1
|
Sharma P, Otto M. Multifunctional nanocomposites modulating the tumor microenvironment for enhanced cancer immunotherapy. Bioact Mater 2024; 31:440-462. [PMID: 37701452 PMCID: PMC10494322 DOI: 10.1016/j.bioactmat.2023.08.022] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 08/09/2023] [Accepted: 08/28/2023] [Indexed: 09/14/2023] Open
Abstract
Cancer immunotherapy has gained momentum for treating malignant tumors over the past decade. Checkpoint blockade and chimeric antigen receptor cell therapy (CAR-T) have shown considerable potency against liquid and solid cancers. However, the tumor microenvironment (TME) is highly immunosuppressive and hampers the effect of currently available cancer immunotherapies on overall treatment outcomes. Advancements in the design and engineering of nanomaterials have opened new avenues to modulate the TME. Progress in the current nanocomposite technology can overcome immunosuppression and trigger robust immunotherapeutic responses by integrating synergistic functions of different molecules. We will review recent advancements in nanomedical applications and discuss specifically designed nanocomposites modulating the TME for cancer immunotherapy. In addition, we provide information on the current landscape of clinical-stage nanocomposites for cancer immunotherapy.
Collapse
Affiliation(s)
- Prashant Sharma
- Department of Child Health, University of Arizona College of Medicine-Phoenix, ABC1 Building, 425 N 5th Street, Phoenix, AZ, 85004, USA
| | - Mario Otto
- Department of Child Health, University of Arizona College of Medicine-Phoenix, ABC1 Building, 425 N 5th Street, Phoenix, AZ, 85004, USA
- Center for Cancer and Blood Disorders (CCBD), Phoenix Children's, 1919 E Thomas Rd, Phoenix, AZ, 85016, USA
| |
Collapse
|
2
|
Qiao H, Li H. PLP2 Could Be a Prognostic Biomarker and Potential Treatment Target in Glioblastoma Multiforme. Pharmgenomics Pers Med 2023; 16:991-1009. [PMID: 37964785 PMCID: PMC10642424 DOI: 10.2147/pgpm.s425251] [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: 07/09/2023] [Accepted: 10/16/2023] [Indexed: 11/16/2023] Open
Abstract
Objective This study aimed to discern the association between PLP2 expression, its biological significance, and the extent of immune infiltration in human GBM. Methods Utilizing the GEPIA2 and TCGA databases, we contrasted the expression levels of PLP2 in GBM against normal tissue. We utilized GEPIA2 and LinkedOmics for survival analysis, recognized genes co-expressed with PLP2 via cBioPortal and GEPIA2, and implemented GO and KEGG analyses. The STRING database facilitated the construction of protein-protein interaction networks. We evaluated the relationship of PLP2 with tumor immune infiltrates using ssGSEA and the TIMER 2.0 database. An IHC assay assessed PLP2 and PDL-1 expression in GBM tissue, and the Drugbank database aided in identifying potential PLP2-targeting compounds. Molecular docking was accomplished using Autodock Vina 1.2.2. Results PLP2 expression was markedly higher in GBM tissues in comparison to normal tissues. High PLP2 expression correlated with a decrease in overall survival across two databases. Functional analyses highlighted a focus of PLP2 functions within leukocyte. Discrepancies in PLP2 expression were evident in immune infiltration, impacting CD4+ T cells, neutrophils, myeloid dendritic cells, and macrophages. There was a concomitant increase in PLP2 and PD-L1 expression in GBM tissues, revealing a link between the two. Molecular docking with ethosuximide and praziquantel yielded scores of -7.441 and -4.295 kcal/mol, correspondingly. Conclusion PLP2's upregulation in GBM may adversely influence the lifespan of GBM patients. The involvement of PLP2 in pathways linked to leukocyte function is suggested. The positive correlation between PLP2 and PD-L1 could provide insights into PLP2's role in glioma modulation. Our research hints at PLP2's potential as a therapeutic target for GBM, with ethosuximide and praziquantel emerging as potential treatment candidates, especially emphasizing the potential of these compounds in GBM treatment targeting PLP2.
Collapse
Affiliation(s)
- Hao Qiao
- The Affiliated Hospital of Qingdao University, Qingdao, Shandong, People’s Republic of China
| | - Huanting Li
- The Affiliated Hospital of Qingdao University, Qingdao, Shandong, People’s Republic of China
| |
Collapse
|
3
|
Kharroubi DM, Nsouli G, Haroun Z. Potential Prognostic and Predictive Role of Monocyte and Lymphocyte Counts on Presentation in Patients Diagnosed With Diffuse Large B-Cell Lymphoma. Cureus 2023; 15:e35654. [PMID: 37009375 PMCID: PMC10065310 DOI: 10.7759/cureus.35654] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/01/2023] [Indexed: 03/05/2023] Open
Abstract
INTRODUCTION Diffuse large B-cell lymphoma (DLBCL) is the most common subtype of non-Hodgkin's lymphoma. Nearly 40% of patients will die of relapsed disease despite the use of rituximab plus cyclophosphamide, doxorubicin, vincristine, and prednisone (R-CHOP) chemotherapy. Many prognostic markers established in the chemotherapy era are no longer valid in the rituximab era. OBJECTIVES We aim to identify whether we can add absolute lymphocyte count (ALC), absolute monocyte count (AMC), and lymphocyte-to-monocyte ratio (LMR) as new prognostic factors for DLBCL treated with R-CHOP. We also aim to find whether a correlation exists between these variables and the revised International Prognostic Index (R-IPI) score. METHODS This is an observational retrospective study done from 2005 to 2015 in Rafic Hariri University Hospital (RHUH), Lebanon, on 42 patients treated with R-CHOP. Patients' data were obtained from medical records. We used receiver operating characteristic (ROC) curve for establishing cutoff values. The chi-square test was used to analyze associations between variables. RESULTS Patients were followed for a median of 42 months (24-96 months). Patients with LMR < 2.53 had a significantly worse outcome than those with LMR ≥ 2.53 (p < 0.0001). This was also true for patients with ALC < 1.47 × 109/L (p = 0.0163) and AMC > 0.603 × 109/L (p = 0.0053). LMR was also able to risk-stratify patients within each R-IPI category into high- and low-risk patients. CONCLUSION ALC, AMC, and LMR, surrogate markers of the host immune system and tumor microenvironment, have prognostic significance in DLBCL patients treated with R-CHOP.
Collapse
|
4
|
Mandal D, Kushwaha K, Gupta J. Emerging nano-strategies against tumour microenvironment (TME): a review. OPENNANO 2023. [DOI: 10.1016/j.onano.2022.100112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
5
|
Yue Y, Cao Y, Wang F, Zhang N, Qi Z, Mao X, Guo S, Li F, Guo Y, Lin Y, Dong W, Huang Y, Gu W. Bortezomib-resistant multiple myeloma patient-derived xenograft is sensitive to anti-CD47 therapy. Leuk Res 2022; 122:106949. [PMID: 36113267 DOI: 10.1016/j.leukres.2022.106949] [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: 05/04/2022] [Revised: 08/28/2022] [Accepted: 09/02/2022] [Indexed: 12/17/2022]
Abstract
Multiple myeloma (MM) remains an incurable hematologic malignancy due to its frequent drug resistance and relapse. Cluster of Differentiation 47 (CD47) is reported to be highly expressed on MM cells, suggesting that the blockade of CD47 signaling pathway could be a potential therapeutic candidate for MM. In this study, we developed a bortezomib-resistant myeloma patient-derived xenograft (PDX) from an extramedullary pleural effusion myeloma patient sample. Notably, anti-CD47 antibody treatments significantly inhibited tumor growth not only in MM cell line-derived models, including MM.1S and NCI-H929, but also in the bortezomib-resistant MM PDX model. Flow cytometric data showed that anti-CD47 therapy promoted the polarization of tumor-associated macrophages from an M2- to an M1-like phenotype. In addition, anti-CD47 therapy decreased the expression of pro-angiogenic factors, increased the expression of anti-angiogenic factors, and improved tumor vascular function, suggesting that anti-CD47 therapy induces tumor vascular normalization. Taken together, these data show that anti-CD47 antibody therapy reconditions the tumor immune microenvironment and inhibits the tumor growth of bortezomib-resistant myeloma PDX. Our findings suggest that CD47 is a potential new target to treat bortezomib-resistant MM.
Collapse
Affiliation(s)
- Yanhua Yue
- Department of Hematology, The First People's Hospital of Changzhou, Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu Province, PR China; Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou, Jiangsu Province, PR China
| | - Yang Cao
- Department of Hematology, The First People's Hospital of Changzhou, Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu Province, PR China
| | - Fei Wang
- Department of Hematology, The First People's Hospital of Changzhou, Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu Province, PR China
| | - Naidong Zhang
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou, Jiangsu Province, PR China
| | - Ziwei Qi
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou, Jiangsu Province, PR China
| | - Xunyuan Mao
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou, Jiangsu Province, PR China
| | - Shuxin Guo
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou, Jiangsu Province, PR China
| | - Feng Li
- Department of Hematology, The First People's Hospital of Changzhou, Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu Province, PR China
| | - Yanting Guo
- Department of Hematology, The First People's Hospital of Changzhou, Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu Province, PR China
| | - Yan Lin
- Department of Hematology, The First People's Hospital of Changzhou, Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu Province, PR China
| | - Weimin Dong
- Department of Hematology, The First People's Hospital of Changzhou, Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu Province, PR China
| | - Yuhui Huang
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou, Jiangsu Province, PR China.
| | - Weiying Gu
- Department of Hematology, The First People's Hospital of Changzhou, Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu Province, PR China.
| |
Collapse
|
6
|
Аnalysis of the cytokine profile of blood serum and tumor supernatants in breast cancer. ACTA BIOMEDICA SCIENTIFICA 2022. [DOI: 10.29413/abs.2022-7.2.15] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Background. A crucial role in the development of cancer is played by the tumor microenvironment (TM) – a microenvironment that is formed as a result of the interaction between the tumor tissue and macroorganism cells. The concentration of TM cytokines in the blood varies depending on the activity of the tumor and the presence of a metastatic process. It is advisable to study the existing mediator imbalance of TM, its characteristic features in the process of tumor development for the diagnosis and prognosis of the tumor process.The aim. To identify markers of tumor progression in the study of tissue and serum cytokines in women diagnosed with breast cancer.Materials and methods. The object of the study is blood serum cytokines and tumor supernatants (MCP-1, VEGF, TNF-α, IFN-γ, TGF-β1, G-CSF, GM-CSF). The study involved 80 patients with breast cancer aged 50–69 years and 26 practically healthy women aged 41 to 62 years. A standard examination of women was conducted; a cytokine profile study was conducted before the appointment of therapy. To study the cytokine profile at the tissue level, tumor biopsies (n = 30) and biopsies of unchanged breast tissue (n = 6) were incubated to determine the production of MCP-1, VEGF, TNF-α, IFN-γ, TGF-β1, G-CSF, GM-CSF.Results. There was a moderate positive correlation between the stage of the disease and the level of TGF-β1, MCP-1 blood serum, a weak one – with G-CSF. In the incubated tumor tissue, a high positive correlation of cytokines on the stage of the disease is observed in growth factors: VEGF (R = 0.79; p > 0.05) and TGF-β1 (R = 0.61; p > 0.05).Conclusion. The study revealed the characteristic features of the cytokine profile of blood serum and tumor tissue in breast cancer at local and widespread stages. The revealed differences in the level of cytokines should be used as additional diagnostic indicators of the degree of activity and prevalence of the tumor process.
Collapse
|
7
|
Souza JCD, Bastos VC, Pereira NB, Dias AAM, Avelar GFD, Gomez RS, Gomes CC. Angiogenesis in patient-derived xenografts of odontogenic myxoma. Int J Exp Pathol 2022; 103:65-69. [PMID: 35225401 PMCID: PMC8961500 DOI: 10.1111/iep.12431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 01/26/2022] [Accepted: 02/05/2022] [Indexed: 12/01/2022] Open
Abstract
Previously, by employing 3D organotypic tissue culture and patient-derived xenograft (PDX) model, oral myxoma response to a MAPK/MEK inhibitor was observed. Gross examination of the tumour fragments obtained after 55 days of PDX grafting revealed increased capsule vascularization. Microscopic analyses showed blood capillaries intermixed with myxoma cells, but the origin of these capillaries, from mice or humans, was not established. This study aimed to investigate whether the endothelial cells observed in the myxoma PDX model are derived from the mouse or from the primary human tumour. Immunohistochemistry was performed on five tumour fragments from the PDX of myxoma after 55 days of implantation in mice. Immunopositivity for antibodies against human (HLA-ABC) and mouse (H2 Db/H2-D1) major histocompatibility complex class I (MHCI) was assessed in the endothelial cells. The endothelial cells in the PDX fragments revealed a membrane staining for the human MHCI protein in the PDX tumour and adjacent connective tissue capsule, indicating that capillaries were derived from the human tumour fragment. Considering the probable human origin of the endothelial cells from capillary blood vessels in the myxoma PDX, we conclude that this PDX model is an interesting model to study myxoma angiogenesis.
Collapse
Affiliation(s)
- Juliana Cristina de Souza
- Department of Pathology, Biological Science Institute (ICB), Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Victor Coutinho Bastos
- Department of Pathology, Biological Science Institute (ICB), Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Núbia Braga Pereira
- Department of Pathology, Biological Science Institute (ICB), Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Adriana Abalen Martins Dias
- Department of General Biology, Biological Science Institute (ICB), Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Gleide Fernandes de Avelar
- Department of Morphology, Biological Science Institute (ICB), Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Ricardo Santiago Gomez
- Department of Oral Surgery and Pathology, School of Dentistry, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Carolina Cavaliéri Gomes
- Department of Pathology, Biological Science Institute (ICB), Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| |
Collapse
|
8
|
Kasherman L, Liu S(L, Karakasis K, Lheureux S. Angiogenesis: A Pivotal Therapeutic Target in the Drug Development of Gynecologic Cancers. Cancers (Basel) 2022; 14:1122. [PMID: 35267430 PMCID: PMC8908988 DOI: 10.3390/cancers14051122] [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: 01/07/2022] [Revised: 02/16/2022] [Accepted: 02/17/2022] [Indexed: 01/27/2023] Open
Abstract
Since the discovery of angiogenesis and its relevance to the tumorigenesis of gynecologic malignancies, a number of therapeutic agents have been developed over the last decade, some of which have become standard treatments in combination with other therapies. Limited clinical activity has been demonstrated with anti-angiogenic monotherapies, and ongoing trials are focused on combination strategies with cytotoxic agents, immunotherapies and other targeted treatments. This article reviews the science behind angiogenesis within the context of gynecologic cancers, the evidence supporting the targeting of these pathways and future directions in clinical trials.
