1
|
Luo J, Liang M, Ma T, Dong B, Jia L, Su M. Identification of angiogenesis-related subtypes and risk models for predicting the prognosis of gastric cancer patients. Comput Biol Chem 2024; 112:108174. [PMID: 39191168 DOI: 10.1016/j.compbiolchem.2024.108174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 08/02/2024] [Accepted: 08/14/2024] [Indexed: 08/29/2024]
Abstract
Gastric cancer (GC) is a leading cause of cancer-related mortality and is characterized by significant heterogeneity, highlighting the need for further studies aimed at personalized treatment strategies. Tumor angiogenesis is critical for tumor development and metastasis, yet its role in molecular subtyping and prognosis prediction remains underexplored. This study aims to identify angiogenesis-related subtypes and develop a prognostic model for GC patients. Using data from The Cancer Genome Atlas (TCGA), we performed consensus cluster analysis on differentially expressed angiogenesis-related genes (ARGs), identifying two patient subtypes with distinct survival outcomes. Differentially expressed genes between the subtypes were analyzed via Cox and LASSO regression, leading to the establishment of a subtype-based prognostic model using a machine learning algorithm. Patients were classified into high- and low-risk groups based on the risk score. Validation was performed using independent datasets (ICGC and GSE15459). We utilized a deconvolution algorithm to investigate the tumor immune microenvironment in different risk groups and conducted analyses on genetic profiling, sensitivity and combination of anti-tumor drug. Our study identified ten prognostic signature genes, enabling the calculation of a risk score to predict prognosis and overall survival. This provides critical data for stratified diagnosis and treatment upon patient admission, monitoring disease progression throughout the entire course, evaluating immunotherapy efficacy, and selecting personalized medications for GC patients.
Collapse
Affiliation(s)
- Jie Luo
- Department of Medical Affairs, Huanggang Central Hospital, Huanggang, China
| | - Mengyun Liang
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Gannan Innovation and Translational Medicine Research Institute, Gannan Medical University, Ganzhou, China
| | - Tengfei Ma
- Clinical Trial Centers, Huanggang Central Hospital, Huanggang, China; Huanggang Institute of Translational Medicine, Huanggang, China
| | - Bizhen Dong
- Huanggang Institute of Translational Medicine, Huanggang, China
| | - Liping Jia
- Department of Respiratory and Critical Care Medicine, Huanggang Central Hospital, Huanggang, China.
| | - Meifang Su
- Department of Hematopathology, Huanggang Central Hospital, Huanggang, China.
| |
Collapse
|
2
|
Wachholz GE, Akbari P, Huijbers EJM, Jalan P, van Beijnum JR, Griffioen AW. Targeting endothelial cell anergy to improve CAR T cell therapy for solid tumors. Biochim Biophys Acta Rev Cancer 2024; 1879:189155. [PMID: 39019408 DOI: 10.1016/j.bbcan.2024.189155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 07/05/2024] [Accepted: 07/10/2024] [Indexed: 07/19/2024]
Abstract
Chimeric antigen receptor (CAR) T cell therapy presents significant results, especially for the treatment of hematologic malignancies. However, there are limitations and challenges to be overcome to achieve similar success for the treatment of solid tumors. These challenges involve selection of the target, infiltration into the tumor microenvironment and maintenance of functionality. The tumor vasculature is a major barrier for leukocytes to enter the tumor parenchyma. Due to the exposure of the vasculature to angiogenic growth factors during tumor progression, the endothelial cells become anergic to inflammatory cytokines, resulting in reduced leukocyte adhesion molecule expression. As such adhesion molecules are a prerequisite for leukocyte extravasation, endothelial cell anergy allows tumors to escape from endogenous immunity, as well as from cellular immunotherapies such as CAR T cells. Hence, overcoming endothelial cell anergy, e.g. through the administration of angiogenesis inhibitors, is believed to restore anti-tumor immunity. Concomitantly, both endogenous immune cells as well as cellular therapeutics such as CAR T cells can permeate into the tumor parenchyma. Here, we discuss how prior or concomitant treatment with an antiangiogenic drug can improve CAR T cell therapy, to become an attractive strategy for the treatment of solid tumors.
