1
|
Götze J, Meißner K, Pereira-Veiga T, Belloum Y, Schneegans S, Kropidlowski J, Gorgulho J, Busch A, Honselmann KC, Schönrock M, Putscher A, Peine S, Nitschke C, Simon R, Spindler V, Izbicki JR, Hackert T, Bokemeyer C, Pantel K, Uzunoglu FG, Sinn M, Harriet Wikman. Identification and characterization of tumor and stromal derived liquid biopsy analytes in pancreatic ductal adenocarcinoma. J Exp Clin Cancer Res 2025; 44:14. [PMID: 39815324 PMCID: PMC11737273 DOI: 10.1186/s13046-024-03262-x] [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: 09/05/2024] [Accepted: 12/19/2024] [Indexed: 01/18/2025] Open
Abstract
BACKGROUND The lack of predictive biomarkers contributes notably to the poor outcomes of patients with pancreatic ductal adenocarcinoma (PDAC). Cancer-associated fibroblasts (CAFs) are the key components of the prominent PDAC stroma. Data on clinical relevance of CAFs entering the bloodstream, known as circulating CAFs (cCAFs) are scarce. Here, we developed a combined liquid biopsy assay to detect cCAFs and circulating tumor cells (CTCs) in metastatic PDAC (mPDAC) and other metastatic gastrointestinal malignancies (mGI). In addition, we evaluated plasma hyaluronan (HA) levels as a complementary surrogate biomarker of the stromal extent in patients with PDAC. METHODS A sequential liquid biopsy assay based on a two step-enrichment, combining marker dependent and independent cell enrichment, was established for cCAF and CTC detection and validated in mPDAC and mGI patients. The enriched cells were identified by multiplex immunofluorescence. HA measurement was performed by ELISA on blood samples from healthy blood donors (HD), localized and late-stage PDAC patients. RESULTS cCAFs (≥ 1cCAFs/7.5 mL blood) were detected in 95.4% of mPDAC and in 78.2% of mGI patients, with significantly higher numbers in mPDAC compared to mGI patients (mean number 22.7 vs. 11.0; P = 0.0318). mPDAC patients with ≥ 15 cCAFs/7.5 mL blood had a significant shorter median overall survival (mOS 3.2 months (95% confidence interval (CI) 0.801-5.855) vs. 14.2 months (95% CI 6.055-22.332); P = 0.013), whereby CTC levels were not associated with mOS. In mGI neither cCAFs nor CTCs had a significant impact on OS. HA plasma levels in mPDAC patients were significantly higher compared to HD (mean 123.0 ng/mL vs. 74.45 ng/mL, P = 0.015). High HA in localized and late-stage PDAC were associated with a significantly shorter mOS (mOSlocalized PDAC: 12.6 months vs. 23.5 months (P = 0.008); mOSmPDAC: 1.8 months vs. 5.3 months (P = 0.004)). CONCLUSIONS Our liquid biopsy assay provides robust detection of cCAFs in mPDAC and mGI patients. The measurement of both circulatory stromal parameters, cCAFs and HA, adds valuable clinical information as they are associated with an unfavorable outcome in PDAC. These results highlight that stromal characteristics unique to PDAC could be leveraged to fill the current gap in discovering predictive biomarkers.
Collapse
Affiliation(s)
- Julian Götze
- Department of Tumor Biology, Center of Experimental Medicine, University Medical Center Hamburg-Eppendorf, Martinistr, 52, 20248, Hamburg, Germany.
- Department of Oncology, Hematology and Bone Marrow Transplantation With Section Pneumology, University Cancer Center Hamburg, Martinistr, 52, 20248, Hamburg, Germany.
| | - Kira Meißner
- Department of Tumor Biology, Center of Experimental Medicine, University Medical Center Hamburg-Eppendorf, Martinistr, 52, 20248, Hamburg, Germany
| | - Thais Pereira-Veiga
- Department of Tumor Biology, Center of Experimental Medicine, University Medical Center Hamburg-Eppendorf, Martinistr, 52, 20248, Hamburg, Germany
| | - Yassine Belloum
- Department of Tumor Biology, Center of Experimental Medicine, University Medical Center Hamburg-Eppendorf, Martinistr, 52, 20248, Hamburg, Germany
| | - Svenja Schneegans
- Department of Tumor Biology, Center of Experimental Medicine, University Medical Center Hamburg-Eppendorf, Martinistr, 52, 20248, Hamburg, Germany
| | - Jolanthe Kropidlowski
- Department of Tumor Biology, Center of Experimental Medicine, University Medical Center Hamburg-Eppendorf, Martinistr, 52, 20248, Hamburg, Germany
| | - Joao Gorgulho
- Department of Oncology, Hematology and Bone Marrow Transplantation With Section Pneumology, University Cancer Center Hamburg, Martinistr, 52, 20248, Hamburg, Germany
| | - Alina Busch
- Department of Oncology, Hematology and Bone Marrow Transplantation With Section Pneumology, University Cancer Center Hamburg, Martinistr, 52, 20248, Hamburg, Germany
| | - Kim Christin Honselmann
- Department of Surgery, University Medical Center Schleswig-Holstein, Campus Luebeck, Luebeck, Germany
| | - Martin Schönrock
- Department of Oncology, Hematology and Bone Marrow Transplantation With Section Pneumology, University Cancer Center Hamburg, Martinistr, 52, 20248, Hamburg, Germany
| | - Arne Putscher
- Department of Tumor Biology, Center of Experimental Medicine, University Medical Center Hamburg-Eppendorf, Martinistr, 52, 20248, Hamburg, Germany
| | - Sven Peine
- Department of Transfusion Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christine Nitschke
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Mildred Scheel Cancer Career Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ronald Simon
- Department of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Volker Spindler
- Institute of Anatomy and Experimental Morphology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jakob Robert Izbicki
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Thilo Hackert
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Carsten Bokemeyer
- Department of Oncology, Hematology and Bone Marrow Transplantation With Section Pneumology, University Cancer Center Hamburg, Martinistr, 52, 20248, Hamburg, Germany
| | - Klaus Pantel
- Department of Tumor Biology, Center of Experimental Medicine, University Medical Center Hamburg-Eppendorf, Martinistr, 52, 20248, Hamburg, Germany
| | - Faik Güntaç Uzunoglu
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Marianne Sinn
- Department of Oncology, Hematology and Bone Marrow Transplantation With Section Pneumology, University Cancer Center Hamburg, Martinistr, 52, 20248, Hamburg, Germany.
| | - Harriet Wikman
- Department of Tumor Biology, Center of Experimental Medicine, University Medical Center Hamburg-Eppendorf, Martinistr, 52, 20248, Hamburg, Germany.
| |
Collapse
|
2
|
Shahhosseini R, Pakmehr S, Elhami A, Shakir MN, Alzahrani AA, Al-Hamdani MM, Abosoda M, Alsalamy A, Mohammadi-Dehcheshmeh M, Maleki TE, Saffarfar H, Ali-Khiavi P. Current biological implications and clinical relevance of metastatic circulating tumor cells. Clin Exp Med 2024; 25:7. [PMID: 39546080 PMCID: PMC11567993 DOI: 10.1007/s10238-024-01518-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 11/04/2024] [Indexed: 11/17/2024]
Abstract
Metastatic disease and cancer recurrence are the primary causes of cancer-related deaths. Circulating tumor cells (CTCs) and disseminated tumor cells (DTCs) are the driving forces behind the spread of cancer cells. The emergence and development of liquid biopsy using rare CTCs as a minimally invasive strategy for early-stage tumor detection and improved tumor management is a promising advancement in recent years. However, before blood sample analysis and clinical translation, precise isolation of CTCs from patients' blood based on their biophysical properties, followed by molecular identification of CTCs using single-cell multi-omics technologies is necessary to understand tumor heterogeneity and provide effective diagnosis and monitoring of cancer progression. Additionally, understanding the origin, morphological variation, and interaction between CTCs and the primary and metastatic tumor niche, as well as and regulatory immune cells, will offer new insights into the development of CTC-based advanced tumor targeting in the future clinical trials.
Collapse
Affiliation(s)
| | - SeyedAbbas Pakmehr
- School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
- Ahvaz Jundishapur University of Medical Sciences Ahvaz, Ahvaz, Iran
| | - Anis Elhami
- School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Maha Noori Shakir
- Department of Medical Laboratories Technology, AL-Nisour University College, Baghdad, Iraq
| | | | | | - Munther Abosoda
- College of Pharmacy, The Islamic University, Najaf, Iraq
- College of Pharmacy, The Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- College of Pharmacy, The Islamic University of Babylon, Babylon, Iraq
| | - Ali Alsalamy
- College of Pharmacy, Imam Ja'afar Al-Sadiq University, Al-Samawa, Al-Muthanna, 66002, Iraq
| | | | | | - Hossein Saffarfar
- Cardiovascular Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| | - Payam Ali-Khiavi
- Medical Faculty, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
3
|
Matsuoka T, Yashiro M. Molecular Mechanism for Malignant Progression of Gastric Cancer Within the Tumor Microenvironment. Int J Mol Sci 2024; 25:11735. [PMID: 39519285 PMCID: PMC11546171 DOI: 10.3390/ijms252111735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 10/23/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024] Open
Abstract
Gastric cancer (GC) is one of the most common cancers worldwide. Most patients are diagnosed at the progressive stage of GC, and progress in the development of effective anti-GC drugs has been insufficient. The tumor microenvironment (TME) regulates various functions of tumor cells, and interactions between the cellular and molecular components of the TME-e.g., inflammatory cells, fibroblasts, vasculature cells, and innate and adaptive immune cells-promote the aggressiveness of cancer cells and dissemination to distant organs. This review summarizes the roles of various TME cells and molecules in regulating the malignant progression and metastasis of GC. We also address the important roles of signaling pathways in mediating the interaction between cancer cells and the different components of the GC TME. Finally, we discuss the implications of these molecular mechanisms for developing novel and effective therapies targeting molecular and cellular components of the GC TME to control the malignant progression of GC.