Collapse
Affiliation(s)
- Lawrence Kasherman
- Department of Medical Oncology, St. George Hospital, Kogarah, NSW 2217, Australia;
- St. George and Sutherland Clinical Schools, University of New South Wales, Sydney, NSW 2052, Australia
- Illawarra Cancer Care Centre, Department of Medical Oncology, Wollongong, NSW 2500, Australia
| | | | | | - Stephanie Lheureux
- Princess Margaret Cancer Centre, Division of Medical Oncology and Hematology, University Health Network, Toronto, ON M5G 2M9, Canada
| |
Collapse
|
9
|
Structural Biology of the Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1350:91-100. [PMID: 34888845 DOI: 10.1007/978-3-030-83282-7_4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cancers can be described as "rogue organs" (Balkwill FR, Capasso M, Hagemann T, J Cell Sci 125:5591-5596, 2012) because they are composed of multiple cell types and tissues. The transformed cells can recruit and alter healthy cells from surrounding tissues for their own benefit. It is these interactions that create the tumor microenvironment (TME). The TME describes the cells, factors, and extracellular matrix proteins that make up the tumor and the area around it; the biology of the TME influences tumor progression. Changes in the TME can lead to the growth and development of the tumor, the death of the tumor, or tumor metastasis. Metastasis is the process by which cancer spreads from its initial site to a different part of the body. Metastasis occurs when cancer cells enter the circulatory system or lymphatic system after they break away from a tumor. Once the cells leave, they can travel to a different part of the body and form new tumors. Therefore, understanding the TME is critical to fully understand cancer and find a way to successfully combat it. Knowledge of the TME can better inform researchers of the ability of potential therapies to reach tumor cells. It can also give researchers potential targets to kill the tumor. Instead of directly killing the cancer cells, therapies can target an aspect of the TME which could then halt tumor development or lead to tumor death. In other cases, targeting another aspect of the TME could make it easier for another therapy to kill the cancer cells, for example, using nanoparticles with collagenases to target the collagen in the surrounding environment to expose the cancer cells to drugs (Zinger A, et al, ACS Nano 13(10):11008-11021, 2019).The TME can be split simply into cells and the structural matrix. Within these groups are fibroblasts, structural proteins, immune cells, lymphocytes, bone marrow-derived inflammatory cells, blood vessels, and signaling molecules (Spill F, et al, Curr Opin Biotechnol 40:41-48, 2016; Del Prete A, et al, Curr Opin Pharmacol 35:40-47, 2017; Arneth B, Medicina (Kaunas) 56(1), 2019). From structure to providing nutrients for growth, each of these components plays a critical role in tumor maintenance. Together these components impact cancer growth, development, and resistance to therapies (Hanahan D, Coussens LM, Cancer Cell 21:309-322, 2012). In this chapter, we will describe the TME and express the importance of the cellular and structural elements of the TME.
Collapse
|
10
|
The molecular feature of macrophages in tumor immune microenvironment of glioma patients. Comput Struct Biotechnol J 2021; 19:4603-4618. [PMID: 34471502 PMCID: PMC8383063 DOI: 10.1016/j.csbj.2021.08.019] [Citation(s) in RCA: 103] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 08/11/2021] [Accepted: 08/12/2021] [Indexed: 12/12/2022] Open
Abstract
Background Gliomas are one of the most common types of primary tumors in central nervous system. Previous studies have found that macrophages actively participate in tumor growth. Methods Weighted gene co-expression network analysis was used to identify meaningful macrophage-related gene genes for clustering. Pamr, SVM, and neural network were applied for validating clustering results. Somatic mutation and methylation were used for defining the features of identified clusters. Differentially expressed genes (DEGs) between the stratified groups after performing elastic regression and principal component analyses were used for the construction of MScores. The expression of macrophage-specific genes were evaluated in tumor microenvironment based on single cell sequencing analysis. A total of 2365 samples from 15 glioma datasets and 5842 pan-cancer samples were used for external validation of MScore. Results Macrophages were identified to be negatively associated with the survival of glioma patients. Twenty-six macrophage-specific DEGs obtained by elastic regression and PCA were highly expressed in macrophages at single-cell level. The prognostic value of MScores in glioma was validated by the active proinflammatory and metabolic profile of infiltrating microenvironment and response to immunotherapies of samples with this signature. MScores managed to stratify patient survival probabilities in 15 external glioma datasets and pan-cancer datasets, which predicted worse survival outcome. Sequencing data and immunohistochemistry of Xiangya glioma cohort confirmed the prognostic value of MScores. A prognostic model based on MScores demonstrated high accuracy rate. Conclusion Our findings strongly support a modulatory role of macrophages, especially M2 macrophages in glioma progression and warrants further experimental studies.
Collapse
Key Words
- ACC, Adrenocortical carcinoma
- BBB, brain blood barrier
- BLCA, Bladder Urothelial Carcinoma
- BRCA, Breast invasive carcinoma
- CDF, cumulative distribution function
- CESC, Cervical squamous cell carcinoma and endocervical adenocarcinoma
- CGGA, Chinese Glioma Genome Atlas
- CHOL, Cholangiocarcinoma
- CNA, copy number alternations
- CNV, copy number variation
- COAD, Colon adenocarcinoma
- CSF-1, colony-stimulating factor-1
- DLBC, Lymphoid Neoplasm Diffuse Large B-cell Lymphoma
- DMP, differentially methylated position
- ESCA, Esophageal carcinoma
- GBM, glioblastoma
- GEO, Gene Expression Omnibus
- GO, gene ontology
- GSEA, gene set enrichment analysis
- GSVA, gene set variation analysis
- Glioma microenvironment
- HNSC, Head and Neck squamous cell carcinoma
- IGR, intergenic region
- IHC, immunohistochemistry
- IL, interleukin
- Immunotherapy
- KEGG, Kyoto Encyclopaedia of Genes and Genomes
- KICH, Kidney Chromophobe
- KIRC, Kidney renal clear cell carcinoma
- KIRP, Kidney renal papillary cell carcinoma
- LGG, low grade glioma
- LIHC, Liver hepatocellular carcinoma
- LUAD, Lung adenocarcinoma
- LUSC, Lung squamous cell carcinoma
- MMP-2, matrix metalloproteinase-2
- MT1, MMP membrane type 1 matrix metalloprotease
- Machine learning
- Macrophage
- OV, Ovarian serous cystadenocarcinoma
- PAAD, Pancreatic adenocarcinoma
- PAM, partition around medoids
- PCA, principal component analysis
- PCPG, Pheochromocytoma and Paraganglioma
- PRAD, Prostate adenocarcinoma
- Prognostic model
- READ, Rectum adenocarcinoma
- SARC, Sarcoma
- SKCM, Skin Cutaneous Melanoma
- SNP, single-nucleotide polymorphism
- SNV, single-nucleotide variant
- STAD, Stomach adenocarcinoma
- SVM, Support Vector Machines
- TAM, tumor associated macrophage
- TCGA, The Cancer Genome Atlas
- TGF-β, tumor growth factor-β
- THCA, Thyroid carcinoma
- THYM, Thymoma
- TIMP-2, tissue inhibitor of metalloproteinase-2
- TLR2, toll-like receptor 2
- TME, tumor microenvironment
- TNFα, tumor necrosis factor α
- TSS, transcription start site
- UCEC, Uterine Corpus Endometrial Carcinoma
- UCS, Uterine Carcinosarcoma
- WGCNA, weighted gene co-expression network analysis
- pamr, prediction analysis for microarrays
Collapse
|
11
|
DNA Methylation-Based Estimates of Circulating Leukocyte Composition for Predicting Colorectal Cancer Survival: A Prospective Cohort Study. Cancers (Basel) 2021; 13:cancers13122948. [PMID: 34204621 PMCID: PMC8231262 DOI: 10.3390/cancers13122948] [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: 05/11/2021] [Revised: 06/06/2021] [Accepted: 06/09/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Inflammation is involved in the evolution of cancer. Leukocytes, of which the proportion can be estimated using epigenome-wide methylation data, may serve as a prognostic marker in colorectal cancer (CRC). Our aim was to investigate whether DNA methylation-based estimates of circulating leukocytes is associated with all-cause and disease-specific mortality in a prospective CRC patients’ cohort. Significant associations with CRC prognosis were observed for CD4+ T cells, CD8+ T cells, B cells, NK cells, and lymphocytes, independent of age, sex, tumor stage, tumor subsite, and therapy. CD4+ T cells outperformed other leukocytes and provided added predictive value in comparison to age, sex, and tumor stage. Although cell counting is commonly used in clinical practice, DNA methylation-estimated cell proportions could be a promising tool in understanding the role of leukocytes as CRC prognostic biomarkers when using stored blood samples. Abstract Leukocytes are involved in the progression of colorectal cancer (CRC). The proportion of six major leukocyte subtypes can be estimated using epigenome-wide DNA methylation (DNAm) data from stored blood samples. Whether the composition of circulating leukocytes can be used as a prognostic factor is unclear. DNAm-based leukocyte proportions were obtained from a prospective cohort of 2206 CRC patients. Multivariate Cox regression models and survival curves were applied to assess associations between leukocyte composition and survival outcomes. A higher proportion of lymphocytes, including CD4+ T cells, CD8+ T cells, B cells, and NK cells, was associated with better survival, while a higher proportion of neutrophils was associated with poorer survival. CD4+ T cells outperformed other leukocytes in estimating the patients’ prognosis. Comparing the highest quantile to the lowest quantile of CD4+ T cells, hazard ratios (95% confidence intervals) of all-cause and CRC-specific mortality were 0.59 (0.48, 0.72) and 0.59 (0.45, 0.77), respectively. Furthermore, the association of CD4+ T cells and prognosis was stronger among patients with early or intermediate CRC or patients with colon cancer. In conclusion, the composition of circulating leukocytes estimated from DNAm, particularly the proportions of CD4+ T cells, could be used as promising independent predictors of CRC survival.
Collapse
|
12
|
Zhang XL, Hu LP, Yang Q, Qin WT, Wang X, Xu CJ, Tian GA, Yang XM, Yao LL, Zhu L, Nie HZ, Li Q, Xu Q, Zhang ZG, Zhang YL, Li J, Wang YH, Jiang SH. CTHRC1 promotes liver metastasis by reshaping infiltrated macrophages through physical interactions with TGF-β receptors in colorectal cancer. Oncogene 2021; 40:3959-3973. [PMID: 33986509 DOI: 10.1038/s41388-021-01827-0] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 04/13/2021] [Accepted: 04/27/2021] [Indexed: 02/03/2023]
Abstract
Metastasis is a major cause of cancer-related deaths. Tumor-intrinsic properties can determine whether tumor metastasis occurs or not. Here, by comparing the gene expression patterns in primary colorectal cancer (CRC) patients with or without metastasis, we found that Collagen Triple Helix Repeat Containing 1 (CTHRC1) in primary CRC served as a metastasis-associated gene. Animal experiments verified that CTHRC1 secreted by CRC cells promoted hepatic metastasis, which was closely correlated with macrophage infiltration. Depletion of macrophages by liposomal clodronate largely abolished the promoting effect of CTHRC1 on CRC liver metastasis. Furthermore, we demonstrated that CTHRC1 modulated macrophage polarization to M2 phenotypes through TGF-β signaling. A mechanistic study revealed that CTHRC1 bound directly to TGF-β receptor II and TGF-β receptor III, stabilized the TGF-β receptor complex, and activated TGF-β signaling. The combination treatment of CTHRC1 monoclonal antibody and anti-PD-1 blocking antibody effectively suppressed CRC hepatic metastasis. Taken together, our data demonstrated that CTHRC1 is an intrinsic marker of CRC metastasis and further revealed that CTHRC1 promoted CRC liver metastasis by reshaping infiltrated macrophages through TGF-β signaling, suggesting that CTHRC1 could be a potential biomarker for the early prediction of and a therapeutic target of CRC hepatic metastasis.
Collapse
Affiliation(s)
- Xue-Li Zhang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Li-Peng Hu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Qin Yang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Wei-Ting Qin
- Department of Radiation Oncology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xu Wang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Chun-Jie Xu
- Department of Gastrointestinal Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Guang-Ang Tian
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiao-Mei Yang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Lin-Li Yao
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Lei Zhu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Hui-Zhen Nie
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Qing Li
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Qing Xu
- Department of Gastrointestinal Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Zhi-Gang Zhang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yan-Li Zhang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Jun Li
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Ya-Hui Wang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Shu-Heng Jiang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
13
|
Stepka P, Vsiansky V, Raudenska M, Gumulec J, Adam V, Masarik M. Metabolic and Amino Acid Alterations of the Tumor Microenvironment. Curr Med Chem 2021; 28:1270-1289. [PMID: 32031065 DOI: 10.2174/0929867327666200207114658] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 01/13/2020] [Accepted: 01/17/2020] [Indexed: 11/22/2022]
Abstract
Metabolic changes driven by the hostile tumor microenvironment surrounding cancer cells and the effect of these changes on tumorigenesis and metastatic potential have been known for a long time. The usual point of interest is glucose and changes in its utilization by cancer cells, mainly in the form of the Warburg effect. However, amino acids, both intra- and extracellular, also represent an important aspect of tumour microenvironment, which can have a significant effect on cancer cell metabolism and overall development of the tumor. Namely, alterations in the metabolism of amino acids glutamine, sarcosine, aspartate, methionine and cysteine have been previously connected to the tumor progression and aggressivity of cancer. The aim of this review is to pinpoint current gaps in our knowledge of the role of amino acids as a part of the tumor microenvironment and to show the effect of various amino acids on cancer cell metabolism and metastatic potential. This review shows limitations and exceptions from the traditionally accepted model of Warburg effect in some cancer tissues, with the emphasis on prostate cancer, because the traditional definition of Warburg effect as a metabolic switch to aerobic glycolysis does not always apply. Prostatic tissue both in a healthy and transformed state significantly differs in many metabolic aspects, including the metabolisms of glucose and amino acids, from the metabolism of other tissues. Findings from different tissues are, therefore, not always interchangeable and have to be taken into account during experimentation modifying the environment of tumor tissue by amino acid supplementation or depletion, which could potentially serve as a new therapeutic approach.