Collapse
Affiliation(s)
- Gabriela E Wachholz
- Angiogenesis Laboratory, Department of Medical Oncology, Amsterdam UMC, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Parvin Akbari
- Angiogenesis Laboratory, Department of Medical Oncology, Amsterdam UMC, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Elisabeth J M Huijbers
- Angiogenesis Laboratory, Department of Medical Oncology, Amsterdam UMC, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Prachi Jalan
- Angiogenesis Laboratory, Department of Medical Oncology, Amsterdam UMC, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Judy R van Beijnum
- Angiogenesis Laboratory, Department of Medical Oncology, Amsterdam UMC, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Arjan W Griffioen
- Angiogenesis Laboratory, Department of Medical Oncology, Amsterdam UMC, Cancer Center Amsterdam, Amsterdam, the Netherlands.
| |
Collapse
|
3
|
Chang H, Li M, Zhang L, Li M, Ong SH, Zhang Z, Zheng J, Xu X, Zhang Y, Wang J, Liu X, Li K, Luo Y, Wang H, Miao Z, Chen X, Zha J, Yu Y. Loss of histone deubiquitinase Bap1 triggers anti-tumor immunity. Cell Oncol (Dordr) 2024:10.1007/s13402-024-00978-y. [PMID: 39141316 DOI: 10.1007/s13402-024-00978-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/26/2024] [Indexed: 08/15/2024] Open
Abstract
PURPOSE Immunotherapy using PD-L1 blockade is effective in only a small group of cancer patients, and resistance is common. This emphasizes the importance of understanding the mechanisms of cancer immune evasion and resistance. METHODS A genome-scale CRISPR-Cas9 screen identified Bap1 as a regulator of PD-L1 expression. To measure tumor size and survival, tumor cells were subcutaneously injected into both syngeneic WT mice and immunocompromised mice. The phenotypic and transcriptional characteristics of Bap1-deleted tumors were examined using flow cytometry, RNA-seq, and CUT&Tag-seq analysis. RESULTS We found that loss of histone deubiquitinase Bap1 in cancer cells activates a cDC1-CD8+ T cell-dependent anti-tumor immunity. The absence of Bap1 leads to an increase in genes associated with anti-tumor immune response and a decrease in genes related to immune evasion. As a result, the tumor microenvironment becomes inflamed, with more cDC1 cells and effector CD8+ T cells, but fewer neutrophils and regulatory T cells. We also found that the elimination of Bap1-deleted tumors depends on the tumor MHCI molecule and Fas-mediated CD8+ T cell cytotoxicity. Our analysis of TCGA data further supports these findings, showing a reverse correlation between BAP1 expression and mRNA signatures of activated DCs and T-cell cytotoxicity in various human cancers. CONCLUSION The histone deubiquitinase Bap1 could be used as a biomarker for tumor stratification and as a potential therapeutic target for cancer immunotherapies.