Collapse
Affiliation(s)
- Tasuku Matsuoka
- Department of Molecular Oncology and Therapeutics, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahi-machi, Abeno-ku, Osaka 5458585, Japan;
- Institute of Medical Genetics, Osaka Metropolitan University, 1-4-3 Asahi-machi, Abeno-ku, Osaka 5458585, Japan
| | - Masakazu Yashiro
- Department of Molecular Oncology and Therapeutics, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahi-machi, Abeno-ku, Osaka 5458585, Japan;
- Institute of Medical Genetics, Osaka Metropolitan University, 1-4-3 Asahi-machi, Abeno-ku, Osaka 5458585, Japan
| |
Collapse
|
4
|
Fabisiewicz A, Szostakowska-Rodzos M, Grzybowska EA. Improving the Prognostic and Predictive Value of Circulating Tumor Cell Enumeration: Is Longitudinal Monitoring the Answer? Int J Mol Sci 2024; 25:10612. [PMID: 39408942 PMCID: PMC11476589 DOI: 10.3390/ijms251910612] [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: 09/27/2024] [Accepted: 09/30/2024] [Indexed: 10/20/2024] Open
Abstract
Circulating tumor cell (CTC) numbers in the blood of cancer patients can indicate the progression and invasiveness of tumors, and their prognostic and predictive value has been repeatedly demonstrated. However, the standard baseline CTC count at the beginning of treatment, while informative, is not completely reliable and may not adequately reflect the state of the disease. A growing number of studies indicate that the long-term monitoring of CTC numbers in the same patient provides more comprehensive prognostic data and should be incorporated into clinical practice, as a factor that contributes to therapeutic decisions. This review describes the current status of CTC enumeration as a prognostic and predictive factor, highlights the shortcomings of current solutions, and advocates for longitudinal CTC analysis as a more effective method of the evaluation of developing disease, treatment efficacy, and the long term-monitoring of the minimal residual disease. As evidenced by the described reports, the longitudinal monitoring of CTCs should provide a better and more sensitive prediction of the course of the disease, and its incorporation in clinical practice should be beneficial.
Collapse
Affiliation(s)
| | | | - Ewa A. Grzybowska
- Department of Molecular and Translational Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, Roentgena 5, 02-781 Warsaw, Poland; (A.F.); (M.S.-R.)
| |
Collapse
|
5
|
Reese KL, Pantel K, Smit DJ. Multibiomarker panels in liquid biopsy for early detection of pancreatic cancer - a comprehensive review. J Exp Clin Cancer Res 2024; 43:250. [PMID: 39218911 PMCID: PMC11367781 DOI: 10.1186/s13046-024-03166-w] [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: 03/18/2024] [Accepted: 08/16/2024] [Indexed: 09/04/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is frequently detected in late stages, which leads to limited therapeutic options and a dismal overall survival rate. To date, no robust method for the detection of early-stage PDAC that can be used for targeted screening approaches is available. Liquid biopsy allows the minimally invasive collection of body fluids (typically peripheral blood) and the subsequent analysis of circulating tumor cells or tumor-associated molecules such as nucleic acids, proteins, or metabolites that may be useful for the early diagnosis of PDAC. Single biomarkers may lack sensitivity and/or specificity to reliably detect PDAC, while combinations of these circulating biomarkers in multimarker panels may improve the sensitivity and specificity of blood test-based diagnosis. In this narrative review, we present an overview of different liquid biopsy biomarkers for the early diagnosis of PDAC and discuss the validity of multimarker panels.
Collapse
Affiliation(s)
- Kim-Lea Reese
- Institute of Tumor Biology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, Hamburg, 20246, Germany
| | - Klaus Pantel
- Institute of Tumor Biology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, Hamburg, 20246, Germany.
| | - Daniel J Smit
- Institute of Tumor Biology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, Hamburg, 20246, Germany.
| |
Collapse
|
6
|
Lopez-Cavestany M, Wright OA, Reckhorn NT, Carter AT, Jayawardana K, Nguyen T, Briggs DP, Koktysh DS, Esteban Linares A, Li D, King MR. Superhydrophobic Array Devices for the Enhanced Formation of 3D Cancer Models. ACS NANO 2024; 18:23637-23654. [PMID: 39150223 PMCID: PMC11363216 DOI: 10.1021/acsnano.4c08132] [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: 06/18/2024] [Revised: 08/07/2024] [Accepted: 08/08/2024] [Indexed: 08/17/2024]
Abstract
During the metastatic cascade, cancer cells travel through the bloodstream as circulating tumor cells (CTCs) to a secondary site. Clustered CTCs have greater shear stress and treatment resistance, yet their biology remains poorly understood. We therefore engineered a tunable superhydrophobic array device (SHArD). The SHArD-C was applied to culture a clinically relevant model of CTC clusters. Using our device, we cultured a model of cancer cell aggregates of various sizes with immortalized cancer cell lines. These exhibited higher E-cadherin expression and are significantly more capable of surviving high fluid shear stress-related forces compared to single cells and model clusters grown using the control method, helping to explain why clustering may provide a metastatic advantage. Additionally, the SHArD-S, when compared with the AggreWell 800 method, provides a more consistent spheroid-forming device culturing reproducible sizes of spheroids for multiple cancer cell lines. Overall, we designed, fabricated, and validated an easily tunable engineered device which grows physiologically relevant three-dimensional (3D) cancer models containing tens to thousands of cells.
Collapse
Affiliation(s)
- Maria Lopez-Cavestany
- Department
of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Olivia A. Wright
- Department
of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Noah T. Reckhorn
- Department
of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Alexandria T. Carter
- Department
of Bioengineering, Rice University, Houston, Texas 77030, United States
| | - Kalana Jayawardana
- Department
of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Tin Nguyen
- Department
of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Dayrl P. Briggs
- Center
for Nanophase Materials Science, Oak Ridge
National Laboratories, Knoxville, Tennessee 37830, United States
| | - Dmitry S. Koktysh
- Vanderbilt
Institute for Nanoscale Science and Engineering, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Alberto Esteban Linares
- Department
of Mechanical Engineering, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Deyu Li
- Department
of Mechanical Engineering, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Michael R. King
- Department
of Bioengineering, Rice University, Houston, Texas 77030, United States
| |
Collapse
|
7
|
Deo A, Sleeman JP, Shaked Y. The role of host response to chemotherapy: resistance, metastasis and clinical implications. Clin Exp Metastasis 2024; 41:495-507. [PMID: 37999904 DOI: 10.1007/s10585-023-10243-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Accepted: 11/06/2023] [Indexed: 11/25/2023]
Abstract
Chemotherapy remains the primary treatment for most metastatic cancers. However, the response to chemotherapy and targeted agents is often transient, and concurrent development of resistance is the primary impediment to effective cancer therapy. Strategies to overcome resistance to treatment have focused on cancer cell intrinsic factors and the tumor microenvironment (TME). Recent evidence indicates that systemic chemotherapy has a significant impact on the host that either facilitates tumor growth, allowing metastatic spread, or renders treatment ineffective. These host responses include the release of bone marrow-derived cells, activation of stromal cells in the TME, and induction of different molecular effectors. Here, we provide an overview of chemotherapy-induced systemic host responses that support tumor aggressiveness and metastasis, and which contribute to therapy resistance. Studying host responses to chemotherapy provides a solid basis for the development of adjuvant strategies to improve treatment outcomes and delay resistance to chemotherapy. This review discusses the emerging field of host response to cancer therapy, and its preclinical and potential clinical implications, explaining how under certain circumstances, these host effects contribute to metastasis and resistance to chemotherapy.
Collapse
Affiliation(s)
- Abhilash Deo
- Department of Cell Biology and Cancer Science, Rappaport Technion Integrated Cancer Center, Technion - Israel Institute of Technology, Haifa, Israel
| | - Jonathan P Sleeman
- European Centre for Angioscience (ECAS), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
- Karlsruhe Institute for Technology (KIT), IBCS-BIP, Campus Nord, 76344, Eggenstein- Leopoldshafen, Germany
| | - Yuval Shaked
- Department of Cell Biology and Cancer Science, Rappaport Technion Integrated Cancer Center, Technion - Israel Institute of Technology, Haifa, Israel.
| |
Collapse
|
8
|
Wiegmans AP, Ivanova E, Naei VY, Monkman J, Fletcher J, Mullally W, Warkiani ME, O'Byrne K, Kulasinghe A. Poor patient outcome correlates with active engulfment of cytokeratin positive CTCs within cancer-associated monocyte population in lung cancer. Clin Exp Metastasis 2024; 41:219-228. [PMID: 38416302 PMCID: PMC11213738 DOI: 10.1007/s10585-024-10270-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Accepted: 01/18/2024] [Indexed: 02/29/2024]
Abstract
High rates of mortality in non-small cell lung cancer lung cancer is due to inherent and acquired resistance to systemic therapies and subsequent metastatic burden. Metastasis is supported by suppression of the immune system at secondary organs and within the circulation. Modulation of the immune system is now being exploited as a therapeutic target with immune checkpoint inhibitors. The tracking of therapeutic efficacy in a real-time can be achieved with liquid biopsy, and evaluation of circulating tumour cells and the associated immune cells. A stable liquid biopsy biomarker for non-small cell lung cancer lung cancer has yet to be approved for clinical use. We performed a cross-sectional single-site study, and collected liquid biopsies from patients diagnosed with early, locally advanced, or metastatic lung cancer, undergoing surgery, or systemic therapy (chemotherapy/checkpoint inhibitors). Evaluation of overall circulating tumour cell counts, or cluster counts did not correlate with patient outcome. Interestingly, the numbers of Pan cytokeratin positive circulating tumour cells engulfed by tumour associated monocytes correlated strongly with patient outcome independent of circulating tumour cell counts and the use of checkpoint inhibitors. We suggest that Pan cytokeratin staining within monocytes is an important indicator of tumour-associated inflammation post-therapy and an effective biomarker with strong prognostic capability for patient outcome.
