451
|
Peña-Romero AC, Orenes-Piñero E. Dual Effect of Immune Cells within Tumour Microenvironment: Pro- and Anti-Tumour Effects and Their Triggers. Cancers (Basel) 2022; 14:1681. [PMID: 35406451 PMCID: PMC8996887 DOI: 10.3390/cancers14071681] [Citation(s) in RCA: 126] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 03/21/2022] [Accepted: 03/23/2022] [Indexed: 02/04/2023] Open
Abstract
Our body is constantly exposed to pathogens or external threats, but with the immune response that our body can develop, we can fight off and defeat possible attacks or infections. Nevertheless, sometimes this threat comes from an internal factor. Situations such as the existence of a tumour also cause our immune system (IS) to be put on alert. Indeed, the link between immunology and cancer is evident these days, with IS being used as one of the important targets for treating cancer. Our IS is able to eliminate those abnormal or damaged cells found in our body, preventing the uncontrolled proliferation of tumour cells that can lead to cancer. However, in several cases, tumour cells can escape from the IS. It has been observed that immune cells, the extracellular matrix, blood vessels, fat cells and various molecules could support tumour growth and development. Thus, the developing tumour receives structural support, irrigation and energy, among other resources, making its survival and progression possible. All these components that accompany and help the tumour to survive and to grow are called the tumour microenvironment (TME). Given the importance of its presence in the tumour development process, this review will focus on one of the components of the TME: immune cells. Immune cells can support anti-tumour immune response protecting us against tumour cells; nevertheless, they can also behave as pro-tumoural cells, thus promoting tumour progression and survival. In this review, the anti-tumour and pro-tumour immunity of several immune cells will be discussed. In addition, the TME influence on this dual effect will be also analysed.
Collapse
Affiliation(s)
| | - Esteban Orenes-Piñero
- Department of Biochemistry and Molecular Biology-A, University of Murcia, 30120 Murcia, Spain;
| |
Collapse
|
452
|
Willumsen N, Jensen C, Green G, Nissen NI, Neely J, Nelson DM, Pedersen RS, Frederiksen P, Chen IM, Boisen MK, Johansen AZ, Madsen DH, Svane IM, Lipton A, Leitzel K, Ali SM, Erler JT, Hurkmans DP, Mathijssen RHJ, Aerts J, Eslam M, George J, Christiansen C, Bissel MJ, Karsdal MA. Fibrotic activity quantified in serum by measurements of type III collagen pro-peptides can be used for prognosis across different solid tumor types. Cell Mol Life Sci 2022; 79:204. [PMID: 35332383 PMCID: PMC8948122 DOI: 10.1007/s00018-022-04226-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 02/15/2022] [Accepted: 02/28/2022] [Indexed: 12/12/2022]
Abstract
Due to activation of fibroblast into cancer-associated fibroblasts, there is often an increased deposition of extracellular matrix and fibrillar collagens, e.g. type III collagen, in the tumor microenvironment (TME) that leads to tumor fibrosis (desmoplasia). Tumor fibrosis is closely associated with treatment response and poor prognosis for patients with solid tumors. To assure that the best possible treatment option is provided for patients, there is medical need for identifying patients with high (or low) fibrotic activity in the TME. Measuring unique collagen fragments such as the pro-peptides released into the bloodstream during fibrillar collagen deposition in the TME can provide a non-invasive measure of the fibrotic activity. Based on data from 8 previously published cohorts, this review provides insight into the prognostic value of quantifying tumor fibrosis by measuring the pro-peptide of type III collagen in serum of a total of 1692 patients with different solid tumor types and discusses the importance of tumor fibrosis for understanding prognosis and for potentially guiding future drug development efforts that aim at overcoming the poor outcome associated with a fibrotic TME.
Collapse
Affiliation(s)
| | - Christina Jensen
- Nordic Bioscience, Herlev Hovedgade 205-207, 2730, Herlev, Denmark
| | | | - Neel I Nissen
- Nordic Bioscience, Herlev Hovedgade 205-207, 2730, Herlev, Denmark
| | | | | | | | | | - Inna M Chen
- Department of Oncology, Herlev and Gentofte Hospital, Copenhagen University Hospital, Herlev, Denmark
| | - Mogens K Boisen
- Department of Oncology, Herlev and Gentofte Hospital, Copenhagen University Hospital, Herlev, Denmark
| | - Astrid Z Johansen
- Department of Oncology, Herlev and Gentofte Hospital, Copenhagen University Hospital, Herlev, Denmark
| | - Daniel H Madsen
- Center for Cancer Immune Therapy, Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
| | - Inge Marie Svane
- Center for Cancer Immune Therapy, Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
| | - Allan Lipton
- Penn State Hershey Medical Center, Hershey, PA, USA
| | - Kim Leitzel
- Penn State Hershey Medical Center, Hershey, PA, USA
| | | | - Janine T Erler
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark
| | - Daan P Hurkmans
- Department of Pathology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Ron H J Mathijssen
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Joachim Aerts
- Department of Pulmonology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Mohammed Eslam
- Storr Liver Centre, Westmead Institute for Medical Research, Westmead Hospital and University of Sydney, Sydney, NSW, Australia
| | - Jacob George
- Storr Liver Centre, Westmead Institute for Medical Research, Westmead Hospital and University of Sydney, Sydney, NSW, Australia
| | | | - Mina J Bissel
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA, 94720, USA
| | - Morten A Karsdal
- Nordic Bioscience, Herlev Hovedgade 205-207, 2730, Herlev, Denmark
| |
Collapse
|
453
|
Targeting HIF1-alpha/miR-326/ITGA5 axis potentiates chemotherapy response in triple-negative breast cancer. Breast Cancer Res Treat 2022; 193:331-348. [PMID: 35338412 DOI: 10.1007/s10549-022-06569-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 03/13/2022] [Indexed: 02/07/2023]
Abstract
PURPOSE Triple-negative breast cancer (TNBC) is the most aggressive subtype of breast cancer that is frequently treated with chemotherapy. However, many patients exhibit either de novo chemoresistance or ultimately develop resistance to chemotherapy, leading to significantly high mortality rates. Therefore, increasing the efficacy of chemotherapy has potential to improve patient outcomes. METHODS Here, we performed whole transcriptome sequencing (both RNA and small RNA-sequencing), coupled with network simulations and patient survival data analyses to build a novel miRNA-mRNA interaction network governing chemoresistance in TNBC. We performed cell proliferation assay, Western blotting, RNAi/miRNA mimic experiments, FN coating, 3D cultures, and ChIP assays to validate the interactions in the network, and their functional roles in chemoresistance. We developed xenograft models to test the therapeutic potential of the identified key miRNA/proteins in potentiating chemoresponse in vivo. We also analyzed several patient datasets to evaluate the clinical relevance of our findings. RESULTS We identified fibronectin (FN1) as a central chemoresistance driver gene. Overexpressing miR-326 reversed FN1-driven chemoresistance by targeting FN1 receptor, ITGA5. miR-326 was downregulated by increased hypoxia/HIF1A and ECM stiffness in chemoresistant tumors, leading to upregulation of ITGA5 and activation of the downstream FAK/Src signaling pathways. Overexpression of miR-326 or inhibition of ITGA5 overcame FN1-driven chemotherapy resistance in vitro by inhibiting FAK/Src pathway and potentiated the efficacy of chemotherapy in vivo. Importantly, lower expression of miR-326 or higher levels of predicted miR-326 target genes was significantly associated with worse overall survival in chemotherapy-treated TNBC patients. CONCLUSION FN1 is central in chemoresistance. In chemoresistant tumors, hypoxia and resulting ECM stiffness repress the expression of the tumor suppressor miRNA, miR-326. Hence, re-expression of miR-326 or inhibition of its target ITGA5 reverses FN1-driven chemoresistance making them attractive therapeutic approaches to enhance chemotherapy response in TNBCs.
Collapse
|
454
|
Ikeda-Imafuku M, Wang LLW, Rodrigues D, Shaha S, Zhao Z, Mitragotri S. Strategies to improve the EPR effect: A mechanistic perspective and clinical translation. J Control Release 2022; 345:512-536. [PMID: 35337939 DOI: 10.1016/j.jconrel.2022.03.043] [Citation(s) in RCA: 131] [Impact Index Per Article: 43.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 03/14/2022] [Accepted: 03/21/2022] [Indexed: 12/12/2022]
Abstract
Many efforts have been made to achieve targeted delivery of anticancer drugs to enhance their efficacy and to reduce their adverse effects. These efforts include the development of nanomedicines as they can selectively penetrate through tumor blood vessels through the enhanced permeability and retention (EPR) effect. The EPR effect was first proposed by Maeda and co-workers in 1986, and since then various types of nanoparticles have been developed to take advantage of the phenomenon with regards to drug delivery. However, the EPR effect has been found to be highly variable and thus unreliable due to the complex tumor microenvironment. Various physical and pharmacological strategies have been explored to overcome this challenge. Here, we review key advances and emerging concepts of such EPR-enhancing strategies. Furthermore, we analyze 723 clinical trials of nanoparticles with EPR enhancers and discuss their clinical translation.
Collapse
Affiliation(s)
- Mayumi Ikeda-Imafuku
- John A. Paulson School of Engineering & Applied Sciences, Harvard University, Cambridge, MA 02138, USA; Wyss Institute for Biologically Inspired Engineering, Cambridge, MA 20138, USA
| | - Lily Li-Wen Wang
- John A. Paulson School of Engineering & Applied Sciences, Harvard University, Cambridge, MA 02138, USA; Wyss Institute for Biologically Inspired Engineering, Cambridge, MA 20138, USA; Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Danika Rodrigues
- John A. Paulson School of Engineering & Applied Sciences, Harvard University, Cambridge, MA 02138, USA; Wyss Institute for Biologically Inspired Engineering, Cambridge, MA 20138, USA
| | - Suyog Shaha
- John A. Paulson School of Engineering & Applied Sciences, Harvard University, Cambridge, MA 02138, USA; Wyss Institute for Biologically Inspired Engineering, Cambridge, MA 20138, USA
| | - Zongmin Zhao
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois at Chicago, Chicago, IL 60612, USA; Translational Oncology Program, University of Illinois Cancer Center, Chicago, IL 60612, USA.
| | - Samir Mitragotri
- John A. Paulson School of Engineering & Applied Sciences, Harvard University, Cambridge, MA 02138, USA; Wyss Institute for Biologically Inspired Engineering, Cambridge, MA 20138, USA.
| |
Collapse
|
455
|
Natural Compounds Targeting Cancer-Associated Fibroblasts against Digestive System Tumor Progression: Therapeutic Insights. Biomedicines 2022; 10:biomedicines10030713. [PMID: 35327514 PMCID: PMC8945097 DOI: 10.3390/biomedicines10030713] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Revised: 03/16/2022] [Accepted: 03/18/2022] [Indexed: 01/27/2023] Open
Abstract
Cancer-associated fibroblasts (CAFs) are critical for cancer occurrence and progression in the tumor microenvironment (TME), due to their versatile roles in extracellular matrix remodeling, tumor–stroma crosstalk, immunomodulation, and angiogenesis. CAFs are the most abundant stromal component in the TME and undergo epigenetic modification and abnormal signaling cascade activation, such as transforming growth factor-β (TGF-β) and Wnt pathways that maintain the distinct phenotype of CAFs, which differs from normal fibroblasts. CAFs have been considered therapeutic targets due to their putative oncogenic functions. Current digestive system cancer treatment strategies often result in lower survival outcomes and fail to prevent cancer progression; therefore, comprehensive characterization of the tumor-promoting and -restraining CAF activities might facilitate the design of new therapeutic approaches. In this review, we summarize the enormous literature on natural compounds that mediate the crosstalk of CAFs with digestive system cancer cells, discuss how the biology and the multifaceted functions of CAFs contribute to cancer progression, and finally, pave the way for CAF-related antitumor therapies.
Collapse
|
456
|
Yeow YL, Wu J, Wang X, Winteringham L, Feindel KW, Tirnitz-Parker JEE, Leedman PJ, Ganss R, Hamzah J. ECM Depletion Is Required to Improve the Intratumoral Uptake of Iron Oxide Nanoparticles in Poorly Perfused Hepatocellular Carcinoma. Front Oncol 2022; 12:837234. [PMID: 35273916 PMCID: PMC8902243 DOI: 10.3389/fonc.2022.837234] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 01/20/2022] [Indexed: 01/04/2023] Open
Abstract
Improving tumor access for drug delivery is challenging, particularly in poorly perfused tumors. The availability of functional tumor blood vessels for systemic access is vital to allow drugs or imaging agents to accumulate in the tumor parenchyma. We subjected mice engineered to develop hepatocellular carcinoma (HCC), to treatment with tumor necrosis factor alpha (TNFα) conjugated to a CSG peptide (CSGRRSSKC). CSG binds to the laminin-nidogen-1 complex of the extracellular matrix (ECM) in HCC. When produced as a recombinant fusion protein, the TNFα-CSG functions as an ECM depletion agent via an immune-mediated mechanism to improve tumor perfusion. Tumor perfusion in HCC was dramatically improved after daily intravenous (i.v.) injection of 5 µg TNFα-CSG for five consecutive days. Following treatment, we assessed the tumor accessibility to accumulate an imaging agent, superparamagnetic iron-oxide nanoparticles (IO-NP). Here, we compared the passive delivery of an i.v. dose of IO-NP in HCC following ECM depletion after TNFα-CSG treatment, to the intratumoral accumulation of a comparable dose of CSG-targeted IO-NP in HCC with intact ECM. Magnetic resonance imaging (MRI) T2-weighted scans and T2 relaxation times indicate that when the tumor ECM is intact, HCC was resistant to the intratumoral uptake of IO-NP, even when the particles were tagged with CSG peptide. In contrast, pre-treatment with TNFα-CSG resulted in the highest IO-NP accumulation in tumors. These findings suggest poorly perfused HCC may be resistant to molecular-targeted imaging agents including CSG-IO-NP. We demonstrate that specific ECM depletion using TNFα-CSG improves nanoparticle delivery into poorly perfused tumors such as HCC.