Collapse
Affiliation(s)
- Hong Chang
- Department of Hematology, Tongji Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, 1239 Siping Road, Shanghai, 200092, China
| | - Mingxia Li
- Department of Hematology, Tongji Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, 1239 Siping Road, Shanghai, 200092, China
| | - Linlin Zhang
- Department of Hematology, Tongji Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, 1239 Siping Road, Shanghai, 200092, China
| | - Meng Li
- Wellcome Sanger Institute, Hinxton, Cambridge, CB10 1HH, UK
| | - Swee Hoe Ong
- Wellcome Sanger Institute, Hinxton, Cambridge, CB10 1HH, UK
| | - Zhiwei Zhang
- Department of Hematology, Tongji Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, 1239 Siping Road, Shanghai, 200092, China
| | - Jie Zheng
- Department of Hematology, Tongji Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, 1239 Siping Road, Shanghai, 200092, China
| | - Xiang Xu
- Department of Hematology, Tongji Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, 1239 Siping Road, Shanghai, 200092, China
| | - Yu Zhang
- Department of Hematology, Tongji Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, 1239 Siping Road, Shanghai, 200092, China
| | - Jing Wang
- Department of Hematology, Tongji Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, 1239 Siping Road, Shanghai, 200092, China
| | - Xingjie Liu
- Department of Hematology, Tongji Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, 1239 Siping Road, Shanghai, 200092, China
| | - Kairui Li
- Department of Hematology, Tongji Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, 1239 Siping Road, Shanghai, 200092, China
| | - Yao Luo
- Department of Hematology, Tongji Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, 1239 Siping Road, Shanghai, 200092, China
| | - Haiyun Wang
- Department of Hematology, Tongji Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, 1239 Siping Road, Shanghai, 200092, China
| | - Zhichao Miao
- Translational Research Institute of Brain and Brain-Like Intelligence, Department of Anesthesiology, Shanghai Fourth People's Hospital Affiliated to Tongji University School of Medicine, Shanghai, 200081, China
- GMU-GIBH Joint School of Life Sciences, The Guangdong-Hong Kong-Macau Joint Laboratory for Cell Fate Regulation and Diseases, Guangzhou Laboratory, Guangzhou Medical University, Guangzhou, China
| | - Xi Chen
- Department of Biology, Southern University of Science and Technology, 1088 Xueyuan Avenue, Shenzhen, 518055, China
| | - Jie Zha
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, China.
- Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, Xiamen, China.
| | - Yong Yu
- Department of Hematology, Tongji Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, 1239 Siping Road, Shanghai, 200092, China.
| |
Collapse
|
4
|
Brandenburg A, Heine A, Brossart P. Next-generation cancer vaccines and emerging immunotherapy combinations. Trends Cancer 2024; 10:749-769. [PMID: 39048489 DOI: 10.1016/j.trecan.2024.06.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 06/06/2024] [Accepted: 06/12/2024] [Indexed: 07/27/2024]
Abstract
Therapeutic cancer vaccines have been a subject of research for several decades as potential new weapons to tackle malignancies. Their goal is to induce a long-lasting and efficient antitumour-directed immune response, capable of mediating tumour regression, preventing tumour progression, and eradicating minimal residual disease, while avoiding major adverse effects. Development of new vaccine technologies and antigen prediction methods has led to significant improvements in cancer vaccine efficacy. However, for their successful clinical application, certain obstacles still need to be overcome, especially tumour-mediated immunosuppression and escape mechanisms. In this review, we introduce therapeutic cancer vaccines and subsequently discuss combination approaches of next-generation cancer vaccines and existing immunotherapies, particularly immune checkpoint inhibitors (ICIs) and adoptive cell transfer/cell-based immunotherapies.
Collapse
Affiliation(s)
- Anne Brandenburg
- Medical Clinic III of Oncology, Hematology, Rheumatology and Immune-Oncology, University Hospital Bonn, Venusberg Campus 1, 53127 Bonn, Germany
| | - Annkristin Heine
- Medical Clinic III of Oncology, Hematology, Rheumatology and Immune-Oncology, University Hospital Bonn, Venusberg Campus 1, 53127 Bonn, Germany
| | - Peter Brossart
- Medical Clinic III of Oncology, Hematology, Rheumatology and Immune-Oncology, University Hospital Bonn, Venusberg Campus 1, 53127 Bonn, Germany.