Collapse
Affiliation(s)
- A P Wiegmans
- Cancer and Ageing Research Program, Centre for Genomics and Personalised Health, Queensland University of Technology, Woolloongabba, QLD, 4102, Australia
- Princess Alexandra Hospital, Oncology, Woolloongabba, QLD, 4102, Australia
| | - E Ivanova
- Cancer and Ageing Research Program, Centre for Genomics and Personalised Health, Queensland University of Technology, Woolloongabba, QLD, 4102, Australia
| | - V Y Naei
- School of Biomedical Engineering, University of Technology Sydney, Sydney, Australia
- Faculty of Medicine, Frazer Institute, The University of Queensland, Brisbane, QLD, Australia
| | - J Monkman
- Faculty of Medicine, Frazer Institute, The University of Queensland, Brisbane, QLD, Australia
| | - J Fletcher
- Princess Alexandra Hospital, Oncology, Woolloongabba, QLD, 4102, Australia
| | - W Mullally
- Princess Alexandra Hospital, Oncology, Woolloongabba, QLD, 4102, Australia
| | - M E Warkiani
- School of Biomedical Engineering, University of Technology Sydney, Sydney, Australia
| | - K O'Byrne
- Cancer and Ageing Research Program, Centre for Genomics and Personalised Health, Queensland University of Technology, Woolloongabba, QLD, 4102, Australia
- Princess Alexandra Hospital, Oncology, Woolloongabba, QLD, 4102, Australia
| | - A Kulasinghe
- Faculty of Medicine, Frazer Institute, The University of Queensland, Brisbane, QLD, Australia.
| |
Collapse
|
9
|
Grayson KA, Greenlee JD, Himmel LE, Hapach LA, Reinhart-King CA, King MR. Spatial distribution of tumor-associated macrophages in an orthotopic prostate cancer mouse model. Pathol Oncol Res 2024; 30:1611586. [PMID: 38689823 PMCID: PMC11058651 DOI: 10.3389/pore.2024.1611586] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Accepted: 04/03/2024] [Indexed: 05/02/2024]
Abstract
Mounting evidence suggests that the immune landscape within prostate tumors influences progression, metastasis, treatment response, and patient outcomes. In this study, we investigated the spatial density of innate immune cell populations within NOD.SCID orthotopic prostate cancer xenografts following microinjection of human DU145 prostate cancer cells. Our laboratory has previously developed nanoscale liposomes that attach to leukocytes via conjugated E-selectin (ES) and kill cancer cells via TNF-related apoptosis inducing ligand (TRAIL). Immunohistochemistry (IHC) staining was performed on tumor samples to identify and quantify leukocyte infiltration for different periods of tumor growth and E-selectin/TRAIL (EST) liposome treatments. We examined the spatial-temporal dynamics of three different immune cell types infiltrating tumors using QuPath image analysis software. IHC staining revealed that F4/80+ tumor-associated macrophages (TAMs) were the most abundant immune cells in all groups, irrespective of time or treatment. The density of TAMs decreased over the course of tumor growth and decreased in response to EST liposome treatments. Intratumoral versus marginal analysis showed a greater presence of TAMs in the marginal regions at 3 weeks of tumor growth which became more evenly distributed over time and in tumors treated with EST liposomes. TUNEL staining indicated that EST liposomes significantly increased cell apoptosis in treated tumors. Additionally, confocal microscopy identified liposome-coated TAMs in both the core and periphery of tumors, highlighting the ability of liposomes to infiltrate tumors by "piggybacking" on macrophages. The results of this study indicate that TAMs represent the majority of innate immune cells within NOD.SCID orthotopic prostate tumors, and spatial density varies widely as a function of tumor size, duration of tumor growth, and treatment of EST liposomes.
Collapse
Affiliation(s)
- Korie A. Grayson
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, United States
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, United States
| | - Joshua D. Greenlee
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, United States
| | - Lauren E. Himmel
- Department of Pathology, Microbiology and Immunology, Translational Pathology Shared Resource, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Lauren A. Hapach
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, United States
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, United States
| | | | - Michael R. King
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, United States
| |
Collapse
|
10
|
Gayan S, Teli A, Sonawane A, Dey T. Impact of Chemotherapeutic Stress Depends on The Nature of Breast Cancer Spheroid and Induce Behavioral Plasticity to Resistant Population. Adv Biol (Weinh) 2024; 8:e2300271. [PMID: 38063815 DOI: 10.1002/adbi.202300271] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 11/20/2023] [Indexed: 04/15/2024]
Abstract
Cellular or tumor dormancy, identified recently as one of the main reasons behind post-therapy recurrence, can be caused by diverse reasons. Chemotherapy has recently been recognized as one of such reasons. However, in-depth studies of chemotherapy-induced dormancy are lacking due to the absence of an in vitro human-relevant model tailor-made for such a scenario. This report utilized multicellular breast cancer spheroid to create a primary platform for establishing a chemotherapy-induced dormancy model. It is observed that extreme chemotherapeutic stress affects invasive and non-invasive spheroids differently. Non-invasive spheroids exhibit more resilience and maintain viability and migrational ability, while invasive spheroids display heightened susceptibility and improved tumorigenic capacity. Heterogenous spheroids exhibit increased tumorigenic capacity while show minimal survival ability. Further probing of chemotherapeutically dormant spheroids is needed to understand the molecular mechanism and identify dormancy-related markers to achieve therapeutic success in the future.
Collapse
Affiliation(s)
- Sukanya Gayan
- Department of Biotechnology (merged with Institute of Bioinformatics and Biotechnology), Savitribai Phule Pune University, Pune, 411007, India
| | - Abhishek Teli
- Department of Biotechnology (merged with Institute of Bioinformatics and Biotechnology), Savitribai Phule Pune University, Pune, 411007, India
| | - Akshay Sonawane
- Department of Biotechnology (merged with Institute of Bioinformatics and Biotechnology), Savitribai Phule Pune University, Pune, 411007, India
| | - Tuli Dey
- Department of Biotechnology (merged with Institute of Bioinformatics and Biotechnology), Savitribai Phule Pune University, Pune, 411007, India
| |
Collapse
|
11
|
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
|
12
|
Nguyen HPQ, Bae WK, Park MS, Chung IJ, Nam TK, Jeong JU, Uong TNT, Cho D, Kim SK, Yoon M. Intensified NK cell therapy in combination with low-dose chemoradiotherapy against human colorectal cancer. Cancer Immunol Immunother 2023; 72:4089-4102. [PMID: 37801126 PMCID: PMC10992501 DOI: 10.1007/s00262-023-03545-w] [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: 08/04/2023] [Accepted: 09/07/2023] [Indexed: 10/07/2023]
Abstract
The therapeutic potential of adoptive natural Killer (NK) cells immunotherapy in combination with chemoradiotherapy, the main treatment modality for colorectal cancer (CRC), has not yet been explored. Here, we aimed to investigate the efficacy of NK cells to potentiate primary tumor control and improve survival outcomes, especially in combination with low-dose chemoradiotherapy. Ex vivo activated NK cells (> 90% purity) from healthy donors were obtained. NK cells were administered intravenously to the CRC-bearing mice and intensified in vivo in combination with low-dose 5-fluorouracil (0.5 mg/kg or 1 mg/Kg) and irradiated tumors with low doses (2 Gy or 4 Gy). Real-time NK cell cytotoxicity demonstrated a synergistic killing effect of a combination of low-dose chemoradiotherapy, mainly through NKp30 and NKG2D, showing a decrease in NK cell degranulation after blocking NKG2D and NKp30. In vivo tumor characteristics after combination treatment showed decreased CD112, CD155, MICA, and MICB expression. Under the combination strategy, 70% of the mice had free lung metastasis and 90% without secondary gross tumors, indicating suppressed distant metastasis to lung and axillary regions. This combination therapy resulted in significantly synergistic antitumor activity against primary solid tumors compared to chemoradiotherapy only. Furthermore, the intensified NK cell administration showed significantly better primary tumor control and survival outcomes than the non-intensified NK cell administration in a human colorectal HT-29 model treated with low-dose chemoradiotherapy. Optimized NK cell therapy combined with low-dose chemoradiotherapy can provide effective therapeutic potential for intractable cold human colorectal cancer.
Collapse
Affiliation(s)
- Huy Phuoc Quang Nguyen
- Department of Radiation Oncology, Chonnam National University Hwasun Hospital, Chonnam National University Medical School, Gwangju, Republic of Korea
- Department of Biomedical Science, Chonnam National University Graduate School, Gwangju, Republic of Korea
| | - Woo Kyun Bae
- Immunotherapy Innovation Center, Chonnam National University Medical School, Hwasun, Republic of Korea.
- Department of Hematology and Oncology, Chonnam National University Medical School and Hwasun Hospital, Hwasun, Republic of Korea.
| | - Myong Suk Park
- Department of Hematology and Oncology, Chonnam National University Medical School and Hwasun Hospital, Hwasun, Republic of Korea
| | - Ik-Joo Chung
- Immunotherapy Innovation Center, Chonnam National University Medical School, Hwasun, Republic of Korea
- Department of Hematology and Oncology, Chonnam National University Medical School and Hwasun Hospital, Hwasun, Republic of Korea
| | - Taek-Keun Nam
- Department of Radiation Oncology, Chonnam National University Hwasun Hospital, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Jae-Uk Jeong
- Department of Radiation Oncology, Chonnam National University Hwasun Hospital, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Tung Nguyen Thanh Uong
- Department of Radiation Oncology, Chonnam National University Hwasun Hospital, Chonnam National University Medical School, Gwangju, Republic of Korea
- Department of Biomedical Science, Chonnam National University Graduate School, Gwangju, Republic of Korea
| | - Duck Cho
- Department of Laboratory Medicine and Genetics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Sang-Ki Kim
- Department of Companion and Laboratory Animal Science, Kongju National University, Yesan, Republic of Korea
| | - Meesun Yoon
- Department of Radiation Oncology, Chonnam National University Hwasun Hospital, Chonnam National University Medical School, Gwangju, Republic of Korea.
- Department of Biomedical Science, Chonnam National University Graduate School, Gwangju, Republic of Korea.
- Immunotherapy Innovation Center, Chonnam National University Medical School, Hwasun, Republic of Korea.
| |
Collapse
|
13
|
Ito Y, Kobuchi S, Kawakita A, Tosaka K, Matsunaga Y, Yoshioka S, Jonan S, Amagase K, Hashimoto K, Kanda M, Saito T, Nakanishi H. Mobilization of Circulating Tumor Cells after Short- and Long-Term FOLFIRINOX and GEM/nab-PTX Chemotherapy in Xenograft Mouse Models of Human Pancreatic Cancer. Cancers (Basel) 2023; 15:5482. [PMID: 38001741 PMCID: PMC10670901 DOI: 10.3390/cancers15225482] [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: 10/23/2023] [Revised: 11/15/2023] [Accepted: 11/17/2023] [Indexed: 11/26/2023] Open
Abstract
Mobilization of CTCs after various types of therapy, such as radiation therapy, has been reported, but systematic study of CTCs after chemotherapy remained quite limited. In this study, we sequentially examined CTC numbers after single-dose and repetitive-dose chemotherapy, including FORFIRINOX (FFX) and Gemcitabine and nab-Paclitaxel (GnP) using two pancreatic cancer xenograft models. CTC was detected by the immunocytology-based microfluidic platform. We further examined the dynamic change in the histology of primary tumor tissues during chemotherapy. We confirmed a transient increase in CTCs 1-2 weeks after single-dose and repetitive-dose of FFX/GnP chemotherapy. Histological examination of the primary tumors revealed that the peak period of CTC at 1-2 weeks after chemotherapy corresponded to the maximal destructive phase consisting of cell cycle arrest, apoptosis of tumor cells, and blood vessel destruction without secondary reparative tissue reactions and regeneration of tumor cells. These findings indicate that mobilization of CTCs early after chemotherapy is mediated by the shedding of degenerated tumor cells into the disrupted blood vessels driven by the pure destructive histological changes in primary tumor tissues. These results suggest that sequential CTC monitoring during chemotherapy can be a useful liquid biopsy diagnostic tool to predict tumor chemosensitivity and resistance in preclinical and clinical settings.