Collapse
Affiliation(s)
- Yen Ling Yeow
- Harry Perkins Institute of Medical Research, Centre for Medical Research, QEII Medical Centre, The University of Western Australia, Nedlands, WA, Australia
| | - Jiansha Wu
- Harry Perkins Institute of Medical Research, Centre for Medical Research, QEII Medical Centre, The University of Western Australia, Nedlands, WA, Australia
| | - Xiao Wang
- Harry Perkins Institute of Medical Research, Centre for Medical Research, QEII Medical Centre, The University of Western Australia, Nedlands, WA, Australia
| | - Louise Winteringham
- Harry Perkins Institute of Medical Research, Centre for Medical Research, QEII Medical Centre, The University of Western Australia, Nedlands, WA, Australia
| | - Kirk W Feindel
- Centre for Microscopy, Characterisation and Analysis, The University of Western Australia, Nedlands, WA, Australia
| | - Janina E E Tirnitz-Parker
- Curtin Medical School and Curtin Health Innovation Research Institute, Curtin University, Bentley, WA, Australia
| | - Peter J Leedman
- Harry Perkins Institute of Medical Research, Centre for Medical Research, QEII Medical Centre, The University of Western Australia, Nedlands, WA, Australia
| | - Ruth Ganss
- Harry Perkins Institute of Medical Research, Centre for Medical Research, QEII Medical Centre, The University of Western Australia, Nedlands, WA, Australia
| | - Juliana Hamzah
- Harry Perkins Institute of Medical Research, Centre for Medical Research, QEII Medical Centre, The University of Western Australia, Nedlands, WA, Australia.,Curtin Medical School and Curtin Health Innovation Research Institute, Curtin University, Bentley, WA, Australia
| |
Collapse
|
457
|
Jiang W, Wei L, Chen B, Luo X, Xu P, Cai J, Hu Y. Platinum prodrug nanoparticles inhibiting tumor recurrence and metastasis by concurrent chemoradiotherapy. J Nanobiotechnology 2022; 20:129. [PMID: 35279133 PMCID: PMC8917711 DOI: 10.1186/s12951-022-01322-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 02/21/2022] [Indexed: 01/13/2023] Open
Abstract
Background Although concurrent chemoradiotherapy (CRT), as one of the most effective antineoplastic therapies in clinic, can successfully inhibit the growth of tumor cells, a risk of developing secondary tumor is still an insurmountable barrier in clinical practice. Results Herein, a new platinum prodrug composed of tannic acid (TA) and Pt2+ (TA-Pt) complex film was synthesized on the surface of Fe2O3 nanoparticles (NPs) with excellent stability and biocompatibility for enhanced CRT. In this system, TA-Pt complex could respond to the tumor acidic microenvironment and damage the DNA of tumor cells. Moreover, the internal iron core not only improved the effect of subsequent radiotherapy (RT), but also disrupted the iron balance in cells, inducing intracellular ferroptosis and eliminating apoptosis-resistant cells. In vitro and vivo experimental results indicated that more than 90% of tumor cells were depleted and more than 75% of the cured tumor-bearing mice evinced no recurrence or metastasis. Conclusions This work offered a new idea for combining the effective chemotherapy, RT and ferroptosis therapy to enhance the curative effect of CRT and inhibit tumor recurrence and metastasis. Graphical Abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s12951-022-01322-y.
Collapse
|
458
|
Yanes B, Rainero E. The Interplay between Cell-Extracellular Matrix Interaction and Mitochondria Dynamics in Cancer. Cancers (Basel) 2022; 14:1433. [PMID: 35326584 PMCID: PMC8946811 DOI: 10.3390/cancers14061433] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 03/01/2022] [Accepted: 03/08/2022] [Indexed: 01/27/2023] Open
Abstract
The tumor microenvironment, in particular the extracellular matrix (ECM), plays a pivotal role in controlling tumor initiation and progression. In particular, the interaction between cancer cells and the ECM promotes cancer cell growth and invasion, leading to the formation of distant metastasis. Alterations in cancer cell metabolism is a key hallmark of cancer, which is often associated with alterations in mitochondrial dynamics. Recent research highlighted that, changes in mitochondrial dynamics are associated with cancer migration and metastasis-these has been extensively reviewed elsewhere. However, less is known about the interplay between the extracellular matrix and mitochondria functions. In this review, we will highlight how ECM remodeling associated with tumorigenesis contribute to the regulation of mitochondrial function, ultimately promoting cancer cell metabolic plasticity, able to fuel cancer invasion and metastasis.
Collapse
Affiliation(s)
| | - Elena Rainero
- School of Biosciences, The University of Sheffield, Western Bank, Sheffield S10 2TN, UK;
| |
Collapse
|
459
|
Qin VM, Haynes NM, D'Souza C, Neeson PJ, Zhu JJ. CAR-T Plus Radiotherapy: A Promising Combination for Immunosuppressive Tumors. Front Immunol 2022; 12:813832. [PMID: 35095911 PMCID: PMC8790144 DOI: 10.3389/fimmu.2021.813832] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 12/22/2021] [Indexed: 12/26/2022] Open
Abstract
Radiotherapy (RT) is the standard-of-care treatment for more than half of cancer patients with localized tumors and is also used as palliative care to facilitate symptom relief in metastatic cancers. In addition, RT can alter the immunosuppressive tumor microenvironment (TME) of solid tumors to augment the anti-tumor immune response of immune checkpoint blockade (ICB). The rationale of this combination therapy can also be extended to other forms of immunotherapy, such as chimeric antigen receptor T cell (CAR-T) therapy. Similar to ICB, the efficacy of CAR-T therapy is also significantly impacted by the immunosuppressive TME, leading to compromised T cell function and/or insufficient T cell infiltration. In this review, we will discuss some of the key barriers to the activity of CAR-T cells in the immunosuppressive TME and focus on how RT can be used to eliminate or bypass these barriers. We will present the challenges to achieving success with this therapeutic partnership. Looking forward, we will also provide strategies currently being investigated to ensure the success of this combination strategy in the clinic.
Collapse
Affiliation(s)
- Vicky Mengfei Qin
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Department of Clinical Pathology, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, VIC, Australia
| | - Nicole M Haynes
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, VIC, Australia
| | - Criselle D'Souza
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, VIC, Australia
| | - Paul J Neeson
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, VIC, Australia
| | - Joe Jiang Zhu
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
460
|
Deciphering Tumour Heterogeneity: From Tissue to Liquid Biopsy. Cancers (Basel) 2022; 14:cancers14061384. [PMID: 35326534 PMCID: PMC8946040 DOI: 10.3390/cancers14061384] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 03/04/2022] [Accepted: 03/05/2022] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Most malignant tumours are highly heterogeneous at molecular and phenotypic levels. Tumour variability poses challenges for the management of patients, as it arises between patients and even evolves in space and time within a single patient. Currently, treatment-decision making usually relies on the molecular characteristics of a limited tumour tissue sample at the time of diagnosis or disease progression but does not take into account the complexity of the bulk tumours and their constant evolution over time. In this review, we explore the extent of tumour heterogeneity and report the mechanisms that promote and sustain this diversity in cancers. We summarise the clinical strikes of tumour diversity in the management of patients with cancer. Finally, we discuss the current material and technological approaches that are relevant to adequately appreciate tumour heterogeneity. Abstract Human solid malignancies harbour a heterogeneous set of cells with distinct genotypes and phenotypes. This heterogeneity is installed at multiple levels. A biological diversity is commonly observed between tumours from different patients (inter-tumour heterogeneity) and cannot be fully captured by the current consensus molecular classifications for specific cancers. To extend the complexity in cancer, there are substantial differences from cell to cell within an individual tumour (intra-tumour heterogeneity, ITH) and the features of cancer cells evolve in space and time. Currently, treatment-decision making usually relies on the molecular characteristics of a limited tumour tissue sample at the time of diagnosis or disease progression but does not take into account the complexity of the bulk tumours and their constant evolution over time. In this review, we explore the extent of tumour heterogeneity with an emphasis on ITH and report the mechanisms that promote and sustain this diversity in cancers. We summarise the clinical strikes of ITH in the management of patients with cancer. Finally, we discuss the current material and technological approaches that are relevant to adequately appreciate ITH.
Collapse
|
461
|
Kokoretsis D, Maniaki EK, Kyriakopoulou K, Koutsakis C, Piperigkou Z, Karamanos NK. Hyaluronan as "Agent Smith" in cancer extracellular matrix pathobiology: Regulatory roles in immune response, cancer progression and targeting. IUBMB Life 2022; 74:943-954. [PMID: 35261139 DOI: 10.1002/iub.2608] [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: 12/30/2021] [Revised: 02/08/2022] [Accepted: 02/14/2022] [Indexed: 12/16/2022]
Abstract
Extracellular matrix (ECM) critically regulates cancer cell behavior by governing cell signaling and properties. Hyaluronan (HA) acts as a structural and functional ECM component that mediates critical properties of cancer cells in a molecular size-dependent manner. HA fragments secreted by cancer-associated fibroblasts (CAFs) reveal the correlation of HA to CAF-mediated matrix remodeling, a key step for the initiation of metastasis. The main goal of this article is to highlight the vital functions of HA in cancer cell initiation and progression as well as HA-mediated paracrine interactions among cancer and stromal cells. Furthermore, the HA implication in mediating immune responses to cancer progression is also discussed. Novel data on the role of HA in the formation of pre-metastatic niche may contribute towards the improvement of current theranostic approaches that benefit cancer management.
Collapse
Affiliation(s)
- Dimitris Kokoretsis
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece
| | - Evangelia-Konstantina Maniaki
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece
| | - Konstantina Kyriakopoulou
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece
| | - Christos Koutsakis
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece
| | - Zoi Piperigkou
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece.,Foundation for Research and Technology-Hellas (FORTH)/Institute of Chemical Engineering Sciences (ICE-HT), Patras, Greece
| | - Nikos K Karamanos
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece.,Foundation for Research and Technology-Hellas (FORTH)/Institute of Chemical Engineering Sciences (ICE-HT), Patras, Greece
| |
Collapse
|
462
|
A Novel Superpixel Approach to the Tumoral Microenvironment in Colorectal Cancer. J Pathol Inform 2022; 13:100009. [PMID: 35223135 PMCID: PMC8855322 DOI: 10.1016/j.jpi.2022.100009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Accepted: 12/29/2021] [Indexed: 01/01/2023] Open
Abstract
Colorectal cancer (CRC) is the most common malignancy of the gastrointestinal tract. The stroma and the tumoral microenvironment (TME) represent ecosystem-like biological networks and are new frontiers in CRC. The present study demonstrates the use of a novel machine learning-based superpixel approach for whole slide images to unravel this biology. Findings of significance include the association of low proportionated stromal area, high immature stromal percentage, and high myxoid stromal ratio (MSR) with worse prognostic outcomes in CRC. Overall, stromal computational markers outperformed all others at predicting clinical outcomes. MSR may be able to prognosticate patients independent of pathological stage, representing an optimal way to effectively prognosticate CRC patients which circumvents the need for more extensive molecular and/or computational profiling. The superpixel approaches to the TME demonstrated here can be performed by a trained pathologist and recorded during synoptic cancer reporting with appropriate quality assurance. Future clinical trials will have the ultimate say in determining whether we can better tailor the need for adjuvant therapy in patients with CRC.
Collapse
|
463
|
Intestinal extracellular matrix hydrogels to generate intestinal organoids for translational applications. J IND ENG CHEM 2022. [DOI: 10.1016/j.jiec.2021.11.044] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
464
|
Holder PG, Lim SA, Huang CS, Sharma P, Dagdas YS, Bulutoglu B, Sockolosky JT. Engineering interferons and interleukins for cancer immunotherapy. Adv Drug Deliv Rev 2022; 182:114112. [PMID: 35085624 DOI: 10.1016/j.addr.2022.114112] [Citation(s) in RCA: 73] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/07/2022] [Accepted: 01/12/2022] [Indexed: 02/08/2023]
Abstract
Cytokines are a class of potent immunoregulatory proteins that are secreted in response to various stimuli and act locally to regulate many aspects of human physiology and disease. Cytokines play important roles in cancer initiation, progression, and elimination, and thus, there is a long clinical history associated with the use of recombinant cytokines to treat cancer. However, the use of cytokines as therapeutics has been limited by cytokine pleiotropy, complex biology, poor drug-like properties, and severe dose-limiting toxicities. Nevertheless, cytokines are crucial mediators of innate and adaptive antitumor immunity and have the potential to enhance immunotherapeutic approaches to treat cancer. Development of immune checkpoint inhibitors and combination immunotherapies has reinvigorated interest in cytokines as therapeutics, and a variety of engineering approaches are emerging to improve the safety and effectiveness of cytokine immunotherapy. In this review we highlight recent advances in cytokine biology and engineering for cancer immunotherapy.
Collapse
|
465
|
Moreira AM, Ferreira RM, Carneiro P, Figueiredo J, Osório H, Barbosa J, Preto J, Pinto-do-Ó P, Carneiro F, Seruca R. Proteomic Identification of a Gastric Tumor ECM Signature Associated With Cancer Progression. Front Mol Biosci 2022; 9:818552. [PMID: 35340765 PMCID: PMC8942767 DOI: 10.3389/fmolb.2022.818552] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 01/18/2022] [Indexed: 12/12/2022] Open
Abstract
The extracellular matrix (ECM) plays an undisputable role in tissue homeostasis and its deregulation leads to altered mechanical and biochemical cues that impact cancer development and progression. Herein, we undertook a novel approach to address the role of gastric ECM in tumorigenesis, which remained largely unexplored. By combining decellularization techniques with a high-throughput quantitative proteomics approach, we have performed an extensive characterization of human gastric mucosa, uncovering its composition and distribution among tumor, normal adjacent and normal distant mucosa. Our results revealed a common ECM signature composed of 142 proteins and indicated that gastric carcinogenesis encompasses ECM remodeling through alterations in the abundance of 24 components, mainly basement membrane proteins. Indeed, we could only identify one de novo tumor-specific protein, the collagen alpha-1(X) chain (COL10A1). Functional analysis of the data demonstrated that gastric ECM remodeling favors tumor progression by activating ECM receptors and cellular processes involved in angiogenesis and cell-extrinsic metabolic regulation. By analyzing mRNA expression in an independent GC cohort available at the TGCA, we validated the expression profile of 12 differentially expressed ECM proteins. Importantly, the expression of COL1A2, LOX and LTBP2 significantly correlated with high tumor stage, with LOX and LTBP2 further impacting patient overall survival. These findings contribute for a better understanding of GC biology and highlight the role of core ECM components in gastric carcinogenesis and their clinical relevance as biomarkers of disease prognosis.
