1
|
Goodarzi K, Sarker P, Rao SS. Regulation of Brain Metastatic Breast Cancer Cell Dormancy versus Proliferation on Hyaluronic Acid Hydrogels via Laminin and Laminin-Derived Peptides. ACS APPLIED BIO MATERIALS 2025; 8:2824-2837. [PMID: 40083120 DOI: 10.1021/acsabm.4c01386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2025]
Abstract
Among the secondary target organs for metastatic breast cancer, brain metastasis is extremely aggressive in nature, resulting in lower survival rates. These metastatic cancer cells have the potential to enter a dormant state in the brain, allowing them to survive for extended time periods. The brain microenvironment plays a key role in controlling the dormant phenotype, yet how various components of this microenvironment influence dormancy is not well understood. In this work, we employed hyaluronic acid (HA)-based hydrogels as a mimetic of the brain tissue environment to study the role of biochemical cues, specifically, the impact of laminin and laminin-derived peptides IKVAV and YIGSR on the regulation of brain metastatic breast cancer cell dormancy versus proliferation. We applied varying protein/peptide concentrations and confirmed functionalization on HA hydrogel surfaces. We then seeded 10,000 cancer cells on the hydrogel surface and cultured them for 5 days. We found that in the presence of laminin or IKVAV, MDA-MB-231Br cells transitioned from a rounded to a spread morphology and exhibited enhanced proliferation as the laminin/IKVAV concentration increased. In contrast, in hydrogels functionalized with YIGSR, these cells maintained a rounded morphology, with no impact on proliferation with varying YIGSR concentrations. We confirmed the involvement of αVβ3 integrin in mediating tumor cell phenotype in hydrogels functionalized with laminin. By evaluating known markers of dormancy and proliferation, we found a direct correlation between the presence of laminin and IKVAV and increased phosphorylated extracellular signal-regulated kinase 1/2 (p-ERK) positivity, along with decreased phosphorylated p38 (p-p38) positivity, while in hydrogels functionalized with YIGSR, the levels of both p-ERK and p-p38 remained unaltered. Finally, we demonstrated that when cells were transferred from IKVAV-deficient to IKVAV-rich hydrogels, the hydrogel induced cellular dormancy was reversible. Collectively, our findings provide insights into how laminin and laminin-derived cues regulate brain metastatic breast cancer cell dormancy versus proliferation.
Collapse
Affiliation(s)
- Kasra Goodarzi
- Department of Chemical and Biological Engineering, The University of Alabama, Tuscaloosa, Alabama United States
| | - Paromita Sarker
- Department of Chemical and Biological Engineering, The University of Alabama, Tuscaloosa, Alabama United States
| | - Shreyas S Rao
- Department of Chemical and Biological Engineering, The University of Alabama, Tuscaloosa, Alabama United States
| |
Collapse
|
2
|
VandenHeuvel SN, Nash LL, Raghavan SA. Dormancy in Metastatic Colorectal Cancer: Tissue Engineering Opportunities for In Vitro Modeling. TISSUE ENGINEERING. PART B, REVIEWS 2025. [PMID: 40195931 DOI: 10.1089/ten.teb.2025.0009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Colorectal cancer (CRC) recurs at a striking rate, specifically in patients with liver metastasis. Dormant CRC cells disseminated following initial primary tumor resection or treatment often resurface years later to form aggressive, therapy-resistant tumors that result in high patient mortality. Routine imaging-based screenings often fail to detect dormant cancer cell clusters, and there are no overt symptomatic presentations, making dormant CRC a major clinical challenge to diagnose and treat. Tissue engineering approaches are ideally suited to model dormant cancer cells and enable the discovery of therapeutic vulnerabilities or unique mechanistic dependencies of dormant CRC. Emerging evidence suggests that tissue-engineered approaches have been successfully used to model dormant breast and lung cancer. With CRC responsible for the second most cancer-related deaths worldwide and CRC patients commonly experiencing recurrence, it is essential to expand dormancy models to understand this phenomenon in the context of CRC. Most published in vitro models of CRC dormancy simplify the complex tumor microenvironment with two-dimensional culture systems to elucidate dormancy-driving mechanisms. Building on this foundation, future research should apply tissue engineering methods to this growing field to generate competent three-dimensional models and increase mechanistic knowledge. This review summarizes the current state of in vitro CRC dormancy models, highlighting the techniques utilized to give rise to dormant CRC cells: nutrient depletion, anticancer drugs, physical extracellular matrix interactions, and genetic manipulation. The metrics used to validate dormancy within each model are also consolidated to demonstrate the lack of established standards and the ambiguity around comparing studies that have been validated differently. The methods of these studies are organized in this review to increase comprehensibility and identify needs and opportunities for future bioengineered in vitro models to address dormancy-driven mortality in patients with CRC liver metastasis. Impact Statement Dormant cancer drives high patient mortality, especially in metastatic colorectal cancer, owing to the clinical inability to identify dormant cells prior to their overt recurrence. Lacking clinical insights, in vitro modeling for mechanistic and therapeutic discovery is hindered. Here, we review models and methods of inducing colorectal cancer dormancy with the goal of consolidating findings for reference. We also highlight the need for advanced, tissue-engineered models to better mimic the organ-specific 3D microenvironment of metastatic colorectal cancer. New models would enable breakthroughs in understanding mechanisms driving dormancy progression and reversal, thereby providing context for therapeutic advances to improve patient survival.
Collapse
Affiliation(s)
| | - Lucia L Nash
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas, USA
| | - Shreya A Raghavan
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas, USA
- Regional Excellence Center in Cancer, Texas A&M University, College Station, Texas, USA
| |
Collapse
|
3
|
Bakhshandeh S, Heras U, Taïeb HM, Varadarajan AR, Lissek SM, Hücker SM, Lu X, Garske DS, Young SAE, Abaurrea A, Caffarel MM, Riestra A, Bragado P, Contzen J, Gossen M, Kirsch S, Warfsmann J, Honarnejad K, Klein CA, Cipitria A. Dormancy-inducing 3D engineered matrix uncovers mechanosensitive and drug-protective FHL2-p21 signaling axis. SCIENCE ADVANCES 2024; 10:eadr3997. [PMID: 39504377 PMCID: PMC11540038 DOI: 10.1126/sciadv.adr3997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 09/26/2024] [Indexed: 11/08/2024]
Abstract
Solid cancers frequently relapse with distant metastasis, despite local and systemic treatment. Cellular dormancy has been identified as an important mechanism underlying drug resistance enabling late relapse. Therefore, relapse from invisible, minimal residual cancer of seemingly disease-free patients call for in vitro models of dormant cells suited for drug discovery. Here, we explore dormancy-inducing 3D engineered matrices, which generate mechanical confinement and induce growth arrest and survival against chemotherapy in cancer cells. We characterized the dormant phenotype of solitary cells by P-ERKlow:P-p38high dormancy signaling ratio, along with Ki67- expression. As underlying mechanism, we identified stiffness-dependent nuclear localization of the four-and-a-half LIM domain 2 (FHL2) protein, leading to p53-independent high p21Cip1/Waf1 nuclear expression, validated in murine and human tissue. Suggestive of a resistance-causing role, cells in the dormancy-inducing matrix became sensitive against chemotherapy upon FHL2 down-regulation. Thus, our biomaterial-based approach will enable systematic screens for previously unidentified compounds suited to eradicate potentially relapsing dormant cancer cells.
Collapse
Affiliation(s)
- Sadra Bakhshandeh
- Department of Biomaterials, Max Planck Institute of Colloids and Interfaces, Potsdam, Germany
| | - Unai Heras
- Group of Bioengineering in Regeneration and Cancer, Biogipuzkoa Health Research Institute, San Sebastian, Spain
- Department of Cell Biology and Histology, Faculty of Medicine and Nursing, University of the Basque Country UPV/EHU, Leioa, Spain
| | - Hubert M. Taïeb
- Department of Biomaterials, Max Planck Institute of Colloids and Interfaces, Potsdam, Germany
| | - Adithi R. Varadarajan
- Division of Personalized Tumor Therapy, Fraunhofer Institute for Toxicology and Experimental Medicine, Regensburg, Germany
| | - Susanna M. Lissek
- Experimental Medicine and Therapy Research, University of Regensburg, Regensburg, Germany
| | - Sarah M. Hücker
- Division of Personalized Tumor Therapy, Fraunhofer Institute for Toxicology and Experimental Medicine, Regensburg, Germany
| | - Xin Lu
- Division of Personalized Tumor Therapy, Fraunhofer Institute for Toxicology and Experimental Medicine, Regensburg, Germany
| | - Daniela S. Garske
- Department of Biomaterials, Max Planck Institute of Colloids and Interfaces, Potsdam, Germany
| | - Sarah A. E. Young
- Department of Biomaterials, Max Planck Institute of Colloids and Interfaces, Potsdam, Germany
| | - Andrea Abaurrea
- Group of Breast Cancer, Biogipuzkoa Health Research Institute, San Sebastian, Spain
| | - Maria M Caffarel
- Group of Breast Cancer, Biogipuzkoa Health Research Institute, San Sebastian, Spain
- IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| | - Ana Riestra
- Department of Pharmacy, Fundación Onkologikoa Fundazioa, San Sebastian, Spain
- Department of Medicine, University of Deusto, Bilbao, Spain
| | - Paloma Bragado
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University of Madrid, Madrid, Spain
- Health Research Institute of the Hospital Clínico San Carlos, Madrid, Spain
| | - Jörg Contzen
- Department of Experimental Neurology, Charité - Universitätsmedizin Berlin, Berlin, Germany
- Institute of Active Polymers, Helmholtz-Zentrum Hereon, Teltow, Germany
- Berlin-Brandenburg Center for Regenerative Therapies, Charité Campus Virchow Klinikum, Berlin, Germany
| | - Manfred Gossen
- Institute of Active Polymers, Helmholtz-Zentrum Hereon, Teltow, Germany
- Berlin-Brandenburg Center for Regenerative Therapies, Charité Campus Virchow Klinikum, Berlin, Germany
| | - Stefan Kirsch
- Division of Personalized Tumor Therapy, Fraunhofer Institute for Toxicology and Experimental Medicine, Regensburg, Germany
| | - Jens Warfsmann
- Division of Personalized Tumor Therapy, Fraunhofer Institute for Toxicology and Experimental Medicine, Regensburg, Germany
| | - Kamran Honarnejad
- Division of Personalized Tumor Therapy, Fraunhofer Institute for Toxicology and Experimental Medicine, Regensburg, Germany
| | - Christoph A. Klein
- Division of Personalized Tumor Therapy, Fraunhofer Institute for Toxicology and Experimental Medicine, Regensburg, Germany
- Experimental Medicine and Therapy Research, University of Regensburg, Regensburg, Germany
| | - Amaia Cipitria
- Department of Biomaterials, Max Planck Institute of Colloids and Interfaces, Potsdam, Germany
- Group of Bioengineering in Regeneration and Cancer, Biogipuzkoa Health Research Institute, San Sebastian, Spain
- IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| |
Collapse
|
4
|
Szewczyk K, Jiang L, Khawaja H, Miranti CK, Zohar Y. Microfluidic Applications in Prostate Cancer Research. MICROMACHINES 2024; 15:1195. [PMID: 39459070 PMCID: PMC11509716 DOI: 10.3390/mi15101195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 09/13/2024] [Accepted: 09/23/2024] [Indexed: 10/28/2024]
Abstract
Prostate cancer is a disease in which cells in the prostate, a gland in the male reproductive system below the bladder, grow out of control and, among men, it is the second-most frequently diagnosed cancer (other than skin cancer). In recent years, prostate cancer death rate has stabilized and, currently, it is the second-most frequent cause of cancer death in men (after lung cancer). Most deaths occur due to metastasis, as cancer cells from the original tumor establish secondary tumors in distant organs. For a long time, classical cell cultures and animal models have been utilized in basic and applied scientific research, including clinical applications for many diseases, such as prostate cancer, since no better alternatives were available. Although helpful in dissecting cellular mechanisms, these models are poor predictors of physiological behavior mainly because of the lack of appropriate microenvironments. Microfluidics has emerged in the last two decades as a technology that could lead to a paradigm shift in life sciences and, in particular, controlling cancer. Microfluidic systems, such as organ-on-chips, have been assembled to mimic the critical functions of human organs. These microphysiological systems enable the long-term maintenance of cellular co-cultures in vitro to reconstitute in vivo tissue-level microenvironments, bridging the gap between traditional cell cultures and animal models. Several reviews on microfluidics for prostate cancer studies have been published focusing on technology advancement and disease progression. As metastatic castration-resistant prostate cancer remains a clinically challenging late-stage cancer, with no curative treatments, we expanded this review to cover recent microfluidic applications related to prostate cancer research. The review includes discussions of the roles of microfluidics in modeling the human prostate, prostate cancer initiation and development, as well as prostate cancer detection and therapy, highlighting potentially major contributions of microfluidics in the continuous march toward eradicating prostate cancer.