Collapse
Affiliation(s)
- Yukako Ito
- Department of Pharmacokinetics, Kyoto Pharmaceutical University, Kyoto 607-8414, Japan; (S.K.); (A.K.); (K.T.); (Y.M.); (S.Y.)
| | - Shinji Kobuchi
- Department of Pharmacokinetics, Kyoto Pharmaceutical University, Kyoto 607-8414, Japan; (S.K.); (A.K.); (K.T.); (Y.M.); (S.Y.)
| | - Amiri Kawakita
- Department of Pharmacokinetics, Kyoto Pharmaceutical University, Kyoto 607-8414, Japan; (S.K.); (A.K.); (K.T.); (Y.M.); (S.Y.)
| | - Kazuki Tosaka
- Department of Pharmacokinetics, Kyoto Pharmaceutical University, Kyoto 607-8414, Japan; (S.K.); (A.K.); (K.T.); (Y.M.); (S.Y.)
| | - Yume Matsunaga
- Department of Pharmacokinetics, Kyoto Pharmaceutical University, Kyoto 607-8414, Japan; (S.K.); (A.K.); (K.T.); (Y.M.); (S.Y.)
| | - Shoma Yoshioka
- Department of Pharmacokinetics, Kyoto Pharmaceutical University, Kyoto 607-8414, Japan; (S.K.); (A.K.); (K.T.); (Y.M.); (S.Y.)
| | - Shizuka Jonan
- Department of Pharmacy, College of Pharmaceutical Sciences, Ritsumeikan University, Shiga 603-8577, Japan; (S.J.); (K.A.)
| | - Kikuko Amagase
- Department of Pharmacy, College of Pharmaceutical Sciences, Ritsumeikan University, Shiga 603-8577, Japan; (S.J.); (K.A.)
| | - Katsunori Hashimoto
- Department of Medical Technology, Faculty of Medical Sciences, Shubun University, Ichinomiya City 491-0938, Japan;
| | - Mitsuro Kanda
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan;
| | - Takuya Saito
- Department of Gastroenterological Surgery, Aichi Medical University, Nagakute City 480-1195, Japan; (T.S.); (H.N.)
| | - Hayao Nakanishi
- Department of Gastroenterological Surgery, Aichi Medical University, Nagakute City 480-1195, Japan; (T.S.); (H.N.)
- Laboratory of Pathology, Okazaki City Hospital, Okazaki 444-0002, Japan
| |
Collapse
|
14
|
Abraham A, Virdi S, Herrero N, Bryant I, Nwakama C, Jacob M, Khaparde G, Jordan D, McCuddin M, McKinley S, Taylor A, Peeples C, Ekpenyong A. Microfluidic Microcirculation Mimetic for Exploring Biophysical Mechanisms of Chemotherapy-Induced Metastasis. MICROMACHINES 2023; 14:1653. [PMID: 37763816 PMCID: PMC10536821 DOI: 10.3390/mi14091653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 08/18/2023] [Accepted: 08/19/2023] [Indexed: 09/29/2023]
Abstract
There is rapidly emerging evidence from pre-clinical studies, patient samples and patient subpopulations that certain chemotherapeutics inadvertently produce prometastatic effects. Prior to this, we showed that doxorubicin and daunorubicin stiffen cells before causing cell death, predisposing the cells to clogging and extravasation, the latter being a step in metastasis. Here, we investigate which other anti-cancer drugs might have similar prometastatic effects by altering the biophysical properties of cells. We treated myelogenous (K562) leukemic cancer cells with the drugs nocodazole and hydroxyurea and then measured their mechanical properties using a microfluidic microcirculation mimetic (MMM) device, which mimics aspects of blood circulation and enables the measurement of cell mechanical properties via transit times through the device. We also quantified the morphological properties of cells to explore biophysical mechanisms underlying the MMM results. Results from MMM measurements show that nocodazole- and hydroxyurea-treated K562 cells exhibit significantly altered transit times. Nocodazole caused a significant (p < 0.01) increase in transit times, implying a stiffening of cells. This work shows the feasibility of using an MMM to explore possible biophysical mechanisms that might contribute to chemotherapy-induced metastasis. Our work also suggests cell mechanics as a therapeutic target for much needed antimetastatic strategies in general.
Collapse
Affiliation(s)
- Ashley Abraham
- Biology Department, Creighton University, Omaha, NE 68178, USA; (A.A.); (N.H.); (M.J.); (G.K.); (D.J.); (M.M.); (S.M.); (A.T.)
| | - Sukhman Virdi
- Physics Department, Creighton University, Omaha, NE 68178, USA; (S.V.); (I.B.); (C.P.)
| | - Nick Herrero
- Biology Department, Creighton University, Omaha, NE 68178, USA; (A.A.); (N.H.); (M.J.); (G.K.); (D.J.); (M.M.); (S.M.); (A.T.)
| | - Israel Bryant
- Physics Department, Creighton University, Omaha, NE 68178, USA; (S.V.); (I.B.); (C.P.)
| | - Chisom Nwakama
- Chemistry Department, Creighton University, Omaha, NE 68178, USA;
| | - Megha Jacob
- Biology Department, Creighton University, Omaha, NE 68178, USA; (A.A.); (N.H.); (M.J.); (G.K.); (D.J.); (M.M.); (S.M.); (A.T.)
| | - Gargee Khaparde
- Biology Department, Creighton University, Omaha, NE 68178, USA; (A.A.); (N.H.); (M.J.); (G.K.); (D.J.); (M.M.); (S.M.); (A.T.)
| | - Destiny Jordan
- Biology Department, Creighton University, Omaha, NE 68178, USA; (A.A.); (N.H.); (M.J.); (G.K.); (D.J.); (M.M.); (S.M.); (A.T.)
| | - Mackenzie McCuddin
- Biology Department, Creighton University, Omaha, NE 68178, USA; (A.A.); (N.H.); (M.J.); (G.K.); (D.J.); (M.M.); (S.M.); (A.T.)
| | - Spencer McKinley
- Biology Department, Creighton University, Omaha, NE 68178, USA; (A.A.); (N.H.); (M.J.); (G.K.); (D.J.); (M.M.); (S.M.); (A.T.)
| | - Adam Taylor
- Biology Department, Creighton University, Omaha, NE 68178, USA; (A.A.); (N.H.); (M.J.); (G.K.); (D.J.); (M.M.); (S.M.); (A.T.)
| | - Conner Peeples
- Physics Department, Creighton University, Omaha, NE 68178, USA; (S.V.); (I.B.); (C.P.)
| | - Andrew Ekpenyong
- Physics Department, Creighton University, Omaha, NE 68178, USA; (S.V.); (I.B.); (C.P.)
| |
Collapse
|
15
|
Muchlińska A, Wenta R, Ścińska W, Markiewicz A, Suchodolska G, Senkus E, Żaczek AJ, Bednarz-Knoll N. Improved Characterization of Circulating Tumor Cells and Cancer-Associated Fibroblasts in One-Tube Assay in Breast Cancer Patients Using Imaging Flow Cytometry. Cancers (Basel) 2023; 15:4169. [PMID: 37627197 PMCID: PMC10453498 DOI: 10.3390/cancers15164169] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 08/01/2023] [Accepted: 08/16/2023] [Indexed: 08/27/2023] Open
Abstract
Circulating tumor cells (CTCs) and circulating cancer-associated fibroblasts (cCAFs) have been individually considered strong indicators of cancer progression. However, technical limitations have prevented their simultaneous analysis in the context of CTC phenotypes different from epithelial. This study aimed to analyze CTCs and cCAFs simultaneously in the peripheral blood of 210 breast cancer patients using DAPI/pan-keratin (K)/vimentin (V)/alpha-SMA/CD29/CD45/CD31 immunofluorescent staining and novel technology-imaging flow cytometry (imFC). Single and clustered CTCs of different sizes and phenotypes (i.e., epithelial phenotype K+/V- and epithelial-mesenchymal transition (EMT)-related CTCs, such as K+/V+, K-/V+, and K-/V-) were detected in 27.6% of the samples and correlated with metastases. EMT-related CTCs interacted more frequently with normal cells and tended to occur in patients with tumors progressing during therapy, while cCAFs coincided with CTCs (mainly K+/V- and K-/V-) in seven (3.3%) patients and seemed to correlate with the presence of metastases, particularly visceral ones. This study emphasizes the advantages of imFC in the field of liquid biopsy and highlights the importance of multimarker-based analysis of different subpopulations and phenotypes of cancer progression-related cells, i.e., CTCs and cCAFs. The co-detection of CTCs and cCAFs might improve the identification of patients at higher risk of progression and their monitoring during therapy.
Collapse
Affiliation(s)
- Anna Muchlińska
- Laboratory of Translational Oncology, Intercollegiate Faculty of Biotechnology, University of Gdańsk and Medical University of Gdańsk, 80-211 Gdańsk, Poland
| | - Robert Wenta
- Laboratory of Translational Oncology, Intercollegiate Faculty of Biotechnology, University of Gdańsk and Medical University of Gdańsk, 80-211 Gdańsk, Poland
| | - Wiktoria Ścińska
- Laboratory of Translational Oncology, Intercollegiate Faculty of Biotechnology, University of Gdańsk and Medical University of Gdańsk, 80-211 Gdańsk, Poland
| | - Aleksandra Markiewicz
- Laboratory of Translational Oncology, Intercollegiate Faculty of Biotechnology, University of Gdańsk and Medical University of Gdańsk, 80-211 Gdańsk, Poland
| | - Grażyna Suchodolska
- Department of Oncology and Radiotherapy, Medical University of Gdańsk, 80-211 Gdańsk, Poland
| | - Elżbieta Senkus
- Department of Oncology and Radiotherapy, Medical University of Gdańsk, 80-211 Gdańsk, Poland
| | - Anna J. Żaczek
- Laboratory of Translational Oncology, Intercollegiate Faculty of Biotechnology, University of Gdańsk and Medical University of Gdańsk, 80-211 Gdańsk, Poland
| | - Natalia Bednarz-Knoll
- Laboratory of Translational Oncology, Intercollegiate Faculty of Biotechnology, University of Gdańsk and Medical University of Gdańsk, 80-211 Gdańsk, Poland
| |
Collapse
|
16
|
Bates ME, Libring S, Reinhart-King CA. Forces exerted and transduced by cancer-associated fibroblasts during cancer progression. Biol Cell 2023; 115:e2200104. [PMID: 37224184 PMCID: PMC10757454 DOI: 10.1111/boc.202200104] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 05/13/2023] [Accepted: 05/22/2023] [Indexed: 05/26/2023]
Abstract
Although it is well-known that cancer-associated fibroblasts (CAFs) play a key role in regulating tumor progression, the effects of mechanical tissue changes on CAFs are understudied. Myofibroblastic CAFs (myCAFs), in particular, are known to alter tumor matrix architecture and composition, heavily influencing the mechanical forces in the tumor microenvironment (TME), but much less is known about how these mechanical changes initiate and maintain the myCAF phenotype. Additionally, recent studies have pointed to the existence of CAFs in circulating tumor cell clusters, indicating that CAFs may be subject to mechanical forces beyond the primary TME. Due to their pivotal role in cancer progression, targeting CAF mechanical regulation may provide therapeutic benefit. Here, we will discuss current knowledge and summarize existing gaps in how CAFs regulate and are regulated by matrix mechanics, including through stiffness, solid and fluid stresses, and fluid shear stress.