Collapse
Affiliation(s)
- Petr Stepka
- Department of Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, CZ-62500 Brno, Czech Republic
| | - Vit Vsiansky
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, CZ-62500 Brno, Czech Republic
| | - Martina Raudenska
- Department of Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, CZ-62500 Brno, Czech Republic
| | - Jaromir Gumulec
- Department of Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, CZ-62500 Brno, Czech Republic
| | - Vojtech Adam
- Central European Institute of Technology, Brno University of Technology, Technicka 3058/10, CZ-61600 Brno, Czech Republic
| | - Michal Masarik
- Central European Institute of Technology, Brno University of Technology, Technicka 3058/10, CZ-61600 Brno, Czech Republic
| |
Collapse
|
14
|
Liang ZW, Ge XX, Xu MD, Qin H, Wu MY, Shen M, Zhang Y, Liu XM, Chen K, Li W, Duan W, Qin S. Tumor-associated macrophages promote the metastasis and growth of non-small-cell lung cancer cells through NF-κB/PP2Ac-positive feedback loop. Cancer Sci 2021; 112:2140-2157. [PMID: 33609307 PMCID: PMC8177805 DOI: 10.1111/cas.14863] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 12/29/2020] [Accepted: 01/24/2021] [Indexed: 01/22/2023] Open
Abstract
Non‐small‐cell lung cancer (NSCLC), with its aggressive biological behavior, is one of the most diagnosed cancers. Tumor‐associated inflammatory cells play important roles in the interaction between chronic inflammation and lung cancer, however the mechanisms involved are far from defined. In the present study, by developing an orthotopic NSCLC mouse model based on chronic inflammation, we proved that an inflammatory microenvironment accelerated the growth of orthotopic xenografts in vivo. Tumor‐associated macrophages, the most abundant population of inflammatory cells, were identified. Treatment with macrophage‐conditioned medium (MCM) promoted the growth and migration of NSCLC cells. Using bioinformatics analysis, we identified downregulated PP2Ac expression in NSCLC cells upon treatment with MCM. We further confirmed that this downregulation was executed in an NF‐κB pathway‐dependent manner. As IκB kinase (IKK) has been proved to be a substrate of PP2Ac, inhibition on PP2Ac could result in amplification of NF‐κB pathway signaling. Overexpression of PP2Ac, or the dominant‐negative forms of IKK or IκB, attenuated the acceleration of growth and metastasis by MCM. Using bioinformatics analysis, we further identified that CXCL1 and COL6A1 could be downstream of NF‐κB/PP2Ac pathway. Luciferase assay and ChIP assay further confirmed the location of response elements on the promoter regions of CXCL1 and COL6A1. Elevated CXCL1 facilitated angiogenesis, whereas upregulated COL6A1 promoted proliferation and migration.
Collapse
Affiliation(s)
- Zhan-Wen Liang
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xin-Xin Ge
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Meng-Dan Xu
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Hualong Qin
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Meng-Yao Wu
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Meng Shen
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yan Zhang
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xiao-Meng Liu
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Kai Chen
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Wei Li
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Weiming Duan
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Songbing Qin
- Department of Radiation Oncology, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
15
|
Mehta AK, Kadel S, Townsend MG, Oliwa M, Guerriero JL. Macrophage Biology and Mechanisms of Immune Suppression in Breast Cancer. Front Immunol 2021; 12:643771. [PMID: 33968034 PMCID: PMC8102870 DOI: 10.3389/fimmu.2021.643771] [Citation(s) in RCA: 122] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 02/17/2021] [Indexed: 12/14/2022] Open
Abstract
Macrophages are crucial innate immune cells that maintain tissue homeostasis and defend against pathogens; however, their infiltration into tumors has been associated with adverse outcomes. Tumor-associated macrophages (TAMs) represent a significant component of the inflammatory infiltrate in breast tumors, and extensive infiltration of TAMs has been linked to poor prognosis in breast cancer. Here, we detail how TAMs impede a productive tumor immunity cycle by limiting antigen presentation and reducing activation of cytotoxic T lymphocytes (CTLs) while simultaneously supporting tumor cell survival, angiogenesis, and metastasis. There is an urgent need to overcome TAM-mediated immune suppression for durable anti-tumor immunity in breast cancer. To date, failure to fully characterize TAM biology and classify multiple subsets has hindered advancement in therapeutic targeting. In this regard, the complexity of TAMs has recently taken center stage owing to their subset diversity and tightly regulated molecular and metabolic phenotypes. In this review, we reveal major gaps in our knowledge of the functional and phenotypic characterization of TAM subsets associated with breast cancer, before and after treatment. Future work to characterize TAM subsets, location, and crosstalk with neighboring cells will be critical to counteract TAM pro-tumor functions and to identify novel TAM-modulating strategies and combinations that are likely to enhance current therapies and overcome chemo- and immuno-therapy resistance.
Collapse
Affiliation(s)
- Anita K Mehta
- Breast Tumor Immunology Laboratory, Dana-Farber Cancer Institute, Boston, MA, United States.,Division of Breast Surgery, Department of Surgery, Brigham and Women's Hospital, Boston, MA, United States
| | - Sapana Kadel
- Breast Tumor Immunology Laboratory, Dana-Farber Cancer Institute, Boston, MA, United States
| | - Madeline G Townsend
- Breast Tumor Immunology Laboratory, Dana-Farber Cancer Institute, Boston, MA, United States.,Division of Breast Surgery, Department of Surgery, Brigham and Women's Hospital, Boston, MA, United States
| | - Madisson Oliwa
- Breast Tumor Immunology Laboratory, Dana-Farber Cancer Institute, Boston, MA, United States.,Division of Breast Surgery, Department of Surgery, Brigham and Women's Hospital, Boston, MA, United States
| | - Jennifer L Guerriero
- Breast Tumor Immunology Laboratory, Dana-Farber Cancer Institute, Boston, MA, United States.,Division of Breast Surgery, Department of Surgery, Brigham and Women's Hospital, Boston, MA, United States
| |
Collapse
|
16
|
Derakhshani A, Rostami Z, Safarpour H, Shadbad MA, Nourbakhsh NS, Argentiero A, Taefehshokr S, Tabrizi NJ, Kooshkaki O, Astamal RV, Singh PK, Taefehshokr N, Alizadeh N, Silvestris N, Baradaran B. From Oncogenic Signaling Pathways to Single-Cell Sequencing of Immune Cells: Changing the Landscape of Cancer Immunotherapy. Molecules 2021; 26:2278. [PMID: 33920054 PMCID: PMC8071039 DOI: 10.3390/molecules26082278] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 04/09/2021] [Accepted: 04/09/2021] [Indexed: 12/19/2022] Open
Abstract
Over the past decade, there have been remarkable advances in understanding the signaling pathways involved in cancer development. It is well-established that cancer is caused by the dysregulation of cellular pathways involved in proliferation, cell cycle, apoptosis, cell metabolism, migration, cell polarity, and differentiation. Besides, growing evidence indicates that extracellular matrix signaling, cell surface proteoglycans, and angiogenesis can contribute to cancer development. Given the genetic instability and vast intra-tumoral heterogeneity revealed by the single-cell sequencing of tumoral cells, the current approaches cannot eliminate the mutating cancer cells. Besides, the polyclonal expansion of tumor-infiltrated lymphocytes in response to tumoral neoantigens cannot elicit anti-tumoral immune responses due to the immunosuppressive tumor microenvironment. Nevertheless, the data from the single-cell sequencing of immune cells can provide valuable insights regarding the expression of inhibitory immune checkpoints/related signaling factors in immune cells, which can be used to select immune checkpoint inhibitors and adjust their dosage. Indeed, the integration of the data obtained from the single-cell sequencing of immune cells with immune checkpoint inhibitors can increase the response rate of immune checkpoint inhibitors, decrease the immune-related adverse events, and facilitate tumoral cell elimination. This study aims to review key pathways involved in tumor development and shed light on single-cell sequencing. It also intends to address the shortcomings of immune checkpoint inhibitors, i.e., their varied response rates among cancer patients and increased risk of autoimmunity development, via applying the data from the single-cell sequencing of immune cells.
Collapse
Affiliation(s)
- Afshin Derakhshani
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 51656-65811, Iran; (A.D.); (M.A.S.); (S.T.); (N.J.T.); (R.V.A.); (N.A.)
- IRCCS Istituto Tumori “Giovanni Paolo II” of Bari, 70124 Bari, Italy;
| | - Zeinab Rostami
- Student Research Committee, Birjand University of Medical Sciences, Birjand 97178-53577, Iran; (Z.R.); (O.K.)
| | - Hossein Safarpour
- Cellular & Molecular Research Center, Birjand University of Medical Sciences, Birjand 97178-53577, Iran;
| | - Mahdi Abdoli Shadbad
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 51656-65811, Iran; (A.D.); (M.A.S.); (S.T.); (N.J.T.); (R.V.A.); (N.A.)
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz 51666-14766, Iran
| | | | | | - Sina Taefehshokr
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 51656-65811, Iran; (A.D.); (M.A.S.); (S.T.); (N.J.T.); (R.V.A.); (N.A.)
| | - Neda Jalili Tabrizi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 51656-65811, Iran; (A.D.); (M.A.S.); (S.T.); (N.J.T.); (R.V.A.); (N.A.)
| | - Omid Kooshkaki
- Student Research Committee, Birjand University of Medical Sciences, Birjand 97178-53577, Iran; (Z.R.); (O.K.)
| | - Reza Vaezi Astamal
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 51656-65811, Iran; (A.D.); (M.A.S.); (S.T.); (N.J.T.); (R.V.A.); (N.A.)
| | - Pankaj Kumar Singh
- Principal Research Technologist, Department of Radiation Oncology, Mayo Clinic, 4500 San Pablo Rd S, Jacksonville, FL 32224, USA;
| | - Nima Taefehshokr
- Department of Microbiology and Immunology, Center for Human Immunology, The University of Western Ontario, London, ON N6A 5C1, Canada;
| | - Nazila Alizadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 51656-65811, Iran; (A.D.); (M.A.S.); (S.T.); (N.J.T.); (R.V.A.); (N.A.)
| | - Nicola Silvestris
- IRCCS Istituto Tumori “Giovanni Paolo II” of Bari, 70124 Bari, Italy;
- Department of Biomedical Sciences and Human Oncology, University of Bari “Aldo Moro”, 70124 Bari, Italy
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 51656-65811, Iran; (A.D.); (M.A.S.); (S.T.); (N.J.T.); (R.V.A.); (N.A.)
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz 51666-14766, Iran
| |
Collapse
|
17
|
Abstract
The Hanahan and Weinberg "hallmarks of cancer" papers provide a useful structure for considering the various mechanisms driving cancer progression, and the same might be useful for wound healing. In this Review, we highlight how tissue repair and cancer share cellular and molecular processes that are regulated in a wound but misregulated in cancer. From sustained proliferative signaling and the activation of invasion and angiogenesis to the promoting role of inflammation, there are many obvious parallels through which one process can inform the other. For some hallmarks, the parallels are more obscure. We propose some new prospective hallmarks that might apply to both cancer and wound healing and discuss how wounding, as in biopsy and surgery, might positively or negatively influence cancer in the clinic.
Collapse
Affiliation(s)
- Lucy MacCarthy-Morrogh
- School of Biochemistry, Biomedical Sciences Building, University of Bristol, Bristol BS8 1TD, UK.
| | - Paul Martin
- School of Biochemistry, Biomedical Sciences Building, University of Bristol, Bristol BS8 1TD, UK.
- School of Physiology, Pharmacology and Neuroscience, Biomedical Sciences Building, University of Bristol, Bristol BS8 1TD, UK
- School of Medicine, Cardiff University, Cardiff CF14 4XN, UK
| |
Collapse
|
18
|
Zhao C, Pu Y, Zhang H, Hu X, Zhang R, He S, Zhao Q, Mu B. IL10-modified Human Mesenchymal Stem Cells inhibit Pancreatic Cancer growth through Angiogenesis Inhibition. J Cancer 2020; 11:5345-5352. [PMID: 32742480 PMCID: PMC7391191 DOI: 10.7150/jca.38062] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 06/15/2020] [Indexed: 12/26/2022] Open
Abstract
In the present study, we constructed the recombinant plasmid IL10-PEGFP-C1 and successfully transfected into human mesenchymal stem cells. After culturing for 72 h, the levels of IL6 and TNF-α in the supernatant of the MSCs-IL10 group were significantly lower than the vector group and the control group (17.6 ± 0.68vs73.8 ± 0.8 and 74.4 ± 1.5) µg/L and (65.05 ± 3.8 vs 203.2 ± 2.4 and 201.3 ± 3.7) µg/L, respectively (p < 0.001) .The animal experiments showed that the volume of subcutaneous tumors in the MSCs-IL10 group in vivo was a significantly less level compared to that in MSC control and the blank control groups (76.84 ± 20.11) mm3 vs (518. 344 ± 48.66) mm3, (576.99± 49.88) mm3, (P < 0. 05) and they have a longer life time. Further we found the mass concentrations of IL6 and TNF-α in the blood serum of MSC-IL10 group were lower than the vector group and the control group (64.42 ± 10.9 vs120.83 ± 15.52 and 122.65 ± 13.71) and (40.05 ± 5.63 vs 126.78 ±1.89 and 105.83 ± 2.16) µg/L respectively (p < 0.001). CD31 immunohistochemistry and alginate encapsulation experiments showed tumor angiogenesis were inhibited in MSCs-IL10 group in comparison to the control and vector group (P < 0.001), FITC-labeled dextran intake was also lower than the other groups (P < 0.01). Collectively, this study suggested IL10 could inhibit the growth of the transplanted tumor in vivo and prolong survival of mice, and the primary mechanism may be the indirect inhibition of pro-inflammatory cytokines IL6 and TNF-α secretion and tumor angiogenesis formation.