Collapse
Affiliation(s)
- Kailie Szewczyk
- Department of Aerospace and Mechanical Engineering, University of Arizona, Tucson, AZ 85721, USA; (K.S.); (L.J.)
| | - Linan Jiang
- Department of Aerospace and Mechanical Engineering, University of Arizona, Tucson, AZ 85721, USA; (K.S.); (L.J.)
| | - Hunain Khawaja
- Cancer Biology Graduate Interdisciplinary Program, University of Arizona, Tucson, AZ 85724, USA;
| | - Cindy K. Miranti
- Department of Molecular and Cellular Biology, University of Arizona, Tucson, AZ 85721, USA;
- University of Arizona Cancer Center, University of Arizona, Tucson, AZ 85724, USA
| | - Yitshak Zohar
- Department of Aerospace and Mechanical Engineering, University of Arizona, Tucson, AZ 85721, USA; (K.S.); (L.J.)
- University of Arizona Cancer Center, University of Arizona, Tucson, AZ 85724, USA
| |
Collapse
|
5
|
Yakati V, Shevde LA, Rao SS. Matrix stiffness influences response to chemo and targeted therapy in brain metastatic breast cancer cells. Biomater Sci 2024; 12:3882-3895. [PMID: 38912649 DOI: 10.1039/d4bm00342j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/25/2024]
Abstract
Breast cancer is the most common malignancy accounting for 12.5% of all newly diagnosed cancer cases across the globe. Breast cancer cells are known to metastasize to distant organs (i.e., brain), wherein they can exhibit a dormant phenotype for extended time periods. These dormant cancer cells exhibit reduced proliferation and therapeutic resistance. However, the mechanisms by which dormant cancer cells exhibit resistance to therapy, in the context of brain metastatic breast cancer (BMBC), is not well understood. Herein, we utilized hyaluronic acid (HA) hydrogels with varying stiffnesses to study drug responsiveness in dormant vs. proliferative BMBC cells. It was found that cells cultured on soft HA hydrogels (∼0.4 kPa) that showed a non-proliferative (dormant) phenotype exhibited resistance to Paclitaxel or Lapatinib. In contrast, cells cultured on stiff HA hydrogels (∼4.5 kPa) that showed a proliferative phenotype exhibited responsiveness to Paclitaxel or Lapatinib. Moreover, dormancy-associated resistance was found to be due to upregulation of the serum/glucocorticoid regulated kinase 1 (SGK1) gene which was mediated, in part, by the p38 signaling pathway. Accordingly, SGK1 inhibition resulted in a dormant-to-proliferative switch and response to therapy. Overall, our study demonstrates that matrix stiffness influences dormancy-associated therapy response mediated, in part, via the p38/SGK1 axis.
Collapse
Affiliation(s)
- Venu Yakati
- Department of Chemical and Biological Engineering, The University of Alabama, Tuscaloosa, AL 35487, USA.
| | - Lalita A Shevde
- Department of Pathology, O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Shreyas S Rao
- Department of Chemical and Biological Engineering, The University of Alabama, Tuscaloosa, AL 35487, USA.
| |
Collapse
|
6
|
Okorafor CC, Shastri S, Wen K, Ebong EE. Mechanisms of triple-negative breast cancer extravasation: Impact of the physical environment and endothelial glycocalyx. FASEB J 2024; 38:e23785. [PMID: 38949120 DOI: 10.1096/fj.202400380r] [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: 03/13/2024] [Revised: 05/27/2024] [Accepted: 06/20/2024] [Indexed: 07/02/2024]
Abstract
Cancer metastasis is the leading cause of death for those afflicted with cancer. In cancer metastasis, the cancer cells break off from the primary tumor, penetrate nearby blood vessels, and attach and extravasate out of the vessels to form secondary tumors at distant organs. This makes extravasation a critical step of the metastatic cascade. Herein, with a focus on triple-negative breast cancer, the role that the prospective secondary tumor microenvironment's mechanical properties play in circulating tumor cells' extravasation is reviewed. Specifically, the effects of the physically regulated vascular endothelial glycocalyx barrier element, vascular flow factors, and subendothelial extracellular matrix mechanical properties on cancer cell extravasation are examined. The ultimate goal of this review is to clarify the physical mechanisms that drive triple-negative breast cancer extravasation, as these mechanisms may be potential new targets for anti-metastasis therapy.
Collapse
Affiliation(s)
- Chinedu C Okorafor
- Department of Chemical Engineering, Northeastern University, Boston, Massachusetts, USA
| | - Sanjana Shastri
- Department of Behavioral Neuroscience, Northeastern University, Boston, Massachusetts, USA
| | - Ke Wen
- Department of Chemical Engineering, Northeastern University, Boston, Massachusetts, USA
| | - Eno E Ebong
- Department of Chemical Engineering, Northeastern University, Boston, Massachusetts, USA
- Department of Bioengineering, Northeastern University, Boston, Massachusetts, USA
- Department of Neuroscience, Albert Einstein College of Medicine, New York, New York, USA
| |
Collapse
|
7
|
Amissah HA, Combs SE, Shevtsov M. Tumor Dormancy and Reactivation: The Role of Heat Shock Proteins. Cells 2024; 13:1087. [PMID: 38994941 PMCID: PMC11240553 DOI: 10.3390/cells13131087] [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: 05/24/2024] [Revised: 06/19/2024] [Accepted: 06/20/2024] [Indexed: 07/13/2024] Open
Abstract
Tumors are a heterogeneous group of cell masses originating in various organs or tissues. The cellular composition of the tumor cell mass interacts in an intricate manner, influenced by humoral, genetic, molecular, and tumor microenvironment cues that dictate tumor growth or suppression. As a result, tumors undergo a period of a dormant state before their clinically discernible stage, which surpasses the clinical dormancy threshold. Moreover, as a genetically imprinted strategy, early-seeder cells, a distinct population of tumor cells, break off to dock nearby or extravasate into blood vessels to secondary tissues, where they form disseminated solitary dormant tumor cells with reversible capacity. Among the various mechanisms underlying the dormant tumor mass and dormant tumor cell formation, heat shock proteins (HSPs) might play one of the most important roles in how the dormancy program plays out. It is known that numerous aberrant cellular processes, such as malignant transformation, cancer cell stemness, tumor invasion, metastasis, angiogenesis, and signaling pathway maintenance, are influenced by the HSPs. An accumulating body of knowledge suggests that HSPs may be involved in the angiogenic switch, immune editing, and extracellular matrix (ECM) remodeling cascades, crucial genetically imprinted strategies important to the tumor dormancy initiation and dormancy maintenance program. In this review, we highlight the biological events that orchestrate the dormancy state and the body of work that has been conducted on the dynamics of HSPs in a tumor mass, as well as tumor cell dormancy and reactivation. Additionally, we propose a conceptual framework that could possibly underlie dormant tumor reactivation in metastatic relapse.
Collapse
Affiliation(s)
- Haneef Ahmed Amissah
- Institute of Life Sciences and Biomedicine, Department of Medical Biology and Medical Biology, FEFU Campus, Far Eastern Federal University, 690922 Vladivostok, Russia;
- Diagnostics Laboratory Department, Trauma and Specialist Hospital, CE-122-2486, Central Region, Winneba P.O. Box 326, Ghana
| | - Stephanie E. Combs
- Department of Radiation Oncology, Technische Universität München (TUM), Klinikum Rechts der Isar, 81675 Munich, Germany;
| | - Maxim Shevtsov
- Department of Radiation Oncology, Technische Universität München (TUM), Klinikum Rechts der Isar, 81675 Munich, Germany;
- Laboratory of Biomedical Nanotechnologies, Institute of Cytology of the Russian Academy of Sciences (RAS), 194064 Saint Petersburg, Russia
- Personalized Medicine Centre, Almazov National Medical Research Centre, 197341 Saint Petersburg, Russia
| |
Collapse
|
8
|
Goodarzi K, Lane R, Rao SS. Varying the RGD concentration on a hyaluronic acid hydrogel influences dormancy versus proliferation in brain metastatic breast cancer cells. J Biomed Mater Res A 2024; 112:710-720. [PMID: 38018303 DOI: 10.1002/jbm.a.37651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 11/10/2023] [Accepted: 11/13/2023] [Indexed: 11/30/2023]
Abstract
A majority of breast cancer deaths occur due to metastasis of cancer cells to distant organs. In particular, brain metastasis is very aggressive with an extremely low survival rate. Breast cancer cells that metastasize to the brain can enter a state of dormancy, which allows them to evade death. The brain microenvironment provides biophysical, biochemical, and cellular cues, and plays an important role in determining the fate of dormant cancer cells. However, how these cues influence dormancy remains poorly understood. Herein, we employed hyaluronic acid (HA) hydrogels with a stiffness of ~0.4 kPa as an in vitro biomimetic platform to investigate the impact of biochemical cues, specifically alterations in RGD concentration, on dormancy versus proliferation in MDA-MB-231Br brain metastatic breast cancer cells. We applied varying concentrations of RGD peptide (0, 1, 2, or 4 mg/mL) to HA hydrogel surfaces and confirmed varying degrees of surface functionalization using a fluorescently labeled RGD peptide. Post functionalization, ~10,000 MDA-MB-231Br cells were seeded on top of the hydrogels and cultured for 5 days. We found that an increase in RGD concentration led to changes in cell morphology, with cells transitioning from a rounded to spindle-like morphology as well as an increase in cell spreading area. Also, an increase in RGD concentration resulted in an increase in cell proliferation. Cellular dormancy was assessed using the ratio of phosphorylated extracellular signal-regulated kinase 1/2 (p-ERK) to phosphorylated p38 (p-p38) positivity, which was significantly lower in hydrogels without RGD and in hydrogels with lowest RGD concentration compared to hydrogels functionalized with higher RGD concentration. We also demonstrated that the HA hydrogel-induced cellular dormancy was reversible. Finally, we demonstrated the involvement of β1 integrin in mediating cell phenotype in our hydrogel platform. Overall, our results provide insight into the role of biochemical cues in regulating dormancy versus proliferation in brain metastatic breast cancer cells.
Collapse
Affiliation(s)
- Kasra Goodarzi
- Department of Chemical and Biological Engineering, The University of Alabama, Tuscaloosa, Alabama, USA
| | - Rachel Lane
- Department of Chemical and Biological Engineering, The University of Alabama, Tuscaloosa, Alabama, USA
| | - Shreyas S Rao
- Department of Chemical and Biological Engineering, The University of Alabama, Tuscaloosa, Alabama, USA
| |
Collapse
|
9
|
Ko J, Song J, Lee Y, Choi N, Kim HN. Understanding organotropism in cancer metastasis using microphysiological systems. LAB ON A CHIP 2024; 24:1542-1556. [PMID: 38192269 DOI: 10.1039/d3lc00889d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2024]
Abstract
Cancer metastasis, the leading cause of cancer-related deaths, remains a complex challenge in medical science. Stephen Paget's "seed and soil theory" introduced the concept of organotropism, suggesting that metastatic success depends on specific organ microenvironments. Understanding organotropism not only offers potential for curbing metastasis but also novel treatment strategies. Microphysiological systems (MPS), especially organ-on-a-chip models, have emerged as transformative tools in this quest. These systems, blending microfluidics, biology, and engineering, grant precise control over cell interactions within organ-specific microenvironments. MPS enable real-time monitoring, morphological analysis, and protein quantification, enhancing our comprehension of cancer dynamics, including tumor migration, vascularization, and pre-metastatic niches. In this review, we explore innovative applications of MPS in investigating cancer metastasis, particularly focusing on organotropism. This interdisciplinary approach converges the field of science, engineering, and medicine, thereby illuminating a path toward groundbreaking discoveries in cancer research.
Collapse
Affiliation(s)
- Jihoon Ko
- Department of BioNano Technology, Gachon University, Seongnam-si, Gyeonggi-do 13120, Republic of Korea.
| | - Jiyoung Song
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea.
| | - Yedam Lee
- Department of BioNano Technology, Gachon University, Seongnam-si, Gyeonggi-do 13120, Republic of Korea.
| | - Nakwon Choi
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea.
- Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology, Seoul 02792, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Korea
| | - Hong Nam Kim
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea.
- Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology, Seoul 02792, Republic of Korea
- School of Mechanical Engineering, Yonsei University, Seoul 03722, Republic of Korea
- Yonsei-KIST Convergence Research Institute, Yonsei University, Seoul 03722, Republic of Korea
| |
Collapse
|
10
|
Richbourg NR, Irakoze N, Kim H, Peyton SR. Outlook and opportunities for engineered environments of breast cancer dormancy. SCIENCE ADVANCES 2024; 10:eadl0165. [PMID: 38457510 PMCID: PMC10923521 DOI: 10.1126/sciadv.adl0165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Accepted: 02/01/2024] [Indexed: 03/10/2024]
Abstract
Dormant, disseminated breast cancer cells resist treatment and may relapse into malignant metastases after decades of quiescence. Identifying how and why these dormant breast cancer cells are triggered into outgrowth is a key unsolved step in treating latent, metastatic breast cancer. However, our understanding of breast cancer dormancy in vivo is limited by technical challenges and ethical concerns with triggering the activation of dormant breast cancer. In vitro models avoid many of these challenges by simulating breast cancer dormancy and activation in well-controlled, bench-top conditions, creating opportunities for fundamental insights into breast cancer biology that complement what can be achieved through animal and clinical studies. In this review, we address clinical and preclinical approaches to treating breast cancer dormancy, how precisely controlled artificial environments reveal key interactions that regulate breast cancer dormancy, and how future generations of biomaterials could answer further questions about breast cancer dormancy.
Collapse
Affiliation(s)
- Nathan R. Richbourg
- Department of Chemical Engineering, University of Massachusetts Amherst, MA 01003, USA
| | - Ninette Irakoze
- Department of Chemical Engineering, University of Massachusetts Amherst, MA 01003, USA
| | - Hyuna Kim
- Molecular and Cellular Biology Graduate Program, University of Massachusetts Amherst, MA 01003, USA
| | - Shelly R. Peyton
- Department of Chemical Engineering, University of Massachusetts Amherst, MA 01003, USA
- Molecular and Cellular Biology Graduate Program, University of Massachusetts Amherst, MA 01003, USA
- Department of Biomedical Engineering, University of Massachusetts Amherst Amherst, MA 01003, USA
| |
Collapse
|
11
|
Singvogel K, Schittek B. Dormancy of cutaneous melanoma. Cancer Cell Int 2024; 24:88. [PMID: 38419052 PMCID: PMC10903048 DOI: 10.1186/s12935-024-03278-5] [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: 10/09/2023] [Accepted: 02/19/2024] [Indexed: 03/02/2024] Open
Abstract
Many cancer-related deaths including melanoma result from metastases that develop months or years after the initial cancer therapy. Even the most effective drugs and immune therapies rarely eradicate all tumor cells. Instead, they strongly reduce cancer burden, permitting dormant cancer cells to persist in niches, where they establish a cellular homeostasis with their host without causing clinical symptoms. Dormant cancers respond poorly to most drugs and therapies since they do not proliferate and hide in niches. It therefore remains a major challenge to develop novel therapies for dormant cancers. In this review we focus on the mechanisms regulating the initiation of cutaneous melanoma dormancy as well as those which are involved in reawakening of dormant cutaneous melanoma cells. In recent years the role of neutrophils and niche components in reawakening of melanoma cells came into focus and indicate possible future therapeutic applications. Sophisticated in vitro and in vivo melanoma dormancy models are needed to make progress in this field and are discussed.
Collapse
Affiliation(s)
- Kathrin Singvogel
- Division of Dermatooncology, Department of Dermatology, University of Tübingen, Liebermeisterstr. 25, D -72076 , Tübingen, Germany
| | - Birgit Schittek
- Division of Dermatooncology, Department of Dermatology, University of Tübingen, Liebermeisterstr. 25, D -72076 , Tübingen, Germany.
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen, Tübingen, Germany.
| |
Collapse
|
12
|
Katti PD, Jasuja H. Current Advances in the Use of Tissue Engineering for Cancer Metastasis Therapeutics. Polymers (Basel) 2024; 16:617. [PMID: 38475301 PMCID: PMC10934711 DOI: 10.3390/polym16050617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 02/16/2024] [Accepted: 02/20/2024] [Indexed: 03/14/2024] Open
Abstract
Cancer is a leading cause of death worldwide and results in nearly 10 million deaths each year. The global economic burden of cancer from 2020 to 2050 is estimated to be USD 25.2 trillion. The spread of cancer to distant organs through metastasis is the leading cause of death due to cancer. However, as of today, there is no cure for metastasis. Tissue engineering is a promising field for regenerative medicine that is likely to be able to provide rehabilitation procedures to patients who have undergone surgeries, such as mastectomy and other reconstructive procedures. Another important use of tissue engineering has emerged recently that involves the development of realistic and robust in vitro models of cancer metastasis, to aid in drug discovery and new metastasis therapeutics, as well as evaluate cancer biology at metastasis. This review covers the current studies in developing tissue-engineered metastasis structures. This article reports recent developments in in vitro models for breast, prostate, colon, and pancreatic cancer. The review also identifies challenges and opportunities in the use of tissue engineering toward new, clinically relevant therapies that aim to reduce the cancer burden.
Collapse
|
13
|
Rutecki S, Leśniewska-Bocianowska A, Chmielewska K, Matuszewska J, Naumowicz E, Uruski P, Radziemski A, Mikuła-Pietrasik J, Tykarski A, Książek K. Serum starvation-based method of ovarian cancer cell dormancy induction and termination in vitro. Biol Methods Protoc 2023; 8:bpad029. [PMID: 38023348 PMCID: PMC10651433 DOI: 10.1093/biomethods/bpad029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 10/26/2023] [Accepted: 11/01/2023] [Indexed: 12/01/2023] Open
Abstract
Awakening and growth reinitiation by dormant cells may contribute to epithelial ovarian cancer (EOC) relapse. The links between these phenomena are loose because of the limited stock of compelling models of EOC dormancy. Here, we show a simple and convenient dormancy research protocol based on serum starvation. This study was conducted on established EOC cell lines A2780, OVCAR-3, and SKOV-3, as well as on primary EOC cells. Cell growth arrest and proliferation were monitored by assessing the Ki67 antigen, PKH26 fluorescence, and cell cycle distribution. In addition, cells were tested for ERK1/2/p38 MAPK activity ratio, apoptosis, and senescence. The study showed that 72-h serum starvation induces G0/G1 growth arrest of a significant fraction of cells, accompanied by reduced Ki67 and ERK1/2/p38 MAPK activity ratio, without signs of apoptosis or cellular senescence. Moreover, providing cells with 72 h of a medium enriched in 5% serum allows the culture to regain its proliferative potential. At the same time, we attempted to induce and terminate dormancy with Mitomycin C addition and withdrawal, which were unsuccessful. In conclusion, serum starvation is a convenient way to reliably induce dormancy in EOC cells, allowing them to be efficiently awakened for further mechanistic research in vitro.
Collapse
Affiliation(s)
- Szymon Rutecki
- Department of Pathophysiology of Ageing and Civilization Diseases, Poznań University of Medical Sciences, Poznań 61-848, Poland
- Poznan University of Medical Sciences Doctoral School, Poznań 60-812, Poland
| | | | - Klaudia Chmielewska
- Department of Pathophysiology of Ageing and Civilization Diseases, Poznań University of Medical Sciences, Poznań 61-848, Poland
| | - Julia Matuszewska
- Department of Pathophysiology of Ageing and Civilization Diseases, Poznań University of Medical Sciences, Poznań 61-848, Poland
| | - Eryk Naumowicz
- General Surgery Ward, Medical Centre HCP, Poznań 61-485, Poland
| | - Paweł Uruski
- Department of Hypertensiology, Poznań University of Medical Sciences, Poznań 61-848, Poland
| | - Artur Radziemski
- Department of Hypertensiology, Poznań University of Medical Sciences, Poznań 61-848, Poland
| | - Justyna Mikuła-Pietrasik
- Department of Pathophysiology of Ageing and Civilization Diseases, Poznań University of Medical Sciences, Poznań 61-848, Poland
| | - Andrzej Tykarski
- Department of Hypertensiology, Poznań University of Medical Sciences, Poznań 61-848, Poland
| | - Krzysztof Książek
- Department of Pathophysiology of Ageing and Civilization Diseases, Poznań University of Medical Sciences, Poznań 61-848, Poland
| |
Collapse
|
14
|
Closset L, Gultekin O, Salehi S, Sarhan D, Lehti K, Gonzalez-Molina J. The extracellular matrix - immune microenvironment crosstalk in cancer therapy: Challenges and opportunities. Matrix Biol 2023; 121:217-228. [PMID: 37524251 DOI: 10.1016/j.matbio.2023.07.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 07/28/2023] [Accepted: 07/28/2023] [Indexed: 08/02/2023]
Abstract
Targeting the tumour immune microenvironment (TIME) by cancer immunotherapy has led to improved patient outcomes. However, response to these treatments is heterogeneous and cancer-type dependant. The therapeutic activity of classical cancer therapies such as chemotherapy, radiotherapy, and surgical oncology is modulated by alterations of the TIME. A major regulator of immune cell function and resistance to both immune and classical therapies is the extracellular matrix (ECM). Concurrently, cancer therapies reshape the TIME as well as the ECM, causing both pro- and anti-tumour responses. Accordingly, the TIME-ECM crosstalk presents attractive opportunities to improve therapy outcomes. Here, we review the molecular crosstalk between the TIME and the ECM in cancer and its implications in cancer progression and clinical intervention. Additionally, we discuss examples and future directions of ECM and TIME co-targeting in combination with oncological therapies including surgery, chemotherapy, and radiotherapy.
Collapse
Affiliation(s)
- Lara Closset
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Solnavägen 9, Stockholm 171 65, Sweden; Saint-Antoine Research center (CRSA), UMR_S 938, INSERM, Sorbonne Université, Paris F-75012, France
| | - Okan Gultekin
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Solnavägen 9, Stockholm 171 65, Sweden
| | - Sahar Salehi
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Solnavägen 9, Stockholm 171 65, Sweden; Department of Women's and Children's Health, Division of Obstetrics and Gynecology, Karolinska Institutet, Stockholm, Sweden; Department of Pelvic Cancer, Theme Cancer, Karolinska University Hospital, Stockholm, Sweden
| | - Dhifaf Sarhan
- Department of Laboratory Medicine, Division of Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Kaisa Lehti
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Solnavägen 9, Stockholm 171 65, Sweden; Department of Biomedical Laboratory Science, Norwegian University of Science and Technology, Trondheim, Norway
| | - Jordi Gonzalez-Molina
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Solnavägen 9, Stockholm 171 65, Sweden.
| |
Collapse
|
15
|
Caianiello S, Bertolaso M, Militello G. Thinking in 3 dimensions: philosophies of the microenvironment in organoids and organs-on-chip. HISTORY AND PHILOSOPHY OF THE LIFE SCIENCES 2023; 45:14. [PMID: 36949354 DOI: 10.1007/s40656-023-00560-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 02/03/2023] [Indexed: 06/18/2023]
Abstract
Organoids and organs-on-a-chip are currently the two major families of 3D advanced organotypic in vitro culture systems, aimed at reconstituting miniaturized models of physiological and pathological states of human organs. Both share the tenets of the so-called "three-dimensional thinking", a Systems Physiology approach focused on recapitulating the dynamic interactions between cells and their microenvironment. We first review the arguments underlying the "paradigm shift" toward three-dimensional thinking in the in vitro culture community. Then, through a historically informed account of the technical affordances and the epistemic commitments of these two approaches, we highlight how they embody two distinct experimental cultures. We finally argue that the current systematic effort for their integration requires not only innovative "synergistic" engineering solutions, but also conceptual integration between different perspectives on biological causality.
Collapse
Affiliation(s)
- Silvia Caianiello
- Institute for the History of Philosophy and Science in the Modern Age (ISPF), Consiglio Nazionale delle Ricerche, Naples, Italy.