| |
Collapse
|
5
|
He B, Hu Y, Cao Q, Li Y, Tang Y, Cao T, Zhou X, Liu S. Progression of unfolded protein response and ferroptosis in angiogenesis. Biomed Pharmacother 2024; 173:116354. [PMID: 38442673 DOI: 10.1016/j.biopha.2024.116354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 02/23/2024] [Accepted: 02/26/2024] [Indexed: 03/07/2024] Open
Abstract
Angiogenesis is the growth of new blood vessels on preexisting ones. It is the outcome of a multifactorial effect involving several cells, which can be brought on by different stress reactions.The accumulation of unfolded proteins in the endoplasmic reticulum occurs when cells are stressed due to environmental changes, where physical or chemical stimuli induce endoplasmic reticulum stress, thereby activating the unfolded protein response (UPR), a homeostasis response designed to re-establish protein balance. Ferroptosis is a planned death of lipid peroxidation and anomalies in metabolism that is dependent on iron. Large concentrations of iron ions accumulate there, along with high concentrations of lipid peroxides and reactive oxygen species, all of which can contribute to the development of several diseases. Through the production of growth factors, adhesion factors, and inflammatory factors that trigger the start of angiogenesis, both UPR and Ferroptosis can be implicated in angiogenesis.To set the stage for further research on angiogenesis, this work concentrated on the effects of Ferroptosis and UPR on angiogenesis, respectively.
Collapse
Affiliation(s)
- Bisha He
- Department of Clinical Laboratory Medicine, Institution of microbiology and infectious diseases, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Yibao Hu
- Department of Clinical Laboratory Medicine, Institution of microbiology and infectious diseases, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Qian Cao
- Department of Clinical Laboratory Medicine, Institution of microbiology and infectious diseases, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Yue Li
- Department of Clinical Laboratory Medicine, Institution of microbiology and infectious diseases, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Yun Tang
- Department of Clinical Laboratory Medicine, Institution of microbiology and infectious diseases, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Ting Cao
- Department of Clinical Laboratory Medicine, Institution of microbiology and infectious diseases, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Xiangping Zhou
- Department of Clinical Laboratory Medicine, Institution of microbiology and infectious diseases, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Shuangquan Liu
- Department of Clinical Laboratory Medicine, Institution of microbiology and infectious diseases, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China.
| |
Collapse
|
6
|
Dudley AC, Griffioen AW. Pathological angiogenesis: mechanisms and therapeutic strategies. Angiogenesis 2023; 26:313-347. [PMID: 37060495 PMCID: PMC10105163 DOI: 10.1007/s10456-023-09876-7] [Citation(s) in RCA: 105] [Impact Index Per Article: 105.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 03/26/2023] [Indexed: 04/16/2023]
Abstract
In multicellular organisms, angiogenesis, the formation of new blood vessels from pre-existing ones, is an essential process for growth and development. Different mechanisms such as vasculogenesis, sprouting, intussusceptive, and coalescent angiogenesis, as well as vessel co-option, vasculogenic mimicry and lymphangiogenesis, underlie the formation of new vasculature. In many pathological conditions, such as cancer, atherosclerosis, arthritis, psoriasis, endometriosis, obesity and SARS-CoV-2(COVID-19), developmental angiogenic processes are recapitulated, but are often done so without the normal feedback mechanisms that regulate the ordinary spatial and temporal patterns of blood vessel formation. Thus, pathological angiogenesis presents new challenges yet new opportunities for the design of vascular-directed therapies. Here, we provide an overview of recent insights into blood vessel development and highlight novel therapeutic strategies that promote or inhibit the process of angiogenesis to stabilize, reverse, or even halt disease progression. In our review, we will also explore several additional aspects (the angiogenic switch, hypoxia, angiocrine signals, endothelial plasticity, vessel normalization, and endothelial cell anergy) that operate in parallel to canonical angiogenesis mechanisms and speculate how these processes may also be targeted with anti-angiogenic or vascular-directed therapies.
Collapse
Affiliation(s)
- Andrew C Dudley
- Department of Microbiology, Immunology and Cancer Biology, The University of Virginia, Charlottesville, VA, 22908, USA.
| | - Arjan W Griffioen
- Angiogenesis Laboratory, Department of Medical Oncology, Amsterdam UMC, Cancer Center Amsterdam, Amsterdam, The Netherlands.
| |
Collapse
|
7
|
Yuan M, Zhai Y, Hui Z. Application basis of combining antiangiogenic therapy with radiotherapy and immunotherapy in cancer treatment. Front Oncol 2022; 12:978608. [PMID: 36439496 PMCID: PMC9681994 DOI: 10.3389/fonc.2022.978608] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Accepted: 10/24/2022] [Indexed: 10/01/2023] Open
Abstract
How to further optimize the combination of radiotherapy and immunotherapy is among the current hot topics in cancer treatment. In addition to adopting the preferred dose-fractionation of radiotherapy or the regimen of immunotherapy, it is also very promising to add antiangiogenic therapy to this combination. We expound the application basis of cancer radiotherapy combined with immunotherapy and antiangiogenic therapy.