Collapse
Affiliation(s)
- Madison E Bates
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, USA
| | - Sarah Libring
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, USA
| | | |
Collapse
|
17
|
Xia J, Zhang J, Xiong Y, Zhao J, Zhou Y, Jiang T, Zhu J. Circulating tumor DNA minimal residual disease in clinical practice of non-small cell lung cancer. Expert Rev Mol Diagn 2023; 23:913-924. [PMID: 37702546 DOI: 10.1080/14737159.2023.2252334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 08/23/2023] [Indexed: 09/14/2023]
Abstract
INTRODUCTION The advance of diagnostics and treatments has greatly improved the prognosis of non-small cell lung cancer (NSCLC) patients. However, relapse and metastasis are still common problems encountered by NSCLC patients who have achieved complete remission. Therefore, overcoming the challenge of relapse and metastasis is particularly important for improving the prognosis of NSCLC patients. Research has shown that minimal residual disease (MRD) was a potential source of tumor relapse and metastasis, and circulating tumor DNA (ctDNA) MRD has obvious advantages in predicting the relapse and metastasis of NSCLC and evaluating treatment effectiveness. Therefore, dynamic monitoring of MRD is of great significance for NSCLC patient management strategies. AREAS COVERED We have reviewed articles related to NSCLC MRD included in PubMed and describes the biological significance and historical context of MRD research, reasons for using ctDNA to evaluate MRD, and potential value and challenges of ctDNA MRD in assessing relapse and metastasis of NSCLC, ultimately guiding clinical therapeutic strategies and management. EXPERT OPINION The standardized scope of ctDNA MRD detection for NSCLC requires more clinical research evidence to minimize study differences, making it possible to include in the clinical staging as a reliable indicator.
Collapse
Affiliation(s)
- Jinghua Xia
- Department of Thoracic Surgery, Tangdu Hospital, Air Force Medical University, Xi'an, China
| | - Jiao Zhang
- Department of Thoracic Surgery, Tangdu Hospital, Air Force Medical University, Xi'an, China
| | - Yanlu Xiong
- Department of Thoracic Surgery, Tangdu Hospital, Air Force Medical University, Xi'an, China
| | - Jinbo Zhao
- Department of Thoracic Surgery, Tangdu Hospital, Air Force Medical University, Xi'an, China
| | - Yinxi Zhou
- Department of Thoracic Surgery, Tangdu Hospital, Air Force Medical University, Xi'an, China
| | - Tao Jiang
- Department of Thoracic Surgery, Tangdu Hospital, Air Force Medical University, Xi'an, China
| | - Jianfei Zhu
- Department of Thoracic Surgery, Tangdu Hospital, Air Force Medical University, Xi'an, China
- Department of Thoracic Surgery, Shaanxi Provincial People's Hospital, Xi'an, China
| |
Collapse
|
18
|
Si H, Esquivel M, Mendoza Mendoza E, Roarty K. The covert symphony: cellular and molecular accomplices in breast cancer metastasis. Front Cell Dev Biol 2023; 11:1221784. [PMID: 37440925 PMCID: PMC10333702 DOI: 10.3389/fcell.2023.1221784] [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: 05/12/2023] [Accepted: 06/16/2023] [Indexed: 07/15/2023] Open
Abstract
Breast cancer has emerged as the most commonly diagnosed cancer and primary cause of cancer-related deaths among women worldwide. Although significant progress has been made in targeting the primary tumor, the effectiveness of systemic treatments to prevent metastasis remains limited. Metastatic disease continues to be the predominant factor leading to fatality in the majority of breast cancer patients. The existence of a prolonged latency period between initial treatment and eventual recurrence in certain patients indicates that tumors can both adapt to and interact with the systemic environment of the host, facilitating and sustaining the progression of the disease. In order to identify potential therapeutic interventions for metastasis, it will be crucial to gain a comprehensive framework surrounding the mechanisms driving the growth, survival, and spread of tumor cells, as well as their interaction with supporting cells of the microenvironment. This review aims to consolidate recent discoveries concerning critical aspects of breast cancer metastasis, encompassing the intricate network of cells, molecules, and physical factors that contribute to metastasis, as well as the molecular mechanisms governing cancer dormancy.
Collapse
Affiliation(s)
- Hongjiang Si
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States
| | - Madelyn Esquivel
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States
| | - Erika Mendoza Mendoza
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States
| | - Kevin Roarty
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX, United States
| |
Collapse
|
19
|
Zhang F, Li Q, Zhang Y, Li N, Rao M, Li S, Ai Z, Yan S, Tian Z. COPS3 inhibition promotes cell proliferation blockage and anoikis via regulating PFKFB3 in osteosarcoma cancer cells. Eur J Pharmacol 2023; 951:175799. [PMID: 37201626 DOI: 10.1016/j.ejphar.2023.175799] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 05/09/2023] [Accepted: 05/16/2023] [Indexed: 05/20/2023]
Abstract
As a key component of the COP9 signalosome complex, which participates in a variety of physiological processes, COPS3 is intimately related to multiple cancers. It promotes cell proliferation, progression and metastasis in several cancer cells. However, whether COPS3 participates in regulating anoikis, a specific kind of apoptosis and functions as an essential modulator of cell metastasis, has not yet been studied. Here, we found COPS3 is highly expressed in several cancers especially in osteosarcoma (OS). Overexpression of COPS3 promoted cell proliferation, cell viability and migration/invasion in both control cells and oxaliplatin (Oxa) treated cells. On the contrary, knockdown of COPS3 further enhanced the cytotoxicity of Oxa. Utilizing bioinformatics analysis, we found that COPS3 was higher expressed in the metastatic group, and associated with the extra-cellular matrix (ECM) receptor interaction pathway, which involve in regulating anoikis. In an anoikis model, COPS3 expression varied and genetic modification of COPS3 influenced the cell death enhanced by Oxa. PFKFB3, an essential modulator of glycolysis, was found to interact with COPS3. Inhibition of PFKFB3 promoted apoptosis and anoikis enhanced by Oxa, and COPS3 overexpression failed to rescue this cell death. On the contrary, in the COPS3 knockdown cells, overexpression of PFKFB3 recovered the anoikis resistance, indicating COPS3 function upstream of PFKFB3. In summary, our results elucidated that COPS3 modulated anoikis via affecting PFKFB3 in OS cancer cells.
Collapse
Affiliation(s)
- Fan Zhang
- Department of Bone and Soft Tissue Oncology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, China
| | - Qianqian Li
- Key Laboratory of Precision Oncology in Universities of Shandong, Institute of Precision Medicine, Jining Medical University, Jining, 272067, China
| | - Yaqin Zhang
- Key Laboratory of Precision Oncology in Universities of Shandong, Institute of Precision Medicine, Jining Medical University, Jining, 272067, China
| | - Na Li
- Key Laboratory of Precision Oncology in Universities of Shandong, Institute of Precision Medicine, Jining Medical University, Jining, 272067, China
| | - Mengjiao Rao
- Key Laboratory of Precision Oncology in Universities of Shandong, Institute of Precision Medicine, Jining Medical University, Jining, 272067, China
| | - Shi Li
- Key Laboratory of Precision Oncology in Universities of Shandong, Institute of Precision Medicine, Jining Medical University, Jining, 272067, China
| | - Zhiying Ai
- Key Laboratory of Precision Oncology in Universities of Shandong, Institute of Precision Medicine, Jining Medical University, Jining, 272067, China
| | - Siyuan Yan
- Key Laboratory of Precision Oncology in Universities of Shandong, Institute of Precision Medicine, Jining Medical University, Jining, 272067, China.
| | - Zhichao Tian
- Department of Bone and Soft Tissue Oncology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, China.
| |
Collapse
|
20
|
Parlani M, Jorgez C, Friedl P. Plasticity of cancer invasion and energy metabolism. Trends Cell Biol 2023; 33:388-402. [PMID: 36328835 PMCID: PMC10368441 DOI: 10.1016/j.tcb.2022.09.009] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 09/26/2022] [Accepted: 09/27/2022] [Indexed: 11/05/2022]
Abstract
Energy deprivation is a frequent adverse event in tumors that is caused by mutations, malperfusion, hypoxia, and nutrition deficit. The resulting bioenergetic stress leads to signaling and metabolic adaptation responses in tumor cells, secures survival, and adjusts migration activity. The kinetic responses of cancer cells to energy deficit were recently identified, including a switch of invasive cancer cells to energy-conservative amoeboid migration and an enhanced capability for distant metastasis. We review the energy programs employed by different cancer invasion modes including collective, mesenchymal, and amoeboid migration, as well as their interconversion in response to energy deprivation, and we discuss the consequences for metastatic escape. Understanding the energy requirements of amoeboid and other dissemination strategies offers rationales for improving therapeutic targeting of metastatic cancer progression.