- Stazione Zoologica "Anton Dohrn", Naples, Italy.
| | - Marta Bertolaso
- Faculty of Science and Technology for Sustainable Development and One Health, Universitá Campus Bio-Medico di Roma, Rome, Italy
| | - Guglielmo Militello
- Faculty of Science and Technology for Sustainable Development and One Health, Universitá Campus Bio-Medico di Roma, Rome, Italy
| |
Collapse
|
16
|
Pradhan L, Moore D, Ovadia EM, Swedzinski SL, Cossette T, Sikes RA, van Golen K, Kloxin AM. Dynamic bioinspired coculture model for probing ER + breast cancer dormancy in the bone marrow niche. SCIENCE ADVANCES 2023; 9:eade3186. [PMID: 36888709 PMCID: PMC9995072 DOI: 10.1126/sciadv.ade3186] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 02/03/2023] [Indexed: 05/28/2023]
Abstract
Late recurrences of breast cancer are hypothesized to arise from disseminated tumor cells (DTCs) that reactivate after dormancy and occur most frequently with estrogen receptor-positive (ER+) breast cancer cells (BCCs) in bone marrow (BM). Interactions between the BM niche and BCCs are thought to play a pivotal role in recurrence, and relevant model systems are needed for mechanistic insights and improved treatments. We examined dormant DTCs in vivo and observed DTCs near bone lining cells and exhibiting autophagy. To study underlying cell-cell interactions, we established a well-defined, bioinspired dynamic indirect coculture model of ER+ BCCs with BM niche cells, human mesenchymal stem cells (hMSCs) and fetal osteoblasts (hFOBs). hMSCs promoted BCC growth, whereas hFOBs promoted dormancy and autophagy, regulated in part by tumor necrosis factor-α and monocyte chemoattractant protein 1 receptor signaling. This dormancy was reversible by dynamically changing the microenvironment or inhibiting autophagy, presenting further opportunities for mechanistic and targeting studies to prevent late recurrence.
Collapse
Affiliation(s)
- Lina Pradhan
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19716, USA
| | - DeVonte Moore
- Department of Chemistry and Biochemistry, University of Delaware, Newark, DE 19716, USA
| | - Elisa M. Ovadia
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19716, USA
| | - Samantha L. Swedzinski
- Department of Materials Science and Engineering, University of Delaware, Newark, DE 19716, USA
| | - Travis Cossette
- Office of Laboratory Animal Medicine, University of Delaware, Newark, DE 19716, USA
| | - Robert A. Sikes
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
| | - Kenneth van Golen
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
| | - April M. Kloxin
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19716, USA
- Department of Materials Science and Engineering, University of Delaware, Newark, DE 19716, USA
| |
Collapse
|
17
|
Kondapaneni RV, Shevde LA, Rao SS. A Biomimetic Hyaluronic Acid Hydrogel Models Mass Dormancy in Brain Metastatic Breast Cancer Spheroids. Adv Biol (Weinh) 2023; 7:e2200114. [PMID: 36354182 DOI: 10.1002/adbi.202200114] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 08/15/2022] [Indexed: 11/11/2022]
Abstract
Approximately 90% of breast cancer related mortalities are due to metastasis to distant organs. At the metastatic sites, cancer cells are capable of evading death by exhibiting cellular or mass dormancy. However, the mechanisms involved in attaining dormancy at the metastatic site are not well understood. This is partly due to the lack of experimental models to study metastatic site-specific interactions, particularly in the context of brain metastatic breast cancer (BMBC). Herein, an in vitro hyaluronic acid (HA) hydrogel-based model is developed to study mass dormancy in BMBC. HA hydrogels with a stiffness of ≈0.4 kPa are utilized to mimic the brain extracellular matrix. MDA-MB-231Br or BT474Br3 BMBC spheroids are prepared and cultured on top of HA hydrogels or in suspension for 7 days. HA hydrogel induced a near mass dormant state in spheroids by achieving a balance between proliferating and dead cells. In contrast, these spheroids displayed growth in suspension cultures. The ratio of %p-ERK to %p-p38 positive cells is significantly lower in HA hydrogels compared to suspension cultures. Further, it is demonstrated that hydrogel induced mass dormant state is reversible. Overall, such models provide useful tools to study dormancy in BMBC and could be employed for drug screening.
Collapse
Affiliation(s)
- Raghu Vamsi Kondapaneni
- Department of Chemical and Biological Engineering, The University of Alabama, Tuscaloosa, AL, 35487, USA
| | - Lalita A Shevde
- Department of Pathology, O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, 35233, USA
| | - Shreyas S Rao
- Department of Chemical and Biological Engineering, The University of Alabama, Tuscaloosa, AL, 35487, USA
| |
Collapse
|
18
|
Kondapaneni RV, Warren R, Rao SS. Low dose chemotherapy induces a dormant state in brain metastatic breast cancer spheroids. AIChE J 2022. [DOI: 10.1002/aic.17858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Raghu Vamsi Kondapaneni
- Department of Chemical and Biological Engineering The University of Alabama Tuscaloosa AL USA
| | - Rachel Warren
- Department of Chemical and Biological Engineering The University of Alabama Tuscaloosa AL USA
| | - Shreyas S. Rao
- Department of Chemical and Biological Engineering The University of Alabama Tuscaloosa AL USA
| |
Collapse
|
19
|
Stromal Co-Cultivation for Modeling Breast Cancer Dormancy in the Bone Marrow. Cancers (Basel) 2022; 14:cancers14143344. [PMID: 35884405 PMCID: PMC9320268 DOI: 10.3390/cancers14143344] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 07/05/2022] [Accepted: 07/05/2022] [Indexed: 11/16/2022] Open
Abstract
Cancers metastasize to the bone marrow before primary tumors can be detected. Bone marrow micrometastases are resistant to therapy, and while they are able to remain dormant for decades, they recur steadily and result in incurable metastatic disease. The bone marrow microenvironment maintains the dormancy and chemoresistance of micrometastases through interactions with multiple cell types and through structural and soluble factors. Modeling dormancy in vitro can identify the mechanisms of these interactions. Modeling also identifies mechanisms able to disrupt these interactions or define novel interactions that promote the reawakening of dormant cells. The in vitro modeling of the interactions of cancer cells with various bone marrow elements can generate hypotheses on the mechanisms that control dormancy, treatment resistance and reawakening in vivo. These hypotheses can guide in vivo murine experiments that have high probabilities of succeeding in order to verify in vitro findings while minimizing the use of animals in experiments. This review outlines the existing data on predominant stromal cell types and their use in 2D co-cultures with cancer cells.
Collapse
|
20
|
Kadonosono T, Miyamoto K, Sakai S, Matsuo Y, Kitajima S, Wang Q, Endo M, Niibori M, Kuchimaru T, Soga T, Hirota K, Kizaka-Kondoh S. AGE/RAGE axis regulates reversible transition to quiescent states of ALK-rearranged NSCLC and pancreatic cancer cells in monolayer cultures. Sci Rep 2022; 12:9886. [PMID: 35701529 PMCID: PMC9198021 DOI: 10.1038/s41598-022-14272-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 06/03/2022] [Indexed: 12/17/2022] Open
Abstract
Cancer recurrence due to tumor cell quiescence after therapy and long-term remission is associated with cancer-related death. Previous studies have used cell models that are unable to return to a proliferative state; thus, the transition between quiescent and proliferative states is not well understood. Here, we report monolayer cancer cell models wherein the human non-small cell lung carcinoma cell line H2228 and pancreatic cancer cell line AsPC-1 can be reversibly induced to a quiescent state under hypoxic and serum-starved (HSS) conditions. Transcriptome and metabolome dual-omics profiles of these cells were compared with those of the human lung adenocarcinoma cell line A549, which was unable to enter a quiescent state under HSS conditions. The quiescence-inducible cells had substantially lower intracellular pyruvate and ATP levels in the quiescent state than in the proliferative state, and their response to sudden demand for energy was dramatically reduced. Furthermore, in quiescence-inducible cells, the transition between quiescent and proliferative states of these cells was regulated by the balance between the proliferation-promoting Ras and Rap1 signaling and the suppressive AGE/RAGE signaling. These cell models elucidate the transition between quiescent and proliferative states, allowing the development of drug-screening systems for quiescent tumor cells.
Collapse
Affiliation(s)
- Tetsuya Kadonosono
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, 226-8501, Japan
| | - Kotaro Miyamoto
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, 226-8501, Japan
| | - Shiori Sakai
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, 226-8501, Japan
| | - Yoshiyuki Matsuo
- Department of Human Stress Response Science, Institute of Biomedical Science, Kansai Medical University, Hirakata, 573-1010, Japan
| | - Shojiro Kitajima
- Institute for Advanced Biosciences, Keio University, Tsuruoka, 997-0052, Japan
| | - Qiannan Wang
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, 226-8501, Japan
| | - Minori Endo
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, 226-8501, Japan
| | - Mizuho Niibori
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, 226-8501, Japan
| | - Takahiro Kuchimaru
- Center for Molecular Medicine, Jichi Medical University, Shimotsuke, 329-0498, Japan
| | - Tomoyoshi Soga
- Institute for Advanced Biosciences, Keio University, Tsuruoka, 997-0052, Japan
| | - Kiichi Hirota
- Department of Human Stress Response Science, Institute of Biomedical Science, Kansai Medical University, Hirakata, 573-1010, Japan
| | - Shinae Kizaka-Kondoh
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, 226-8501, Japan.
| |
Collapse
|
21
|
Pally D, Goutham S, Bhat R. Extracellular matrix as a driver for intratumoral heterogeneity. Phys Biol 2022; 19. [PMID: 35545075 DOI: 10.1088/1478-3975/ac6eb0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Accepted: 05/11/2022] [Indexed: 11/12/2022]
Abstract
The architecture of an organ is built through interactions between its native cells and its connective tissue consisting of stromal cells and the extracellular matrix (ECM). Upon transformation through tumorigenesis, such interactions are disrupted and replaced by a new set of intercommunications between malignantly transformed parenchyma, an altered stromal cell population, and a remodeled ECM. In this perspective, we propose that the intratumoral heterogeneity of cancer cell phenotypes is an emergent property of such reciprocal intercommunications, both biochemical and mechanical-physical, which engender and amplify the diversity of cell behavioral traits. An attempt to assimilate such findings within a framework of phenotypic plasticity furthers our understanding of cancer progression.
Collapse
Affiliation(s)
- Dharma Pally
- Molecular Reproduction Development and Genetics, Indian Institute of Science, GA 07, Bangalore, Karnataka, 560012, INDIA
| | - Shyamili Goutham
- Molecular Reproduction Development and Genetics, Indian Institute of Science, GA 07, Bangalore, Karnataka, 560012, INDIA
| | - Ramray Bhat
- Molecular Reproduction Development and Genetics, Indian Institute of Science, GA 07, Bangalore, Karnataka, 560012, INDIA
| |
Collapse
|
22
|
Farino Reyes CJ, Slater JH. Tuning Hydrogel Adhesivity and Degradability to Model the Influence of Premetastatic Niche Matrix Properties on Breast Cancer Dormancy and Reactivation. Adv Biol (Weinh) 2022; 6:e2200012. [PMID: 35277951 PMCID: PMC9090988 DOI: 10.1002/adbi.202200012] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 01/01/2000] [Indexed: 01/27/2023]
Abstract
Dormant, disseminated tumor cells (DTCs) can persist for decades in secondary tissues before being reactivated to form tumors. The properties of the premetastatic niche can influence the DTC phenotype. To better understand how matrix properties of premetastatic niches influence DTC behavior, three hydrogel formulations are implemented to model a permissive niche and two nonpermissive niches. Poly(ethylene glycol) (PEG)-based hydrogels with varying adhesivity ([RGDS]) and degradability ([N-vinyl pyrrolidinone]) are implemented to mimic a permissive niche with high adhesivity and degradability and two nonpermissive niches, one with moderate adhesivity and degradability and one with no adhesivity and high degradability. The influence of matrix properties on estrogen receptor positive (ER+ ) breast cancer cells (MCF7s) is determined via a multimetric analysis. MCF7s cultured in the permissive niche adopted a growth state, while those in the nonpermissive niche with reduced adhesivity and degradability underwent tumor mass dormancy. Complete removal of adhesivity while maintaining high degradability induced single cell dormancy. The ability to mimic reactivation of dormant cells through a dynamic increase in [RGDS] is also demonstrated. This platform provides the capability of inducing growth, dormancy, and reactivation of ER+ breast cancer and can be useful in understanding how premetastatic niche properties influence cancer cell fate.
Collapse
Affiliation(s)
- Cindy J. Farino Reyes
- Department of Biomedical Engineering University of Delaware 590 Avenue 1743, Biomedical Engineering Newark DE 19713 USA
| | - John H. Slater
- Department of Biomedical Engineering University of Delaware 590 Avenue 1743, Biomedical Engineering Newark DE 19713 USA
| |
Collapse
|
23
|
Parker AL, Benguigui M, Fornetti J, Goddard E, Lucotti S, Insua-Rodríguez J, Wiegmans AP. Current challenges in metastasis research and future innovation for clinical translation. Clin Exp Metastasis 2022; 39:263-277. [PMID: 35072851 PMCID: PMC8971179 DOI: 10.1007/s10585-021-10144-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 12/28/2021] [Indexed: 02/06/2023]
Abstract
While immense strides have been made in understanding tumor biology and in developing effective treatments that have substantially improved the prognosis of cancer patients, metastasis remains the major cause of cancer-related death. Improvements in the detection and treatment of primary tumors are contributing to a growing, detailed understanding of the dynamics of metastatic progression. Yet challenges remain in detecting metastatic dissemination prior to the establishment of overt metastases and in predicting which patients are at the highest risk of developing metastatic disease. Further improvements in understanding the mechanisms governing metastasis have great potential to inform the adaptation of existing therapies and the development of novel approaches to more effectively control metastatic disease. This article presents a forward-looking perspective on the challenges that remain in the treatment of metastasis, and the exciting emerging approaches that promise to transform the treatment of metastasis in cancer patients.