Collapse
Affiliation(s)
- Ana M. Moreira
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Porto, Portugal
- Doctoral Program on Cellular and Molecular Biotechnology Applied to Health Sciences, School of Medicine and Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal
| | - Rui M. Ferreira
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Porto, Portugal
| | - Patrícia Carneiro
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Porto, Portugal
| | - Joana Figueiredo
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Porto, Portugal
| | - Hugo Osório
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Porto, Portugal
- Faculty of Medicine, University of Porto, Porto, Portugal
| | - José Barbosa
- Faculty of Medicine, University of Porto, Porto, Portugal
- Department of General Surgery, Centro Hospitalar Universitário de São João, Porto, Portugal
| | - John Preto
- Faculty of Medicine, University of Porto, Porto, Portugal
- Department of General Surgery, Centro Hospitalar Universitário de São João, Porto, Portugal
| | - Perpétua Pinto-do-Ó
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Institute of Biomedical Engineering (INEB), University of Porto, Porto, Portugal
- School of Medicine and Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal
| | - Fátima Carneiro
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Porto, Portugal
- Faculty of Medicine, University of Porto, Porto, Portugal
- Department of Pathology, Centro Hospitalar Universitário de São João, Porto, Portugal
| | - Raquel Seruca
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Porto, Portugal
- Faculty of Medicine, University of Porto, Porto, Portugal
- *Correspondence: Raquel Seruca,
| |
Collapse
|
466
|
Fertal SA, Poterala JE, Ponik SM, Wisinski KB. Stromal Characteristics and Impact on New Therapies for Metastatic Triple-Negative Breast Cancer. Cancers (Basel) 2022; 14:1238. [PMID: 35267548 PMCID: PMC8909697 DOI: 10.3390/cancers14051238] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 02/22/2022] [Accepted: 02/25/2022] [Indexed: 12/25/2022] Open
Abstract
The heterogenous nature of triple-negative breast cancer (TNBC) is an underlying factor in therapy resistance, metastasis, and overall poor patient outcome. The lack of hormone and growth factor receptors lends to the use of chemotherapy as the first-line treatment for TNBC. However, the failure of chemotherapy demonstrates the need to develop novel immunotherapies, antibody-drug conjugates (ADCs), and other tumor- and stromal-targeted therapeutics for TNBC patients. The potential for stromal-targeted therapy is driven by studies indicating that the interactions between tumor cells and the stromal extracellular matrix (ECM) activate mechanisms of therapy resistance. Here, we will review recent outcomes from clinical trials targeting metastatic TNBC with immunotherapies aimed at programed death ligand-receptor interactions, and ADCs specifically linked to trophoblast cell surface antigen 2 (Trop-2). We will discuss how biophysical and biochemical cues from the ECM regulate the pathophysiology of tumor and stromal cells toward a pro-tumor immune environment, therapy resistance, and poor TNBC patient outcome. Moreover, we will highlight how ECM-mediated resistance is motivating the development of new stromal-targeted therapeutics with potential to improve therapy for this disease.
Collapse
Affiliation(s)
- Shelby A. Fertal
- University of Wisconsin (UW) Carbone Cancer Center, Madison, WI 53705, USA; (S.A.F.); (J.E.P.); (S.M.P.)
- Department of Cell and Regenerative Biology, UW School of Medicine and Public Health, Madison, WI 53705, USA
| | - Johanna E. Poterala
- University of Wisconsin (UW) Carbone Cancer Center, Madison, WI 53705, USA; (S.A.F.); (J.E.P.); (S.M.P.)
- Department of Medicine, UW School of Medicine and Public Health, Madison, WI 53726, USA
| | - Suzanne M. Ponik
- University of Wisconsin (UW) Carbone Cancer Center, Madison, WI 53705, USA; (S.A.F.); (J.E.P.); (S.M.P.)
- Department of Cell and Regenerative Biology, UW School of Medicine and Public Health, Madison, WI 53705, USA
| | - Kari B. Wisinski
- University of Wisconsin (UW) Carbone Cancer Center, Madison, WI 53705, USA; (S.A.F.); (J.E.P.); (S.M.P.)
- Department of Medicine, UW School of Medicine and Public Health, Madison, WI 53726, USA
| |
Collapse
|
467
|
Cancer-Associated Fibroblasts: Mechanisms of Tumor Progression and Novel Therapeutic Targets. Cancers (Basel) 2022; 14:cancers14051231. [PMID: 35267539 PMCID: PMC8909913 DOI: 10.3390/cancers14051231] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 02/17/2022] [Accepted: 02/24/2022] [Indexed: 12/14/2022] Open
Abstract
Simple Summary The tumor microenvironment plays an important role in determining the biological behavior of several of the more aggressive malignancies. Among the various cell types evident in the tumor “field”, cancer-associated fibroblasts (CAFs) are a heterogenous collection of activated fibroblasts secreting a wide repertoire of factors that regulate tumor development and progression, inflammation, drug resistance, metastasis and recurrence. Insensitivity to chemotherapeutics and metastatic spread are the major contributors to cancer patient mortality. This review discusses the complex interactions between CAFs and the various populations of normal and neoplastic cells that interact within the dynamic confines of the tumor microenvironment with a focus on the involved pathways and genes. Abstract Cancer-associated fibroblasts (CAFs) are a heterogenous population of stromal cells found in solid malignancies that coexist with the growing tumor mass and other immune/nonimmune cellular elements. In certain neoplasms (e.g., desmoplastic tumors), CAFs are the prominent mesenchymal cell type in the tumor microenvironment, where their presence and abundance signal a poor prognosis in multiple cancers. CAFs play a major role in the progression of various malignancies by remodeling the supporting stromal matrix into a dense, fibrotic structure while secreting factors that lead to the acquisition of cancer stem-like characteristics and promoting tumor cell survival, reduced sensitivity to chemotherapeutics, aggressive growth and metastasis. Tumors with high stromal fibrotic signatures are more likely to be associated with drug resistance and eventual relapse. Clarifying the molecular basis for such multidirectional crosstalk among the various normal and neoplastic cell types present in the tumor microenvironment may yield novel targets and new opportunities for therapeutic intervention. This review highlights the most recent concepts regarding the complexity of CAF biology including CAF heterogeneity, functionality in drug resistance, contribution to a progressively fibrotic tumor stroma, the involved signaling pathways and the participating genes.
Collapse
|
468
|
Garcia-Arranz M, Villarejo-Campos P, Barambio J, Garcia Gomez-Heras S, Vega-Clemente L, Guadalajara H, García-Olmo D. Toxicity study in a pig model of intraperitoneal collagenase as an "enzymatic scalpel" directed to break stroma in order to generate a new perspective for peritoneal carcinomatosis approach: an experimental research. World J Surg Oncol 2022; 20:53. [PMID: 35216593 PMCID: PMC8881860 DOI: 10.1186/s12957-022-02524-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 02/05/2022] [Indexed: 11/22/2022] Open
Abstract
Background This study aimed to measure the toxicity resulting from collagenase administration to the peritoneal cavity in a pig model as a preliminary step to break down the stroma surrounding tumors. Methods Eight pigs were treated with 2 different collagenase concentrations previously tested in rats by our group. Time and temperature were controlled using a peritoneal lavage system (PRS System, Combat Medical Ltd.) identical to that used in human surgeries through hyperthermic intraperitoneal chemotherapy (HIPEC); 2 additional pigs were treated with peritoneal lavage only. Samples of blood and peritoneal fluid were collected pre-treatment, immediately after treatment, and 24 h postoperatively. In addition, histological studies and blood collagenase levels were measured. Results No complications were observed during the surgeries. Intraoperative images evidenced the release of peritoneal tissue during collagenase treatment. After surgery, the animals showed no signs of pain. Diet and mobility were normal at 4 h postoperatively, and there were no significant differences in hematologic or biochemical parameters. Quantification of MMP1 and MMP2 in all samples as measured by absorbance showed no differences in blood collagenase levels between pre-treatment, post-treatment, and 24 h postoperatively. None of the animals treated with collagenase showed peritoneal adhesions during the second surgery. Histologically, peritoneal organs and serous structures did not show any microscopic alterations associated with collagenase treatment in any group. Conclusion Lavage of the peritoneal cavity with doses of up to 100,000 collagen digestion units/animal for 30 min is safe and removes connective tissue from the peritoneal cavity.
Collapse
Affiliation(s)
- M Garcia-Arranz
- New Therapies Laboratory, Health Research Institute-Fundación Jiménez Díaz University Hospital (IIS-FJD), Avda. Reyes Católicos, 2, 28040, Madrid, Spain. .,Department of Surgery, Universidad Autónoma de Madrid, C/ Arzobispo Morcillo s/n, 28034, Madrid, Spain.
| | - P Villarejo-Campos
- Department of Surgery, Fundación Jiménez Díaz University Hospital, Avda. Reyes Católicos, 2, 28040, Madrid, Spain
| | - J Barambio
- Department of Surgery, Fundación Jiménez Díaz University Hospital, Avda. Reyes Católicos, 2, 28040, Madrid, Spain
| | - S Garcia Gomez-Heras
- Department of Human Histology, Universidad Rey Juan Carlos, Avda de Atenas s/n, 28922, Alcorcón, Spain
| | - L Vega-Clemente
- New Therapies Laboratory, Health Research Institute-Fundación Jiménez Díaz University Hospital (IIS-FJD), Avda. Reyes Católicos, 2, 28040, Madrid, Spain
| | - H Guadalajara
- Department of Surgery, Universidad Autónoma de Madrid, C/ Arzobispo Morcillo s/n, 28034, Madrid, Spain.,Department of Surgery, Fundación Jiménez Díaz University Hospital, Avda. Reyes Católicos, 2, 28040, Madrid, Spain
| | - D García-Olmo
- New Therapies Laboratory, Health Research Institute-Fundación Jiménez Díaz University Hospital (IIS-FJD), Avda. Reyes Católicos, 2, 28040, Madrid, Spain.,Department of Surgery, Universidad Autónoma de Madrid, C/ Arzobispo Morcillo s/n, 28034, Madrid, Spain.,Department of Surgery, Fundación Jiménez Díaz University Hospital, Avda. Reyes Católicos, 2, 28040, Madrid, Spain
| |
Collapse
|
469
|
Lepucki A, Orlińska K, Mielczarek-Palacz A, Kabut J, Olczyk P, Komosińska-Vassev K. The Role of Extracellular Matrix Proteins in Breast Cancer. J Clin Med 2022; 11:jcm11051250. [PMID: 35268340 PMCID: PMC8911242 DOI: 10.3390/jcm11051250] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 01/16/2022] [Accepted: 02/22/2022] [Indexed: 12/12/2022] Open
Abstract
The extracellular matrix is a structure composed of many molecules, including fibrillar (types I, II, III, V, XI, XXIV, XXVII) and non-fibrillar collagens (mainly basement membrane collagens: types IV, VIII, X), non-collagenous glycoproteins (elastin, laminin, fibronectin, thrombospondin, tenascin, osteopontin, osteonectin, entactin, periostin) embedded in a gel of negatively charged water-retaining glycosaminoglycans (GAGs) such as non-sulfated hyaluronic acid (HA) and sulfated GAGs which are linked to a core protein to form proteoglycans (PGs). This highly dynamic molecular network provides critical biochemical and biomechanical cues that mediate the cell–cell and cell–matrix interactions, influence cell growth, migration and differentiation and serve as a reservoir of cytokines and growth factors’ action. The breakdown of normal ECM and its replacement with tumor ECM modulate the tumor microenvironment (TME) composition and is an essential part of tumorigenesis and metastasis, acting as key driver for malignant progression. Abnormal ECM also deregulate behavior of stromal cells as well as facilitating tumor-associated angiogenesis and inflammation. Thus, the tumor matrix modulates each of the classically defined hallmarks of cancer promoting the growth, survival and invasion of the cancer. Moreover, various ECM-derived components modulate the immune response affecting T cells, tumor-associated macrophages (TAM), dendritic cells and cancer-associated fibroblasts (CAF). This review article considers the role that extracellular matrix play in breast cancer. Determining the detailed connections between the ECM and cellular processes has helped to identify novel disease markers and therapeutic targets.
Collapse
Affiliation(s)
- Arkadiusz Lepucki
- Department of Community Pharmacy, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia in Katowice, 41-200 Sosnowiec, Poland; (A.L.); (K.O.)
| | - Kinga Orlińska
- Department of Community Pharmacy, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia in Katowice, 41-200 Sosnowiec, Poland; (A.L.); (K.O.)
| | - Aleksandra Mielczarek-Palacz
- Department of Immunology and Serology, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 41-200 Sosnowiec, Poland; (A.M.-P.); (J.K.)
| | - Jacek Kabut
- Department of Immunology and Serology, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 41-200 Sosnowiec, Poland; (A.M.-P.); (J.K.)
| | - Pawel Olczyk
- Department of Community Pharmacy, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia in Katowice, 41-200 Sosnowiec, Poland; (A.L.); (K.O.)
- Correspondence:
| | - Katarzyna Komosińska-Vassev
- Department of Clinical Chemistry and Laboratory Diagnostics, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 41-200 Sosnowiec, Poland;
| |
Collapse
|
470
|
Lagunas-Rangel FA, Liu W, Schiöth HB. Can Exposure to Environmental Pollutants Be Associated with Less Effective Chemotherapy in Cancer Patients? INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:ijerph19042064. [PMID: 35206262 PMCID: PMC8871977 DOI: 10.3390/ijerph19042064] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 02/07/2022] [Accepted: 02/09/2022] [Indexed: 02/05/2023]
Abstract
Since environmental pollutants are ubiquitous and many of them are resistant to degradation, we are exposed to many of them on a daily basis. Notably, these pollutants can have harmful effects on our health and be linked to the development of disease. Epidemiological evidence together with a better understanding of the mechanisms that link toxic substances with the development of diseases, suggest that exposure to some environmental pollutants can lead to an increased risk of developing cancer. Furthermore, several studies have raised the role of low-dose exposure to environmental pollutants in cancer progression. However, little is known about how these compounds influence the treatments given to cancer patients. In this work, we present a series of evidences suggesting that environmental pollutants such as bisphenol A (BPA), benzo[a]pyrene (BaP), persistent organic pollutants (POPs), aluminum chloride (AlCl3), and airborne particulate matter may reduce the efficacy of some common chemotherapeutic drugs used in different types of cancer. We discuss the potential underlying molecular mechanisms that lead to the generation of this chemoresistance, such as apoptosis evasion, DNA damage repair, activation of pro-cancer signaling pathways, drug efflux and action of antioxidant enzymes, among others.