Collapse
Affiliation(s)
- Maria Parlani
- Department of Cell Biology, Radboud University Medical Centre, Nijmegen 6525GA, The Netherlands
| | - Carolina Jorgez
- David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Peter Friedl
- Department of Cell Biology, Radboud University Medical Centre, Nijmegen 6525GA, The Netherlands; David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Cancer Genomics Center, 3584 CG Utrecht, The Netherlands.
| |
Collapse
|
21
|
Lu T, Oomens L, Terstappen LWMM, Prakash J. In Vivo Detection of Circulating Cancer-Associated Fibroblasts in Breast Tumor Mouse Xenograft: Impact of Tumor Stroma and Chemotherapy. Cancers (Basel) 2023; 15:cancers15041127. [PMID: 36831470 PMCID: PMC9954095 DOI: 10.3390/cancers15041127] [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: 11/30/2022] [Revised: 02/01/2023] [Accepted: 02/08/2023] [Indexed: 02/12/2023] Open
Abstract
Cancer-associated fibroblasts (CAFs) are important drivers in the tumor microenvironment and facilitate the growth and survival of tumor cells, as well as metastasis formation. They may travel together with tumor cells to support their survival and aid in the formation of a metastatic niche. In this study, we aimed to study circulating CAFs (cCAFs) and circulating tumor cells (CTCs) in a preclinical breast tumor model in mice in order to understand the effect of chemotherapy on cCAFs and CTC formation. Tumors with MDA-MB-231 human breast tumor cells with/without primary human mammary fibroblasts (representing CAFs) were coinjected in SCID mice to develop tumors. We found that the tumors with CAFs grew faster than tumors without CAFs. To study the effect of the stroma on CTCs and cCAFs, we isolated cells using microsieve filtration technology and established ITGA5 as a new cCAF biomarker, which showed good agreement with the CAF markers FAP and α-SMA. We found that ITGA5+ cCAFs shed in the blood of mice bearing stroma-rich coinjection-based tumors, while there was no difference in CTC formation. Although treatment with liposomal doxorubicin reduced tumor growth, it increased the numbers of both cCAFs and CTCs in blood. Moreover, cCAFs and CTCs were found to form clusters in the chemotherapy-treated mice. Altogether, these findings indicate that the tumor stroma supports tumor growth and the formation of cCAFs. Furthermore, chemotherapy may exacerbate the formation of cCAFs and CTCs, which may eventually support the formation of a metastasis niche in breast cancer.
Collapse
Affiliation(s)
- Tao Lu
- Engineered Therapeutics, Department of Advanced Organ Bioengineering and Therapeutics, TechMed Centre, Faculty of Science and Technology, University of Twente, Drienerlolaan 5, 7500 AE Enschede, The Netherlands
| | - Lisa Oomens
- VyCAP B.V., Capitool 41, 7521 PL Enschede, The Netherlands
| | - Leon W. M. M. Terstappen
- Medical Cell BioPhysics, Faculty of Science and Technology, University of Twente, Hallenweg 23, 7522 NH Enschede, The Netherlands
| | - Jai Prakash
- Engineered Therapeutics, Department of Advanced Organ Bioengineering and Therapeutics, TechMed Centre, Faculty of Science and Technology, University of Twente, Drienerlolaan 5, 7500 AE Enschede, The Netherlands
- Correspondence:
| |
Collapse
|
22
|
Elevation of Cytoplasmic Calcium Suppresses Microtentacle Formation and Function in Breast Tumor Cells. Cancers (Basel) 2023; 15:cancers15030884. [PMID: 36765843 PMCID: PMC9913253 DOI: 10.3390/cancers15030884] [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: 11/21/2022] [Revised: 01/17/2023] [Accepted: 01/27/2023] [Indexed: 02/05/2023] Open
Abstract
Cytoskeletal remodeling in circulating tumor cells (CTCs) facilitates metastatic spread. Previous oncology studies examine sustained aberrant calcium (Ca2+) signaling and cytoskeletal remodeling scrutinizing long-term phenotypes such as tumorigenesis and metastasis. The significance of acute Ca2+ signaling in tumor cells that occur within seconds to minutes is overlooked. This study investigates rapid cytoplasmic Ca2+ elevation in suspended cells on actin and tubulin cytoskeletal rearrangements and the metastatic microtentacle (McTN) phenotype. The compounds Ionomycin and Thapsigargin acutely increase cytoplasmic Ca2+, suppressing McTNs in the metastatic breast cancer cell lines MDA-MB-231 and MDA-MB-436. Functional decreases in McTN-mediated reattachment and cell clustering during the first 24 h of treatment are not attributed to cytotoxicity. Rapid cytoplasmic Ca2+ elevation was correlated to Ca2+-induced actin cortex contraction and rearrangement via myosin light chain 2 and cofilin activity, while the inhibition of actin polymerization with Latrunculin A reversed Ca2+-mediated McTN suppression. Preclinical and phase 1 and 2 clinical trial data have established Thapsigargin derivatives as cytotoxic anticancer agents. The results from this study suggest an alternative molecular mechanism by which these compounds act, and proof-of-principle Ca2+-modulating compounds can rapidly induce morphological changes in free-floating tumor cells to reduce metastatic phenotypes.
Collapse
|
23
|
Zarubova J, Hasani-Sadrabadi MM, Norris SCP, Majedi FS, Xiao C, Kasko AM, Li S. Cell-Taxi: Mesenchymal Cells Carry and Transport Clusters of Cancer Cells. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2203515. [PMID: 36307906 PMCID: PMC9772300 DOI: 10.1002/smll.202203515] [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: 06/10/2022] [Revised: 09/09/2022] [Indexed: 06/16/2023]
Abstract
Cell clusters that collectively migrate from primary tumors appear to be far more potent in forming distant metastases than single cancer cells. A better understanding of the collective cell migration phenomenon and the involvement of various cell types during this process is needed. Here, an in vitro platform based on inverted-pyramidal microwells to follow and quantify the collective migration of hundreds of tumor cell clusters at once is developed. These results indicate that mesenchymal stromal cells (MSCs) or cancer-associated fibroblasts (CAFs) in the heterotypic tumor cell clusters may facilitate metastatic dissemination by transporting low-motile cancer cells in a Rac-dependent manner and that extracellular vesicles secreted by mesenchymal cells only play a minor role in this process. Furthermore, in vivo studies show that cancer cell spheroids containing MSCs or CAFs have faster spreading rates. These findings highlight the active role of co-traveling stromal cells in the collective migration of tumor cell clusters and may help in developing better-targeted therapies.
Collapse
Affiliation(s)
- Jana Zarubova
- Department of Bioengineering, University of California, 420 Westwood Plaza, 5121 Engineering V, Los Angeles, CA, 90095-1600, USA
- Department of Biomaterials and Tissue Engineering, Institute of Physiology of the Czech Academy of Sciences, Prague, 14220, Czech Republic
| | - Mohammad Mahdi Hasani-Sadrabadi
- Department of Bioengineering, University of California, 420 Westwood Plaza, 5121 Engineering V, Los Angeles, CA, 90095-1600, USA
| | - Sam C P Norris
- Department of Bioengineering, University of California, 420 Westwood Plaza, 5121 Engineering V, Los Angeles, CA, 90095-1600, USA
| | - Fatemeh Sadat Majedi
- Department of Bioengineering, University of California, 420 Westwood Plaza, 5121 Engineering V, Los Angeles, CA, 90095-1600, USA
| | - Crystal Xiao
- Department of Bioengineering, University of California, 420 Westwood Plaza, 5121 Engineering V, Los Angeles, CA, 90095-1600, USA
| | - Andrea M Kasko
- Department of Bioengineering, University of California, 420 Westwood Plaza, 5121 Engineering V, Los Angeles, CA, 90095-1600, USA
| | - Song Li
- Department of Bioengineering, University of California, 420 Westwood Plaza, 5121 Engineering V, Los Angeles, CA, 90095-1600, USA
| |
Collapse
|
24
|
Tumor-promoting aftermath post-chemotherapy: A focus on breast cancer. Life Sci 2022; 310:121125. [DOI: 10.1016/j.lfs.2022.121125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 10/14/2022] [Accepted: 10/22/2022] [Indexed: 11/09/2022]
|
25
|
Yang L, Qiao P, Zhang J, Chen X, Hu A, Huang S. Crosstalk between ROCK1 and PYROXD1 regulates CAFs activation and promotes laryngeal squamous cell carcinoma metastasis. Discov Oncol 2022; 13:120. [PMID: 36334145 PMCID: PMC9637080 DOI: 10.1007/s12672-022-00578-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 09/15/2022] [Indexed: 11/06/2022] Open
Abstract
We previously found that the Rho-associated kinase 1 (ROCK1) activated Cancer-associated fibroblasts (CAFs) to promote LSCC metastasis. Accumulating evidence indicates that pyridine nucleotide-disulfide oxidoreductase domain 1 (PYROXD1) is an oncogene; however, the crosstalk between ROCK1 and PYROXD1 in LSCC metastasis remains largely unknown. Here, we found that ROCK1 could target PYROXD1. The knockdown of ROCK1 expression reduces the expression of PYROXD1, while the knockdown of PYROXD1 expression did not alter the expression of ROCK1 indicating that ROCK1 is upstream of PYROXD1. Further, LSCC cells cocultured with PYROXD1 knocked-down CAFs exhibited lower proliferation, migration, invasion and metastasis abilities. Conversely, LSCC cells cocultured with PYROXD1-overexpressing CAFs showed opposite results. In conclusion, the crosstalk between ROCK1 and PYROXD1 regulated CAFs activation and promoted LSCC metastasis.
Collapse
Affiliation(s)
- Liyun Yang
- Department of Otolaryngology, Gongli Hospital, The Second Military Medical University, Shanghai, 200135, China
| | - Peipei Qiao
- Department of Otolaryngology, Gongli Hospital, The Second Military Medical University, Shanghai, 200135, China
| | - Jianwei Zhang
- Department of Otolaryngology, Gongli Hospital, The Second Military Medical University, Shanghai, 200135, China
| | - Xiaoping Chen
- Department of Otolaryngology, Gongli Hospital, The Second Military Medical University, Shanghai, 200135, China
| | - An Hu
- Department of Otolaryngology, Gongli Hospital, The Second Military Medical University, Shanghai, 200135, China.
| | - Shuixian Huang
- Department of Otolaryngology, Gongli Hospital, The Second Military Medical University, Shanghai, 200135, China.
| |
Collapse
|
26
|
Cakan-Akdogan G, Ersoz E, Sozer SC, Gelinci E. An in vivo zebrafish model reveals circulating tumor cell targeting capacity of serum albumin nanoparticles. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2022]
|
27
|
Greenlee JD, Liu K, Lopez-Cavestany M, King MR. Piezo1 Mechano-Activation Is Augmented by Resveratrol and Differs between Colorectal Cancer Cells of Primary and Metastatic Origin. Molecules 2022; 27:5430. [PMID: 36080197 PMCID: PMC9458129 DOI: 10.3390/molecules27175430] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 08/09/2022] [Accepted: 08/20/2022] [Indexed: 11/24/2022] Open
Abstract
Cancer cells must survive aberrant fluid shear stress (FSS) in the circulation to metastasize. Herein, we investigate the role that FSS has on colorectal cancer cell apoptosis, proliferation, membrane damage, calcium influx, and therapeutic sensitization. We tested this using SW480 (primary tumor) and SW620 cells (lymph node metastasis) derived from the same patient. The cells were exposed to either shear pulses, modeling millisecond intervals of high FSS seen in regions of turbulent flow, or sustained shear to model average magnitudes experienced by circulating tumor cells. SW480 cells were significantly more sensitive to FSS-induced death than their metastatic counterparts. Shear pulses caused significant cell membrane damage, while constant shear decreased cell proliferation and increased the expression of CD133. To investigate the role of mechanosensitive ion channels, we treated cells with the Piezo1 agonist Yoda1, which increased intracellular calcium. Pretreatment with resveratrol further increased the calcium influx via the lipid-raft colocalization of Piezo1. However, minimal changes in apoptosis were observed due to calcium saturation, as predicted via a computational model of apoptosis. Furthermore, SW480 cells had increased levels of Piezo1, calcium influx, and TRAIL-mediated apoptosis compared to SW620 cells, highlighting differences in the mechano-activation of metastatic cells, which may be a necessary element for successful dissemination in vivo.