Collapse
Affiliation(s)
- Chunyan Zhao
- Sicuhan Key Laboratory of Medical Imaging, Affiliated Hospital of North Sichuan Medical University 637000, Nanchong, Sichuan Province, China
| | - Yu Pu
- Sicuhan Key Laboratory of Medical Imaging, Affiliated Hospital of North Sichuan Medical University 637000, Nanchong, Sichuan Province, China
| | - Haidi Zhang
- School of Preclinical Medicine, North Sichuan Medical University 637000, Nanchong, Sichuan Province, China
| | - Xianhua Hu
- School of Preclinical Medicine, North Sichuan Medical University 637000, Nanchong, Sichuan Province, China
| | - Rendan Zhang
- School of Preclinical Medicine, North Sichuan Medical University 637000, Nanchong, Sichuan Province, China
| | - Shuai He
- Department of Clinical Medicine, North Sichuan Medical University 637000, Nanchong, Sichuan Province, China
| | - Qi Zhao
- Department of Clinical Medicine, North Sichuan Medical University 637000, Nanchong, Sichuan Province, China
| | - Bo Mu
- Sicuhan Key Laboratory of Medical Imaging, Affiliated Hospital of North Sichuan Medical University 637000, Nanchong, Sichuan Province, China.,School of Preclinical Medicine, North Sichuan Medical University 637000, Nanchong, Sichuan Province, China
| |
Collapse
|
19
|
Saman H, Raza SS, Uddin S, Rasul K. Inducing Angiogenesis, a Key Step in Cancer Vascularization, and Treatment Approaches. Cancers (Basel) 2020; 12:cancers12051172. [PMID: 32384792 PMCID: PMC7281705 DOI: 10.3390/cancers12051172] [Citation(s) in RCA: 96] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Revised: 04/10/2020] [Accepted: 04/17/2020] [Indexed: 12/27/2022] Open
Abstract
Angiogenesis is a term that describes the formation of new blood and lymphatic vessels from a pre-existing vasculature. This allows tumour cells to acquire sustenance in the form of nutrients and oxygen and the ability to evacuate metabolic waste. As one of the hallmarks of cancer, angiogenesis has been studied extensively in animal and human models to enable better understanding of cancer biology and the development of new anti-cancer treatments. Angiogenesis plays a crucial role in the process of tumour genesis, because solid tumour need a blood supply if they are to grow beyond a few millimeters in size. On the other hand, there is growing evidence that some solid tumour exploit existing normal blood supply and do not require a new vessel formation to grow and to undergo metastasis. This review of the literature will present the current understanding of this intricate process and the latest advances in the use of angiogenesis-targeting therapies in the fight against cancer.
Collapse
Affiliation(s)
- Harman Saman
- Barts Cancer Institute, Queen Mary University of London, London E1 4NS, UK
- Department of Medicine, Hazm Maubrairek Hospital, Ar-Rayyan PO Box 305, Qatar
- Correspondence: or ; Tel.: +97-466506781
| | - Syed Shadab Raza
- Department of Stem Cell Biology and Regenerative Medicine, ERA University, Lucknow 226003, India;
| | - Shahab Uddin
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar;
| | - Kakil Rasul
- National Cancer Care and Research, Hamad Medical Corporation, Doha 3050, Qatar;
| |
Collapse
|
20
|
Oplawski M, Dziobek K, Zmarzły N, Grabarek B, Halski T, Januszyk P, Kuś-Kierach A, Adwent I, Dąbruś D, Kiełbasiński K, Boroń D. Expression Profile of VEGF-C, VEGF-D, and VEGFR-3 in Different Grades of Endometrial Cancer. Curr Pharm Biotechnol 2020; 20:1004-1010. [PMID: 31333122 DOI: 10.2174/1389201020666190718164431] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 03/22/2019] [Accepted: 07/08/2019] [Indexed: 12/15/2022]
Abstract
BACKGROUND Vascular endothelial growth factor (VEGF)-C, -D, and VEGF receptor-3 are proteins characterized as crucial for tumor lymphangiogenesis. It is accompanied by angiogenesis during wound healing, but also in the neoplastic process. The research studies have shown that the lymphatic system plays a key role in the progression of carcinogenesis. OBJECTIVE The aim of this study was to evaluate changes in the expression of VEGF-C, VEGF-D and VEGFR-3 in different grades of endometrial cancer (G1-G3). METHODS The study included 45 patients diagnosed with endometrial cancer (G1=17; G2=15; G3=13) and 15 patients without neoplastic changes. The expression of VEGF-C, VEGF-D, and VEGFR-3 was assessed using microarray technique and immunohistochemistry. Statistical analysis was performed using the one-way ANOVA and Tukey's post-hoc test. RESULTS Statistically significant changes in the expression at the transcriptome level were found only in the case of VEGF-C (G1 vs. C, fold change - FC = -1.15; G2 vs. C, FC = -2.33; G3 vs. C, FC = - 1.68). However, VEGF-D and VEGFR-3 were expressed at the protein level. Analysis of VEGF-D expression showed that the optical density of the reaction product in G1 reached 101.7, while the values in G2 and G3 were 142.7 and 184.4, respectively. For VEGF-R3, the optical density of the reaction product reached the following levels: 72 in control, 118.77 in G1, 145.8 in G2, and 170.9 in G3. CONCLUSION An increase in VEGF-D and VEGFR-3 levels may indicate that VEGF-D-dependent processes are intensified along with the dedifferentiation of tumor cells. The lack of VEGF-C expression in endometrial cancer samples may suggest that this tumor is characterized by a different mechanism of metastasis than EMT. Our study emphasizes that when analyzing the metastatic potential of cancer, the expression of more than one factor should be taken into account.
Collapse
Affiliation(s)
- Marcin Oplawski
- Department of Gynecology and Obstetrics with Gynecologic Oncology, Ludwik Rydygier Memorial Specialized Hospital, Cracow, Poland
| | - Konrad Dziobek
- Center of Oncology, M. Sklodowska-Curie Memorial Institute, Cracow Branch, Warsaw, Poland
| | - Nikola Zmarzły
- Katowice School of Technology, The University of Science and Art in Katowice, Katowice, Poland.,Department of Molecular Biology, School of Pharmacy with the Division of Laboratory Medicine in Sosnowiec, Medical University of Silesia in Katowice, Katowice, Poland
| | - Beniamin Grabarek
- Center of Oncology, M. Sklodowska-Curie Memorial Institute, Cracow Branch, Warsaw, Poland.,Katowice School of Technology, The University of Science and Art in Katowice, Katowice, Poland
| | - Tomasz Halski
- Faculty of Health Science, Public Higher Medical Professional School in Opole, Opole, Poland
| | - Piotr Januszyk
- Department of Gynecology and Obstetrics with Gynecologic Oncology, Ludwik Rydygier Memorial Specialized Hospital, Cracow, Poland
| | - Agnieszka Kuś-Kierach
- Faculty of Health Science, Public Higher Medical Professional School in Opole, Opole, Poland
| | - Iwona Adwent
- Faculty of Health Science, Public Higher Medical Professional School in Opole, Opole, Poland
| | - Dariusz Dąbruś
- Faculty of Health Science, Public Higher Medical Professional School in Opole, Opole, Poland
| | | | - Dariusz Boroń
- Department of Gynecology and Obstetrics with Gynecologic Oncology, Ludwik Rydygier Memorial Specialized Hospital, Cracow, Poland.,Katowice School of Technology, The University of Science and Art in Katowice, Katowice, Poland.,Faculty of Health Science, Public Higher Medical Professional School in Opole, Opole, Poland
| |
Collapse
|
21
|
Lopes-Coelho F, Silva F, Gouveia-Fernandes S, Martins C, Lopes N, Domingues G, Brito C, Almeida AM, Pereira SA, Serpa J. Monocytes as Endothelial Progenitor Cells (EPCs), Another Brick in the Wall to Disentangle Tumor Angiogenesis. Cells 2020; 9:cells9010107. [PMID: 31906296 PMCID: PMC7016533 DOI: 10.3390/cells9010107] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 12/22/2019] [Accepted: 12/30/2019] [Indexed: 12/12/2022] Open
Abstract
Bone marrow contains endothelial progenitor cells (EPCs) that, upon pro-angiogenic stimuli, migrate and differentiate into endothelial cells (ECs) and contribute to re-endothelialization and neo-vascularization. There are currently no reliable markers to characterize EPCs, leading to their inaccurate identification. In the past, we showed that, in a panel of tumors, some cells on the vessel wall co-expressed CD14 (monocytic marker) and CD31 (EC marker), indicating a putative differentiation route of monocytes into ECs. Herein, we disclosed monocytes as potential EPCs, using in vitro and in vivo models, and also addressed the cancer context. Monocytes acquired the capacity to express ECs markers and were able to be incorporated into blood vessels, contributing to cancer progression, by being incorporated in tumor neo-vasculature. Reactive oxygen species (ROS) push monocytes to EC differentiation, and this phenotype is reverted by cysteine (a scavenger and precursor of glutathione), which indicates that angiogenesis is controlled by the interplay between the oxidative stress and the scavenging capacity of the tumor microenvironment.
Collapse
Affiliation(s)
- Filipa Lopes-Coelho
- CEDOC, Chronic Diseases Research Centre, NOVA Medical School|Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria, 130, 1169-056 Lisboa, Portugal; (F.L.-C.); (F.S.); (S.G.-F.); (G.D.); (S.A.P.)
- Instituto Português de Oncologia de Lisboa Francisco Gentil (IPOLFG), Rua Prof. Lima Basto 1099-023 Lisboa, Portugal; (C.M.); (A.M.A.)
| | - Fernanda Silva
- CEDOC, Chronic Diseases Research Centre, NOVA Medical School|Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria, 130, 1169-056 Lisboa, Portugal; (F.L.-C.); (F.S.); (S.G.-F.); (G.D.); (S.A.P.)
- Instituto Português de Oncologia de Lisboa Francisco Gentil (IPOLFG), Rua Prof. Lima Basto 1099-023 Lisboa, Portugal; (C.M.); (A.M.A.)
| | - Sofia Gouveia-Fernandes
- CEDOC, Chronic Diseases Research Centre, NOVA Medical School|Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria, 130, 1169-056 Lisboa, Portugal; (F.L.-C.); (F.S.); (S.G.-F.); (G.D.); (S.A.P.)
- Instituto Português de Oncologia de Lisboa Francisco Gentil (IPOLFG), Rua Prof. Lima Basto 1099-023 Lisboa, Portugal; (C.M.); (A.M.A.)
| | - Carmo Martins
- Instituto Português de Oncologia de Lisboa Francisco Gentil (IPOLFG), Rua Prof. Lima Basto 1099-023 Lisboa, Portugal; (C.M.); (A.M.A.)
| | - Nuno Lopes
- Instituto de Biologia Experimental e Tecnológica, Avenida da República, Estação Agronómica, 2780-157 Oeiras, Portugal; (N.L.); (C.B.)
| | - Germana Domingues
- CEDOC, Chronic Diseases Research Centre, NOVA Medical School|Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria, 130, 1169-056 Lisboa, Portugal; (F.L.-C.); (F.S.); (S.G.-F.); (G.D.); (S.A.P.)
- Instituto Português de Oncologia de Lisboa Francisco Gentil (IPOLFG), Rua Prof. Lima Basto 1099-023 Lisboa, Portugal; (C.M.); (A.M.A.)
| | - Catarina Brito
- Instituto de Biologia Experimental e Tecnológica, Avenida da República, Estação Agronómica, 2780-157 Oeiras, Portugal; (N.L.); (C.B.)
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal
| | - António M Almeida
- Instituto Português de Oncologia de Lisboa Francisco Gentil (IPOLFG), Rua Prof. Lima Basto 1099-023 Lisboa, Portugal; (C.M.); (A.M.A.)
- Hospital da Luz, Av. Lusíada 100, 1500-650 Lisboa, Portugal
| | - Sofia A Pereira
- CEDOC, Chronic Diseases Research Centre, NOVA Medical School|Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria, 130, 1169-056 Lisboa, Portugal; (F.L.-C.); (F.S.); (S.G.-F.); (G.D.); (S.A.P.)
| | - Jacinta Serpa
- CEDOC, Chronic Diseases Research Centre, NOVA Medical School|Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria, 130, 1169-056 Lisboa, Portugal; (F.L.-C.); (F.S.); (S.G.-F.); (G.D.); (S.A.P.)
- Instituto Português de Oncologia de Lisboa Francisco Gentil (IPOLFG), Rua Prof. Lima Basto 1099-023 Lisboa, Portugal; (C.M.); (A.M.A.)
- Correspondence: ; Tel.: +350-217-229-800; Fax: +351-217-248-756
| |
Collapse
|
22
|
Dziobek K, Opławski M, Grabarek BO, Zmarzły N, Tomala B, Halski T, Leśniak E, Januszyk K, Brus R, Kiełbasiński R, Boroń D. Changes in the Expression Profile of VEGF-A, VEGF-B, VEGFR-1, VEGFR-2 in Different Grades of Endometrial Cancer. Curr Pharm Biotechnol 2019; 20:955-963. [PMID: 31322068 PMCID: PMC7403754 DOI: 10.2174/1389201020666190717092448] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 02/15/2019] [Accepted: 06/26/2019] [Indexed: 01/15/2023]
Abstract
Background VEGF-A, VEGF-B, VEGFR-1 and VEGFR-2 are important proteins involved in the induction and development of a new blood vessel network through which the tumor is properly nourished and oxygenated. Objectives The aim of the study was to evaluate changes in VEGF-A, VEGF-B, VEGFR-1 and VEGFR-2 expression in endometrial cancer depending on its grade and to determine the VEGFR-1 to VEGFR-2 concentration ratio. Methods The study group consisted of 45 patients diagnosed with endometrial cancer (G1, 17; G2, 15; G3, 13). The control group included 15 patients. VEGF-A, VEGF-B, VEGF-R1, VEGFR-2 expression was assessed using the immunohistochemical method. Statistical analysis was carried out using the Statistica 12 PL program (StatSoft, Cracow, Poland). It included the one-way ANOVA and Tukey's post-hoc test (p<0.05). Results Statistically significant differences in the level of VEGF-A, VEGF-B, VEGF-R1, VEGFR-2 were observed between the majority of analyzed groups (except for VEGF-B; G3 vs. G1, p=0.997700). The expression pattern of VEGF-A, VEGF-R1, VEGFR-2 was as follows: G3>G2>G1>C; VEGF-B: G2> G3> G1>C. A lower concentration of VEGFR-1 than VEGFR-2 was found regardless of the cancer grade. Conclusion VEGF-A, VEGF-B, VEGF-R1, VEGFR-2 are key proteins involved in tumor angiogenesis. The analysis of the entire panel of proteins participating in a given process is an important element of modern diagnostics. The concentration ratio of VEGFR-1 to VEGFR-2 appears to be a determining factor in the patients' survival prognosis.