Collapse
Affiliation(s)
- Francisco Alejandro Lagunas-Rangel
- Department of Surgical Sciences, Functional Pharmacology and Neuroscience, Uppsala University, BMC Box 593, Husargatan 3, 75124 Uppsala, Sweden;
- Correspondence: (F.A.L.-R.); (H.B.S.)
| | - Wen Liu
- Department of Surgical Sciences, Functional Pharmacology and Neuroscience, Uppsala University, BMC Box 593, Husargatan 3, 75124 Uppsala, Sweden;
| | - Helgi B. Schiöth
- Department of Surgical Sciences, Functional Pharmacology and Neuroscience, Uppsala University, BMC Box 593, Husargatan 3, 75124 Uppsala, Sweden;
- Institute of Translational Medicine and Biotechnology, I. M. Sechenov First Moscow State Medical University, 8-2 Trubetskaya Str. Moscow, 119991 Moscow, Russia
- Correspondence: (F.A.L.-R.); (H.B.S.)
| |
Collapse
|
471
|
Multimerin-1 and cancer: a review. Biosci Rep 2022; 42:230760. [PMID: 35132992 PMCID: PMC8881648 DOI: 10.1042/bsr20211248] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 01/29/2022] [Accepted: 02/01/2022] [Indexed: 11/21/2022] Open
Abstract
Multimerin-1 (MMRN1) is a platelet protein with a role in haemostasis and coagulation. It is also present in endothelial cells (ECs) and the extracellular matrix (ECM), where it may be involved in cell adhesion, but its molecular functions and protein–protein interactions in these cellular locations have not been studied in detail yet. In recent years, MMRN1 has been identified as a differentially expressed gene (DEG) in various cancers and it has been proposed as a possible cancer biomarker. Some evidence suggest that MMRN1 expression is regulated by methylation, protein interactions, and non-coding RNAs (ncRNAs) in different cancers. This raises the questions if a functional role of MMRN1 is being targeted during cancer development, and if MMRN1’s differential expression pattern correlates with cancer progression. As a result, it is timely to review the current state of what is known about MMRN1 to help inform future research into MMRN1’s molecular mechanisms in cancer.
Collapse
|
472
|
Andrea AE, Chiron A, Mallah S, Bessoles S, Sarrabayrouse G, Hacein-Bey-Abina S. Advances in CAR-T Cell Genetic Engineering Strategies to Overcome Hurdles in Solid Tumors Treatment. Front Immunol 2022; 13:830292. [PMID: 35211124 PMCID: PMC8861853 DOI: 10.3389/fimmu.2022.830292] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 01/18/2022] [Indexed: 12/15/2022] Open
Abstract
During this last decade, adoptive transfer of T lymphocytes genetically modified to express chimeric antigen receptors (CARs) emerged as a valuable therapeutic strategy in hematological cancers. However, this immunotherapy has demonstrated limited efficacy in solid tumors. The main obstacle encountered by CAR-T cells in solid malignancies is the immunosuppressive tumor microenvironment (TME). The TME impedes tumor trafficking and penetration of T lymphocytes and installs an immunosuppressive milieu by producing suppressive soluble factors and by overexpressing negative immune checkpoints. In order to overcome these hurdles, new CAR-T cells engineering strategies were designed, to potentiate tumor recognition and infiltration and anti-cancer activity in the hostile TME. In this review, we provide an overview of the major mechanisms used by tumor cells to evade immune defenses and we critically expose the most optimistic engineering strategies to make CAR-T cell therapy a solid option for solid tumors.
Collapse
Affiliation(s)
- Alain E. Andrea
- Laboratoire de Biochimie et Thérapies Moléculaires, Faculté de Pharmacie, Université Saint Joseph de Beyrouth, Beirut, Lebanon
| | - Andrada Chiron
- Université de Paris, CNRS, INSERM, UTCBS, Unité des technologies Chimiques et Biologiques pour la Santé, Paris, France
- Clinical Immunology Laboratory, Groupe Hospitalier Universitaire Paris-Sud, Hôpital Kremlin-Bicêtre, Assistance Publique-Hôpitaux de Paris, Le-Kremlin-Bicêtre, France
| | - Sarah Mallah
- Faculty of Arts and Sciences, Lebanese American University, Beirut, Lebanon
| | - Stéphanie Bessoles
- Université de Paris, CNRS, INSERM, UTCBS, Unité des technologies Chimiques et Biologiques pour la Santé, Paris, France
| | - Guillaume Sarrabayrouse
- Université de Paris, CNRS, INSERM, UTCBS, Unité des technologies Chimiques et Biologiques pour la Santé, Paris, France
| | - Salima Hacein-Bey-Abina
- Université de Paris, CNRS, INSERM, UTCBS, Unité des technologies Chimiques et Biologiques pour la Santé, Paris, France
- Clinical Immunology Laboratory, Groupe Hospitalier Universitaire Paris-Sud, Hôpital Kremlin-Bicêtre, Assistance Publique-Hôpitaux de Paris, Le-Kremlin-Bicêtre, France
| |
Collapse
|
473
|
FAK in Cancer: From Mechanisms to Therapeutic Strategies. Int J Mol Sci 2022; 23:ijms23031726. [PMID: 35163650 PMCID: PMC8836199 DOI: 10.3390/ijms23031726] [Citation(s) in RCA: 74] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 01/28/2022] [Accepted: 01/30/2022] [Indexed: 01/25/2023] Open
Abstract
Focal adhesion kinase (FAK), a non-receptor tyrosine kinase, is overexpressed and activated in many cancer types. FAK regulates diverse cellular processes, including growth factor signaling, cell cycle progression, cell survival, cell motility, angiogenesis, and the establishment of immunosuppressive tumor microenvironments through kinase-dependent and kinase-independent scaffolding functions in the cytoplasm and nucleus. Mounting evidence has indicated that targeting FAK, either alone or in combination with other agents, may represent a promising therapeutic strategy for various cancers. In this review, we summarize the mechanisms underlying FAK-mediated signaling networks during tumor development. We also summarize the recent progress of FAK-targeted small-molecule compounds for anticancer activity from preclinical and clinical evidence.
Collapse
|
474
|
LeSavage BL, Suhar RA, Broguiere N, Lutolf MP, Heilshorn SC. Next-generation cancer organoids. NATURE MATERIALS 2022; 21:143-159. [PMID: 34385685 DOI: 10.1038/s41563-021-01057-5] [Citation(s) in RCA: 216] [Impact Index Per Article: 72.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 06/21/2021] [Indexed: 05/13/2023]
Abstract
Organotypic models of patient-specific tumours are revolutionizing our understanding of cancer heterogeneity and its implications for personalized medicine. These advancements are, in part, attributed to the ability of organoid models to stably preserve genetic, proteomic, morphological and pharmacotypic features of the parent tumour in vitro, while also offering unprecedented genomic and environmental manipulation. Despite recent innovations in organoid protocols, current techniques for cancer organoid culture are inherently uncontrolled and irreproducible, owing to several non-standardized facets including cancer tissue sources and subsequent processing, medium formulations, and animal-derived three-dimensional matrices. Given the potential for cancer organoids to accurately recapitulate the intra- and intertumoral biological heterogeneity associated with patient-specific cancers, eliminating the undesirable technical variability accompanying cancer organoid culture is necessary to establish reproducible platforms that accelerate translatable insights into patient care. Here we describe the current challenges and recent multidisciplinary advancements and opportunities for standardizing next-generation cancer organoid systems.
Collapse
Affiliation(s)
- Bauer L LeSavage
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Riley A Suhar
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA
| | - Nicolas Broguiere
- Laboratory of Stem Cell Bioengineering, Institute of Bioengineering, School of Life Sciences and School of Engineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
- Institute of Chemical Sciences and Engineering, School of Basic Science, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Matthias P Lutolf
- Laboratory of Stem Cell Bioengineering, Institute of Bioengineering, School of Life Sciences and School of Engineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
- Institute of Chemical Sciences and Engineering, School of Basic Science, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Sarah C Heilshorn
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA.
| |
Collapse
|
475
|
Enhancing the anticancer effect of paclitaxel by using polymeric nanoparticles decorated with colorectal cancer targeting CPKSNNGVC-peptide. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103125] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
|
476
|
He ZX, Zhao SB, Fang X, E JF, Fu HY, Song YH, Wu JY, Pan P, Gu L, Xia T, Liu YL, Li ZS, Wang SL, Bai Y. Prognostic and Predictive Value of BGN in Colon Cancer Outcomes and Response to Immunotherapy. Front Oncol 2022; 11:761030. [PMID: 35096572 PMCID: PMC8790701 DOI: 10.3389/fonc.2021.761030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 12/15/2021] [Indexed: 11/13/2022] Open
Abstract
Background Colon cancer is one of the most frequent malignancies and causes high mortality worldwide. Exploring the tumor-immune interactions in the tumor microenvironment and identifying new prognostic and therapeutic biomarkers will assist in decoding the novel mechanism of tumor immunotherapy. BGN is a typical extracellular matrix protein that was previously validated as a signaling molecule regulating multiple processes of tumorigenesis. However, its role in tumor immunity requires further investigation. Methods The differentially expressed genes in three GEO datasets were analyzed, and BGN was identified as the target gene by intersection analysis of PPIs. The relevance between clinical outcomes and BGN expression levels was evaluated using data from the GEO database, TCGA and tissue microarray of colon cancer samples. Univariable and multivariable Cox regression models were conducted for identifying the risk factors correlated with clinical prognosis of colon cancer patients. Next, the association between BGN expression levels and the infiltration of immune cells as well as the process of the immune response was analyzed. Finally, we predicted the immunotherapeutic response rates in the subgroups of low and high BGN expression by TIS score, ImmuCellAI and TIDE algorithms. Results BGN expression demonstrated a statistically significant upregulation in colon cancer tissues than in normal tissues. Elevated BGN was associated with shorter overall survival as well as unfavorable clinicopathological features, including tumor size, serosa invasion and length of hospitalization. Mechanistically, pathway enrichment and functional analysis demonstrated that BGN was positively correlated with immune and stromal scores in the TME and primarily involved in the regulation of immune response. Further investigation revealed that BGN was strongly expressed in the immunosuppressive phenotype and tightly associated with the infiltration of multiple immune cells in colon cancer, especially M2 macrophages and induced Tregs. Finally, we demonstrated that high BGN expression presented a better immunotherapeutic response in colon cancer patients. Conclusion BGN is an encouraging predictor of diagnosis, prognosis and immunotherapeutic response in patients with colon cancer. Assessment of BGN expression represents a novel approach with great promise for identifying patients who may potentially benefit from immunotherapy.
Collapse
Affiliation(s)
- Zi-Xuan He
- Department of Gastroenterology, Changhai Hospital, Second Military Medical University/Naval Medical University, Shanghai, China
| | - Sheng-Bing Zhao
- Department of Gastroenterology, Changhai Hospital, Second Military Medical University/Naval Medical University, Shanghai, China
| | - Xue Fang
- Department of Gastroenterology, Changhai Hospital, Second Military Medical University/Naval Medical University, Shanghai, China
| | - Ji-Fu E
- Department of Colorectal Surgery, Changhai Hospital, Second Military Medical University/Naval Medical University, Shanghai, China
| | - Hong-Yu Fu
- Department of Gastroenterology, Changhai Hospital, Second Military Medical University/Naval Medical University, Shanghai, China
| | - Yi-Hang Song
- Department of Gastroenterology, Changhai Hospital, Second Military Medical University/Naval Medical University, Shanghai, China
| | - Jia-Yi Wu
- Department of Gastroenterology, Changhai Hospital, Second Military Medical University/Naval Medical University, Shanghai, China
| | - Peng Pan
- Department of Gastroenterology, Changhai Hospital, Second Military Medical University/Naval Medical University, Shanghai, China
| | - Lun Gu
- Department of Gastroenterology, Changhai Hospital, Second Military Medical University/Naval Medical University, Shanghai, China
| | - Tian Xia
- Department of Gastroenterology, Changhai Hospital, Second Military Medical University/Naval Medical University, Shanghai, China
| | - Yi-Long Liu
- College of Basic Medicine Sciences, Second Military Medical University/Naval Medical University, Shanghai, China
| | - Zhao-Shen Li
- Department of Gastroenterology, Changhai Hospital, Second Military Medical University/Naval Medical University, Shanghai, China
| | - Shu-Ling Wang
- Department of Gastroenterology, Changhai Hospital, Second Military Medical University/Naval Medical University, Shanghai, China
| | - Yu Bai
- Department of Gastroenterology, Changhai Hospital, Second Military Medical University/Naval Medical University, Shanghai, China
| |
Collapse
|
477
|
Sudarsa IW, Aryanti C. Duration of Tumor-infiltrating Lymphocytes Assessment with Significant Overall Survival Prognostic Value in Locally Advanced Breast Cancer. Open Access Maced J Med Sci 2022. [DOI: 10.3889/oamjms.2022.7297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Background Tumor microenvironment, represented by tumor-infiltrating lymphocytes, was dynamically evolving throughout the time. Thus, in the interpretation of TIL as a prognostic factor overall survival of breast cancer, it is important to note the duration of the TIL examination until neoadjuvant chemotherapy is performed.
Aim To determine whether the assessment of TIL at a certain duration still has value as a prognostic factor for the overall survival of breast cancer and its cut off point.
Methods This was a retrospective cohort study that included subjects with locally advanced breast cancer in Sanglah General Hospital, registered in Bali’s Cancer Registry. The study has been approved by the institutional review board of Udayana University, Denpasar, Bali. Data collected were age, breast cancer subtype, value of TIL, time of NAC start, and duration of survival. Missing data were obtained from electronic medical records. TIL is then grouped into groups with low TIL (negative and +1) and high TIL (+2 and +3). Data analysis were done with Statistical Package for the Social Sciences 25.0.
Results As many as 150 subjects with locally advanced breast cancer who survived and died in 2011-2020 were analyzed. The mean age of subjects in this study was 48.7 (SD 9.3) years with a median survival of 47 months. The mean duration of the TIL asessment to NAC was 46.3 days (SD 24.5). The duration cut off point of TIL assessment to NAC that is valuable as breast cancer’s overall prognostic value was 31 days (AUC 0.716, Sensitivity 64.1%, Specificity 35.5%). In subjects with TIL examined for less than 31 days, it was found that TIL could significantly prognosticated the overall survival of breast cancer (p = 0.005). High TIL was associated with better overall survival and low TIL is associated with poor overall survival.