Collapse
Affiliation(s)
- Amelia L Parker
- Matrix and Metastasis Lab, Kinghorn Cancer Centre, Garvin Institute of Medical Research, Darlinghurst, NSW, 2010, Australia.
- St Vincent's Clinical School, UNSW Sydney, Sydney, 2052, Australia.
| | - Madeleine Benguigui
- Cell Biology and Cancer Science, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, 31096, Haifa, Israel
| | - Jaime Fornetti
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake, UT, USA
| | - Erica Goddard
- Public Health Sciences Division/Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Serena Lucotti
- Children's Cancer and Blood Foundation Laboratories, Departments of Pediatrics, and Cell and Developmental Biology, Drukier Institute for Children's Health, Meyer Cancer Center, Weill Cornell Medicine, NY, New York, USA
| | - Jacob Insua-Rodríguez
- Department of Physiology and Biophysics, Department of Biological Chemistry, Chao Family Comprehensive Cancer Centre, University of California, Irvine, CA, USA
| | - Adrian P Wiegmans
- Cancer and Ageing Research Program, Centre for Genomics and Personalised Health, Queensland University of Technology (QUT), Translational Research Institute, Woolloongabba, QLD, 4121, Australia
| |
Collapse
|
24
|
3D microgels to quantify tumor cell properties and therapy response dynamics. Biomaterials 2022; 283:121417. [DOI: 10.1016/j.biomaterials.2022.121417] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 01/18/2022] [Accepted: 02/15/2022] [Indexed: 12/21/2022]
|
25
|
Abstract
Dormancy is an evolutionarily conserved protective mechanism widely observed in nature. A pathological example is found during cancer metastasis, where cancer cells disseminate from the primary tumor, home to secondary organs, and enter a growth-arrested state, which could last for decades. Recent studies have pointed toward the microenvironment being heavily involved in inducing, preserving, or ceasing this dormant state, with a strong focus on identifying specific molecular mechanisms and signaling pathways. Increasing evidence now suggests the existence of an interplay between intracellular as well as extracellular biochemical and mechanical cues in guiding such processes. Despite the inherent complexities associated with dormancy, proliferation, and growth of cancer cells and tumor tissues, viewing these phenomena from a physical perspective allows for a more global description, independent from many details of the systems. Building on the analogies between tissues and fluids and thermodynamic phase separation concepts, we classify a number of proposed mechanisms in terms of a thermodynamic metastability of the tumor with respect to growth. This can be governed by interaction with the microenvironment in the form of adherence (wetting) to a substrate or by mechanical confinement of the surrounding extracellular matrix. By drawing parallels with clinical and experimental data, we advance the notion that the local energy minima, or metastable states, emerging in the tissue droplet growth kinetics can be associated with a dormant state. Despite its simplicity, the provided framework captures several aspects associated with cancer dormancy and tumor growth.
Collapse
|
26
|
Janjua KA, Shahzad R, Shehzad A. Development of Novel Cancer Biomarkers for Diagnosis and Prognosis. CANCER BIOMARKERS IN DIAGNOSIS AND THERAPEUTICS 2022:277-343. [DOI: 10.1007/978-981-16-5759-7_11] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
27
|
Knitsch R, AlWahsh M, Raschke H, Lambert J, Hergenröder R. In Vitro Spatio-Temporal NMR Metabolomics of Living 3D Cell Models. Anal Chem 2021; 93:13485-13494. [PMID: 34478621 DOI: 10.1021/acs.analchem.1c02221] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Three-dimensional cell cultures are of growing importance in biochemical research as they represent tissue features more accurately than standard two-dimensional systems, but to investigate these challenging new models an adaptation of established analytical techniques is required. Spatially resolved data for living organoids are needed to gain insight into transport processes and biochemical characteristics of domains with different nutrient supply and waste product removal. Within this work, we present an NMR-based approach to obtain dynamically radial metabolite profiles for cell spheroids, one of the most frequently used 3D models. Our approach combines an easy to reproduce custom-made measurement design, maintaining physiological conditions without inhibition of the NMR experiment, with spatially selective NMR pulse sequences. To overcome the inherently low sensitivity of NMR spectroscopy we excited slices instead of smaller cube-like voxels in combination with an efficient interleaved measurement approach and employed a commercially available cryogenic NMR probe. Finally, radial metabolite profiles could be obtained via double Abel inversion of the measured one-dimensional intensity profiles. Applying this method to Ty82 cancer cell spheroids demonstrates the achieved spatial resolution, for instance confirming exceedingly high lactic acid and strongly decreased glucose concentrations in the oxygen-depleted core of the spheroid. Furthermore, our approach can be employed to investigate fast and slow metabolic changes in single spheroids simultaneously, which is shown as an example of a spheroid degrading over several days after stopping the nutrient supply.
Collapse
Affiliation(s)
- Robert Knitsch
- Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V., Bunsen-Kirchhoff-Str. 11, 44139 Dortmund, Germany
| | - Mohammad AlWahsh
- Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V., Bunsen-Kirchhoff-Str. 11, 44139 Dortmund, Germany.,Institute of Pathology and Medical Research Center (ZMF), University Medical Center Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1, 68167 Mannheim, Germany
| | - Hannes Raschke
- Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V., Bunsen-Kirchhoff-Str. 11, 44139 Dortmund, Germany
| | - Jörg Lambert
- Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V., Bunsen-Kirchhoff-Str. 11, 44139 Dortmund, Germany
| | - Roland Hergenröder
- Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V., Bunsen-Kirchhoff-Str. 11, 44139 Dortmund, Germany
| |
Collapse
|
28
|
Uzuner D, Akkoç Y, Peker N, Pir P, Gözüaçık D, Çakır T. Transcriptional landscape of cellular networks reveal interactions driving the dormancy mechanisms in cancer. Sci Rep 2021; 11:15806. [PMID: 34349126 PMCID: PMC8339123 DOI: 10.1038/s41598-021-94005-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 06/28/2021] [Indexed: 11/22/2022] Open
Abstract
Primary cancer cells exert unique capacity to disseminate and nestle in distant organs. Once seeded in secondary sites, cancer cells may enter a dormant state, becoming resistant to current treatment approaches, and they remain silent until they reactivate and cause overt metastases. To illuminate the complex mechanisms of cancer dormancy, 10 transcriptomic datasets from the literature enabling 21 dormancy–cancer comparisons were mapped on protein–protein interaction networks and gene-regulatory networks to extract subnetworks that are enriched in significantly deregulated genes. The genes appearing in the subnetworks and significantly upregulated in dormancy with respect to proliferative state were scored and filtered across all comparisons, leading to a dormancy–interaction network for the first time in the literature, which includes 139 genes and 1974 interactions. The dormancy interaction network will contribute to the elucidation of cellular mechanisms orchestrating cancer dormancy, paving the way for improvements in the diagnosis and treatment of metastatic cancer.
Collapse
Affiliation(s)
- Dilara Uzuner
- Department of Bioengineering, Gebze Technical University, 41400, Kocaeli, Turkey
| | - Yunus Akkoç
- Koç University Research Center for Translational Medicine (KUTTAM), Zeytinburnu, 34010, Istanbul, Turkey
| | - Nesibe Peker
- Koç University Research Center for Translational Medicine (KUTTAM), Zeytinburnu, 34010, Istanbul, Turkey
| | - Pınar Pir
- Department of Bioengineering, Gebze Technical University, 41400, Kocaeli, Turkey
| | - Devrim Gözüaçık
- Koç University Research Center for Translational Medicine (KUTTAM), Zeytinburnu, 34010, Istanbul, Turkey.,Koç University School of Medicine, Sarıyer , 34450, Istanbul, Turkey.,SUNUM Nanotechnology Research and Application Center, Tuzla, 34956, Istanbul, Turkey
| | - Tunahan Çakır
- Department of Bioengineering, Gebze Technical University, 41400, Kocaeli, Turkey.
| |
Collapse
|
29
|
Clark AM, Allbritton NL, Wells A. Integrative microphysiological tissue systems of cancer metastasis to the liver. Semin Cancer Biol 2021; 71:157-169. [PMID: 32580025 PMCID: PMC7750290 DOI: 10.1016/j.semcancer.2020.06.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 06/10/2020] [Accepted: 06/10/2020] [Indexed: 02/07/2023]
Abstract
The liver is the most commonly involved organ in metastases from a wide variety of solid tumors. The use of biologically and cellularly complex liver tissue systems have shown that tumor cell behavior and therapeutic responses are modulated within the liver microenvironment and in ways distinct from the behaviors in the primary locations. These microphysiological systems have provided unexpected and powerful insights into the tumor cell biology of metastasis. However, neither the tumor nor the liver exist in an isolated tissue situation, having to function within a complete body and respond to systemic events as well as those in other organs. To examine the influence of one organ on the function of other tissues, microphysiological systems are being linked. Herein, we discuss extending this concept to tumor metastases by integrating complex models of the primary tumor with the liver metastatic environment. In addition, inflammatory organs and the immune system can be incorporated into these multi-organ systems to probe the effects on tumor behavior and cancer treatments.
Collapse
Affiliation(s)
- Amanda M Clark
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15261, USA; VA Pittsburgh Healthcare System, Pittsburgh, PA 15213, USA; UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA 15213, USA.
| | - Nancy L Allbritton
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
| | - Alan Wells
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15261, USA; VA Pittsburgh Healthcare System, Pittsburgh, PA 15213, USA; UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA 15213, USA; Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15260, USA; Department of Computational & Systems Biology, University of Pittsburgh, Pittsburgh, PA 15260, USA
| |
Collapse
|
30
|
Farino Reyes CJ, Pradhan S, Slater JH. The Influence of Ligand Density and Degradability on Hydrogel Induced Breast Cancer Dormancy and Reactivation. Adv Healthc Mater 2021; 10:e2002227. [PMID: 33929776 PMCID: PMC8555704 DOI: 10.1002/adhm.202002227] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 03/26/2021] [Indexed: 01/07/2023]
Abstract
The role of hydrogel properties in regulating the phenotype of triple negative metastatic breast cancer is investigated using four cell lines: the MDA-MB-231 parental line and three organotropic sublines BoM-1833 (bone-tropic), LM2-4175 (lung-tropic), and BrM2a-831 (brain-tropic). Each line is encapsulated and cultured for 15 days in three poly(ethylene glycol) (PEG)-based hydrogel formulations composed of proteolytically degradable PEG, integrin-ligating RGDS, and the non-degradable crosslinker N-vinyl pyrrolidone. Dormancy-associated metrics including viable cell density, proliferation, metabolism, apoptosis, chemoresistance, phosphorylated-ERK and -p38, and morphological characteristics are quantified. A multimetric classification approach is implemented to categorize each hydrogel-induced phenotype as: 1) growth, 2) balanced tumor dormancy, 3) balanced cellular dormancy, or 4) restricted survival, cellular dormancy. Hydrogels with high adhesivity and degradability promote growth. Hydrogels with no adhesivity, but high degradability, induce restricted survival, cellular dormancy in the parental line and balanced cellular dormancy in the organotropic lines. Hydrogels with reduced adhesivity and degradability induce balanced cellular dormancy in the parental and lung-tropic lines and balanced tumor mass dormancy in bone- and brain-tropic lines. The ability to induce escape from dormancy via dynamic incorporation of RGDS is also presented. These results demonstrate that ECM properties and organ-tropism synergistically regulate cancer cell phenotype and dormancy.