Collapse
Affiliation(s)
- Konrad Dziobek
- Center of Oncology, M. Sklodowska-Curie Memorial Institute, Cracow Branch, Poland
| | - Marcin Opławski
- Department of Gynecology and Obstetrics with Gynecologic Oncology, Ludwik Rydygier Memorial Specialized Hospital, Krakow, Poland
| | - Beniamin Oskar Grabarek
- Department of Molecular Biology, School of Pharmacy with the Division of Laboratory Medicine in Sosnowiec, Medical University of Silesia in Katowice, Poland
| | - Nikola Zmarzły
- Department of Molecular Biology, School of Pharmacy with the Division of Laboratory Medicine in Sosnowiec, Medical University of Silesia in Katowice, Poland
| | - Barbara Tomala
- Faculty of Health Science, Public Higher Medical Professional School in Opole, Poland
| | - Tomasz Halski
- Faculty of Health Science, Public Higher Medical Professional School in Opole, Poland
| | - Ewa Leśniak
- Faculty of Health Science, Public Higher Medical Professional School in Opole, Poland
| | - Krzysztof Januszyk
- Faculty of Health Science, Public Higher Medical Professional School in Opole, Poland
| | - Ryszard Brus
- Department of Nurse, High School of Strategic Planning, Koscielna 6, 41-303, Dabrowa Gornicza, Poland
| | - Robert Kiełbasiński
- Department of Obstetrics & Gynaecology ward, Health Center in Mikolow, Mikolow, Poland
| | - Dariusz Boroń
- Center of Oncology, M. Sklodowska-Curie Memorial Institute, Cracow Branch, Poland.,Department of Gynecology and Obstetrics with Gynecologic Oncology, Ludwik Rydygier Memorial Specialized Hospital, Krakow, Poland.,Faculty of Health Science, Public Higher Medical Professional School in Opole, Poland.,Department of Histology and Cell Pathology, School of Medicine with the Division of Dentistry in Zabrze, Medical University of Silesia in Katowice, Katowice, Poland.,Katowice School of Technology, The University of Science and Art in Katowice, Katowice, Poland
| |
Collapse
|
23
|
Abstract
OBJECTIVE Identify the molecular mechanism of inflammatory stimuli induced pancreatic cancer progression. METHODS RNA-seq, microarray assay and bioinformatics analyses were used to identify differentially expressed genes. Immunohistochemical staining was performed to evaluate CD68, CD163, β-catenin, CD103, CCL3 markers. Quantitative real-time polymerase chain reaction (qRT-PCR), luciferase reporter assay, apoptosis assay, wound healing assay and immunofluorescence were performed to study the relationship of inflammatory stimuli and WNT/β-catenin pathway. RESULTS Differentially expressed genes of macrophage-conditioned medium-treated pancreatic cancer cells were related with WNT/β-catenin pathway. Inflammatory stimuli could activate WNT/β-catenin signaling pathway. In 106 pancreatic cancer patients, nuclear β-catenin expression of CD68-high group was much higher than CD68-low group (P < 0.05), as same as CD163 (P < 0.05). Inflammatory stimuli downregulated the expression of CCL3 via WNT/β-catenin pathway and inhibited the chemotaxis of CD103 dendritic cells. Six pancreatic cancer prognosis associating genes were upregulated by inflammatory stimuli via WNT/β-catenin pathway. Transforming growth factor-β promoted malignant biological behavior of pancreatic cancer cells through WNT/β-catenin pathway-dependent mechanism. CONCLUSIONS Our present study provided a novel mechanism involved in the inflammation-driven cancer progression through tumor immune escape and downstream gene regulation of WNT/β-catenin pathway-dependent manner.
Collapse
|
24
|
Azodyes as markers for tumor hypoxia imaging and therapy: An up-to-date review. Chem Biol Interact 2019; 307:91-104. [DOI: 10.1016/j.cbi.2019.04.034] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 04/21/2019] [Accepted: 04/28/2019] [Indexed: 12/20/2022]
|
25
|
Abstract
Research over the last decades has provided strong evidence for the pivotal role of the tumor-associated blood and lymphatic vasculature in supporting immunoevasion and in subverting T cell-mediated immunosurveillance. Conversely, tumor blood and lymphatic vessel growth is in part regulated by the immune system, with infiltrating innate as well as adaptive immune cells providing both immunosuppressive and various angiogenic signals. Thus, tumor angiogenesis and escape of immunosurveillance are two cancer hallmarks that are tightly linked and interregulated by cell constituents from compartments secreting both chemokines and cytokines. In this review, we discuss the implication and regulation of innate and adaptive immune cells in regulating blood and lymphatic angiogenesis in tumor progression and metastases. Moreover, we also highlight novel therapeutic approaches that target the tumor vasculature as well as the immune compartment to sustain and improve therapeutic efficacy in cancer.
Collapse
Affiliation(s)
- Massimiliano Mazzone
- VIB-Center for Cancer Biology and Department of Oncology, KU Leuven, Leuven B-3000 Belgium;
| | - Gabriele Bergers
- VIB-Center for Cancer Biology and Department of Oncology, KU Leuven, Leuven B-3000 Belgium;
- Department of Neurological Surgery, UCSF Comprehensive Cancer Center, San Francisco, California 94158, USA;
| |
Collapse
|
26
|
Cozzi S, Escarpa AS, Parra DL, Jamal DN, Mitjana JMC, R JMP, Edo FG, Miguelez CG. Association between inflammatory bowel disease and uveal melanoma: case report of two young adults and a literature review. Rep Pract Oncol Radiother 2018; 24:56-59. [PMID: 30455614 DOI: 10.1016/j.rpor.2018.10.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Accepted: 10/17/2018] [Indexed: 12/16/2022] Open
Abstract
Introduction Uveal melanoma is a rare tumour caused by genetic factors and alterations in the immune response. Inflammatory bowel disease (IBD) is a multifactorial chronic inflammatory disorder characterized by an inappropriate or excessive immune response. The two main types of IBD are Crohn's disease (CD) and ulcerative colitis (UC). A diagnosis of IBD and the use of immunosuppressive drugs are both independently associated with an increased risk of developing skin melanoma. The association between IBD and uveal melanoma (UM) has not been previously described. Cases description Two young Caucasian men, aged 24 and 28, developed UM 3 and 15 years, respectively, after being diagnosed with IBD. Both received long-term treatment with immunomodulatory drugs, with periodic switching among the drugs due to the refractory nature of IBD. In both cases, melanoma was treated by brachytherapy with iodine-125 COMS plaque implant at a dose of 75 Gy. Discussion Chronic inflammation can promote cell proliferation and growth. The use of immunomodulatory drugs is associated with an increased risk of developing melanoma and non-melanoma skin cancer. The two patients described in this report both had long-standing IBD treated with immunomodulatory drugs. It seems reasonable to suggest that these two factors may have promoted the development of uveal melanoma. More studies are warranted to investigate and confirm this possible association.
Collapse
Affiliation(s)
- Salvatore Cozzi
- Radiation Oncology Department, University Hospital "Maggiore della Caritá", Novara, Italy
| | - Andrea Slocker Escarpa
- Radiation Oncology Department, Catalan Institute of Oncology, University of Barcelona, L'Hospitalet de Llobregat, Barcelona, Spain
| | | | - Dina Najjari Jamal
- Radiation Oncology Department, Catalan Institute of Oncology, University of Barcelona, L'Hospitalet de Llobregat, Barcelona, Spain
| | | | | | - Ferran Guedea Edo
- Radiation Oncology Department, Catalan Institute of Oncology, University of Barcelona, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Cristina Gutierrez Miguelez
- Radiation Oncology Department, Catalan Institute of Oncology, University of Barcelona, L'Hospitalet de Llobregat, Barcelona, Spain
| |
Collapse
|
27
|
Wunderlich R, Rühle PF, Deloch L, Rödel F, Fietkau R, Gaipl US, Frey B. Ionizing radiation reduces the capacity of activated macrophages to induce T-cell proliferation, but does not trigger dendritic cell-mediated non-targeted effects. Int J Radiat Biol 2018; 95:33-43. [DOI: 10.1080/09553002.2018.1490037] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
Affiliation(s)
- Roland Wunderlich
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Research Unit Radiation Cytogenetics, Helmholtz Center Munich, Neuherberg, Germany
| | - Paul Friedrich Rühle
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Lisa Deloch
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Franz Rödel
- Department of Radiotherapy and Oncology, Goethe-University Frankfurt am Main, Frankfurt, Germany
| | - Rainer Fietkau
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Udo S. Gaipl
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Benjamin Frey
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
28
|
Role of mTOR Signaling in Tumor Microenvironment: An Overview. Int J Mol Sci 2018; 19:ijms19082453. [PMID: 30126252 PMCID: PMC6121402 DOI: 10.3390/ijms19082453] [Citation(s) in RCA: 103] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Revised: 08/06/2018] [Accepted: 08/15/2018] [Indexed: 12/31/2022] Open
Abstract
The mammalian target of rapamycin (mTOR) pathway regulates major processes by integrating a variety of exogenous cues, including diverse environmental inputs in the tumor microenvironment (TME). In recent years, it has been well recognized that cancer cells co-exist and co-evolve with their TME, which is often involved in drug resistance. The mTOR pathway modulates the interactions between the stroma and the tumor, thereby affecting both the tumor immunity and angiogenesis. The activation of mTOR signaling is associated with these pro-oncogenic cellular processes, making mTOR a promising target for new combination therapies. This review highlights the role of mTOR signaling in the characterization and the activity of the TME’s elements and their implications in cancer immunotherapy.
Collapse
|
29
|
Mathivet T, Bouleti C, Van Woensel M, Stanchi F, Verschuere T, Phng LK, Dejaegher J, Balcer M, Matsumoto K, Georgieva PB, Belmans J, Sciot R, Stockmann C, Mazzone M, De Vleeschouwer S, Gerhardt H. Dynamic stroma reorganization drives blood vessel dysmorphia during glioma growth. EMBO Mol Med 2018; 9:1629-1645. [PMID: 29038312 PMCID: PMC5709745 DOI: 10.15252/emmm.201607445] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Glioma growth and progression are characterized by abundant development of blood vessels that are highly aberrant and poorly functional, with detrimental consequences for drug delivery efficacy. The mechanisms driving this vessel dysmorphia during tumor progression are poorly understood. Using longitudinal intravital imaging in a mouse glioma model, we identify that dynamic sprouting and functional morphogenesis of a highly branched vessel network characterize the initial tumor growth, dramatically changing to vessel expansion, leakage, and loss of branching complexity in the later stages. This vascular phenotype transition was accompanied by recruitment of predominantly pro‐inflammatory M1‐like macrophages in the early stages, followed by in situ repolarization to M2‐like macrophages, which produced VEGF‐A and relocate to perivascular areas. A similar enrichment and perivascular accumulation of M2 versus M1 macrophages correlated with vessel dilation and malignancy in human glioma samples of different WHO malignancy grade. Targeting macrophages using anti‐CSF1 treatment restored normal blood vessel patterning and function. Combination treatment with chemotherapy showed survival benefit, suggesting that targeting macrophages as the key driver of blood vessel dysmorphia in glioma progression presents opportunities to improve efficacy of chemotherapeutic agents. We propose that vessel dysfunction is not simply a general feature of tumor vessel formation, but rather an emergent property resulting from a dynamic and functional reorganization of the tumor stroma and its angiogenic influences.