Discussion Tumor immune microenvironment played an important role in tumor progression and supression. High levels of TIL has been generally accepted as an indicator for a more robust anti-tumor immune response, thus yielding favorable outcome. Timing of TIL analysis was important to determine as microenvironment dynamically progressing. In this study, we proved that timing matters and only short-term duration of TIL assessment to NAC meaningful as a prognostic value for breast cancer’s overall survival.
Conclusion The duration of TIL to NAC assessment was important in determine the meaningful of TIL value in the prognostication overall survival of locally advanced breast cancer. The short-term TIL assessment duration (<31 days) predicted well, but not in the long term assessment duration. In the future, it is hoped that clinicians will be more critical in the interpretation of TIL, especially the duration of TIL assessment before NAC.
Collapse
|
478
|
Cristi F, Gutiérrez T, Hitt MM, Shmulevitz M. Genetic Modifications That Expand Oncolytic Virus Potency. Front Mol Biosci 2022; 9:831091. [PMID: 35155581 PMCID: PMC8826539 DOI: 10.3389/fmolb.2022.831091] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 01/06/2022] [Indexed: 12/20/2022] Open
Abstract
Oncolytic viruses (OVs) are a promising type of cancer therapy since they selectively replicate in tumor cells without damaging healthy cells. Many oncolytic viruses have progressed to human clinical trials, however, their performance as monotherapy has not been as successful as expected. Importantly, recent literature suggests that the oncolytic potential of these viruses can be further increased by genetically modifying the viruses. In this review, we describe genetic modifications to OVs that improve their ability to kill tumor cells directly, to dismantle the tumor microenvironment, or to alter tumor cell signaling and enhance anti-tumor immunity. These advances are particularly important to increase virus spread and reduce metastasis, as demonstrated in animal models. Since metastasis is the principal cause of mortality in cancer patients, having OVs designed to target metastases could transform cancer therapy. The genetic alterations reported to date are only the beginning of all possible improvements to OVs. Modifications described here could be combined together, targeting multiple processes, or with other non-viral therapies with potential to provide a strong and lasting anti-tumor response in cancer patients.
Collapse
Affiliation(s)
- Francisca Cristi
- Shmulevitz Laboratory, Department of Medical Microbiology and Immunology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Tomás Gutiérrez
- Goping Laboratory, Department of Biochemistry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Mary M. Hitt
- Hitt Laboratory, Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
- *Correspondence: Mary M. Hitt, ; Maya Shmulevitz,
| | - Maya Shmulevitz
- Shmulevitz Laboratory, Department of Medical Microbiology and Immunology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
- *Correspondence: Mary M. Hitt, ; Maya Shmulevitz,
| |
Collapse
|
479
|
Kahle ER, Han B, Chandrasekaran P, Phillips ER, Mulcahey MK, Lu XL, Marcolongo MS, Han L. Molecular Engineering of Pericellular Microniche via Biomimetic Proteoglycans Modulates Cell Mechanobiology. ACS NANO 2022; 16:1220-1230. [PMID: 35015500 PMCID: PMC9271520 DOI: 10.1021/acsnano.1c09015] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Molecular engineering of biological tissues using synthetic mimics of native matrix molecules can modulate the mechanical properties of the cellular microenvironment through physical interactions with existing matrix molecules, and in turn, mediate the corresponding cell mechanobiology. In articular cartilage, the pericellular matrix (PCM) is the immediate microniche that regulates cell fate, signaling, and metabolism. The negatively charged osmo-environment, as endowed by PCM proteoglycans, is a key biophysical cue for cell mechanosensing. This study demonstrated that biomimetic proteoglycans (BPGs), which mimic the ultrastructure and polyanionic nature of native proteoglycans, can be used to molecularly engineer PCM micromechanics and cell mechanotransduction in cartilage. Upon infiltration into bovine cartilage explant, we showed that localization of BPGs in the PCM leads to increased PCM micromodulus and enhanced chondrocyte intracellular calcium signaling. Applying molecular force spectroscopy, we revealed that BPGs integrate with native PCM through augmenting the molecular adhesion of aggrecan, the major PCM proteoglycan, at the nanoscale. These interactions are enabled by the biomimetic "bottle-brush" ultrastructure of BPGs and facilitate the integration of BPGs within the PCM. Thus, this class of biomimetic molecules can be used for modulating molecular interactions of pericellular proteoglycans and harnessing cell mechanosensing. Because the PCM is a prevalent feature of various cell types, BPGs hold promising potential for improving regeneration and disease modification for not only cartilage-related healthcare but many other tissues and diseases.
Collapse
Affiliation(s)
- Elizabeth R. Kahle
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States
| | - Biao Han
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States
| | - Prashant Chandrasekaran
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States
| | - Evan R. Phillips
- Department of Materials Science and Engineering, Drexel University, Philadelphia, PA 19104, United States
| | - Mary K. Mulcahey
- Department of Orthopaedic Surgery, Tulane University School of Medicine, New Orleans, LA 70112, United States
| | - X. Lucas Lu
- Department of Mechanical Engineering, University of Delaware, Newark, DE 19716, United States
| | - Michele S. Marcolongo
- Department of Materials Science and Engineering, Drexel University, Philadelphia, PA 19104, United States
- Department of Mechanical Engineering, Villanova University, Villanova, PA 19085, United States
| | - Lin Han
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States
| |
Collapse
|
480
|
Takabe P, Siiskonen H, Rönkä A, Kainulainen K, Pasonen-Seppänen S. The Impact of Hyaluronan on Tumor Progression in Cutaneous Melanoma. Front Oncol 2022; 11:811434. [PMID: 35127523 PMCID: PMC8813769 DOI: 10.3389/fonc.2021.811434] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 12/31/2021] [Indexed: 12/21/2022] Open
Abstract
The incidence of cutaneous melanoma is rapidly increasing worldwide. Cutaneous melanoma is an aggressive type of skin cancer, which originates from malignant transformation of pigment producing melanocytes. The main risk factor for melanoma is ultraviolet (UV) radiation, and thus it often arises from highly sun-exposed skin areas and is characterized by a high mutational burden. In addition to melanoma-associated mutations such as BRAF, NRAS, PTEN and cell cycle regulators, the expansion of melanoma is affected by the extracellular matrix surrounding the tumor together with immune cells. In the early phases of the disease, hyaluronan is the major matrix component in cutaneous melanoma microenvironment. It is a high-molecular weight polysaccharide involved in several physiological and pathological processes. Hyaluronan is involved in the inflammatory reactions associated with UV radiation but its role in melanomagenesis is still unclear. Although abundant hyaluronan surrounds epidermal and dermal cells in normal skin and benign nevi, its content is further elevated in dysplastic lesions and local tumors. At this stage hyaluronan matrix may act as a protective barrier against melanoma progression, or alternatively against immune cell attack. While in advanced melanoma, the content of hyaluronan decreases due to altered synthesis and degradation, and this correlates with poor prognosis. This review focuses on hyaluronan matrix in cutaneous melanoma and how the changes in hyaluronan metabolism affect the progression of melanoma.
Collapse
Affiliation(s)
- Piia Takabe
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| | - Hanna Siiskonen
- Department of Clinical Pathology, Kuopio University Hospital, Kuopio, Finland
| | - Aino Rönkä
- Department of Oncology, Kuopio University Hospital, Kuopio, Finland
| | - Kirsi Kainulainen
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| | - Sanna Pasonen-Seppänen
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
- *Correspondence: Sanna Pasonen-Seppänen,
| |
Collapse
|
481
|
Nilsen A, Hillestad T, Skingen VE, Aarnes E, Fjeldbo CS, Hompland T, Evensen TS, Stokke T, Kristensen GB, Grallert B, Lyng H. miR-200a/b/-429 downregulation is a candidate biomarker of tumor radioresistance and independent of hypoxia in locally advanced cervical cancer. Mol Oncol 2022; 16:1402-1419. [PMID: 35064630 PMCID: PMC8936520 DOI: 10.1002/1878-0261.13184] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 12/21/2021] [Accepted: 01/19/2022] [Indexed: 11/07/2022] Open
Abstract
Many patients with locally advanced cervical cancer experience recurrence within the radiation field after chemoradiotherapy. Biomarkers of tumor radioresistance are required to identify patients in need of intensified treatment. Here, the biomarker potential of miR-200 family members was investigated in this disease. Also, involvement of tumor hypoxia in the radioresistance mechanism was determined, using a previously defined 6-gene hypoxia classifier. miR-200 expression was measured in pre-treatment tumor biopsies of an explorative cohort (n=90) and validation cohort 1 (n=110) by RNA sequencing. Publicly available miR-200 data of 79 patients were included for validation of prognostic significance. A score based on expression of the miR-200a/b/-429 (miR-200a, miR-200b and miR-429) cluster showed prognostic significance in all cohorts. The score was significant in multivariate analysis of central pelvic recurrence. No association with distant recurrence or hypoxia status was found. Potential miRNA target genes were identified from gene expression profiles and showed enrichment of genes in extracellular matrix organization and cell adhesion. miR-200a/b/-429 overexpression had a pronounced radiosensitizing effect in tumor xenografts, whereas the effect was minor in vitro. In conclusion, miR-200a/b/-429 downregulation is a candidate biomarker of central pelvic recurrence and seems to predict cell-adhesion-mediated tumor radioresistance independent of clinical markers and hypoxia.
Collapse
Affiliation(s)
- Anja Nilsen
- Department of Radiation BiologyNorwegian Radium HospitalOslo University HospitalNorway
| | - Tiril Hillestad
- Department of Core FacilitiesNorwegian Radium HospitalOslo University HospitalNorway
| | - Vilde E. Skingen
- Department of Radiation BiologyNorwegian Radium HospitalOslo University HospitalNorway
| | - Eva‐Katrine Aarnes
- Department of Radiation BiologyNorwegian Radium HospitalOslo University HospitalNorway
| | - Christina S. Fjeldbo
- Department of Radiation BiologyNorwegian Radium HospitalOslo University HospitalNorway
| | - Tord Hompland
- Department of Radiation BiologyNorwegian Radium HospitalOslo University HospitalNorway
- Department of Core FacilitiesNorwegian Radium HospitalOslo University HospitalNorway
| | - Tina Sandø Evensen
- Department of Core FacilitiesNorwegian Radium HospitalOslo University HospitalNorway
| | - Trond Stokke
- Department of Core FacilitiesNorwegian Radium HospitalOslo University HospitalNorway
| | - Gunnar B. Kristensen
- Department of Gynecological OncologyNorwegian Radium HospitalOslo University HospitalNorway
- Institute of Cancer Genetics and InformaticsOslo University HospitalNorway
| | - Beata Grallert
- Department of Radiation BiologyNorwegian Radium HospitalOslo University HospitalNorway
| | - Heidi Lyng
- Department of Radiation BiologyNorwegian Radium HospitalOslo University HospitalNorway
- Department of PhysicsUniversity of OsloNorway
| |
Collapse
|
482
|
Deng J, Fleming JB. Inflammation and Myeloid Cells in Cancer Progression and Metastasis. Front Cell Dev Biol 2022; 9:759691. [PMID: 35127700 PMCID: PMC8814460 DOI: 10.3389/fcell.2021.759691] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 12/24/2021] [Indexed: 12/13/2022] Open
Abstract
To date, the most immunotherapy drugs act upon T cell surface proteins to promote tumoricidal T cell activity. However, this approach has to date been unsuccessful in certain solid tumor types including pancreatic, prostate cancer and glioblastoma. Myeloid-related innate immunity can promote tumor progression through direct and indirect effects on T cell activity; improved understanding of this field may provide another therapeutic avenue for patients with these tumors. Myeloid cells can differentiate into both pro-inflammatory and anti-inflammatory mature form depending upon the microenvironment. Most cancer type exhibit oncogenic activating point mutations (ex. P53 and KRAS) that trigger cytokines production. In addition, tumor environment (ex. Collagen, Hypoxia, and adenosine) also regulated inflammatory signaling cascade. Both the intrinsic and extrinsic factor driving the tumor immune microenvironment and regulating the differentiation and function of myeloid cells, T cells activity and tumor progression. In this review, we will discuss the relationship between cancer cells and myeloid cells-mediated tumor immune microenvironment to promote cancer progression and immunotherapeutic resistance. Furthermore, we will describe how cytokines and chemokines produced by cancer cells influence myeloid cells within immunosuppressive environment. Finally, we will comment on the development of immunotherapeutic strategies with respect to myeloid-related innate immunity.
Collapse
Affiliation(s)
- Jenying Deng
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Jason B. Fleming
- H. Lee Moffitt Cancer Center, Department of Gastrointestinal Oncology, Tampa, FL, United States
- *Correspondence: Jason B. Fleming,
| |
Collapse
|
483
|
Enkhbat M, Zhong B, Chang R, Geng J, Lu LS, Chen YJ, Wang PY. Harnessing Focal Adhesions to Accelerate p53 Accumulation and Anoikis of A549 Cells Using Colloidal Self-Assembled Patterns (cSAPs). ACS APPLIED BIO MATERIALS 2022; 5:322-333. [PMID: 35034455 DOI: 10.1021/acsabm.1c01109] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Extracellular matrix (ECM) of the tumor microenvironment (TME), including topography and biological molecules, is crucial in cancer cell attachment, growth, and even the sensitivity to the chemo and cell drugs treatment. This study hypothesizes that mimic ECM structures can alter the attachment and drug sensitivity of cancer cells. A family of artificial ECM called colloidal self-assembled patterns (cSAPs) was fabricated to mimic tumor ECM structures. Cell adhesion, proliferation, and drug sensitivity of the A549 non-small cell lung cancer (NSCLC) cells were studied on 24 cSAPs, named cSAP#1-cSAP#24, where surface topography and wettability were distinct. The results showed that cell adhesion and cell spreading were generally reduced on cSAPs compared to the flat controls. In addition, the synergistic effect of cSAPs and several chemo drugs on cell survival was investigated. Interestingly, A549 cells were more sensitive to the combination of doxorubicin and cSAP#4. Under this condition, the focal adhesion kinase (FAK) signaling was downregulated while p53 signaling was upregulated, confirmed by real-time PCR and western blot analysis. It indicates that the specific surface structure could induce higher drug sensitivity and in vitro anoikis of A549 cells. A serum alternative, human platelet lysate (hPL), and different cSAPs were examined to verify our hypothesis. The result further confirmed that cell adhesion strongly affected the drug sensitivity of A549 cells. This study demonstrates that the tumor ECM is vital in cancer cell activity and drug sensitivity; therefore, it should be considered in drug discovery and therapeutic regimens.