Collapse
Affiliation(s)
- Meng Yuan
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yirui Zhai
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhouguang Hui
- Department of VIP Medical Services, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
8
|
Hashemi G, Dight J, Khosrotehrani K, Sormani L. Melanoma Tumour Vascularization and Tissue-Resident Endothelial Progenitor Cells. Cancers (Basel) 2022; 14:4216. [PMID: 36077754 PMCID: PMC9454996 DOI: 10.3390/cancers14174216] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 08/17/2022] [Accepted: 08/24/2022] [Indexed: 11/20/2022] Open
Abstract
The aggressiveness of solid cancers, such as melanoma, relies on their metastatic potential. It has become evident that this key cause of mortality is largely conferred by the tumour-associated stromal cells, especially endothelial cells. In addition to their essential role in the formation of the tumour vasculature, endothelial cells significantly contribute to the establishment of the tumour microenvironment, thus enabling the dissemination of cancer cells. Melanoma tumour vascularization occurs through diverse biological processes. Vasculogenesis is the formation of de novo blood vessels from endothelial progenitor cells (EPCs), and recent research has shown the role of EPCs in melanoma tumour vascularization. A more detailed understanding of the complex role of EPCs and how they contribute to the abnormal vessel structures in tumours is of importance. Moreover, anti-angiogenic drugs have a limited effect on melanoma tumour vascularization, and the role of these drugs on EPCs remains to be clarified. Overall, targeting cancer vasculature remains a challenge, and the role of anti-angiogenic drugs and combination therapies in melanoma, a focus of this review, is an area of extensive exploration.
Collapse
Affiliation(s)
| | | | - Kiarash Khosrotehrani
- Experimental Dermatology Group, Dermatology Research Centre, The UQ Diamantina Institute, The University of Queensland, Woolloongabba, QLD 4102, Australia
| | - Laura Sormani
- Experimental Dermatology Group, Dermatology Research Centre, The UQ Diamantina Institute, The University of Queensland, Woolloongabba, QLD 4102, Australia
| |
Collapse
|
9
|
Kozin SV. Vascular damage in tumors: a key player in stereotactic radiation therapy? Trends Cancer 2022; 8:806-819. [PMID: 35835699 DOI: 10.1016/j.trecan.2022.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 05/23/2022] [Accepted: 06/01/2022] [Indexed: 11/17/2022]
Abstract
The use of stereotactic radiation therapy (SRT) for cancer treatment has grown in recent years, showing excellent results for some tumors. The greatly increased doses per fraction in SRT compared to conventional radiotherapy suggest a 'new biology' that determines treatment outcome. Proposed mechanisms include significant damage to tumor blood vessels and enhanced antitumor immune responses, which are also vasculature-dependent. These ideas are mostly based on the results of radiation studies in animal models because direct observations in humans are limited. However, even preclinical findings are somewhat incomplete and result in ambiguous conclusions. Current evidence of vasculature-related mechanisms of SRT is reviewed. Understanding them could result in better optimization of SRT alone or in combination with immune or other cancer therapies.