Collapse
Affiliation(s)
- Thomas Mathivet
- Vascular Patterning Lab, Center for Cancer Biology, VIB, Leuven, Belgium .,Vascular Patterning Lab, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Claire Bouleti
- Vascular Patterning Lab, Center for Cancer Biology, VIB, Leuven, Belgium.,Vascular Patterning Lab, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Matthias Van Woensel
- Department of Neurosciences, Laboratory of Experimental Neurosurgery and Neuroanatomy, KU Leuven, Leuven, Belgium
| | - Fabio Stanchi
- Vascular Patterning Lab, Center for Cancer Biology, VIB, Leuven, Belgium.,Vascular Patterning Lab, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Tina Verschuere
- Department of Neurosciences, Laboratory of Experimental Neurosurgery and Neuroanatomy, KU Leuven, Leuven, Belgium
| | - Li-Kun Phng
- Vascular Patterning Lab, Center for Cancer Biology, VIB, Leuven, Belgium.,Vascular Patterning Lab, Department of Oncology, KU Leuven, Leuven, Belgium.,Laboratory for Vascular Morphogenesis, RIKEN Center for Developmental Biology, Kobe, Japan
| | - Joost Dejaegher
- Department of Neurosciences, Laboratory of Experimental Neurosurgery and Neuroanatomy, KU Leuven, Leuven, Belgium
| | - Marly Balcer
- Vascular Patterning Lab, Center for Cancer Biology, VIB, Leuven, Belgium.,Vascular Patterning Lab, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Ken Matsumoto
- Vascular Patterning Lab, Center for Cancer Biology, VIB, Leuven, Belgium.,Vascular Patterning Lab, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Petya B Georgieva
- Vascular Patterning Lab, Center for Cancer Biology, VIB, Leuven, Belgium.,Vascular Patterning Lab, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Jochen Belmans
- Department of Neurosciences, Laboratory of Experimental Neurosurgery and Neuroanatomy, KU Leuven, Leuven, Belgium
| | - Raf Sciot
- Department of Pathology, KU Leuven and University Hospitals Leuven, Leuven, Belgium
| | - Christian Stockmann
- UMR 970, Paris Cardiovascular Research Center, Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, France
| | - Massimiliano Mazzone
- Lab of Molecular Oncology and Angiogenesis, Department of Oncology, KU Leuven, Leuven, Belgium.,Lab of Molecular Oncology and Angiogenesis, Center for Cancer Biology, VIB, Leuven, Belgium
| | - Steven De Vleeschouwer
- Department of Neurosciences, Laboratory of Experimental Neurosurgery and Neuroanatomy, KU Leuven, Leuven, Belgium.,Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Holger Gerhardt
- Vascular Patterning Lab, Center for Cancer Biology, VIB, Leuven, Belgium .,Vascular Patterning Lab, Department of Oncology, KU Leuven, Leuven, Belgium.,Integrative Vascular Biology Laboratory, Max-Delbrück-Center for Molecular Medicine, Helmholtz Association (MDC), Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany
| |
Collapse
|
30
|
Effect of scar-producing moxibustion at the acupoints Zusanli (ST 36) and Feishu (BL 13) on neutrophil-to-lymphocyte ratio and quality of life in patients with non-small-cell lung cancer: A randomized, controlled trial. J TRADIT CHIN MED 2018. [DOI: 10.1016/s0254-6272(18)30636-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
31
|
Xu H, Zeng Y, Liu L, Gao Q, Jin S, Lan Q, Lai W, Luo X, Wu H, Huang Y, Chu Z. PRL-3 improves colorectal cancer cell proliferation and invasion through IL-8 mediated glycolysis metabolism. Int J Oncol 2017; 51:1271-1279. [DOI: 10.3892/ijo.2017.4090] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Accepted: 07/19/2017] [Indexed: 11/06/2022] Open
|
32
|
Tao M, Liu L, Shen M, Zhi Q, Gong FR, Zhou BP, Wu Y, Liu H, Chen K, Shen B, Wu MY, Shou LM, Li W. Inflammatory stimuli promote growth and invasion of pancreatic cancer cells through NF-κB pathway dependent repression of PP2Ac. Cell Cycle 2016; 15:381-93. [PMID: 26761431 DOI: 10.1080/15384101.2015.1127468] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Previous studies have indicated that inflammatory stimulation represses protein phosphatase 2A (PP2A), a well-known tumor suppressor. However, whether PP2A repression participates in pancreatic cancer progression has not been verified. We used lipopolysaccharide (LPS) and macrophage-conditioned medium (MCM) to establish in vitro inflammation models, and investigated whether inflammatory stimuli affect pancreatic cancer cell growth and invasion PP2A catalytic subunit (PP2Ac)-dependently. Via nude mouse models of orthotopic tumor xenografts and dibutyltin dichloride (DBTC)-induced chronic pancreatitis, we evaluated the effect of an inflammatory microenvironment on PP2Ac expression in vivo. We cloned the PP2Acα and PP2Acβ isoform promoters to investigate the PP2Ac transcriptional regulation mechanisms. MCM accelerated pancreatic cancer cell growth; MCM and LPS promoted cell invasion. DBTC promoted xenograft growth and metastasis, induced tumor-associated macrophage infiltration, promoted angiogenesis, activated the nuclear factor-κB (NF-κB) pathway, and repressed PP2Ac expression. In vitro, LPS and MCM downregulated PP2Ac mRNA and protein. PP2Acα overexpression attenuated JNK, ERK, PKC, and IKK phosphorylation, and impaired LPS/MCM-stimulated cell invasion and MCM-promoted cell growth. LPS and MCM activated the NF-κB pathway in vitro. LPS and MCM induced IKK and IκB phosphorylation, leading to p65/RelA nuclear translocation and transcriptional activation. Overexpression of the dominant negative forms of IKKα attenuated LPS and MCM downregulation of PP2Ac, suggesting inflammatory stimuli repress PP2Ac expression NF-κB pathway-dependently. Luciferase reporter gene assay verified that LPS and MCM downregulated PP2Ac transcription through an NF-κB-dependent pathway. Our study presents a new mechanism in inflammation-driven cancer progression through NF-κB pathway-dependent PP2Ac repression.
Collapse
Affiliation(s)
- Min Tao
- a Department of Oncology , the First Affiliated Hospital of Soochow University , Suzhou , China.,b PREMED Key Laboratory for Precision Medicine, Soochow University , Suzhou , China.,c Jiangsu Institute of Clinical Immunology , Suzhou , China.,d Institute of Medical Biotechnology, Soochow University , Suzhou , China
| | - Lu Liu
- a Department of Oncology , the First Affiliated Hospital of Soochow University , Suzhou , China
| | - Meng Shen
- e Department of General Surgery , the First Affiliated Hospital of Soochow University , Suzhou , China
| | - Qiaoming Zhi
- e Department of General Surgery , the First Affiliated Hospital of Soochow University , Suzhou , China
| | - Fei-Ran Gong
- f Department of Hematology , the First Affiliated Hospital of Soochow University , Suzhou , China
| | - Binhua P Zhou
- g Markey Cancer Center, University of Kentucky College of Medicine , Lexington , KY , USA.,h Departments of Molecular and Cellular Biochemistry , University of Kentucky College of Medicine , Lexington , KY , USA
| | - Yadi Wu
- g Markey Cancer Center, University of Kentucky College of Medicine , Lexington , KY , USA.,i Molecular and Biomedical Pharmacology, University of Kentucky College of Medicine , Lexington , KY , USA
| | - Haiyan Liu
- j Laboratory of Cellular and Molecular Tumor Immunology, Institute of Biology and Medical Sciences, Soochow University , Suzhou , Jiangsu Province , China
| | - Kai Chen
- a Department of Oncology , the First Affiliated Hospital of Soochow University , Suzhou , China
| | - Bairong Shen
- k Center for Systems Biology, Soochow University , Suzhou , China
| | - Meng-Yao Wu
- a Department of Oncology , the First Affiliated Hospital of Soochow University , Suzhou , China
| | - Liu-Mei Shou
- a Department of Oncology , the First Affiliated Hospital of Soochow University , Suzhou , China.,l Department of Oncology , the First Affiliated Hospital of Zhejiang Chinese Medicine University , Hangzhou , China
| | - Wei Li
- a Department of Oncology , the First Affiliated Hospital of Soochow University , Suzhou , China.,b PREMED Key Laboratory for Precision Medicine, Soochow University , Suzhou , China.,c Jiangsu Institute of Clinical Immunology , Suzhou , China.,k Center for Systems Biology, Soochow University , Suzhou , China
| |
Collapse
|
33
|
Vlachos C, Gaitanis G, Katsanos KH, Christodoulou DK, Tsianos E, Bassukas ID. Psoriasis and inflammatory bowel disease: links and risks. PSORIASIS-TARGETS AND THERAPY 2016; 6:73-92. [PMID: 29387596 PMCID: PMC5683131 DOI: 10.2147/ptt.s85194] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Psoriasis and the spectrum of inflammatory bowel diseases (IBD) are chronic, inflammatory, organotropic conditions. The epidemiologic coexistence of these diseases is corroborated by findings at the level of disease, biogeography, and intrafamilial and intrapatient coincidence. The identification of shared susceptibility loci and DNA polymorphisms has confirmed this correlation at a genetic level. The pathogenesis of both diseases implicates the innate and adaptive segments of the immune system. Increased permeability of the epidermal barrier in skin and intestine underlies the augmented interaction of allergens and pathogens with inflammatory receptors of immune cells. The immune response between psoriasis and IBD is similar and comprises phagocytic, dendritic, and natural killer cell, along with a milieu of cytokines and antimicrobial peptides that stimulate T-cells. The interplay between dendritic cells and Th17 cells appears to be the core dysregulated immune pathway in all these conditions. The distinct similarities in the pathogenesis are also reflected in the wide overlapping of their therapeutic approaches. Small-molecule pharmacologic immunomodulators have been applied, and more recently, biologic treatments that target proinflammatory interleukins have been introduced or are currently being evaluated. However, the fact that some treatments are quite selective for either skin or gut conditions also highlights their crucial pathophysiologic differences. In the present review, a comprehensive comparison of risk factors, pathogenesis links, and therapeutic strategies for psoriasis and IBD is presented. Specific emphasis is placed on the role of the immune cell species and inflammatory mediators participating in the pathogenesis of these diseases.
Collapse
Affiliation(s)
| | | | - Konstantinos H Katsanos
- Division of Gastroenterology, Faculty of Medicine, School of Health Sciences, University of Ioannina, Ioannina, Greece
| | - Dimitrios K Christodoulou
- Division of Gastroenterology, Faculty of Medicine, School of Health Sciences, University of Ioannina, Ioannina, Greece
| | - Epameinondas Tsianos
- Division of Gastroenterology, Faculty of Medicine, School of Health Sciences, University of Ioannina, Ioannina, Greece
| | | |
Collapse
|
34
|
Banday MZ, Balkhi HM, Hamid Z, Sameer AS, Chowdri NA, Haq E. Tumor necrosis factor-α (TNF-α)-308G/A promoter polymorphism in colorectal cancer in ethnic Kashmiri population - A case control study in a detailed perspective. Meta Gene 2016; 9:128-36. [PMID: 27331018 PMCID: PMC4908285 DOI: 10.1016/j.mgene.2016.06.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Accepted: 06/01/2016] [Indexed: 12/31/2022] Open
Abstract
Background Inflammation constitutes one of the important components of colorectal cancer (CRC) pathogenesis. Tumor necrosis factor-α (TNF-α), a cytokine and an important inflammatory mediator plays a pivotal role in the malignant cellular proliferation, angiogenesis, tissue invasion and metastasis in CRC. The studies on association of various polymorphisms in human TNF-α gene including TNF-α-308G/A single nucleotide polymorphism (SNP) are limited, mixed and inconclusive. Materials and methods The aim of this study was to analyze the association of TNF-α-308G/A promoter SNP with colorectal cancer (CRC) susceptibility and development risk and also to evaluate the modifying effects of possible TNF-α-308G/A genotypes on different risk factors of CRC in ethnic population of Kashmir, India through a case–control setup. The genotype frequencies of TNF-α-308G/A promoter SNP were compared between 142 CRC patients and 184 individually matched healthy controls by using polymerase chain reaction and restriction fragment length polymorphism (PCR-RFLP) method. The associations between the TNF-α-308G/A SNP and CRC risk were examined through conditional logistic regression models adjusted for multiple possible confounding (third) variables. Further, the associations between this SNP and various clinico-pathological parameters, demographic variables and environmental factors within the case group subjects with regard to CRC risk were also evaluated. Results The association between the TNF-α-308G/A SNP and the modulation of risk of CRC was not found to be significant (p value = 0.156). The effect of less common TNF-α-308A allele on the risk of colorectal cancer was also not found to be significant (p value = 0.175). The variant genotype (AA) was nonexistent in the study population. Further, we found no significant effect modulation of CRC risk by wild and heterozygous TNF-α-308G/A SNP genotypes in presence of different possible risk factors (p > 0.05). We also found no significant association of TNF-α-308G/A SNP with the subsets of various characteristics of the case group subjects under study (p > 0.05). Conclusions This study indicates that there is no significant association between the TNF-α-308G/A promoter SNP and the risk of developing CRC in ethnic Kashmiri population. However, in order to substantiate our findings, this study needs to be replicated with bigger sample size and should involve other ethnically defined populations with high CRC risk.
Collapse
Affiliation(s)
- Mujeeb Zafar Banday
- Department of Biotechnology, University of Kashmir, Hazratbal, Srinagar, Kashmir, India
| | - Henah Mehraj Balkhi
- Department of Biotechnology, University of Kashmir, Hazratbal, Srinagar, Kashmir, India
| | - Zeenat Hamid
- Department of Biotechnology, University of Kashmir, Hazratbal, Srinagar, Kashmir, India
| | - Aga Syed Sameer
- Department of Basic Medical Sciences, College of Medicine, King Saud Bin Abdulaziz University for Health Sciences, Jeddah, Saudi Arabia
| | - Nissar A. Chowdri
- Department of Surgery, Sher-I-Kashmir Institute of Medical Sciences, Soura, Srinagar, Kashmir, India
| | - Ehtishamul Haq
- Department of Biotechnology, University of Kashmir, Hazratbal, Srinagar, Kashmir, India
- Corresponding author.
| |
Collapse
|
35
|
Garner EF, Beierle EA. Cancer Stem Cells and Their Interaction with the Tumor Microenvironment in Neuroblastoma. Cancers (Basel) 2015; 8:cancers8010005. [PMID: 26729169 PMCID: PMC4728452 DOI: 10.3390/cancers8010005] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2015] [Revised: 12/16/2015] [Accepted: 12/21/2015] [Indexed: 12/26/2022] Open
Abstract
Neuroblastoma, a solid tumor arising from neural crest cells, accounts for over 15% of all pediatric cancer deaths. The interaction of neuroblastoma cancer-initiating cells with their microenvironment likely plays an integral role in the maintenance of resistant disease and tumor relapse. In this review, we discuss the interaction between neuroblastoma cancer-initiating cells and the elements of the tumor microenvironment and how these interactions may provide novel therapeutic targets for this difficult to treat disease.