Collapse
Affiliation(s)
- Myagmartsend Enkhbat
- Oujiang Laboratory, Wenzhou, Zhejiang 325000, China.,Shenzhen Key Laboratory of Biomimetic Materials and Cellular Immunomodulation, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
| | - Boya Zhong
- Shenzhen Key Laboratory of Biomimetic Materials and Cellular Immunomodulation, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
| | - Ray Chang
- Shenzhen Key Laboratory of Biomimetic Materials and Cellular Immunomodulation, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
| | - Jin Geng
- Shenzhen Key Laboratory of Biomimetic Materials and Cellular Immunomodulation, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
| | - Long-Sheng Lu
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 110, Taiwan
| | - Yin-Ju Chen
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 110, Taiwan
| | - Peng-Yuan Wang
- Oujiang Laboratory, Wenzhou, Zhejiang 325000, China.,Shenzhen Key Laboratory of Biomimetic Materials and Cellular Immunomodulation, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
| |
Collapse
|
484
|
Tabebi M, Kumar Dutta R, Skoglund C, Söderkvist P, Gimm O. Loss of SDHB Induces a Metabolic Switch in the hPheo1 Cell Line toward Enhanced OXPHOS. Int J Mol Sci 2022; 23:560. [PMID: 35008989 PMCID: PMC8745660 DOI: 10.3390/ijms23010560] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 12/26/2021] [Accepted: 12/27/2021] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Enzymes of tricarboxylic acid (TCA) have recently been recognized as tumor suppressors. Mutations in the SDHB subunit of succinate dehydrogenase (SDH) cause pheochromocytomas and paragangliomas (PCCs/PGLs) and predispose patients to malignant disease with poor prognosis. METHODS Using the human pheochromocytoma cell line (hPheo1), we knocked down SDHB gene expression using CRISPR-cas9 technology. RESULTS Microarray gene expression analysis showed that >500 differentially expressed gene targets, about 54%, were upregulated in response to SDHB knock down. Notably, genes involved in glycolysis, hypoxia, cell proliferation, and cell differentiation were up regulated, whereas genes involved in oxidative phosphorylation (OXPHOS) were downregulated. In vitro studies show that hPheo1 proliferation is not affected negatively and the cells that survive by shifting their metabolism to the use of glutamine as an alternative energy source and promote OXPHOS activity. Knock down of SDHB expression results in a significant increase in GLUD1 expression in hPheo1 cells cultured as monolayer or as 3D culture. Analysis of TCGA data confirms the enhancement of GLUD1 in SDHB mutated/low expressed PCCs/PGLs. CONCLUSIONS Our data suggest that the downregulation of SDHB in PCCs/PGLs results in increased GLUD1 expression and may represent a potential biomarker and therapeutic target in SDHB mutated tumors and SDHB loss of activity-dependent diseases.
Collapse
Affiliation(s)
- Mouna Tabebi
- Department of Biomedical and Clinical Sciences (BKV), Linköping University, 581 83 Linköping, Sweden; (R.K.D.); (C.S.); (P.S.)
| | - Ravi Kumar Dutta
- Department of Biomedical and Clinical Sciences (BKV), Linköping University, 581 83 Linköping, Sweden; (R.K.D.); (C.S.); (P.S.)
| | - Camilla Skoglund
- Department of Biomedical and Clinical Sciences (BKV), Linköping University, 581 83 Linköping, Sweden; (R.K.D.); (C.S.); (P.S.)
| | - Peter Söderkvist
- Department of Biomedical and Clinical Sciences (BKV), Linköping University, 581 83 Linköping, Sweden; (R.K.D.); (C.S.); (P.S.)
- Clinical Genomics Linköping, Science for Life Laboratory, Linköping University, 581 83 Linköping, Sweden
| | - Oliver Gimm
- Department of Surgery and Department of Biomedical and Clinical Sciences (BKV), Linköping University, 581 83 Linköping, Sweden;
| |
Collapse
|
485
|
Lorenzo-Gómez R, Miranda-Castro R, de Los Toyos JR, de-Los-Santos-Álvarez N, Lobo-Castañón MJ. Aptamers targeting a tumor-associated extracellular matrix component: The human mature collagen XIα1. Anal Chim Acta 2022; 1189:339206. [PMID: 34815029 DOI: 10.1016/j.aca.2021.339206] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 10/16/2021] [Accepted: 10/19/2021] [Indexed: 12/11/2022]
Abstract
The extracellular matrix (ECM) plays an essential role in tumor progression and invasion through its continuous remodeling. The growth of most carcinomas is associated with an excessive collagen deposition that provides the proper environment for tumor development and chemoresistance. The α1 chain of a minor human collagen, type XI, is overexpressed in some tumor stroma, but not found in normal stroma. To test the clinical utility of this collagen as a cancer biomarker, specific receptors are needed. Available antibodies do not show enough selectivity or are directed toward the propeptide region that is cleaved when the protein is released to the ECM. Here we show the selection of an aptamer for the specific C-telopeptide region using a 16-mer peptide as the target for the SELEX. The aptamer selected with a Kd of ∼25 nM was able to capture the collagen XI from cell lysates. It was also used for target detection in a mixed antibody-aptamer sandwich assay showing it can be useful for diagnostic purposes in biological fluids.
Collapse
Affiliation(s)
- Ramón Lorenzo-Gómez
- Departamento de Química Física y Analítica, Facultad de Química, Universidad de Oviedo, Av. Julián Clavería 8, 33006, Oviedo, Spain; Instituto de Investigación Sanitaria del Principado de Asturias, Avenida de Roma, 33011, Oviedo, Spain.
| | - Rebeca Miranda-Castro
- Departamento de Química Física y Analítica, Facultad de Química, Universidad de Oviedo, Av. Julián Clavería 8, 33006, Oviedo, Spain; Instituto de Investigación Sanitaria del Principado de Asturias, Avenida de Roma, 33011, Oviedo, Spain.
| | - Juan R de Los Toyos
- Instituto de Investigación Sanitaria del Principado de Asturias, Avenida de Roma, 33011, Oviedo, Spain; Departamento de Inmunología, Facultad de Medicina y Ciencias de La Salud, Universidad de Oviedo, Av. Julián Clavería 6, 33006, Oviedo, Spain.
| | - Noemí de-Los-Santos-Álvarez
- Departamento de Química Física y Analítica, Facultad de Química, Universidad de Oviedo, Av. Julián Clavería 8, 33006, Oviedo, Spain; Instituto de Investigación Sanitaria del Principado de Asturias, Avenida de Roma, 33011, Oviedo, Spain.
| | - María Jesús Lobo-Castañón
- Departamento de Química Física y Analítica, Facultad de Química, Universidad de Oviedo, Av. Julián Clavería 8, 33006, Oviedo, Spain; Instituto de Investigación Sanitaria del Principado de Asturias, Avenida de Roma, 33011, Oviedo, Spain.
| |
Collapse
|
486
|
Thomas SC, Madaan T, Kamble NS, Siddiqui NA, Pauletti GM, Kotagiri N. Engineered Bacteria Enhance Immunotherapy and Targeted Therapy through Stromal Remodeling of Tumors. Adv Healthc Mater 2022; 11:e2101487. [PMID: 34738725 PMCID: PMC8770579 DOI: 10.1002/adhm.202101487] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 10/28/2021] [Indexed: 01/03/2023]
Abstract
Desmoplastic solid tumors are characterized by the rapid build-up of extracellular matrix (ECM) macromolecules, such as hyaluronic acid (HA). The resulting physiological barrier prevents the infiltration of immune cells and also impedes the delivery of anticancer agents. The development of a hypervesiculating Escherichia coli Nissle (ΔECHy) based tumor targeting bacterial system capable of distributing a fusion peptide, cytolysin A (ClyA)-hyaluronidase (Hy) via outer membrane vesicles (OMVs) is reported. The capability of targeting hypoxic tumors, manufacturing recombinant proteins in situ and the added advantage of an on-site OMV based distribution system makes the engineered bacterial vector a unique candidate for peptide delivery. The HA degrading potential of Hy for stromal modulation is combined with the cytolytic activity of ClyA followed by testing it within syngeneic cancer models. ΔECHy is combined with immune checkpoint antibodies and tyrosine kinase inhibitors (TKIs) to demonstrate that remodeling the tumor stroma results in the improvement of immunotherapy outcomes and enhancing the efficacy of biological signaling inhibitors. The biocompatibility of ΔECHy is also investigated to show that the engineered bacteria are effectively cleared, elicit minimal inflammatory and immune responses, and therefore could be a reliable candidate as a live biotherapeutic.
Collapse
Affiliation(s)
- Shindu C. Thomas
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, 231 Albert Sabin Way, Cincinnati, OH 45267, USA
| | - Tushar Madaan
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, 231 Albert Sabin Way, Cincinnati, OH 45267, USA
| | - Nitin S. Kamble
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, 231 Albert Sabin Way, Cincinnati, OH 45267, USA
| | - Nabil A. Siddiqui
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, 231 Albert Sabin Way, Cincinnati, OH 45267, USA
| | - Giovanni M. Pauletti
- Department of Pharmaceutical and Administrative Sciences, University of Health Sciences and Pharmacy in St. Louis, 1 Pharmacy Place, St. Louis, MO 63110, USA
| | - Nalinikanth Kotagiri
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, 231 Albert Sabin Way, Cincinnati, OH 45267, USA
| |
Collapse
|
487
|
DePalma TJ, Sivakumar H, Skardal A. Strategies for developing complex multi-component in vitro tumor models: Highlights in glioblastoma. Adv Drug Deliv Rev 2022; 180:114067. [PMID: 34822927 PMCID: PMC10560581 DOI: 10.1016/j.addr.2021.114067] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 11/05/2021] [Accepted: 11/18/2021] [Indexed: 02/06/2023]
Abstract
In recent years, many research groups have begun to utilize bioengineered in vitro models of cancer to study mechanisms of disease progression, test drug candidates, and develop platforms to advance personalized drug treatment options. Due to advances in cell and tissue engineering over the last few decades, there are now a myriad of tools that can be used to create such in vitro systems. In this review, we describe the considerations one must take when developing model systems that accurately mimic the in vivo tumor microenvironment (TME) and can be used to answer specific scientific questions. We will summarize the importance of cell sourcing in models with one or multiple cell types and outline the importance of choosing biomaterials that accurately mimic the native extracellular matrix (ECM) of the tumor or tissue that is being modeled. We then provide examples of how these two components can be used in concert in a variety of model form factors and conclude by discussing how biofabrication techniques such as bioprinting and organ-on-a-chip fabrication can be used to create highly reproducible complex in vitro models. Since this topic has a broad range of applications, we use the final section of the review to dive deeper into one type of cancer, glioblastoma, to illustrate how these components come together to further our knowledge of cancer biology and move us closer to developing novel drugs and systems that improve patient outcomes.
Collapse
Affiliation(s)
- Thomas J DePalma
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA
| | - Hemamylammal Sivakumar
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA
| | - Aleksander Skardal
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA; The Ohio State University and Arthur G. James Comprehensive Cancer Center, Columbus, OH 43210, USA
| |
Collapse
|
488
|
Taeb S, Ashrafizadeh M, Zarrabi A, Rezapoor S, Musa AE, Farhood B, Najafi M. Role of Tumor Microenvironment in Cancer Stem Cells Resistance to Radiotherapy. Curr Cancer Drug Targets 2021; 22:18-30. [PMID: 34951575 DOI: 10.2174/1568009622666211224154952] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 07/29/2021] [Accepted: 08/24/2021] [Indexed: 11/22/2022]
Abstract
Cancer is a chronic disorder that involves several elements of both the tumor and the host stromal cells. At present, the complex relationship between the various factors presents in the tumor microenvironment (TME) and tumor cells, as well as immune cells located within the TME, is still poorly known. Within the TME, the crosstalk of these factors and immune cells essentially determines how a tumor reacts to the treatment and how the tumor can ultimately be destroyed, remain dormant, or develop and metastasize. Also, in TME, reciprocal crosstalk between cancer-associated fibroblasts (CAFs), extracellular matrix (ECM), hypoxia-inducible factor (HIF) intensifies the proliferation capacity of cancer stem cells (CSCs). CSCs are subpopulation of cells that reside within the tumor bulk and have the capacity to self-renew, differentiate, and repair DNA damage. These characteristics make CSCs develop resistance to a variety of treatments, such as radiotherapy (RT). RT is a frequent and often curative treatment for local cancer which mediates tumor elimination by cytotoxic actions. Also, cytokines and growth factors that are released into TME, have been involved in the activation of tumor radioresistance and the induction of different immune cells, altering local immune responses. In this review, we discuss the pivotal role of TME in resistance of CSCs to RT.
Collapse
Affiliation(s)
- Shahram Taeb
- Department of Radiology, School of Paramedical Sciences, Guilan University of Medical Sciences, Rasht, Iran
| | - Milad Ashrafizadeh
- Faculty of Engineering and Natural Sciences, Sabanci University, Orta Mahalle, Üniversite Caddesi No. 27, Orhanlı, Tuzla, 34956 , Turkey
| | - Ali Zarrabi
- Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla 34956, Turkey
| | - Saeed Rezapoor
- Department of Radiology, Faculty of Paramedical, Tehran University of Medical Sciences, Iran
| | - Ahmed Eleojo Musa
- Department of Medical Physics, Tehran University of Medical Sciences (International Campus), Iran
| | - Bagher Farhood
- Department of Medical Physics and Radiology, Faculty of Paramedical Sciences, Kashan University of Medical Sciences., Iraq
| | - Masoud Najafi
- Medical Technology Research Center, Institute of Health Technology, Kermanshah University of Medical Sciences, Iran
| |
Collapse
|
489
|
Lee YE, Yuk CM, Lee M, Han KC, Jun E, Kim TS, Ku JL, Im SG, Lee E, Jang M. Facile discovery of a therapeutic agent for NK-mediated synergistic antitumor effects using a patient-derived 3D platform. Biomater Sci 2021; 10:678-691. [PMID: 34940764 DOI: 10.1039/d1bm01699g] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Despite the essential roles of natural killer (NK) cells in cancer treatment, the physical barrier and biological cues of the tumor microenvironment (TME) may induce NK cell dysfunction, causing their poor infiltration into tumors. The currently available two-dimensional (2D) cancer-NK co-culture systems hardly represent the characteristics of TME and are not suitable for tracking the infiltration of immune cells and assessing the efficacy of immunotherapy. This study aims to monitor NK-mediated cancer cell killing using a polymer thin film-based, 3D assay platform that contains highly tumorigenic cancer spheroids. A poly(cyclohexyl methacrylate) (pCHMA)-coated surface enables the generation of tumorigenic spheroids from pancreatic cancer patient-derived cancer cells, showing considerable amounts of extracellular matrix (ECM) proteins and cancer stem cell (CSC)-like characteristics. The 3D spheroid-based assay platform allows rapid discovery of a therapeutic agent for synergistic NK-mediated cytotoxicity through imaging-based high-content screening. In detail, the small molecule C19, known as a multi-epithelial-mesenchymal transition pathway inhibitor, is shown to enhance NK activation and infiltration via modulation of the ECM, resulting in synergistic cytotoxicity against cancer spheroids. This 3D biomimetic co-culture assay platform provides promising applications for predicting patient-specific responses to immunotherapy through advanced therapeutic combinations involving a chemical drug and immune cells.