Collapse
Affiliation(s)
- Cindy J Farino Reyes
- Department of Biomedical Engineering, University of Delaware, 590 Avenue 1743, Biomedical Engineering, Newark, DE, 19713, USA
| | - Shantanu Pradhan
- Department of Biomedical Engineering, University of Delaware, 590 Avenue 1743, Biomedical Engineering, Newark, DE, 19713, USA
| | - John H Slater
- Department of Biomedical Engineering, University of Delaware, 590 Avenue 1743, Biomedical Engineering, Newark, DE, 19713, USA
| |
Collapse
|
31
|
Preciado J, Lam T, Azarin SM, Lou E, Aksan A. Induction of dormancy by confinement: An agarose-silica biomaterial for isolating and analyzing dormant cancer cells. J Biomed Mater Res B Appl Biomater 2021; 109:2117-2130. [PMID: 33983681 DOI: 10.1002/jbm.b.34859] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 04/12/2021] [Accepted: 04/24/2021] [Indexed: 11/07/2022]
Abstract
The principal cause of cancer deaths is the residual disease, which eventually results in metastases. Certain metastases are induced by disseminated dormancy-capable single cancer cells that can reside within the body undetected for months to years. Awakening of the dormant cells starts a cascade resulting in the patient's demise. Despite its established clinical significance, dormancy research and its clinical translation have been hindered by lack of in vitro models that can identify, isolate, and analyze dormancy-capable cells. We have previously shown that immobilization of cells in a stiff microenvironment induces dormancy in dormancy-capable cell lines. In this communication, we present a novel biomaterial and an in vitro immobilization method to isolate, analyze, and efficiently recover dormancy-capable cancer cells. MCF-7, MDA-MB-231, and MDA-MB-468 cells were individually coated with agarose using a microfluidic flow-focusing device. Coated cells were then immobilized in a rigid and porous silica gel. Dormancy induction by this process was validated by decreased Ki-67 expression, increased p38/ERK activity ratio, and reduced expression of CDK-2, cyclins D1, and E1. We showed that we can reliably and repeatedly induce dormancy in dormancy-capable MCF-7 cells and enhance the dormancy-capable sub-population in MDA-MB-231 cells. As expected, dormancy-resistant MDA-MB-468 cells did not survive immobilization. The dormant cells could be awakened on demand, by digesting the agarose gel in situ, and efficiently recovered by magnetically separating the silica gel, making the cells available for downstream analysis and testing. The awakened cells were shown to regain motility immediately, proliferating, and migrating normally.
Collapse
Affiliation(s)
- Julian Preciado
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota, USA
| | - Tiffany Lam
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, Minnesota, USA
| | - Samira M Azarin
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, Minnesota, USA
| | - Emil Lou
- Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, Minnesota, USA
| | - Alptekin Aksan
- Department of Mechanical Engineering, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
32
|
Tumor Dormancy: Implications for Invasion and Metastasis. Int J Mol Sci 2021; 22:ijms22094862. [PMID: 34064392 PMCID: PMC8124645 DOI: 10.3390/ijms22094862] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/25/2021] [Accepted: 04/28/2021] [Indexed: 12/14/2022] Open
Abstract
Tumor dormancy refers to a critical stage of cancer development when tumor cells are present, but cancer does not progress. It includes both the concept of cellular dormancy, indicating the reversible switch of a cancer cell to a quiescent state, and that of tumor mass dormancy, indicating the presence of neoplastic masses that have reached cell population equilibrium via balanced growth/apoptosis rates. Tumor dormancy provides the conceptual framework, potentially explaining a major challenge in clinical oncology, tumor recurrence, which may occur years after cancer diagnosis. The mechanisms by which tumors are kept dormant, and what triggers their reawakening, are fundamental questions in cancer biology. It seems that a plethora of intracellular pathways and extracellular factors are involved in this process, rewiring the cells to plastically alter their metabolic and proliferative status. This phenomenon is highly dynamic in space and time. Mechanistic insights into both cellular and tumor dormancy have provided the rationale for targeting this otherwise stable period of cancer development, in order to prevent recurrence and maximize therapeutic benefit.
Collapse
|
33
|
Haase K, Offeddu GS, Gillrie MR, Kamm RD. Endothelial Regulation of Drug Transport in a 3D Vascularized Tumor Model. ADVANCED FUNCTIONAL MATERIALS 2020; 30:2002444. [PMID: 33692661 PMCID: PMC7939067 DOI: 10.1002/adfm.202002444] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Indexed: 05/06/2023]
Abstract
Drug discovery and efficacy in cancer treatments are limited by the inability of pre-clinical models to predict successful outcomes in humans. Limitations remain partly due to their lack of a physiologic tumor microenvironment (TME), which plays a considerable role in drug delivery and tumor response to therapy. Chemotherapeutics and immunotherapies rely on transport through the vasculature, via the smallest capillaries and stroma to the tumor, where passive and active transport processes are at play. Here, a 3D vascularized tumor on-chip is used to examine drug delivery in a relevant TME within a large bed of perfusable vasculature. This system demonstrates highly localized pathophysiological effects of two tumor spheroids (Skov3 and A549) which cause significant changes in vessel density and barrier function. Paclitaxel (Taxol) uptake is examined through diffusivity measurements, functional efflux assays and accumulation of the fluorescent-conjugated drug within the TME. Due to vascular and stromal contributions, differences in the response of vascularized tumors to Taxol (shrinkage and CD44 expression) are apparent compared with simpler models. This model specifically allows for examination of spatially resolved tumor-associated endothelial dysfunction, likely improving the representation of in vivo drug distribution, and has potential for development into a more predictable model of drug delivery.
Collapse
Affiliation(s)
- Kristina Haase
- Massachusetts Institute of Technology, Massachusetts, 02139, USA
| | | | - Mark R Gillrie
- Massachusetts Institute of Technology, Massachusetts, 02139, USA; University of Calgary, Calgary, T2N 1N4, Canada
| | - Roger D Kamm
- Massachusetts Institute of Technology, Massachusetts, 02139, USA
| |
Collapse
|
34
|
Islam MS, Dasgupta H, Basu M, Roy A, Alam N, Roychoudhury S, Kumar Panda C. Reduction of nuclear Y654-p-β-catenin expression through SH3GL2-meditated downregulation of EGFR in chemotolerance TNBC: Clinical and prognostic importance. J Cell Physiol 2020; 235:8114-8128. [PMID: 31960967 DOI: 10.1002/jcp.29466] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 01/07/2020] [Indexed: 12/11/2022]
Abstract
Triple negative breast cancer (TNBC) originates from a less differentiated ductal cell of breast, which is less sensitive to chemotherapy. The chemotolerance mechanism of TNBC has not yet been studied in detail. For this reason, molecular profiles (expression/genetic/epigenetic) of Y654-p-β-catenin (active) and its kinase epidermal growth factor receptor (EGFR) along with SH3GL2 (regulator of EGFR homeostasis) were compared between neoadjuvant chemotherapy treated (NACT) and pretherapeutic TNBC samples. Reduced nuclear expression of Y654-p-β-catenin protein with low proliferation index and CD44 prevalence showed concordance with reduced expression of EGFR/Y1045-p-EGFR proteins in the NACT samples than the pretherapeutic TNBC samples. Infrequent messenger RNA expression, gene amplification (10-32.5%), and mutation (1%) of EGFR were seen in the TNBC samples irrespective of therapy, suggesting the importance of EGFR protein stabilization in this tumor. The upregulation of SH3GL2 seen in the NACT samples in contrast to the pretherapeutic samples might be due to its promoter hypomethylation, as seen in the quantitative methylation assay. A similar trend of upregulation of SH3GL2 and downregulation of EGFR, Y1045-p-EGFR, Y654-p-β-catenin were seen in the MDA-MB-231 cell line using antharacycline antitumor drugs (doxorubicin/nogalamycin). The NACT patients with reduced expression of Y654-p-β-catenin and/or EGFR and high expression of SH3GL2 showed comparatively better prognosis than the pretherapeutic patients. Thus, our study showed that reduced nuclear expression of Y654-p-β-catenin in NACT samples due to downregulation of EGFR protein through promoter hypomethylation-mediated upregulation of SH3GL2, resulting in low proliferation index/CD44 prevalence with better prognosis of the NACT patients, might have an important role in the chemotolerance of TNBC.
Collapse
Affiliation(s)
- Md Saimul Islam
- Department of Oncogene Regulation, Chittaranjan National Cancer Institute, Kolkata, West Bengal, India
| | - Hemantika Dasgupta
- Department of Oncogene Regulation, Chittaranjan National Cancer Institute, Kolkata, West Bengal, India
| | - Mukta Basu
- Department of Oncogene Regulation, Chittaranjan National Cancer Institute, Kolkata, West Bengal, India
| | - Anup Roy
- Department of Pathology, Nil Ratan Sircar Medical College and Hospital, Kolkata, West Bengal, India
| | - Neyaz Alam
- Department of Surgical Oncology, Chittaranjan National Cancer Institute, Kolkata, West Bengal, India
| | - Susanta Roychoudhury
- Molecular Biology and Basic Research Division, Saroj Gupta Cancer Centre and Research Institute, Kolkata, West Bengal, India
| | - Chinmay Kumar Panda
- Department of Oncogene Regulation, Chittaranjan National Cancer Institute, Kolkata, West Bengal, India
| |
Collapse
|
35
|
Sistigu A, Musella M, Galassi C, Vitale I, De Maria R. Tuning Cancer Fate: Tumor Microenvironment's Role in Cancer Stem Cell Quiescence and Reawakening. Front Immunol 2020; 11:2166. [PMID: 33193295 PMCID: PMC7609361 DOI: 10.3389/fimmu.2020.02166] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 08/10/2020] [Indexed: 12/12/2022] Open
Abstract
Cancer cell dormancy is a common feature of human tumors and represents a major clinical barrier to the long-term efficacy of anticancer therapies. Dormant cancer cells, either in primary tumors or disseminated in secondary organs, may reawaken and relapse into a more aggressive disease. The mechanisms underpinning dormancy entry and exit strongly resemble those governing cancer cell stemness and include intrinsic and contextual cues. Cellular and molecular components of the tumor microenvironment persistently interact with cancer cells. This dialog is highly dynamic, as it evolves over time and space, strongly cooperates with intrinsic cell nets, and governs cancer cell features (like quiescence and stemness) and fate (survival and outgrowth). Therefore, there is a need for deeper insight into the biology of dormant cancer (stem) cells and the mechanisms regulating the equilibrium quiescence-versus-proliferation are vital in our pursuit of new therapeutic opportunities to prevent cancer from recurring. Here, we review and discuss microenvironmental regulations of cancer dormancy and its parallels with cancer stemness, and offer insights into the therapeutic strategies adopted to prevent a lethal recurrence, by either eradicating resident dormant cancer (stem) cells or maintaining them in a dormant state.
Collapse
Affiliation(s)
- Antonella Sistigu
- Istituto di Patologia Generale, Università Cattolica del Sacro Cuore, Rome, Italy.,Tumor Immunology and Immunotherapy Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Martina Musella
- Istituto di Patologia Generale, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Claudia Galassi
- Istituto di Patologia Generale, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Ilio Vitale
- IIGM - Italian Institute for Genomic Medicine, c/o IRCSS Candiolo (TO), Candiolo, Italy.,Candiolo Cancer Institute, FPO - IRCCS, Candiolo, Italy
| | - Ruggero De Maria
- Istituto di Patologia Generale, Università Cattolica del Sacro Cuore, Rome, Italy.,Fondazione Policlinico Universitario "A. Gemelli" - IRCCS, Rome, Italy
| |
Collapse
|
36
|
Rajan N, Khanal T, Ringel MD. Progression and dormancy in metastatic thyroid cancer: concepts and clinical implications. Endocrine 2020; 70:24-35. [PMID: 32779092 PMCID: PMC7530083 DOI: 10.1007/s12020-020-02453-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 08/01/2020] [Indexed: 02/07/2023]
Abstract
Distant metastasis classically has been defined as a late-stage event in cancer progression. However, it has become clear that metastases also may occur early in the "lifetime" of a cancer and that they may remain stable at distant sites. This stability of metastatic cancer deposits has been termed "metastatic dormancy" or, as we term it, "metastatic progression dormancy" as the progression either may reflect growth of already existing metastases or new cancer spread. Biologically, dormancy is the presence of nongrowing, static metastatic cells that survive over time. Clinically, dormancy is defined by stability in tumor markers, imaging, and clinical course. Metastatic well-differentiated thyroid cancer offers an excellent tumor type to understand these processes for several reasons: (1) primary therapy often includes removal of the entire gland with ablation of residual normal tissue thereby removing one source for new metastases; (2) the presence of a sensitive biochemical and radiographic monitoring tests enabling monitoring of metastasis throughout the progression process; and (3) its tendency toward prolonged clinical dormancy that can last for years or decades be followed by progression. This latter factor provides opportunities to define therapeutic targets and/or markers of progression. In this review, we will discuss concepts of metastatic progression dormancy and the factors that drive both long-term stability and loss of dormancy with a focus on thyroid cancer.