Collapse
Affiliation(s)
| | | | | | - Michael R. King
- Department of Biomedical Engineering, Vanderbilt University, PMB 351631, 2301 Vanderbilt Place, Nashville, TN 37235-1631, USA
| |
Collapse
|
28
|
Chen Q, Zou J, He Y, Pan Y, Yang G, Zhao H, Huang Y, Zhao Y, Wang A, Chen W, Lu Y. A narrative review of circulating tumor cells clusters: A key morphology of cancer cells in circulation promote hematogenous metastasis. Front Oncol 2022; 12:944487. [PMID: 36059616 PMCID: PMC9434215 DOI: 10.3389/fonc.2022.944487] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Accepted: 07/18/2022] [Indexed: 11/28/2022] Open
Abstract
Circulating tumor cells (CTCs) that survive in the blood are playing an important role in the metastasis process of tumor. In addition, they have become a tool for tumor diagnosis, prognosis and recurrence monitoring. CTCs can exist in the blood as individual cells or as clumps of aggregated cells. In recent years, more and more studies have shown that clustered CTCs have stronger metastasis ability compared to single CTCs. With the deepening of studies, scholars have found that cancer cells can combine not only with each other, but also with non-tumor cells present in the blood, such as neutrophils, platelets, etc. At the same time, it was confirmed that non-tumor cells bound to CTCs maintain the survival and proliferation of cancer cells through a variety of ways, thus promoting the occurrence and development of tumor. In this review, we collected information on tumorigenesis induced by CTC clusters to make a summary and a discussion about them. Although CTC clusters have recently been considered as a key role in the transition process, many characteristics of them remain to be deeply explored. A detailed understanding of their vulnerability can prospectively pave the way for new inhibitors for metastasis.
Collapse
Affiliation(s)
- Qiong Chen
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jueyao Zou
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yong He
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yanhong Pan
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
- Department of Pharmacy, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Gejun Yang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Han Zhao
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Ying Huang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yang Zhao
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine (TCM) Prevention and Treatment of Tumor, Nanjing, China
| | - Aiyun Wang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine (TCM) Prevention and Treatment of Tumor, Nanjing, China
| | - Wenxing Chen
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine (TCM) Prevention and Treatment of Tumor, Nanjing, China
| | - Yin Lu
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine (TCM) Prevention and Treatment of Tumor, Nanjing, China
| |
Collapse
|
29
|
Götze J, Nitschke C, Uzunoglu FG, Pantel K, Sinn M, Wikman H. Tumor-Stroma Interaction in PDAC as a New Approach for Liquid Biopsy and its Potential Clinical Implications. Front Cell Dev Biol 2022; 10:918795. [PMID: 35712663 PMCID: PMC9197075 DOI: 10.3389/fcell.2022.918795] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 05/05/2022] [Indexed: 12/29/2022] Open
Abstract
The extremely poor prognosis for patients with pancreatic ductal adenocarcinoma (PDAC) has remained unchanged for decades. As a hallmark of PDAC histology, the distinct desmoplastic response in the tumor microenvironment is considered a key factor exerting pro- and antitumor effects. Increasing emphasis has been placed on cancer-associated fibroblasts (CAFs), whose heterogeneity and functional diversity is reflected in the numerous subtypes. The myofibroblastic CAFs (myCAFs), inflammatory CAFs (iCAFs) and antigen presenting CAFs (apCAFs) are functionally divergent CAF subtypes with tumor promoting as well as repressing effects. Precise knowledge of the underlying interactions is the basis for a variety of treatment approaches, which are subsumed under the term antistromal therapy. Clinical implementation is still pending due to the lack of benefit-as well as paradoxical preclinical findings. While the prominent significance of CAFs in the immediate environment of the tumor is becoming clear, less is known about the circulating (c)CAFs. cCAFs are of particular interest as they seem not only to be potential new liquid biopsy biomarkers but also to support the survival of circulating tumor cells (CTC) in the bloodstream. In PDAC, CTCs correlate with an unfavorable outcome and can also be employed to monitor treatment response, but the current clinical relevance is limited. In this review, we discuss CTCs, cCAFs, secretomes that include EVs or fragments of collagen turnover as liquid biopsy biomarkers, and clinical approaches to target tumor stroma in PDAC.
Collapse
Affiliation(s)
- Julian Götze
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, University Cancer Center Hamburg, Hamburg, Germany.,Institute of Tumor Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christine Nitschke
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Faik G Uzunoglu
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Klaus Pantel
- Institute of Tumor Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Marianne Sinn
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, University Cancer Center Hamburg, Hamburg, Germany
| | - Harriet Wikman
- Institute of Tumor Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
30
|
Weiss F, Lauffenburger D, Friedl P. Towards targeting of shared mechanisms of cancer metastasis and therapy resistance. Nat Rev Cancer 2022; 22:157-173. [PMID: 35013601 PMCID: PMC10399972 DOI: 10.1038/s41568-021-00427-0] [Citation(s) in RCA: 138] [Impact Index Per Article: 46.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/22/2021] [Indexed: 02/07/2023]
Abstract
Resistance to therapeutic treatment and metastatic progression jointly determine a fatal outcome of cancer. Cancer metastasis and therapeutic resistance are traditionally studied as separate fields using non-overlapping strategies. However, emerging evidence, including from in vivo imaging and in vitro organotypic culture, now suggests that both programmes cooperate and reinforce each other in the invasion niche and persist upon metastatic evasion. As a consequence, cancer cell subpopulations exhibiting metastatic invasion undergo multistep reprogramming that - beyond migration signalling - supports repair programmes, anti-apoptosis processes, metabolic adaptation, stemness and survival. Shared metastasis and therapy resistance signalling are mediated by multiple mechanisms, such as engagement of integrins and other context receptors, cell-cell communication, stress responses and metabolic reprogramming, which cooperate with effects elicited by autocrine and paracrine chemokine and growth factor cues present in the activated tumour microenvironment. These signals empower metastatic cells to cope with therapeutic assault and survive. Identifying nodes shared in metastasis and therapy resistance signalling networks should offer new opportunities to improve anticancer therapy beyond current strategies, to eliminate both nodular lesions and cells in metastatic transit.
Collapse
Affiliation(s)
- Felix Weiss
- Department of Cell Biology, RIMLS, Radboud University Medical Center, Nijmegen, Netherlands
| | - Douglas Lauffenburger
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Peter Friedl
- Department of Cell Biology, RIMLS, Radboud University Medical Center, Nijmegen, Netherlands.
- David H. Koch Center for Applied Research of Genitourinary Cancers, Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Cancer Genomics Center, Utrecht, Netherlands.
| |
Collapse
|
31
|
Simon T, Salhia B. Cancer-Associated Fibroblast Subpopulations With Diverse and Dynamic Roles in the Tumor Microenvironment. Mol Cancer Res 2022; 20:183-192. [PMID: 34670861 PMCID: PMC9306405 DOI: 10.1158/1541-7786.mcr-21-0282] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 08/21/2021] [Accepted: 10/15/2021] [Indexed: 01/07/2023]
Abstract
Close interactions between cancer cells and cancer-associated fibroblasts (CAF) have repeatedly been reported to support tumor progression. Yet, targeting CAFs has so far failed to show a real benefit in cancer treatment, as preclinical studies have shown that such a strategy can enhance tumor growth. Accordingly, recent paradigm-shifting data suggest that certain CAF subpopulations could also show tumor-inhibitory capabilities. The present review aims to provide an in-depth description of the cellular heterogeneity of the CAF compartment in tumors. Through combining information from different cancer types, here we define 4 main CAF subpopulations that might cohabitate in any tumor microenvironment (TME). In addition, a model for the evolution of CAFs during tumor development is introduced. Moreover, the presence of tumor-inhibitory CAFs in the TME as well as their molecular characteristics are extensively discussed. Finally, the potential cellular origins of these distinct CAF subpopulations are reviewed. To our knowledge, this is the first attempt at establishing a broad but comprehensive classification of CAF subpopulations. Altogether, the present manuscript aims to provide the latest developments and innovative insights that could help refine future therapeutic targeting of CAFs for cancer treatment.
Collapse
Affiliation(s)
- Thomas Simon
- Department of Translational Genomics, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Bodour Salhia
- Department of Translational Genomics, Keck School of Medicine, University of Southern California, Los Angeles, California.,Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California.,Corresponding Author: Bodour Salhia, Department of Translational Genomics, Keck School of Medicine, University of Southern California, 1450 Biggy Street, Los Angeles, CA 90033. Phone: 323-442-3099; E-mail:
| |
Collapse
|
32
|
Circulating Tumour Cells (CTCs) in NSCLC: From Prognosis to Therapy Design. Pharmaceutics 2021; 13:pharmaceutics13111879. [PMID: 34834295 PMCID: PMC8619417 DOI: 10.3390/pharmaceutics13111879] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 10/27/2021] [Accepted: 10/30/2021] [Indexed: 02/08/2023] Open
Abstract
Designing optimal (neo)adjuvant therapy is a crucial aspect of the treatment of non-small-cell lung carcinoma (NSCLC). Standard methods of chemotherapy, radiotherapy, and immunotherapy represent effective strategies for treatment. However, in some cases with high metastatic activity and high levels of circulating tumour cells (CTCs), the efficacy of standard treatment methods is insufficient and results in treatment failure and reduced patient survival. CTCs are seen not only as an isolated phenomenon but also a key inherent part of the formation of metastasis and a key factor in cancer death. This review discusses the impact of NSCLC therapy strategies based on a meta-analysis of clinical studies. In addition, possible therapeutic strategies for repression when standard methods fail, such as the administration of low-toxicity natural anticancer agents targeting these phenomena (curcumin and flavonoids), are also discussed. These strategies are presented in the context of key mechanisms of tumour biology with a strong influence on CTC spread and metastasis (mechanisms related to tumour-associated and -infiltrating cells, epithelial–mesenchymal transition, and migration of cancer cells).