Collapse
Affiliation(s)
- Young Eun Lee
- Theragnosis Research Center, Biomedical Research Division, Korea Institute of Science and Technology, Seongbuk-Gu, Seoul 02792, Republic of Korea. .,Department of Life Sciences, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea
| | - Chae Min Yuk
- Theragnosis Research Center, Biomedical Research Division, Korea Institute of Science and Technology, Seongbuk-Gu, Seoul 02792, Republic of Korea.
| | - Minseok Lee
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea.
| | - Ki-Cheol Han
- Theragnosis Research Center, Biomedical Research Division, Korea Institute of Science and Technology, Seongbuk-Gu, Seoul 02792, Republic of Korea.
| | - Eunsung Jun
- Department of Convergence Medicine, Asan Institute for Life Sciences, University of Ulsan College of Medicine and Asan Medical Center, Seoul 05505, Republic of Korea.,Division of Hepato-Biliary and Pancreatic Surgery, Department of Surgery, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505, Republic of Korea
| | - Tae Sung Kim
- Department of Life Sciences, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea
| | - Ja-Lok Ku
- Korean Cell Line Bank, Laboratory of Cell Biology, Cancer Research Institute, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Sung G Im
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea.
| | - Eunjung Lee
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea.
| | - Mihue Jang
- Theragnosis Research Center, Biomedical Research Division, Korea Institute of Science and Technology, Seongbuk-Gu, Seoul 02792, Republic of Korea. .,KHU-KIST Department of Converging Science and Technology, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
490
|
Chen Y, Zheng X, Wu C. The Role of the Tumor Microenvironment and Treatment Strategies in Colorectal Cancer. Front Immunol 2021; 12:792691. [PMID: 34925375 PMCID: PMC8674693 DOI: 10.3389/fimmu.2021.792691] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 11/15/2021] [Indexed: 12/17/2022] Open
Abstract
Colorectal cancer (CRC) has the second highest mortality rate among all cancers worldwide. Surgery, chemotherapy, radiotherapy, molecular targeting and other treatment methods have significantly prolonged the survival of patients with CRC. Recently, the emergence of tumor immunotherapy represented by immune checkpoint inhibitors (ICIs) has brought new immunotherapy options for the treatment of advanced CRC. As the efficacy of ICIs is closely related to the tumor immune microenvironment (TME), it is necessary to clarify the relationship between the immune microenvironment of CRC and the efficacy of immunotherapy to ensure that the appropriate drugs are selected. We herein review the latest research progress in the immune microenvironment and strategies related to immunotherapy for CRC. We hope that this review helps in the selection of appropriate treatment strategies for CRC patients.
Collapse
Affiliation(s)
- Yaping Chen
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Xiao Zheng
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Changping Wu
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, China.,Department of Oncology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| |
Collapse
|
491
|
Prognostic Impact of Sarcopenia in Patients with Metastatic Hormone-Sensitive Prostate Cancer. Cancers (Basel) 2021; 13:cancers13246345. [PMID: 34944964 PMCID: PMC8699789 DOI: 10.3390/cancers13246345] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 12/14/2021] [Accepted: 12/15/2021] [Indexed: 12/24/2022] Open
Abstract
The clinical value of sarcopenia has not been determined yet in metastatic hormone-sensitive prostate cancer (mHSPC). We retrospectively evaluated data of 70 consecutive patients with mHSPC receiving treatment with either early docetaxel (n = 42) or abiraterone acetate (n = 28) between July 2018 and April 2021. Skeletal muscle index was calculated from cross-sectional areas of skeletal muscle on baseline computed tomography (CT), defining sarcopenia as a skeletal muscle index of ≤52.4 cm2/m2. Failure-free survival (FFS), radiographic progression-free survival, and time to prostate-specific antigen (PSA) progression were estimated using the Kaplan-Meier method, and differences in survival probability were compared using the log-rank test. Cox proportional hazards regression analysis was conducted to identify the predictors of clinical outcomes. Patients with sarcopenia (n = 47) had shorter FFS than those without sarcopenia (n = 23) (median, 20.1 months vs. not reached; log-rank p < 0.001). Sarcopenia was independently associated with shorter FFS (hazard ratio (HR), 6.69; 95% confidence interval (CI), 1.57-28.49; p = 0.010) and time to PSA progression (HR, 12.91; 95% CI, 1.08-153.85; p = 0.043). In conclusion, sarcopenia is an independent prognostic factor for poor FFS and time to PSA progression in patients with mHSPC who receive early docetaxel or abiraterone acetate treatment.
Collapse
|
492
|
Cienfuegos-Jimenez O, Vazquez-Garza E, Rojas-Martinez A. CAR-NK Cells for Cancer Therapy: Molecular Redesign of the Innate Antineoplastic Response. Curr Gene Ther 2021; 22:303-318. [PMID: 34923939 DOI: 10.2174/1566523222666211217091724] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 10/14/2021] [Accepted: 10/22/2021] [Indexed: 11/22/2022]
Abstract
The Chimeric Antigen Receptor (CAR) has arisen as a powerful synthetic biology-based technology with demonstrated versatility for implementation in T and NK cells. Despite CAR T cell successes in clinical trials, several challenges remain to be addressed regarding adverse events and long-term efficacy. NK cells present an attractive alternative with intrinsic advantages over T cells for treating solid and liquid tumors. Early preclinical and clinical trials suggest at least two major advantages: improved safety and an off-the-shelf application in patients due to its HLA independence. Due to the early stages of CAR NK translation to clinical trials, limited data is currently available. By analyzing these results, it seems that CAR NK cells could offer a reduced probability of Cytokine Release Syndrome (CRS) or Graft versus Host Disease (GvHD) in cancer patients, reducing safety concerns. Furthermore, NK cell therapy approaches may be boosted by combining it with immunological checkpoint inhibitors and by implementing genetic circuits to direct CAR-bearing cell behavior. This review provides a description of the CAR technology for modifying NK cells and the translation from preclinical studies to early clinical trials in this new field of immunotherapy.
Collapse
Affiliation(s)
- Oscar Cienfuegos-Jimenez
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud. Monterrey, CP64710, Mexico
| | - Eduardo Vazquez-Garza
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud. Monterrey, CP64710, Mexico
| | - Augusto Rojas-Martinez
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud. Monterrey, CP64710, Mexico
| |
Collapse
|
493
|
Carvalho TMA, Di Molfetta D, Greco MR, Koltai T, Alfarouk KO, Reshkin SJ, Cardone RA. Tumor Microenvironment Features and Chemoresistance in Pancreatic Ductal Adenocarcinoma: Insights into Targeting Physicochemical Barriers and Metabolism as Therapeutic Approaches. Cancers (Basel) 2021; 13:6135. [PMID: 34885243 PMCID: PMC8657427 DOI: 10.3390/cancers13236135] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 11/26/2021] [Accepted: 12/01/2021] [Indexed: 12/14/2022] Open
Abstract
Currently, the median overall survival of PDAC patients rarely exceeds 1 year and has an overall 5-year survival rate of about 9%. These numbers are anticipated to worsen in the future due to the lack of understanding of the factors involved in its strong chemoresistance. Chemotherapy remains the only treatment option for most PDAC patients; however, the available therapeutic strategies are insufficient. The factors involved in chemoresistance include the development of a desmoplastic stroma which reprograms cellular metabolism, and both contribute to an impaired response to therapy. PDAC stroma is composed of immune cells, endothelial cells, and cancer-associated fibroblasts embedded in a prominent, dense extracellular matrix associated with areas of hypoxia and acidic extracellular pH. While multiple gene mutations are involved in PDAC initiation, this desmoplastic stroma plays an important role in driving progression, metastasis, and chemoresistance. Elucidating the mechanisms underlying PDAC resistance are a prerequisite for designing novel approaches to increase patient survival. In this review, we provide an overview of the stromal features and how they contribute to the chemoresistance in PDAC treatment. By highlighting new paradigms in the role of the stromal compartment in PDAC therapy, we hope to stimulate new concepts aimed at improving patient outcomes.
Collapse
Affiliation(s)
- Tiago M. A. Carvalho
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, 70126 Bari, Italy; (D.D.M.); (M.R.G.); (S.J.R.); (R.A.C.)
| | - Daria Di Molfetta
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, 70126 Bari, Italy; (D.D.M.); (M.R.G.); (S.J.R.); (R.A.C.)
| | - Maria Raffaella Greco
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, 70126 Bari, Italy; (D.D.M.); (M.R.G.); (S.J.R.); (R.A.C.)
| | | | - Khalid O. Alfarouk
- Al-Ghad International College for Applied Medical Sciences, Al-Madinah Al-Munwarah 42316, Saudi Arabia;
| | - Stephan J. Reshkin
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, 70126 Bari, Italy; (D.D.M.); (M.R.G.); (S.J.R.); (R.A.C.)
| | - Rosa A. Cardone
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, 70126 Bari, Italy; (D.D.M.); (M.R.G.); (S.J.R.); (R.A.C.)
| |
Collapse
|
494
|
Gupta P, Neupane YR, Parvez S, Kohli K. Recent advances in targeted nanotherapeutic approaches for breast cancer management. Nanomedicine (Lond) 2021; 16:2605-2631. [PMID: 34854336 DOI: 10.2217/nnm-2021-0281] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Breast cancer is the most commonly occurring tumor disease worldwide. Breast cancer is currently managed by conventional chemotherapy, which is inadequate in curbing this heterogeneous disease and results in off-site toxic effects, suggesting effective treatment approaches with better therapeutic profiles are needed. This review, therefore, focuses on the recent advancements in delivering therapeutics to the target site using passive and/or active targeted nanodrug-delivery systems to ameliorate endolysosomal escape. In addition, recent strategies in targeting breast cancer stem cells are discussed. The role of naturally cell-secreted nanovesicles (exosomes) in the management of triple-negative breast cancer is also discussed.
Collapse
Affiliation(s)
- Priya Gupta
- Department of Pharmaceutics, School of Pharmaceutical Education & Research, Jamia Hamdard, New Delhi, 110062, India
| | - Yub Raj Neupane
- Department of Pharmacy, National University of Singapore, Singapore, 117559
| | - Suhel Parvez
- Department of Toxicology, School of Chemical & Life Sciences, Jamia Hamdard, New Delhi, 110062, India
| | - Kanchan Kohli
- Department of Pharmaceutics, School of Pharmaceutical Education & Research, Jamia Hamdard, New Delhi, 110062, India.,Lloyd Institute of Management & Technology (Pharm.), Plot No. 11, Knowledge Park-II, Greater Noida, 201308, Uttar Pradesh, India
| |
Collapse
|
495
|
Aghlara-Fotovat S, Nash A, Kim B, Krencik R, Veiseh O. Targeting the extracellular matrix for immunomodulation: applications in drug delivery and cell therapies. Drug Deliv Transl Res 2021; 11:2394-2413. [PMID: 34176099 DOI: 10.1007/s13346-021-01018-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/10/2021] [Indexed: 12/12/2022]
Abstract
Host immune cells interact bi-directionally with their extracellular matrix (ECM) to receive and deposit molecular signals, which orchestrate cellular activation, proliferation, differentiation, and function to maintain healthy tissue homeostasis. In response to pathogens or damage, immune cells infiltrate diseased sites and synthesize critical ECM molecules such as glycoproteins, proteoglycans, and glycosaminoglycans to promote healing. When the immune system misidentifies pathogens or fails to survey damaged cells effectively, maladies such as chronic inflammation, autoimmune diseases, and cancer can develop. In these conditions, it is essential to restore balance to the body through modulation of the immune system and the ECM. This review details the components of dysregulated ECM implicated in pathogenic environments and therapeutic approaches to restore tissue homeostasis. We evaluate emerging strategies to overcome inflamed, immune inhibitory, and otherwise diseased microenvironments, including mechanical stimulation, targeted proteases, adoptive cell therapy, mechanomedicine, and biomaterial-based cell therapeutics. We highlight various strategies that have produced efficacious responses in both pre-clinical and human trials and identify additional opportunities to develop next-generation interventions. Significantly, we identify a need for therapies to address dense or fibrotic tissue for the treatment of organ tissue damage and various cancer subtypes. Finally, we conclude that therapeutic techniques that disrupt, evade, or specifically target the pathogenic microenvironment have a high potential for improving therapeutic outcomes and should be considered a priority for immediate exploration. A schematic showing the various methods of extracellular matrix disruption/targeting in both fibrotic and cancerous environments. a Biomaterial-based cell therapy can be used to deliver anti-inflammatory cytokines, chemotherapeutics, or other factors for localized, slow release of therapeutics. b Mechanotherapeutics can be used to inhibit the deposition of molecules such as collagen that affect stiffness. c Ablation of the ECM and target tissue can be accomplished via mechanical degradation such as focused ultrasound. d Proteases can be used to improve the distribution of therapies such as oncolytic virus. e Localization of therapeutics such as checkpoint inhibitors can be improved with the targeting of specific ECM components, reducing off-target effects and toxicity.