Collapse
Affiliation(s)
- Sergey V Kozin
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA.
| |
Collapse
|
10
|
The Multi-Kinase Inhibitor Lucitanib Enhances the Antitumor Activity of Coinhibitory and Costimulatory Immune Pathway Modulators in Syngeneic Models. J Immunother 2022; 45:335-348. [PMID: 35791438 DOI: 10.1097/cji.0000000000000427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 05/26/2022] [Indexed: 11/26/2022]
Abstract
Lucitanib is a multi-tyrosine kinase inhibitor whose targets are associated with angiogenesis and other key cancer and immune pathways. Its antiangiogenic properties are understood, but lucitanib's immunomodulatory activity is heretofore unknown. Lucitanib exhibited such activity in vivo, increasing CD3+, CD8+, and CD4+ T cells and decreasing dendritic cells and monocyte-derived suppressor cells in mouse spleens. Depletion of CD8+ T cells from syngeneic MC38 colon tumor-bearing mice reduced the antitumor efficacy of lucitanib and revealed a CD8+ T-cell-dependent component of lucitanib's activity. The combination of lucitanib and costimulatory immune pathway agonists targeting 4-1BB, glucocorticoid-induced TNFR (GITR), inducible T-cell co-stimulator (ICOS), or OX40 exhibited enhanced antitumor activity compared with each single agent in immunocompetent tumor models. Lucitanib combined with blockade of cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) or programmed cell death protein-1 (PD-1) coinhibitory immune pathways also showed enhanced antitumor activity over the single agents in multiple models. In CT26 tumors, lucitanib, alone or combined with anti-PD-1, reduced CD31+ vessels and depleted F4/80+ macrophages. Combination treatment also increased the number of intratumoral T cells. Gene expression in pathways associated with immune activity was upregulated by lucitanib in MC38 tumors and further potentiated by combination with anti-PD-1. Accordingly, lucitanib, alone or combined with anti-PD-1, increased intratumoral CD8+ T-cell abundance. Lucitanib's antitumor and pharmacodynamic activity, alone or combined with anti-PD-1, was not recapitulated by specific vascular endothelial growth factor receptor-2 (VEGFR2) inhibition. These data indicate that lucitanib can modulate vascular and immune components of the tumor microenvironment and cooperate with immunotherapy to enhance antitumor efficacy. They support the clinical development of lucitanib combined with immune pathway modulators to treat cancer.
Collapse
|
11
|
Hickman A, Koetsier J, Kurtanich T, Nielsen MC, Winn G, Wang Y, Bentebibel SE, Shi L, Punt S, Williams L, Haymaker C, Chesson CB, Fa'ak F, Dominguez A, Jones R, Kuiatse I, Caivano AR, Khounlo S, Warier ND, Marathi U, Market RV, Biediger RJ, Craft JW, Hwu P, Davies MA, Woodside DG, Vanderslice P, Diab A, Overwijk WW, Hailemichael Y. LFA-1 activation enriches tumor-specific T cells in a cold tumor model and synergizes with CTLA-4 blockade. J Clin Invest 2022; 132:154152. [PMID: 35552271 PMCID: PMC9246385 DOI: 10.1172/jci154152] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 05/10/2022] [Indexed: 12/02/2022] Open
Abstract
The inability of CD8+ effector T cells (Teffs) to reach tumor cells is an important aspect of tumor resistance to cancer immunotherapy. The recruitment of these cells to the tumor microenvironment (TME) is regulated by integrins, a family of adhesion molecules that are expressed on T cells. Here, we show that 7HP349, a small-molecule activator of lymphocyte function–associated antigen-1 (LFA-1) and very late activation antigen-4 (VLA-4) integrin cell-adhesion receptors, facilitated the preferential localization of tumor-specific T cells to the tumor and improved antitumor response. 7HP349 monotherapy had modest effects on anti–programmed death 1–resistant (anti–PD-1–resistant) tumors, whereas combinatorial treatment with anti–cytotoxic T lymphocyte–associated protein 4 (anti–CTLA-4) increased CD8+ Teff intratumoral sequestration and synergized in cooperation with neutrophils in inducing cancer regression. 7HP349 intratumoral CD8+ Teff enrichment activity depended on CXCL12. We analyzed gene expression profiles using RNA from baseline and on treatment tumor samples of 14 melanoma patients. We identified baseline CXCL12 gene expression as possibly improving the likelihood or response to anti–CTLA-4 therapies. Our results provide a proof-of-principle demonstration that LFA-1 activation could convert a T cell–exclusionary TME to a T cell–enriched TME through mechanisms involving cooperation with innate immune cells.