Collapse
Affiliation(s)
- Neel Rajan
- Division of Endocrinology, Diabetes, and Metabolism, Arthur G. James Comprehensive Center, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Tilak Khanal
- Division of Endocrinology, Diabetes, and Metabolism, Arthur G. James Comprehensive Center, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Matthew D Ringel
- Division of Endocrinology, Diabetes, and Metabolism, Arthur G. James Comprehensive Center, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA.
| |
Collapse
|
37
|
Farino CJ, Pradhan S, Slater JH. The Influence of Matrix-Induced Dormancy on Metastatic Breast Cancer Chemoresistance. ACS APPLIED BIO MATERIALS 2020; 3:5832-5844. [DOI: 10.1021/acsabm.0c00549] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Cindy J. Farino
- Department of Biomedical Engineering, University of Delaware, 150 Academy Street, 161 Colburn Lab, Newark, Delaware 19716, United States
| | - Shantanu Pradhan
- Department of Biomedical Engineering, University of Delaware, 150 Academy Street, 161 Colburn Lab, Newark, Delaware 19716, United States
| | - John H. Slater
- Department of Biomedical Engineering, University of Delaware, 150 Academy Street, 161 Colburn Lab, Newark, Delaware 19716, United States
- Department of Material Science and Engineering, University of Delaware, 201 DuPont Hall, Newark, Delaware 19716, United States
- Delaware Biotechnology Institute, 15 Innovation Way, Newark, Delaware 19711, United States
| |
Collapse
|
38
|
Buschhaus JM, Humphries BA, Eckley SS, Robison TH, Cutter AC, Rajendran S, Haley HR, Bevoor AS, Luker KE, Luker GD. Targeting disseminated estrogen-receptor-positive breast cancer cells in bone marrow. Oncogene 2020; 39:5649-5662. [PMID: 32678295 PMCID: PMC7442734 DOI: 10.1038/s41388-020-01391-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 06/08/2020] [Accepted: 07/07/2020] [Indexed: 12/19/2022]
Abstract
Estrogen receptor-positive (ER+) breast cancer can recur up to 20 years after initial diagnosis. Delayed recurrences arise from disseminated tumors cells (DTCs) in sites such as bone marrow that remain quiescent during endocrine therapy and subsequently proliferate to produce clinically detectable metastases. Identifying therapies that eliminate DTCs and/or effectively target cells transitioning to proliferation promises to reduce risk of recurrence. To tackle this problem, we utilized a 3D co-culture model incorporating ER+ breast cancer cells and bone marrow mesenchymal stem cells to represent DTCs in a bone marrow niche. 3D co-cultures maintained cancer cells in a quiescent, viable state as measured by both single-cell and population-scale imaging. Single-cell imaging methods for metabolism by fluorescence lifetime (FLIM) of NADH and signaling by kinases Akt and ERK revealed that breast cancer cells utilized oxidative phosphorylation and signaling by Akt to a greater extent both in 3D co-cultures and a mouse model of ER+ breast cancer cells in bone marrow. Using our 3D co-culture model, we discovered that combination therapies targeting oxidative phosphorylation via the thioredoxin reductase (TrxR) inhibitor, D9, and the Akt inhibitor, MK-2206, preferentially eliminated breast cancer cells without altering viability of bone marrow stromal cells. Treatment of mice with disseminated ER+ human breast cancer showed that D9 plus MK-2206 blocked formation of new metastases more effectively than tamoxifen. These data establish an integrated experimental system to investigate DTCs in bone marrow and identify combination therapy against metabolic and kinase targets as a promising approach to effectively target these cells and reduce risk of recurrence in breast cancer.
Collapse
Affiliation(s)
- Johanna M Buschhaus
- Department of Biomedical Engineering, University of Michigan, 2200 Bonisteel, Blvd., Ann Arbor, MI, 48109-2099, USA
- Center for Molecular Imaging, Department of Radiology, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA
| | - Brock A Humphries
- Center for Molecular Imaging, Department of Radiology, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA
| | - Samantha S Eckley
- Unit for Laboratory Animal Medicine, University of Michigan, 412 Victor Vaughan, Ann Arbor, MI, 48109-2200, USA
- Office of Animal Resources, University of Iowa, Iowa City, IA, USA
| | - Tanner H Robison
- Department of Biomedical Engineering, University of Michigan, 2200 Bonisteel, Blvd., Ann Arbor, MI, 48109-2099, USA
- Center for Molecular Imaging, Department of Radiology, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA
| | - Alyssa C Cutter
- Center for Molecular Imaging, Department of Radiology, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA
| | - Shrila Rajendran
- Center for Molecular Imaging, Department of Radiology, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA
| | - Henry R Haley
- Center for Molecular Imaging, Department of Radiology, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA
| | - Avinash S Bevoor
- Center for Molecular Imaging, Department of Radiology, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA
| | - Kathryn E Luker
- Center for Molecular Imaging, Department of Radiology, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA
| | - Gary D Luker
- Department of Biomedical Engineering, University of Michigan, 2200 Bonisteel, Blvd., Ann Arbor, MI, 48109-2099, USA.
- Center for Molecular Imaging, Department of Radiology, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA.
- Department of Microbiology and Immunology, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA.
| |
Collapse
|
39
|
Islam S, Dasgupta H, Basu M, Roy A, Alam N, Roychoudhury S, Panda CK. Downregulation of beta-catenin in chemo-tolerant TNBC through changes in receptor and antagonist profiles of the WNT pathway: Clinical and prognostic implications. Cell Oncol (Dordr) 2020; 43:725-741. [PMID: 32430683 DOI: 10.1007/s13402-020-00525-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 03/30/2020] [Accepted: 04/16/2020] [Indexed: 02/01/2023] Open
Abstract
PURPOSE In approximately 30% of triple-negative breast cancer (TNBC) patients a complete pathological response is achieved. However, after neo-adjuvant chemotherapy treatment (NACT) residual tumour cells can be intrinsically resistant to chemotherapy. In this study, associations of the WNT/beta-catenin pathway with chemo-tolerance of NACT treated TNBC patients were compared to that of pre-treatment TNBC patients. METHODS Expression analyses were performed in both pre-treatment and NACT treated TNBC samples using immunohistochemistry and qRT-PCR, along with DNA copy number variation (CNV) and promoter methylation analyses to elucidate the mechanism(s) underlying chemo-tolerance. In addition, in vitro validation experiments were performed in TNBC cells followed by in vivo clinicopathological correlation analyses. RESULTS A reduced expression (41.1%) of nuclear beta-catenin together with a low proliferation index was observed in NACT samples, whereas a high expression (59.0%) was observed in pre-treatment samples. The reduced nuclear expression of beta-catenin in the NACT samples showed concordance with reduced expression levels (47-52.9%) of its associated receptors (FZD7 and LRP6) and increased expression levels (35.2-41.1%) of its antagonists (SFRP1, SFRP2, DKK1) compared to those in the pre-treatment samples. The expression levels of the receptors showed no concordance with its respective gene copy number/mRNA expression statuses, regardless treatment. Interestingly, however, significant increases in promoter hypomethylation of the antagonists were observed in the NACT samples compared to the pre-treatment samples. Similar expression patterns of the antagonists, receptors and beta-catenin were observed in the TNBC-derived cell line MDA-MB-231 using the anthracyclines doxorubicin and nogalamycin, suggesting the importance of promoter hypomethylation in chemotolerance. NACT patients showing reduced receptor and/or beta-catenin expression levels and high antagonist expression levels exhibited a comparatively better prognosis than the pre-treatment patients. CONCLUSIONS Our data suggest that reduced nuclear expression of beta-catenin in NACT TNBC samples, due to downregulation of its receptors and upregulation of its antagonists through promoter hypomethylation of the WNT pathway, plays an important role in chemo-tolerance.
Collapse
Affiliation(s)
- Saimul Islam
- Department of Oncogene Regulation, Chittaranjan National Cancer Institute, 37, S.P. Mukherjee Road, Kolkata, West Bengal, 700026, India
| | - Hemantika Dasgupta
- Department of Oncogene Regulation, Chittaranjan National Cancer Institute, 37, S.P. Mukherjee Road, Kolkata, West Bengal, 700026, India
| | - Mukta Basu
- Department of Oncogene Regulation, Chittaranjan National Cancer Institute, 37, S.P. Mukherjee Road, Kolkata, West Bengal, 700026, India
| | - Anup Roy
- Department of Pathology, Nil Ratan Sircar Medical College and Hospital, 138, Acharya Jagadish Chandra Bose Rd, 700014, Kolkata, India
| | - Neyaz Alam
- Department of Surgical Oncology, Chittaranjan National Cancer Institute, 37, S.P. Mukherjee Road, Kolkata, West Bengal, 700026, India
| | - Susanta Roychoudhury
- Saroj Gupta Cancer Centre and Research Institute, Thakurpukur, Kolkata, 700 063, India
| | - Chinmay Kumar Panda
- Department of Oncogene Regulation, Chittaranjan National Cancer Institute, 37, S.P. Mukherjee Road, Kolkata, West Bengal, 700026, India.
| |
Collapse
|
40
|
Valcourt DM, Kapadia CH, Scully MA, Dang MN, Day ES. Best Practices for Preclinical In Vivo Testing of Cancer Nanomedicines. Adv Healthc Mater 2020; 9:e2000110. [PMID: 32367687 PMCID: PMC7473451 DOI: 10.1002/adhm.202000110] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 03/17/2020] [Indexed: 01/06/2023]
Abstract
Significant advances have been made in the development of nanoparticles for cancer treatment in recent years. Despite promising results in preclinical animal models, cancer nanomedicines often fail in clinical trials. This failure rate could be reduced by defining stringent criteria for testing and quality control during the design and development stages, and by performing carefully planned preclinical studies in relevant animal models. This article discusses best practices for the evaluation of nanomedicines in murine tumor models. First, a recommended set of experiments to perform is introduced, including discussion of the types of data to collect during these studies. This is followed by an outline of various tumor models and their clinical relevance. Next, different routes of nanoparticle administration are overviewed, followed by a summary of important controls to include in in vivo studies of nanomedicine. Finally, animal welfare considerations are discussed, and an overview of the steps involved in achieving US Food and Drug Administration approval after animal studies are completed is provided. Researchers should use this report as a guideline for effective preclinical evaluation of cancer nanomedicine. As the community adopts best practices for in vivo testing, the rate of clinical translation of cancer nanomedicines is likely to improve.
Collapse
Affiliation(s)
- Danielle M Valcourt
- Department of Biomedical Engineering, University of Delaware, 161 Colburn Lab, Newark, DE, 19716, USA
| | - Chintan H Kapadia
- Department of Biomedical Engineering, University of Delaware, 161 Colburn Lab, Newark, DE, 19716, USA
| | - Mackenzie A Scully
- Department of Biomedical Engineering, University of Delaware, 161 Colburn Lab, Newark, DE, 19716, USA
| | - Megan N Dang
- Department of Biomedical Engineering, University of Delaware, 161 Colburn Lab, Newark, DE, 19716, USA
| | - Emily S Day
- Department of Biomedical Engineering, University of Delaware, 161 Colburn Lab, Newark, DE, 19716, USA
- Department of Materials Science & Engineering, University of Delaware, 201 DuPont Hall, Newark, DE, 19716, USA
- Helen F. Graham Cancer Center & Research Institute, 4701 Ogletown Stanton Road, Newark, DE, 19713, USA
| |
Collapse
|
41
|
Narkhede AA, Crenshaw JH, Crossman DK, Shevde LA, Rao SS. An in vitro hyaluronic acid hydrogel based platform to model dormancy in brain metastatic breast cancer cells. Acta Biomater 2020; 107:65-77. [PMID: 32119920 DOI: 10.1016/j.actbio.2020.02.039] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 02/19/2020] [Accepted: 02/25/2020] [Indexed: 01/07/2023]
Abstract
Breast cancer cells (BCCs) can remain dormant at the metastatic site, which when revoked leads to formation of metastasis several years after the treatment of primary tumor. Particularly, awakening of dormant BCCs in the brain results in breast cancer brain metastasis (BCBrM) which marks the most advanced stage of the disease with a median survival period of ~4-16 months. However, our understanding of dormancy associated with BCBrM remains obscure, in part, due to the lack of relevant in vitro platforms to model dormancy associated with BCBrM. To address this need, we developed an in vitro hyaluronic acid (HA) hydrogel platform to model dormancy in brain metastatic BCCs via exploiting the bio-physical cues provided by HA hydrogels while bracketing the normal brain and metastatic brain malignancy relevant stiffness range. In this system, we observed that MDA-MB-231Br and BT474Br3 brain metastatic BCCs exhibited a dormant phenotype when cultured on soft (0.4 kPa) HA hydrogel compared to stiff (4.5 kPa) HA hydrogel as characterized by significantly lower EdU and Ki67 positivity. Further, we demonstrated the nuclear localization of p21 and p27 (markers associated with dormancy) in dormant MDA-MB-231Br cells contrary to their cytoplasmic localization in the proliferative population. We also demonstrated that the stiffness-based dormancy in MDA-MB-231Br cells was reversible and was, in part, mediated by focal adhesion kinases and the initial cell seeding density. Finally, RNA sequencing confirmed the dormant phenotype in MDA-MB-231Br cells. This platform could further our understanding of dormancy in BCBrM and could be adapted for anti-metastatic drug screening. STATEMENT OF SIGNIFICANCE: Our understanding of dormancy associated with BCBrM remains obscure, in part, due to the lack of relevant in vitro platforms to model dormancy associated with BCBrM. Herein, we present a HA hydrogel-based platform to model dormancy in brain metastatic BCCs while recapitulating key aspects of brain microenvironment. We demonstrated that the biophysical cues provided the HA hydrogel mediates dormancy in brain metastatic BCCs by assessing both proliferation and cell cycle arrest markers. We also established the role of focal adhesion kinases and initial cell seeding density in the stiffness-mediated dormancy in brain metastatic BCCs. Further, RNA-seq. confirmed the dormant phenotype in brain metastatic BCCs. This platform could be utilized to further our understanding of microenvironmental regulation of dormancy in BCBrM.