Collapse
|
33
|
Greenlee JD, Lopez-Cavestany M, Ortiz-Otero N, Liu K, Subramanian T, Cagir B, King MR. Oxaliplatin resistance in colorectal cancer enhances TRAIL sensitivity via death receptor 4 upregulation and lipid raft localization. eLife 2021; 10:e67750. [PMID: 34342264 PMCID: PMC8331188 DOI: 10.7554/elife.67750] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 07/12/2021] [Indexed: 11/13/2022] Open
Abstract
Colorectal cancer (CRC) remains a leading cause of cancer death, and its mortality is associated with metastasis and chemoresistance. We demonstrate that oxaliplatin-resistant CRC cells are sensitized to TRAIL-mediated apoptosis. Oxaliplatin-resistant cells exhibited transcriptional downregulation of caspase-10, but this had minimal effects on TRAIL sensitivity following CRISPR-Cas9 deletion of caspase-10 in parental cells. Sensitization effects in oxaliplatin-resistant cells were found to be a result of increased DR4, as well as significantly enhanced DR4 palmitoylation and translocation into lipid rafts. Raft perturbation via nystatin and resveratrol significantly altered DR4/raft colocalization and TRAIL sensitivity. Blood samples from metastatic CRC patients were treated with TRAIL liposomes, and a 57% reduction of viable circulating tumor cells (CTCs) was observed. Increased DR4/lipid raft colocalization in CTCs was found to correspond with increased oxaliplatin resistance and increased efficacy of TRAIL liposomes. To our knowledge, this is the first study to investigate the role of lipid rafts in primary CTCs.
Collapse
Affiliation(s)
- Joshua D Greenlee
- Vanderbilt University, Department of Biomedical Engineering PMBNashvilleUnited States
| | - Maria Lopez-Cavestany
- Vanderbilt University, Department of Biomedical Engineering PMBNashvilleUnited States
| | - Nerymar Ortiz-Otero
- Vanderbilt University, Department of Biomedical Engineering PMBNashvilleUnited States
| | - Kevin Liu
- Vanderbilt University, Department of Biomedical Engineering PMBNashvilleUnited States
| | - Tejas Subramanian
- Vanderbilt University, Department of Biomedical Engineering PMBNashvilleUnited States
| | - Burt Cagir
- Donald Guthrie Foundation (DGF) for Research and Education SayreSayreUnited States
| | - Michael R King
- Vanderbilt University, Department of Biomedical Engineering PMBNashvilleUnited States
| |
Collapse
|
34
|
Wang Z, Liu J, Huang H, Ye M, Li X, Wu R, Liu H, Song Y. Metastasis-associated fibroblasts: an emerging target for metastatic cancer. Biomark Res 2021; 9:47. [PMID: 34112258 PMCID: PMC8194104 DOI: 10.1186/s40364-021-00305-9] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Accepted: 05/27/2021] [Indexed: 12/12/2022] Open
Abstract
Metastasis suggests a poor prognosis for cancer patients, and treatment strategies for metastatic cancer are still very limited. Numerous studies have shown that cancer-associated fibroblasts (CAFs), a large component of the tumor microenvironment, contribute to tumor metastasis. Stromal fibroblasts at metastatic sites are different from CAFs within primary tumors and can be termed metastasis-associated fibroblasts (MAFs), and they also make great contributions to the establishment of metastatic lesions and the therapeutic resistance of metastatic tumors. MAFs are capable of remodeling the extracellular matrix of metastatic tumors, modulating immune cells in the tumor microenvironment, promoting angiogenesis and enhancing malignant tumor phenotypes. Thus, MAFs can help establish premetastatic niches and mediate resistance to therapeutic strategies, including immunotherapy and antiangiogenic therapy. The results of preclinical studies suggest that targeting MAFs can alleviate the progression of metastatic cancer and mitigate therapeutic resistance, indicating that MAFs are a promising target for metastatic cancer. Here, we comprehensively summarize the existing evidence on MAFs and discuss their origins, generation, functions and related therapeutic strategies in an effort to provide a better understanding of MAFs and offer treatment perspectives for metastatic cancer.
Collapse
Affiliation(s)
- Zimu Wang
- Department of Respiratory Medicine, Jinling Hospital, Nanjing University School of Medicine, #305, East Zhongshan Road, 210002, Nanjing, Jiangsu, China
| | - Jiaxin Liu
- Department of Respiratory Medicine, Jinling Hospital, Nanjing University School of Medicine, #305, East Zhongshan Road, 210002, Nanjing, Jiangsu, China
| | - Hairong Huang
- Department of Cardiothoracic Surgery, Jinling Hospital, 210002, Nanjing, China
| | - Mingxiang Ye
- Department of Respiratory Medicine, Jinling Hospital, Nanjing University School of Medicine, #305, East Zhongshan Road, 210002, Nanjing, Jiangsu, China
| | - Xinying Li
- Department of Respiratory Medicine, Nanjing Drum Tower Hospital, Nanjing University School of Medicine, 210008, Nanjing, Jiangsu, China
| | - Ranpu Wu
- Department of Respiratory Medicine, Jinling Hospital, Southeast University of Medicine, 210009, Nanjing, Jiangsu, China
| | - Hongbing Liu
- Department of Respiratory Medicine, Jinling Hospital, Nanjing University School of Medicine, #305, East Zhongshan Road, 210002, Nanjing, Jiangsu, China.
| | - Yong Song
- Department of Respiratory Medicine, Jinling Hospital, Nanjing University School of Medicine, #305, East Zhongshan Road, 210002, Nanjing, Jiangsu, China.
| |
Collapse
|
35
|
Sznurkowska MK, Aceto N. The gate to metastasis: key players in cancer cell intravasation. FEBS J 2021; 289:4336-4354. [PMID: 34077633 PMCID: PMC9546053 DOI: 10.1111/febs.16046] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 04/19/2021] [Accepted: 06/01/2021] [Indexed: 02/06/2023]
Abstract
Metastasis is a leading cause of cancer‐related death and consists of a sequence of events including tumor expansion, intravasation of cancer cells into the circulation, survival in the bloodstream, extravasation at distant sites, and subsequent organ colonization. Particularly, intravasation is a process whereby cancer cells transverse the endothelium and leave the primary tumor site, pioneering the metastatic cascade. The identification of those mechanisms that trigger the entry of cancer cells into the bloodstream may reveal fundamentally novel ways to block metastasis at its start. Multiple factors have been implicated in cancer progression, yet, signals that unequivocally provoke the detachment of cancer cells from the primary tumor are still under investigation. Here, we discuss the role of intrinsic properties of cancer cells, tumor microenvironment, and mechanical cues in the intravasation process, outlining studies that suggest the involvement of various factors and highlighting current understanding and open questions in the field.
Collapse
Affiliation(s)
- Magdalena K Sznurkowska
- Department of Biomedicine, Cancer Metastasis Laboratory, University of Basel and University Hospital Basel, Switzerland
| | - Nicola Aceto
- Department of Biomedicine, Cancer Metastasis Laboratory, University of Basel and University Hospital Basel, Switzerland.,Department of Biology, Institute of Molecular Health Sciences, Swiss Federal Institute of Technology (ETH) Zurich, Switzerland
| |
Collapse
|
36
|
Badia-Ramentol J, Linares J, Gómez-Llonin A, Calon A. Minimal Residual Disease, Metastasis and Immunity. Biomolecules 2021; 11:130. [PMID: 33498251 PMCID: PMC7909268 DOI: 10.3390/biom11020130] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 01/12/2021] [Accepted: 01/13/2021] [Indexed: 12/11/2022] Open
Abstract
Progression from localized to metastatic disease requires cancer cells spreading to distant organs through the bloodstream. Only a small proportion of these circulating tumor cells (CTCs) survives dissemination due to anoikis, shear forces and elimination by the immune system. However, all metastases originate from CTCs capable of surviving and extravasating into distant tissue to re-initiate a tumor. Metastasis initiation is not always immediate as disseminated tumor cells (DTCs) may enter a non-dividing state of cell dormancy. Cancer dormancy is a reversible condition that can be maintained for many years without being clinically detectable. Subsequently, late disease relapses are thought to be due to cancer cells ultimately escaping from dormant state. Cancer dormancy is usually associated with minimal residual disease (MRD), where DTCs persist after intended curative therapy. Thus, MRD is commonly regarded as an indicator of poor prognosis in all cancers. In this review, we examine the current understanding of MRD and immunity during cancer progression to metastasis and discuss clinical perspectives for oncology.
Collapse
Affiliation(s)
| | | | | | - Alexandre Calon
- Cancer Research Program, Hospital del Mar Medical Research Institute (IMIM), 08003 Barcelona, Spain; (J.B.-R.); (J.L.); (A.G.-L.)
| |
Collapse
|
37
|
Zhang Z, Patel SB, King MR. Micelle-in-Liposomes for Sustained Delivery of Anticancer Agents That Promote Potent TRAIL-Induced Cancer Cell Apoptosis. Molecules 2020; 26:E157. [PMID: 33396409 PMCID: PMC7795772 DOI: 10.3390/molecules26010157] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 12/22/2020] [Accepted: 12/29/2020] [Indexed: 11/16/2022] Open
Abstract
Tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) induces cancer cell-specific apoptosis and has garnered intense interest as a promising agent for cancer treatment. However, the development of TRAIL has been hampered in part because most human cancer cells are resistant to TRAIL. A few small molecules including natural compounds such as piperlongumine (PL) have been reported to sensitize cancer cells to TRAIL. We prepared a novel type of nanomaterial, micelle-in-liposomes (MILs) for solubilization and delivery of PL. PL-loaded MILs were used to sensitize cancer cells to TRAIL. As visualized by cryo-TEM, micelles were successfully loaded inside the aqueous core of liposomes. The MILs increased the water solubility of PL by ~20 fold. A sustained PL release from MILs in physiologically relevant buffer over 7 days was achieved, indicating that the liposomes prevented premature drug release from the micelles in the MILs. Also demonstrated is a potent synergistic apoptotic effect in cancer cells by PL MILs in conjunction with liposomal TRAIL. MILs provide a new formulation and delivery vehicle for hydrophobic anticancer agents, which can be used alone or in combination with TRAIL to promote cancer cell death.
Collapse
Affiliation(s)
| | | | - Michael R. King
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37212, USA; (Z.Z.); (S.B.P.)
| |
Collapse
|