Collapse
Affiliation(s)
- Evan F Garner
- Department of Surgery, Division of Pediatric Surgery, University of Alabama, Birmingham, AL 35233, USA.
| | - Elizabeth A Beierle
- Department of Surgery, Division of Pediatric Surgery, University of Alabama, Birmingham, AL 35233, USA.
| |
Collapse
|
36
|
Singh M, Close DA, Mukundan S, Johnston PA, Sant S. Production of Uniform 3D Microtumors in Hydrogel Microwell Arrays for Measurement of Viability, Morphology, and Signaling Pathway Activation. Assay Drug Dev Technol 2015; 13:570-83. [PMID: 26274587 DOI: 10.1089/adt.2015.662] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Despite significant investments in cancer research and drug discovery/development, the rate of new cancer drug approval is ≤5% and most cases of metastatic cancer remain incurable. Ninety-five percent of new cancer drugs fail in clinical development because of a lack of therapeutic efficacy and/or unacceptable toxicity. One of the major factors responsible for the low success rate of anticancer drug development is the failure of preclinical models to adequately recapitulate the complexity and heterogeneity of human cancer. For throughput and capacity reasons, high-throughput screening growth inhibition assays almost exclusively use two-dimensional (2D) monolayers of tumor cell lines cultured on tissue culture-treated plastic/glass surfaces in serum-containing medium. However, these 2D tumor cell line cultures fail to recapitulate the three-dimensional (3D) context of cells in solid tumors even though the tumor microenvironment has been shown to have a profound effect on anticancer drug responses. Tumor spheroids remain the best characterized and most widely used 3D models; however, spheroid sizes tend to be nonuniform, making them unsuitable for high-throughput drug testing. To circumvent this challenge, we have developed defined size microwell arrays using nonadhesive hydrogels that are applicable to a wide variety of cancer cell lines to fabricate size-controlled 3D microtumors. We demonstrate that the hydrogel microwell array platform can be applied successfully to generate hundreds of uniform microtumors within 3-6 days from many cervical and breast, as well as head and neck squamous cell carcinoma (HNSCC) cells. Moreover, controlling size of the microwells in the hydrogel array allows precise control over the size of the microtumors. Finally, we demonstrate the application of this platform technology to probe activation as well as inhibition of epidermal growth factor receptor (EGFR) signaling in 3D HNSCC microtumors in response to EGF and cetuximab treatments, respectively. We believe that the ability to generate large numbers of HNSCC microtumors of uniform size and 3D morphology using hydrogel arrays will provide more physiological in vitro 3D tumor models to investigate how tumor size influences signaling pathway activation and cancer drug efficacy.
Collapse
Affiliation(s)
- Manjulata Singh
- 1 Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - David A Close
- 1 Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Shilpaa Mukundan
- 1 Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Paul A Johnston
- 1 Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh , Pittsburgh, Pennsylvania.,2 University of Pittsburgh Cancer Institute, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Shilpa Sant
- 1 Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh , Pittsburgh, Pennsylvania.,3 Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh , Pittsburgh, Pennsylvania.,4 McGowan Institute for Regenerative Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania
| |
Collapse
|
37
|
CD14-expressing cancer cells establish the inflammatory and proliferative tumor microenvironment in bladder cancer. Proc Natl Acad Sci U S A 2015; 112:4725-30. [PMID: 25825750 DOI: 10.1073/pnas.1424795112] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Nonresolving chronic inflammation at the neoplastic site is consistently associated with promoting tumor progression and poor patient outcomes. However, many aspects behind the mechanisms that establish this tumor-promoting inflammatory microenvironment remain undefined. Using bladder cancer (BC) as a model, we found that CD14-high cancer cells express higher levels of numerous inflammation mediators and form larger tumors compared with CD14-low cells. CD14 antigen is a glycosyl-phosphatidylinositol (GPI)-linked glycoprotein and has been shown to be critically important in the signaling pathways of Toll-like receptor (TLR). CD14 expression in this BC subpopulation of cancer cells is required for increased cytokine production and increased tumor growth. Furthermore, tumors formed by CD14-high cells are more highly vascularized with higher myeloid cell infiltration. Inflammatory factors produced by CD14-high BC cells recruit and polarize monocytes and macrophages to acquire immune-suppressive characteristics. In contrast, CD14-low BC cells have a higher baseline cell division rate than CD14-high cells. Importantly, CD14-high cells produce factors that further increase the proliferation of CD14-low cells. Collectively, we demonstrate that CD14-high BC cells may orchestrate tumor-promoting inflammation and drive tumor cell proliferation to promote tumor growth.
Collapse
|
38
|
Wunderlich R, Ernst A, Rödel F, Fietkau R, Ott O, Lauber K, Frey B, Gaipl US. Low and moderate doses of ionizing radiation up to 2 Gy modulate transmigration and chemotaxis of activated macrophages, provoke an anti-inflammatory cytokine milieu, but do not impact upon viability and phagocytic function. Clin Exp Immunol 2015; 179:50-61. [PMID: 24730395 PMCID: PMC4260896 DOI: 10.1111/cei.12344] [Citation(s) in RCA: 96] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/04/2014] [Indexed: 12/31/2022] Open
Abstract
Benign painful and inflammatory diseases have been treated for decades with low/moderate doses of ionizing radiation (LD-X-irradiation). Tissue macrophages regulate initiation and resolution of inflammation by the secretion of cytokines and by acting as professional phagocytes. Having these pivotal functions, we were interested in how activated macrophages are modulated by LD-X-irradiation, also with regard to radiation protection issues and carcinogenesis. We set up an ex-vivo model in which lipopolysaccharide pre-activated peritoneal macrophages (pMΦ) of radiosensitive BALB/c mice, mimicking activated macrophages under inflammatory conditions, were exposed to X-irradiation from 0·01 Gy up to 2 Gy. Afterwards, the viability of the pMΦ, their transmigration and chemotaxis, the phagocytic behaviour, the secretion of inflammatory cytokines and underlying signalling pathways were determined. Exposure of pMΦ up to a single dose of 2 Gy did not influence their viability and phagocytic function, an important fact regarding radiation protection. However, significantly reduced migration, but increased chemotaxis of pMΦ after exposure to 0·1 or 0·5 Gy, was detected. Both might relate to the resolution of inflammation. Cytokine analyses revealed that, in particular, the moderate dose of 0·5 Gy applied in low-dose radiotherapy for inflammatory diseases results in an anti-inflammatory cytokine microenvironment of pMΦ, as the secretion of the proinflammatory cytokine interleukin (IL)-1β was reduced and that of the anti-inflammatory cytokine transforming growth factor (TGF)-β increased. Further, the reduced secretion of IL-1β correlated with reduced nuclear translocation of nuclear factor (NF)-κB p65, starting at exposure of pMΦ to 0·5 Gy of X-irradiation. We conclude that inflammation is modulated by LD-X-irradiation via changing the inflammatory phenotype of macrophages.
Collapse
Affiliation(s)
- R Wunderlich
- Department of Radiation Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Ghaffari A, Hoskin V, Szeto A, Hum M, Liaghati N, Nakatsu K, LeBrun D, Madarnas Y, Sengupta S, Elliott BE. A novel role for ezrin in breast cancer angio/lymphangiogenesis. Breast Cancer Res 2014; 16:438. [PMID: 25231728 PMCID: PMC4303119 DOI: 10.1186/s13058-014-0438-2] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2013] [Accepted: 09/01/2014] [Indexed: 01/08/2023] Open
Abstract
INTRODUCTION Recent evidence suggests that tumour lymphangiogenesis promotes lymph node metastasis, a major prognostic factor for survival of breast cancer patients. However, signaling mechanisms involved in tumour-induced lymphangiogenesis remain poorly understood. The expression of ezrin, a membrane cytoskeletal crosslinker and Src substrate, correlates with poor outcome in a diversity of cancers including breast. Furthermore, ezrin is essential in experimental invasion and metastasis models of breast cancer. Ezrin acts cooperatively with Src in the regulation of the Src-induced malignant phenotype and metastasis. However, it remains unclear if ezrin plays a role in Src-induced tumour angio/lymphangiogenesis. METHODS The effects of ezrin knockdown and mutation on angio/lymphangiogenic potential of human MDA-MB-231 and mouse AC2M2 mammary carcinoma cell lines were examined in the presence of constitutively active or wild-type (WT) Src. In vitro assays using primary human lymphatic endothelial cells (hLEC), an ex vivo aortic ring assay, and in vivo tumour engraftment were utilized to assess angio/lymphangiogenic activity of cancer cells. RESULTS Ezrin-deficient cells expressing activated Src displayed significant reduction in endothelial cell branching in the aortic ring assay in addition to reduced hLEC migration, tube formation, and permeability compared to the controls. Intravital imaging and microvessel density (MVD) analysis of tumour xenografts revealed significant reductions in tumour-induced angio/lymphangiogenesis in ezrin-deficient cells when compared to the WT or activated Src-expressing cells. Moreover, syngeneic tumours derived from ezrin-deficient or Y477F ezrin-expressing (non-phosphorylatable by Src) AC2M2 cells further confirmed the xenograft results. Immunoblotting analysis provided a link between ezrin expression and a key angio/lymphangiogenesis signaling pathway by revealing that ezrin regulates Stat3 activation, VEGF-A/-C and IL-6 expression in breast cancer cell lines. Furthermore, high expression of ezrin in human breast tumours significantly correlated with elevated Src expression and the presence of lymphovascular invasion. CONCLUSIONS The results describe a novel function for ezrin in the regulation of tumour-induced angio/lymphangiogenesis promoted by Src in breast cancer. The combination of Src/ezrin might prove to be a beneficial prognostic/predictive biomarker for early-stage metastatic breast cancer.
Collapse
|
40
|
The role of cancer-associated fibroblasts, solid stress and other microenvironmental factors in tumor progression and therapy resistance. Cancer Cell Int 2014; 14:41. [PMID: 24883045 PMCID: PMC4038849 DOI: 10.1186/1475-2867-14-41] [Citation(s) in RCA: 141] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2013] [Accepted: 05/09/2014] [Indexed: 12/16/2022] Open
Abstract
Tumors are not merely masses of neoplastic cells but complex tissues composed of cellular and noncellular elements. This review provides recent data on the main components of a dynamic system, such as carcinoma associated fibroblasts that change the extracellular matrix (ECM) topology, induce stemness and promote metastasis-initiating cells. Altered production and characteristics of collagen, hyaluronan and other ECM proteins induce increased matrix stiffness. Stiffness along with tumor growth-induced solid stress and increased interstitial fluid pressure contribute to tumor progression and therapy resistance. Second, the role of immune cells, cytokines and chemokines is outlined. We discuss other noncellular characteristics of the tumor microenvironment such as hypoxia and extracellular pH in relation to neoangiogenesis. Overall, full understanding of the events driving the interactions between tumor cells and their environment is of crucial importance in overcoming treatment resistance and improving patient outcome.
Collapse
|
41
|
Zhang P, Dong L, Long H, Yang TT, Zhou Y, Fan QY, Ma BA. Homologous mesenchymal stem cells promote the emergence and growth of pulmonary metastases of the rat osteosarcoma cell line UMR-106. Oncol Lett 2014; 8:127-132. [PMID: 24959232 PMCID: PMC4063563 DOI: 10.3892/ol.2014.2127] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2012] [Accepted: 03/28/2014] [Indexed: 12/27/2022] Open
Abstract
Osteosarcoma (OS) is the most frequent primary bone sarcoma and tends to develop pulmonary metastasis. Studies have shown that mesenchymal stem cells (MSCs) are involved in OS growth and metastasis, but the mechanism remains unclear. The aim of the present study was to identify whether homologous MSCs could promote the growth and metastasis of OS in rats with a normal immune system. The OS cell line, UMR-106, which originally derives from a Sprague-Dawley (SD) rat-transplantable osteogenic sarcoma with an osteoblastic phenotype, has a strong carcinogenic capability and a high lung metastasis. Xenotransplanted models of UMR-106 with or without MSCs injected through the tibia (IT) or caudal vein (IV) were established. SD rats were randomly divided into six groups: Control, UMR-106 (IT), MSCs (IV), UMR-106 (IT) + MSCs (IV), UMR-106 (IV) and UMR-106 (IV) + MSCs (IV). Following injection, all rats were sacrificed at week 5, and the volume and quantity of metastatic sarcoma and the serum alkaline phosphatase levels were measured. There was no metastatic sarcoma in the liver, spleen and kidney in all groups. The rats in the MSCs (IV) + UMR-106 (IV) group showed a significantly higher volume and number of pulmonary metastatic tumors than those of the UMR-106 (IV) group. In pulmonary metastatic tissues, MSCs were found in the MSCs (IV) + UMR-106 (IV) group, but not in the UMR-106 (IT) + MSCs (IV) group. Notably, the expression of vascular endothelial growth factor (VEGF) was increased in the MSCs + UMR-106 cells co-culture system. The present study indicated that MSCs can significantly promote the pulmonary metastasis of the rat OS cell line, UMR-106, with a normal immune system, and VEGF was involved in MSC-promoted UMR-106 emergence and growth of pulmonary metastasis.
Collapse
Affiliation(s)
- Peng Zhang
- Department of Orthopedic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China ; Department of Orthopedic Surgery, Urumqi General Hospital, Urumqi, Xinjiang 830000, P.R. China
| | - Ling Dong
- Department of Physiology, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Hua Long
- Department of Orthopedic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Tong-Tao Yang
- Department of Orthopedic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Yong Zhou
- Department of Orthopedic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Qing-Yu Fan
- Department of Orthopedic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Bao-An Ma
- Department of Orthopedic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| |
Collapse
|
42
|
Dalton HJ, Armaiz-Pena GN, Gonzalez-Villasana V, Lopez-Berestein G, Bar-Eli M, Sood AK. Monocyte subpopulations in angiogenesis. Cancer Res 2014; 74:1287-93. [PMID: 24556724 DOI: 10.1158/0008-5472.can-13-2825] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Growing understanding of the role of the tumor microenvironment in angiogenesis has brought monocyte-derived cells into focus. Monocyte subpopulations are an increasingly attractive therapeutic target in many pathologic states, including cancer. Before monocyte-directed therapies can be fully harnessed for clinical use, understanding of monocyte-driven angiogenesis in tissue development and homeostasis, as well as malignancy, is required. Here, we provide an overview of the mechanisms by which monocytic subpopulations contribute to angiogenesis in tissue and tumor development, highlight gaps in our existing knowledge, and discuss opportunities to exploit these cells for clinical benefit.
Collapse
Affiliation(s)
- Heather J Dalton
- Authors' Affiliations: Departments of Gynecologic Oncology and Reproductive Medicine, Experimental Therapeutics, and Cancer Biology; and Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | | | | | | | | |
Collapse
|
43
|
Krstic J, Santibanez JF. Transforming growth factor-beta and matrix metalloproteinases: functional interactions in tumor stroma-infiltrating myeloid cells. ScientificWorldJournal 2014; 2014:521754. [PMID: 24578639 PMCID: PMC3918721 DOI: 10.1155/2014/521754] [Citation(s) in RCA: 133] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Accepted: 10/28/2013] [Indexed: 01/05/2023] Open
Abstract
Transforming growth factor-beta (TGF-β) is a pleiotropic factor with several different roles in health and disease. In tumorigenesis, it may act as a protumorigenic factor and have a profound impact on the regulation of the immune system response. Matrix metalloproteinases (MMPs) are a family that comprises more than 25 members, which have recently been proposed as important regulators acting in tumor stroma by regulating the response of noncellular and cellular microenvironment. Tumor stroma consists of several types of resident cells and infiltrating cells derived from bone marrow, which together play crucial roles in the promotion of tumor growth and metastasis. In cancer cells, TGF-β regulates MMPs expression, while MMPs, produced by either cancer cells or residents' stroma cells, activate latent TGF-β in the extracellular matrix, together facilitating the enhancement of tumor progression. In this review we will focus on the compartment of myeloid stroma cells, such as tumor-associated macrophages, neutrophils, and dendritic and mast cells, which are potently regulated by TGF-β and produce large amounts of MMPs. Their interplay and mutual implications in the generation of pro-tumorigenic cancer microenvironment will be analyzed.