Collapse
Affiliation(s)
| | - Amanda Nash
- Department of Bioengineering, Rice University, Houston, TX, 77030, USA
| | - Boram Kim
- Department of Bioengineering, Rice University, Houston, TX, 77030, USA
| | - Robert Krencik
- Center for Neuroregeneration, Department of Neurosurgery, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Omid Veiseh
- Department of Bioengineering, Rice University, Houston, TX, 77030, USA.
| |
Collapse
|
496
|
Chung SW, Xie Y, Suk JS. Overcoming physical stromal barriers to cancer immunotherapy. Drug Deliv Transl Res 2021; 11:2430-2447. [PMID: 34351575 PMCID: PMC8571040 DOI: 10.1007/s13346-021-01036-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/19/2021] [Indexed: 12/17/2022]
Abstract
Immunotherapy has emerged as an unprecedented hope for the treatment of notoriously refractory cancers. Numerous investigational drugs and immunotherapy-including combination regimens are under preclinical and clinical investigation. However, only a small patient subpopulation across different types of cancer responds to the therapy due to the presence of several mechanisms of resistance. There have been extensive efforts to overcome this limitation and to expand the patient population that could be benefited by this state-of-the-art therapeutic modality. Among various causes of the resistance, we here focus on physical stromal barriers that impede the access of immunotherapeutic drug molecules and/or native and engineered immune cells to cancer tissues and cells. Two primary stromal barriers that contribute to the resistance include aberrant tumor vasculatures and excessive extracellular matrix build-ups that restrict extravasation and infiltration, respectively, of molecular and cellular immunotherapeutic agents into tumor tissues. Here, we review the features of these barriers that limit the efficacy of immunotherapy and discuss recent advances that could potentially help immunotherapy overcome the barriers and improve therapeutic outcomes.
Collapse
Affiliation(s)
- Seung Woo Chung
- Center for Nanomedicine, The Johns Hopkins University School of Medicine, 400 N. Broadway, Baltimore, MD, 602921231, USA
- Department of Ophthalmology, The Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA
| | - Yunxuan Xie
- Center for Nanomedicine, The Johns Hopkins University School of Medicine, 400 N. Broadway, Baltimore, MD, 602921231, USA
- Department of Ophthalmology, The Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, 21218, USA
| | - Jung Soo Suk
- Center for Nanomedicine, The Johns Hopkins University School of Medicine, 400 N. Broadway, Baltimore, MD, 602921231, USA.
- Department of Ophthalmology, The Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA.
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, 21218, USA.
| |
Collapse
|
497
|
Tang R, Dang M, Zhang X, Tao J, Shi W, Lu W, Yu R, Su X, Tang Y, Teng Z. Disrupting stromal barriers to enhance photothermal-chemo therapy using a halofuginone-loaded Janus mesoporous nanoplatform. J Colloid Interface Sci 2021; 610:313-320. [PMID: 34923269 DOI: 10.1016/j.jcis.2021.11.190] [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: 09/14/2021] [Revised: 11/29/2021] [Accepted: 11/29/2021] [Indexed: 12/23/2022]
Abstract
Dense tumor stroma is the physiological barrier in drug delivery that prevents anticancer drugs from entering the tumor, thereby seriously limiting the drugs' therapeutic effect. In this study, a Janus nanoplatform consisting of periodic mesoporous organosilica-coated platinum nanoplatforms (JPMO-Pt) and anti-stroma drug halofuginone (HF) (denoted as JPMO-Pt-HF), was developed to deplete the tumor stroma and synergistically treat breast cancer in BALB/c mice. The prepared JPMO-Pt had a uniform size of 245 nm, a good dispersion, an excellent in vitro and in vivo biocompatibility, and a high loading capacity for HF (up to 50 μg/mg). The antitumor experiments showed that the survival rate of 4 T1 cells exhibited an obvious downward trend when the cells were incubated with the JPMO-Pt-HF and irradiated with 808 nm laser. Moreover, the cell survival rate was only about 10% at 48 h when the HF concentration was 2.0 μg/mL. Notably, JPMO-Pt-HF under irradiation had an excellent synergistic therapeutic effect on tumor cells. In vivo antitumor experiment further showed that the JPMO-Pt-HF, in combination with laser irradiation, could minimize tumor growth, showing significantly better effects than those observed for the case of monotherapy involving photothermal therapy (PTT) (152 vs. 670 mm3, p < 0.0001) and HF (152 vs. 419 mm3, p = 0.0208). In addition, immunohistochemistry of tumor tissues indicated that JPMO-Pt-HF obviously reduced the relative collagen and α-smooth muscle actin (α-SMA) area fraction. Taken together, this research designs a new platform that not only possesses the ability to degrade the tumor matrix but also combines PTT and chemotherapeutic effects, and holds promise for effective tumor treatment.
Collapse
Affiliation(s)
- Rui Tang
- Key Laboratory for Organic Electronics and Information Displays and Institute of Advanced Materials, Jiangsu Key Laboratory for Biosensors, Jiangsu National Synergetic Innovation Centre for Advanced Materials, Nanjing University of Posts and Telecommunications, Nanjing, 210046 Jiangsu, PR China
| | - Meng Dang
- Key Laboratory for Organic Electronics and Information Displays and Institute of Advanced Materials, Jiangsu Key Laboratory for Biosensors, Jiangsu National Synergetic Innovation Centre for Advanced Materials, Nanjing University of Posts and Telecommunications, Nanjing, 210046 Jiangsu, PR China
| | - Xiaojun Zhang
- Department of Medical Imaging, Children's hospital of Nanjing Medical University, 210008 Jiangsu, PR China
| | - Jun Tao
- Key Laboratory for Organic Electronics and Information Displays and Institute of Advanced Materials, Jiangsu Key Laboratory for Biosensors, Jiangsu National Synergetic Innovation Centre for Advanced Materials, Nanjing University of Posts and Telecommunications, Nanjing, 210046 Jiangsu, PR China
| | - Wenhui Shi
- Key Laboratory for Organic Electronics and Information Displays and Institute of Advanced Materials, Jiangsu Key Laboratory for Biosensors, Jiangsu National Synergetic Innovation Centre for Advanced Materials, Nanjing University of Posts and Telecommunications, Nanjing, 210046 Jiangsu, PR China
| | - Wei Lu
- Key Laboratory for Organic Electronics and Information Displays and Institute of Advanced Materials, Jiangsu Key Laboratory for Biosensors, Jiangsu National Synergetic Innovation Centre for Advanced Materials, Nanjing University of Posts and Telecommunications, Nanjing, 210046 Jiangsu, PR China
| | - Ruifa Yu
- Key Laboratory for Organic Electronics and Information Displays and Institute of Advanced Materials, Jiangsu Key Laboratory for Biosensors, Jiangsu National Synergetic Innovation Centre for Advanced Materials, Nanjing University of Posts and Telecommunications, Nanjing, 210046 Jiangsu, PR China
| | - Xiaodan Su
- Key Laboratory for Organic Electronics and Information Displays and Institute of Advanced Materials, Jiangsu Key Laboratory for Biosensors, Jiangsu National Synergetic Innovation Centre for Advanced Materials, Nanjing University of Posts and Telecommunications, Nanjing, 210046 Jiangsu, PR China
| | - Yuxia Tang
- Department of Medical Imaging, Jinling Hospital, School of Medicine, Nanjing University, 210002 Jiangsu, PR China.
| | - Zhaogang Teng
- Key Laboratory for Organic Electronics and Information Displays and Institute of Advanced Materials, Jiangsu Key Laboratory for Biosensors, Jiangsu National Synergetic Innovation Centre for Advanced Materials, Nanjing University of Posts and Telecommunications, Nanjing, 210046 Jiangsu, PR China; State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210093, PR China.
| |
Collapse
|
498
|
Aiba T, Hattori C, Sugisaka J, Shimizu H, Ono H, Domeki Y, Saito R, Kawana S, Kawashima Y, Terayama K, Toi Y, Nakamura A, Yamanda S, Kimura Y, Suzuki Y, Niida A, Sugawara S. Gene expression signatures as candidate biomarkers of response to PD-1 blockade in non-small cell lung cancers. PLoS One 2021; 16:e0260500. [PMID: 34843570 PMCID: PMC8629226 DOI: 10.1371/journal.pone.0260500] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 11/10/2021] [Indexed: 11/18/2022] Open
Abstract
Although anti-PD-1/PD-L1 monotherapy has achieved clinical success in non-small cell lung cancer (NSCLC), definitive predictive biomarkers remain to be elucidated. In this study, we performed whole-transcriptome sequencing of pretreatment tumor tissue samples and pretreatment and on-treatment whole blood samples (WB) samples obtained from a clinically annotated cohort of NSCLC patients (n = 40) treated with nivolumab (anti-PD-1) monotherapy. Using a single-sample gene set enrichment scoring method, we found that the tumors of responders with lung adenocarcinoma (LUAD, n = 20) are inherently immunogenic to promote antitumor immunity, whereas those with lung squamous cell carcinoma (LUSC, n = 18) have a less immunosuppressive tumor microenvironment. These findings suggested that nivolumab may function as a molecular targeted agent in LUAD and as an immunomodulating agent in LUSC. In addition, our study explains why the reliability of PD-L1 expression on tumor cells as a predictive biomarker for the response to nivolumab monotherapy is quite different between LUAD and LUSC.
Collapse
Affiliation(s)
- Tomoiki Aiba
- Department of Pulmonary Medicine, Sendai Kousei Hospital, Sendai, Japan
- * E-mail:
| | - Chieko Hattori
- Department of Pulmonary Medicine, Sendai Kousei Hospital, Sendai, Japan
| | - Jun Sugisaka
- Department of Pulmonary Medicine, Sendai Kousei Hospital, Sendai, Japan
| | - Hisashi Shimizu
- Department of Pulmonary Medicine, Sendai Kousei Hospital, Sendai, Japan
| | - Hirotaka Ono
- Department of Pulmonary Medicine, Sendai Kousei Hospital, Sendai, Japan
| | - Yutaka Domeki
- Department of Pulmonary Medicine, Sendai Kousei Hospital, Sendai, Japan
| | - Ryohei Saito
- Department of Pulmonary Medicine, Sendai Kousei Hospital, Sendai, Japan
| | - Sachiko Kawana
- Department of Pulmonary Medicine, Sendai Kousei Hospital, Sendai, Japan
| | - Yosuke Kawashima
- Department of Pulmonary Medicine, Sendai Kousei Hospital, Sendai, Japan
| | - Keisuke Terayama
- Department of Pulmonary Medicine, Sendai Kousei Hospital, Sendai, Japan
| | - Yukihiro Toi
- Department of Pulmonary Medicine, Sendai Kousei Hospital, Sendai, Japan
| | - Atsushi Nakamura
- Department of Pulmonary Medicine, Sendai Kousei Hospital, Sendai, Japan
| | - Shinsuke Yamanda
- Department of Pulmonary Medicine, Sendai Kousei Hospital, Sendai, Japan
| | - Yuichiro Kimura
- Department of Pulmonary Medicine, Sendai Kousei Hospital, Sendai, Japan
| | - Yutaka Suzuki
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Chiba, Japan
| | - Atsushi Niida
- Human Genome Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Shunichi Sugawara
- Department of Pulmonary Medicine, Sendai Kousei Hospital, Sendai, Japan
| |
Collapse
|
499
|
Diehl V, Huber LS, Trebicka J, Wygrecka M, Iozzo RV, Schaefer L. The Role of Decorin and Biglycan Signaling in Tumorigenesis. Front Oncol 2021; 11:801801. [PMID: 34917515 PMCID: PMC8668865 DOI: 10.3389/fonc.2021.801801] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 11/09/2021] [Indexed: 12/12/2022] Open
Abstract
The complex and adaptive nature of malignant neoplasm constitute a major challenge for the development of effective anti-oncogenic therapies. Emerging evidence has uncovered the pivotal functions exerted by the small leucine-rich proteoglycans, decorin and biglycan, in affecting tumor growth and progression. In their soluble forms, decorin and biglycan act as powerful signaling molecules. By receptor-mediated signal transduction, both proteoglycans modulate key processes vital for tumor initiation and progression, such as autophagy, inflammation, cell-cycle, apoptosis, and angiogenesis. Despite of their structural homology, these two proteoglycans interact with distinct cell surface receptors and thus modulate distinct signaling pathways that ultimately affect cancer development. In this review, we summarize growing evidence for the complex roles of decorin and biglycan signaling in tumor biology and address potential novel therapeutic implications.
Collapse
Affiliation(s)
- Valentina Diehl
- Institute of Pharmacology and Toxicology, Goethe University, Frankfurt, Germany
| | - Lisa Sophie Huber
- Institute of Pharmacology and Toxicology, Goethe University, Frankfurt, Germany
| | - Jonel Trebicka
- Department of Internal Medicine I, Goethe University, Frankfurt, Germany
| | - Malgorzata Wygrecka
- Center for Infection and Genomics of the Lung, Member of the German Center for Lung Research, University of Giessen and Marburg Lung Center, Giessen, Germany
| | - Renato V. Iozzo
- Department of Pathology, Anatomy and Cell Biology and the Translational Cellular Oncology Program, Sidney Kimmel Cancer Center, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA, United States
| | - Liliana Schaefer
- Institute of Pharmacology and Toxicology, Goethe University, Frankfurt, Germany
| |
Collapse
|
500
|
Kim S, Kim SA, Han J, Kim IS. Rho-Kinase as a Target for Cancer Therapy and Its Immunotherapeutic Potential. Int J Mol Sci 2021; 22:ijms222312916. [PMID: 34884721 PMCID: PMC8657458 DOI: 10.3390/ijms222312916] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 11/24/2021] [Accepted: 11/24/2021] [Indexed: 02/07/2023] Open
Abstract
Cancer immunotherapy is fast rising as a prominent new pillar of cancer treatment, harnessing the immune system to fight against numerous types of cancer. Rho-kinase (ROCK) pathway is involved in diverse cellular activities, and is therefore the target of interest in various diseases at the cellular level including cancer. Indeed, ROCK is well-known for its involvement in the tumor cell and tumor microenvironment, especially in its ability to enhance tumor cell progression, migration, metastasis, and extracellular matrix remodeling. Importantly, ROCK is also considered to be a novel and effective modulator of immune cells, although further studies are needed. In this review article, we describe the various activities of ROCK and its potential to be utilized in cancer treatment, particularly in cancer immunotherapy, by shining a light on its activities in the immune system.
Collapse
Affiliation(s)
- Seohyun Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Korea; (S.K.); (S.A.K.); (J.H.)
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea
| | - Seong A. Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Korea; (S.K.); (S.A.K.); (J.H.)
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea
| | - Jihoon Han
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Korea; (S.K.); (S.A.K.); (J.H.)
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea
| | - In-San Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Korea; (S.K.); (S.A.K.); (J.H.)
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea
- Correspondence:
| |
Collapse
|