverse of the chemo-resistance. Consistent to many repurposing drugs such as pirfenidone, metformin, losartan, tranilast, dexamethasone and pentoxifylline are used to decrease immune-suppression by abrogation of TME inhibitory signal that stimulates the immune system and increases efficiency and reduces resistance to chemotherapy drugs. To overcome immunosuppression based on fibroblast/fibrosis remodeling, in this review, we focus on inhibitory signal transduction, which is the physical barrier, alleviates mechanical stress and prevents mechano-metabolic activation.
Collapse
Affiliation(s)
- Mahboubeh Tajaldini
- Ischemic Disorder Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Amirhoushang Poorkhani
- Ischemic Disorder Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Taghi Amiriani
- Ischemic Disorder Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Amirhossein Amiriani
- Ischemic Disorder Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Hossein Javid
- Department of Medical Laboratory Sciencess, Catastega Institue of Medical Sciences, Mashhad, Iran
| | - Parham Aref
- Ischemic Disorder Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Farahnazsadat Ahmadi
- Ischemic Disorder Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Somayeh Sadani
- Ischemic Disorder Research Center, Golestan University of Medical Sciences, Gorgan, Iran.
| | - Vahid Khori
- Ischemic Disorder Research Center, Golestan University of Medical Sciences, Gorgan, Iran.
| |
Collapse
|
50
|
Ildiz ES, Gvozdenovic A, Kovacs WJ, Aceto N. Travelling under pressure - hypoxia and shear stress in the metastatic journey. Clin Exp Metastasis 2023; 40:375-394. [PMID: 37490147 PMCID: PMC10495280 DOI: 10.1007/s10585-023-10224-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 07/05/2023] [Indexed: 07/26/2023]
Abstract
Cancer cell invasion, intravasation and survival in the bloodstream are early steps of the metastatic process, pivotal to enabling the spread of cancer to distant tissues. Circulating tumor cells (CTCs) represent a highly selected subpopulation of cancer cells that tamed these critical steps, and a better understanding of their biology and driving molecular principles may facilitate the development of novel tools to prevent metastasis. Here, we describe key research advances in this field, aiming at describing early metastasis-related processes such as collective invasion, shedding, and survival of CTCs in the bloodstream, paying particular attention to microenvironmental factors like hypoxia and mechanical stress, considered as important influencers of the metastatic journey.
Collapse
Affiliation(s)
- Ece Su Ildiz
- Department of Biology, Institute of Molecular Health Sciences, Swiss Federal Institute of Technology Zurich (ETH Zurich), Zurich, Switzerland
| | - Ana Gvozdenovic
- Department of Biology, Institute of Molecular Health Sciences, Swiss Federal Institute of Technology Zurich (ETH Zurich), Zurich, Switzerland
| | - Werner J Kovacs
- Department of Biology, Institute of Molecular Health Sciences, Swiss Federal Institute of Technology Zurich (ETH Zurich), Zurich, Switzerland
| | - Nicola Aceto
- Department of Biology, Institute of Molecular Health Sciences, Swiss Federal Institute of Technology Zurich (ETH Zurich), Zurich, Switzerland.
| |
Collapse
|