Collapse
Affiliation(s)
- Jelena Krstic
- Laboratory for Experimental Hematology and Stem Cells, Institute for Medical Research, University of Belgrade, Dr Subotića 4, 11129 Belgrade, Serbia
| | - Juan F. Santibanez
- Laboratory for Experimental Hematology and Stem Cells, Institute for Medical Research, University of Belgrade, Dr Subotića 4, 11129 Belgrade, Serbia
| |
Collapse
|
44
|
Wlodkowic D, Cooper JM. Microfluidic cell arrays in tumor analysis: new prospects for integrated cytomics. Expert Rev Mol Diagn 2014; 10:521-30. [DOI: 10.1586/erm.10.28] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
45
|
Jiao SF, Sun K, Chen XJ, Zhao X, Cai N, Liu YJ, Xu LM, Kong XM, Wei LX. Inhibition of tumor necrosis factor alpha reduces the outgrowth of hepatic micrometastasis of colorectal tumors in a mouse model of liver ischemia-reperfusion injury. J Biomed Sci 2014; 21:1. [PMID: 24397824 PMCID: PMC3902418 DOI: 10.1186/1423-0127-21-1] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2013] [Accepted: 01/04/2014] [Indexed: 12/13/2022] Open
Abstract
Background Patients with colorectal cancer (CRC) often develop liver metastases, in which case surgery is considered the only potentially curative treatment option. However, liver surgery is associated with a risk of ischemia-reperfusion (IR) injury, which is thought to promote the growth of colorectal liver metastases. The influence of IR-induced tumor necrosis factor alpha (TNF-α) elevation in the process still is unknown. To investigate the role of TNF-α in the growth of pre-existing micrometastases in the liver following IR, we used a mouse model of colorectal liver metastases. In this model, mice received IR treatment seven days after intrasplenic injections of colorectal CT26 cells. Prior to IR treatment, either TNF-α blocker Enbrel or low-dose TNF-α, which could inhibit IR-induced TNF-α elevation, was administered by intraperitoneal injection. Results Hepatic IR treatment significantly promoted CT26 tumor growth in the liver, but either Enbrel or low-dose TNF-α pretreatment reversed this trend. Further studies showed that the CT26 + IR group prominently increased the levels of ALT and AST, liver necrosis, inflammatory infiltration and the expressions of hepatic IL-6, MMP9 and E-selectin compared to those of CT26 group. Inhibition of TNF-α elevation remarkably attenuated the increases of these liver inflammatory damage indicators and tumor-promoting factors. Conclusion These findings suggested that inhibition of TNF-α elevation delayed the IR-enhanced outgrowth of colorectal liver metastases by reducing IR-induced inflammatory damage and the formation of tumor-promoting microenvironments. Both Enbrel and low-dose TNF-α represented the potential therapeutic approaches for the protection of colorectal liver metastatic patients against IR injury-induced growth of liver micrometastases foci.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Xian-Ming Kong
- Medical Sciences Research Center, Renji hospital, School of Medicine, Shanghai Jiaotong University, 1630 Dongfang Road, Shanghai 200127, China.
| | | |
Collapse
|
46
|
Rivera L, Pandika M, Bergers G. Escape mechanisms from antiangiogenic therapy: an immune cell's perspective. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 772:83-99. [PMID: 24272355 DOI: 10.1007/978-1-4614-5915-6_4] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Neovascularization, the formation of new blood vessels, has become a well-established hallmark of cancer. Its functional importance for the manifestation and progression of tumors has been validated further by the beneficial therapeutic effects of angiogenesis inhibitors, most notably those targeting vascular endothelial growth factor signaling pathways. However, with the transient and short-lived nature of patient response, it has become evident that tumors have the ability to adapt to the pressures of vascular growth restriction. Observations made both in the clinic and at the bench suggest the existence of several escape mechanisms that either reestablish neovascularization in tumors or change tumor behavior to enable propagation and progression without obligate neovascularization. Some of these bypass mechanisms are regulated by low oxygen conditions (hypoxia) caused by therapy-induced vessel regression. Induction of hypoxia and hypoxia-inducible factors regulate a wide range of tumor-promoting pathways, including those of neovascularization, that can upregulate additional proangiogenic factors and drive the recruitment of various bone marrow-derived cells that have the capacity to express proangiogenic factors or directly contribute to neovasculature.
Collapse
Affiliation(s)
- Lee Rivera
- Departments of Neurological Surgery, University of California, Helen Diller Family Cancer Research Center, 1450 3rd Street, MC 0520, San Francisco, CA, 94158-9001, USA
| | | | | |
Collapse
|
47
|
Guo C, Buranych A, Sarkar D, Fisher PB, Wang XY. The role of tumor-associated macrophages in tumor vascularization. Vasc Cell 2013; 5:20. [PMID: 24314323 PMCID: PMC3913793 DOI: 10.1186/2045-824x-5-20] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Accepted: 11/25/2013] [Indexed: 12/15/2022] Open
Abstract
Tumor vascularization is a highly complex process that involves the interaction between tumors and their surrounding stroma, as well as many distinct angiogenesis-regulating factors. Tumor associated macrophages (TAMs) represent one of the most abundant cell components in the tumor environment and key contributors to cancer-related inflammation. A large body of evidence supports the notion that TAMs play a critical role in promoting the formation of an abnormal tumor vascular network and subsequent tumor progression and invasion. Clinical and experimental evidence has shown that high levels of infiltrating TAMs are associated with poor patient prognosis and tumor resistance to therapies. In addition to stimulating angiogenesis during tumor growth, TAMs enhance tumor revascularization in response to cytotoxic therapy (e.g., radiotherapy), thereby causing cancer relapse. In this review, we highlight the emerging data related to the phenotype and polarization of TAMs in the tumor microenvironment, as well as the underlying mechanisms of macrophage function in the regulation of the angiogenic switch and tumor vascularization. Additionally, we discuss the potential of targeting pro-angiogenic TAMs, or reprograming TAMs toward a tumoricidal and angiostatic phenotype, to promote normalization of the tumor vasculature to enhance the outcome of cancer therapies.
Collapse
Affiliation(s)
- Chunqing Guo
- Department of Human & Molecular Genetics, Virginia Commonwealth University School of Medicine, PO BOX 980033, Richmond VA23298, USA
| | - Annicole Buranych
- Department of Human & Molecular Genetics, Virginia Commonwealth University School of Medicine, PO BOX 980033, Richmond VA23298, USA
| | - Devanand Sarkar
- Department of Human & Molecular Genetics, Virginia Commonwealth University School of Medicine, PO BOX 980033, Richmond VA23298, USA
- VCU Institute of Molecular Medicine, Virginia Commonwealth University School of Medicine, Richmond VA23298, USA
- VCU Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond VA23298, USA
| | - Paul B Fisher
- Department of Human & Molecular Genetics, Virginia Commonwealth University School of Medicine, PO BOX 980033, Richmond VA23298, USA
- VCU Institute of Molecular Medicine, Virginia Commonwealth University School of Medicine, Richmond VA23298, USA
- VCU Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond VA23298, USA
| | - Xiang-Yang Wang
- Department of Human & Molecular Genetics, Virginia Commonwealth University School of Medicine, PO BOX 980033, Richmond VA23298, USA
- VCU Institute of Molecular Medicine, Virginia Commonwealth University School of Medicine, Richmond VA23298, USA
- VCU Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond VA23298, USA
| |
Collapse
|
48
|
Circulating fibrocytes stabilize blood vessels during angiogenesis in a paracrine manner. THE AMERICAN JOURNAL OF PATHOLOGY 2013; 184:556-71. [PMID: 24300950 DOI: 10.1016/j.ajpath.2013.10.021] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2013] [Revised: 10/29/2013] [Accepted: 10/31/2013] [Indexed: 12/12/2022]
Abstract
Accumulating evidence supports that circulating fibrocytes play important roles in angiogenesis. However, the specific role of fibrocytes in angiogenesis and the underlying mechanisms remain unclear. In this study, we found that fibrocytes stabilized newly formed blood vessels in a mouse wound-healing model by inhibiting angiogenesis during the proliferative phase and inhibiting blood vessel regression during the remodeling phase. Fibrocytes also inhibited angiogenesis in a Matrigel mouse model. In vitro study showed that fibrocytes inhibited both the apoptosis and proliferation of vascular endothelial cells (VECs) in a permeable support (Transwell) co-culture system. In a three-dimensional collagen gel, fibrocytes stabilized the VEC tubes by decreasing VEC tube density on stimulation with growth factors and preventing VEC tube regression on withdrawal of growth factors. Further mechanistic investigation revealed that fibrocytes expressed many prosurvival factors that are responsible for the prosurvival effect of fibrocytes on VECs and blood vessels. Fibrocytes also expressed angiogenesis inhibitors, including thrombospondin-1 (THBS1). THBS1 knockdown partially blocked the fibrocyte-induced inhibition of VEC proliferation in the Transwell co-culture system and recovered the fibrocyte-induced decrease of VEC tube density in collagen gel. Purified fibrocytes transfected with THBS1 siRNA partially recovered the fibrocyte-induced inhibition of angiogenesis in both the wound-healing and Matrigel models. In conclusion, our findings reveal that fibrocytes stabilize blood vessels via prosurvival factors and anti-angiogenic factors, including THBS1.
Collapse
|
49
|
Rigual NR, Shafirstein G, Frustino J, Seshadri M, Cooper M, Wilding G, Sullivan MA, Henderson B. Adjuvant intraoperative photodynamic therapy in head and neck cancer. JAMA Otolaryngol Head Neck Surg 2013; 139:706-11. [PMID: 23868427 DOI: 10.1001/jamaoto.2013.3387] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
IMPORTANCE There is an immediate need to develop local intraoperative adjuvant treatment strategies to improve outcomes in patients with cancer who undergo head and neck surgery. OBJECTIVES To determine the safety of photodynamic therapy with 2-(1-hexyloxyethyl)-2-devinyl pyropheophorbide-a (HPPH) in combination with surgery in patients with head and neck squamous cell carcinoma. DESIGN, SETTING, AND PARTICIPANTS Nonrandomized, single-arm, single-site, phase 1 study at a comprehensive cancer center among 16 adult patients (median age, 65 years) with biopsy-proved primary or recurrent resectable head and neck squamous cell carcinoma. INTERVENTIONS Intravenous injection of HPPH (4.0 mg/m2), followed by activation with 665-nm laser light in the surgical bed immediately after tumor resection. MAIN OUTCOMES AND MEASURES Adverse events and highest laser light dose. RESULTS Fifteen patients received the full course of treatment, and 1 patient received HPPH without intraoperative laser light because of an unrelated myocardial infarction. Disease sites included larynx (7 patients), oral cavity (6 patients), skin (1 patient), ear canal (1 patient), and oropharynx (1 patient, who received HPPH only). The most frequent adverse events related to photodynamic therapy were mild to moderate edema (9 patients) and pain (3 patients). One patient developed a grade 3 fistula after salvage laryngectomy, and another patient developed a grade 3 wound infection and mandibular fracture. Phototoxicity reactions included 1 moderate photophobia and 2 mild to moderate skin burns (2 due to operating room spotlights and 1 due to the pulse oximeter). The highest laser light dose was 75 J/cm2. CONCLUSIONS AND RELEVANCE The adjuvant use of HPPH-photodynamic therapy and surgery for head and neck squamous cell carcinoma seems safe and deserves further study. TRIAL REGISTRATION clinicaltrials.gov Identifier: NCT00470496.
Collapse
Affiliation(s)
- Nestor R Rigual
- Department of Head and Neck Surgery, Roswell Park Cancer Institute, Buffalo, New York, USA.
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Beyaert R, Beaugerie L, Van Assche G, Brochez L, Renauld JC, Viguier M, Cocquyt V, Jerusalem G, Machiels JP, Prenen H, Masson P, Louis E, De Keyser F. Cancer risk in immune-mediated inflammatory diseases (IMID). Mol Cancer 2013; 12:98. [PMID: 23987103 PMCID: PMC3765952 DOI: 10.1186/1476-4598-12-98] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2013] [Accepted: 08/22/2013] [Indexed: 02/07/2023] Open
Abstract
Inflammation and cancer have a profound yet ambiguous relationship. Inflammation - especially chronic inflammation - has protumorigenic effects, but inflammatory cells also mediate an immune response against the tumor and immunosuppression is known to increase the risk for certain tumors.This article reviews current literature on the role of inflammation in cancer and the cancer risk in immune-mediated inflammatory diseases (IMIDs). We discuss the effect on cancer risk of different drug classes used in the treatment of IMIDs treatment, including biologicals such as tumor necrosis factor (TNF) inhibitors.Overall cancer incidence and mortality risk are similar to the general population in inflammatory bowel disease (IBD), and slightly increased for rheumatoid arthritis and psoriasis, with risk profiles differing for different tumor types. Increased risk for non-melanoma skin cancer is associated with thiopurine treatment in IBD, with the combination of anti-TNF and methotrexate in rheumatoid arthritis and with PUVA, cyclosporine and anti-TNF treatment in psoriasis. Data on the safety of using biologic or immunosuppressant therapy in IMID patients with a history of cancer are scarce.This review provides clinicians with a solid background to help them in making decisions about treatment of immune-mediated diseases in patients with a tumor history.This article is related to another review article in Molecular Cancer: http://www.molecular-cancer.com/content/12/1/86.
Collapse
Affiliation(s)
- Rudi Beyaert
- Department of Rheumatology, Ghent University, 0K12, De Pintelaan 185, Ghent B-9000, Belgium.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|