Collapse
Affiliation(s)
- Akshay A Narkhede
- Department of Chemical and Biological Engineering, The University of Alabama, Tuscaloosa, AL 35487-0203, USA
| | - James H Crenshaw
- Department of Chemical and Biological Engineering, The University of Alabama, Tuscaloosa, AL 35487-0203, USA
| | - David K Crossman
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Lalita A Shevde
- Department of Pathology, O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Shreyas S Rao
- Department of Chemical and Biological Engineering, The University of Alabama, Tuscaloosa, AL 35487-0203, USA.
| |
Collapse
|
42
|
Montagner M, Sahai E. In vitro Models of Breast Cancer Metastatic Dormancy. Front Cell Dev Biol 2020; 8:37. [PMID: 32195244 PMCID: PMC7062644 DOI: 10.3389/fcell.2020.00037] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 01/15/2020] [Indexed: 12/12/2022] Open
Abstract
Delayed relapses at distant sites are a common clinical observation for certain types of cancers after removal of primary tumor, such as breast and prostate cancer. This evidence has been explained by postulating a long period during which disseminated cancer cells (DCCs) survive in a foreign environment without developing into overt metastasis. Because of the asymptomatic nature of this phenomenon, isolation, and analysis of disseminated dormant cancer cells from clinically disease-free patients is ethically and technically highly problematic and currently these data are largely limited to the bone marrow. That said, detecting, profiling and treating indolent metastatic lesions before the onset of relapse is the imperative. To overcome this major limitation many laboratories developed in vitro models of the metastatic niche for different organs and different types of cancers. In this review we focus specifically on in vitro models designed to study metastatic dormancy of breast cancer cells (BCCs). We provide an overview of the BCCs employed in the different organotypic systems and address the components of the metastatic microenvironment that have been shown to impact on the dormant phenotype: tissue architecture, stromal cells, biochemical environment, oxygen levels, cell density. A brief description of the organ-specific in vitro models for bone, liver, and lung is provided. Finally, we discuss the strategies employed so far for the validation of the different systems.
Collapse
Affiliation(s)
- Marco Montagner
- Department of Molecular Medicine, School of Medicine and Surgery, University of Padua, Padua, Italy
| | - Erik Sahai
- The Francis Crick Institute, London, United Kingdom
| |
Collapse
|
43
|
Kondapaneni RV, Rao SS. Matrix stiffness and cluster size collectively regulate dormancy versus proliferation in brain metastatic breast cancer cell clusters. Biomater Sci 2020; 8:6637-6646. [DOI: 10.1039/d0bm00969e] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Dormant versus proliferative phenotypes in metastatic tumor cell clusters are mediated via matrix stiffness and cluster size.
Collapse
Affiliation(s)
| | - Shreyas S. Rao
- Department of Chemical and Biological Engineering
- The University of Alabama
- Tuscaloosa
- USA
| |
Collapse
|
44
|
Pradhan S, Slater JH. Fabrication, characterization, and implementation of engineered hydrogels for controlling breast cancer cell phenotype and dormancy. MethodsX 2019; 6:2744-2766. [PMID: 31828024 PMCID: PMC6889770 DOI: 10.1016/j.mex.2019.11.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 11/07/2019] [Indexed: 01/13/2023] Open
Abstract
A better understanding of how microenvironmental factors regulate cancer dormancy is needed for development of new therapeutic strategies to control metastatic recurrence and disease progression. Modeling cancer dormancy using engineered, in vitro platforms is necessary for investigation under well-defined and well-controlled microenvironments. We present methods and protocols to fabricate, characterize, and implement engineered hydrogels with well-defined biochemical and physical properties for control over breast cancer cell phenotype in three-dimensional (3D) culture. Changes in hydrogel adhesivity, crosslink density, and degradability induce a range of phenotypic behaviors in breast cancer cells including: (1) high growth, (2) moderate growth, (3) single cell, restricted survival dormancy, and (4) balanced dormancy. We describe a method of classifying hydrogel formulations that support each of these phenotypic states. We also describe a method to phenotypically switch cancer cells from single cell dormancy to high growth by dynamically modulating ligand density, thereby recapitulating reactivation and metastatic recurrence.
Collapse
Affiliation(s)
- Shantanu Pradhan
- Department of Biomedical Engineering, University of Delaware, Newark, DE, USA
| | - John H Slater
- Department of Biomedical Engineering, University of Delaware, Newark, DE, USA.,Department of Materials Science & Engineering, University of Delaware, Newark, DE, USA.,Delaware Biotechnology Institute, Newark, DE, USA
| |
Collapse
|
45
|
Butturini E, Carcereri de Prati A, Boriero D, Mariotto S. Tumor Dormancy and Interplay with Hypoxic Tumor Microenvironment. Int J Mol Sci 2019; 20:ijms20174305. [PMID: 31484342 PMCID: PMC6747268 DOI: 10.3390/ijms20174305] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 08/28/2019] [Accepted: 08/30/2019] [Indexed: 12/16/2022] Open
Abstract
The tumor microenvironment is a key factor in disease progression, local resistance, immune-escaping, and metastasis. The rapid proliferation of tumor cells and the aberrant structure of the blood vessels within tumors result in a marked heterogeneity in the perfusion of the tumor tissue with regions of hypoxia. Although most of the tumor cells die in these hypoxic conditions, a part of them can adapt and survive for many days or months in a dormant state. Dormant tumor cells are characterized by cell cycle arrest in G0/G1 phase as well as a low metabolism, and are refractive to common chemotherapy, giving rise to metastasis. Despite these features, the cells retain their ability to proliferate when conditions improve. An understanding of the regulatory machinery of tumor dormancy is essential for identifying early cancer biomarkers and could provide a rationale for the development of novel agents to target dormant tumor cell populations. In this review, we examine the current knowledge of the mechanisms allowing tumor dormancy and discuss the crucial role of the hypoxic microenvironment in this process.
Collapse
Affiliation(s)
- Elena Butturini
- Department of Neuroscience, Biomedicine and Movement Sciences, Section of Biological Chemistry, University of Verona, 37134 Verona, Italy.
| | - Alessandra Carcereri de Prati
- Department of Neuroscience, Biomedicine and Movement Sciences, Section of Biological Chemistry, University of Verona, 37134 Verona, Italy.
| | - Diana Boriero
- Department of Neuroscience, Biomedicine and Movement Sciences, Section of Biological Chemistry, University of Verona, 37134 Verona, Italy.
| | - Sofia Mariotto
- Department of Neuroscience, Biomedicine and Movement Sciences, Section of Biological Chemistry, University of Verona, 37134 Verona, Italy.
| |
Collapse
|
46
|
Pradhan S, Slater JH. Tunable hydrogels for controlling phenotypic cancer cell states to model breast cancer dormancy and reactivation. Biomaterials 2019; 215:119177. [PMID: 31176804 PMCID: PMC6592634 DOI: 10.1016/j.biomaterials.2019.04.022] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 04/16/2019] [Accepted: 04/18/2019] [Indexed: 12/14/2022]
Abstract
During metastasis, disseminated tumor cells (DTCs) from the primary tumor infiltrate secondary organs and reside there for varying lengths of time prior to forming new tumors. The time delay between infiltration and active proliferation, known as dormancy, mediates the length of the latency period. DTCs may undergo one of four fates post-infiltration: death, cellular dormancy, dormant micrometastasis, or invasive growth which, is in part, mediated by extracellular matrix (ECM) properties. Recapitulation of these cell states using engineered hydrogels could facilitate the systematic and controlled investigation of the mechanisms by which ECM properties influence DTC fate. Toward this goal, we implemented a set of sixteen hydrogels with systematic variations in chemical (ligand (RGDS) density and enzymatic degradability) and mechanical (elasticity, swelling, mesh size) properties to investigate their influence on the fate of encapsulated metastatic breast cancer cells, MDA-MB-231. Cell viability, apoptosis, proliferation, metabolic activity, and morphological measurements were acquired at five-day intervals over fifteen days in culture. Analysis of the phenotypic metrics indicated the presence of four different cell states that were classified as: (1) high growth, (2) moderate growth, (3) single cell, restricted survival, dormancy, or (4) balanced dormancy. Correlating hydrogel properties with the resultant cancer cell state indicated that ligand (RGDS) density and enzymatic degradability likely had the most influence on cell fate. Furthermore, we demonstrate the ability to reactivate cells from the single cell, dormant state to the high growth state through a dynamic increase in ligand (RGDS) density after forty days in culture. This tunable engineered hydrogel platform offers insight into matrix properties regulating tumor dormancy, and the dormancy-proliferation switch, and may provide future translational benefits toward development of anti-dormancy therapeutic strategies.
Collapse
Affiliation(s)
- Shantanu Pradhan
- Department of Biomedical Engineering, University of Delaware, 150 Academy Street, 161 Colburn Lab, Newark, DE, 19716, USA
| | - John H Slater
- Department of Biomedical Engineering, University of Delaware, 150 Academy Street, 161 Colburn Lab, Newark, DE, 19716, USA.
| |
Collapse
|
47
|
Jahanban-Esfahlan R, Seidi K, Manjili MH, Jahanban-Esfahlan A, Javaheri T, Zare P. Tumor Cell Dormancy: Threat or Opportunity in the Fight against Cancer. Cancers (Basel) 2019; 11:cancers11081207. [PMID: 31430951 PMCID: PMC6721805 DOI: 10.3390/cancers11081207] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 08/11/2019] [Accepted: 08/13/2019] [Indexed: 12/15/2022] Open
Abstract
Tumor dormancy, a clinically undetectable state of cancer, makes a major contribution to the development of multidrug resistance (MDR), minimum residual disease (MRD), tumor outgrowth, cancer relapse, and metastasis. Despite its high incidence, the whole picture of dormancy-regulated molecular programs is far from clear. That is, it is unknown when and which dormant cells will resume proliferation causing late relapse, and which will remain asymptomatic and harmless to their hosts. Thus, identification of dormancy-related culprits and understanding their roles can help predict cancer prognosis and may increase the probability of timely therapeutic intervention for the desired outcome. Here, we provide a comprehensive review of the dormancy-dictated molecular mechanisms, including angiogenic switch, immune escape, cancer stem cells, extracellular matrix (ECM) remodeling, metabolic reprogramming, miRNAs, epigenetic modifications, and stress-induced p38 signaling pathways. Further, we analyze the possibility of leveraging these dormancy-related molecular cues to outmaneuver cancer and discuss the implications of such approaches in cancer treatment.
Collapse
Affiliation(s)
- Rana Jahanban-Esfahlan
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz 9841, Iran
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz 9841, Iran
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz 9841, Iran
| | - Khaled Seidi
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz 9841, Iran
| | - Masoud H Manjili
- Department of Microbiology & Immunology, VCU School of Medicine, Massey Cancer Center, Richmond, VA 23298, USA
| | | | - Tahereh Javaheri
- Ludwig Boltzmann Institute for Cancer Research, 1090 Vienna, Austria.
| | - Peyman Zare
- Faculty of Medicine, Cardinal Stefan Wyszyński University in Warsaw, 01-938 Warsaw, Poland.
| |
Collapse
|