Collapse
Affiliation(s)
- Amber Hickman
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, United States of America
| | - Joost Koetsier
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, United States of America
| | - Trevin Kurtanich
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, United States of America
| | - Michael C Nielsen
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, United States of America
| | - Glenn Winn
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, United States of America
| | - Yunfei Wang
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, United States of America
| | - Salah-Eddine Bentebibel
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, United States of America
| | - Leilei Shi
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, United States of America
| | - Simone Punt
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, United States of America
| | - Leila Williams
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, United States of America
| | - Cara Haymaker
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, United States of America
| | - Charles B Chesson
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, United States of America
| | - Faisal Fa'ak
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, United States of America
| | - Ana Dominguez
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, United States of America
| | - Richard Jones
- Department of Lymphoma & Myeloma, The University of Texas MD Anderson Cancer Center, Houston, United States of America
| | - Isere Kuiatse
- Department of Lymphoma & Myeloma, The University of Texas MD Anderson Cancer Center, Houston, United States of America
| | - Amy R Caivano
- Molecular Cardiology Research Laboratories, Texas Heart Institute, Houston, United States of America
| | - Sayadeth Khounlo
- Molecular Cardiology Research Laboratories, Texas Heart Institute, Houston, United States of America
| | - Navin D Warier
- Molecular Cardiology Research Laboratories, Texas Heart Institute, Houston, United States of America
| | | | - Robert V Market
- Molecular Cardiology Research Laboratories, Texas Heart Institute, Houston, United States of America
| | - Ronald J Biediger
- Molecular Cardiology Research Laboratories, Texas Heart Institute, Houston, United States of America
| | - John W Craft
- Department of Biology and Chemistry, University of Houston, Houston, United States of America
| | - Patrick Hwu
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, United States of America
| | - Michael A Davies
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, United States of America
| | - Darren G Woodside
- Molecular Cardiology Research Laboratories, Texas Heart Institute, Houston, United States of America
| | - Peter Vanderslice
- Molecular Cardiology Research Laboratories, Texas Heart Institute, Houston, United States of America
| | - Adi Diab
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, United States of America
| | - Willem W Overwijk
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, United States of America
| | - Yared Hailemichael
- The University of Texas MD Anderson Cancer Center, Houston, United States of America
| |
Collapse
|
12
|
Kasherman L, Siu DHW, Woodford R, Harris CA. Angiogenesis Inhibitors and Immunomodulation in Renal Cell Cancers: The Past, Present, and Future. Cancers (Basel) 2022; 14:1406. [PMID: 35326557 PMCID: PMC8946206 DOI: 10.3390/cancers14061406] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 03/07/2022] [Indexed: 12/12/2022] Open
Abstract
Angiogenesis inhibitors have been adopted into the standard armamentarium of therapies for advanced-stage renal cell carcinomas (RCC), but more recently, combination regimens with immune checkpoint inhibitors have demonstrated better outcomes. Despite this, the majority of affected patients still eventually experience progressive disease due to therapeutic resistance mechanisms, and there remains a need to develop novel therapeutic strategies. This article will review the synergistic mechanisms behind angiogenesis and immunomodulation in the tumor microenvironment and discuss the pre-clinical and clinical evidence for both clear-cell and non-clear-cell RCC, exploring opportunities for future growth in this exciting area of drug development.
Collapse
Affiliation(s)
- Lawrence Kasherman
- Department of Medical Oncology, St. George Hospital, Kogarah, NSW 2217, Australia; (D.H.W.S.); (R.W.); (C.A.H.)
- St. George and Sutherland Clinical Schools, University of New South Wales, Sydney, NSW 2217, Australia
- Department of Medical Oncology, Illawarra Cancer Care Centre, Wollongong, NSW 2500, Australia
| | - Derrick Ho Wai Siu
- Department of Medical Oncology, St. George Hospital, Kogarah, NSW 2217, Australia; (D.H.W.S.); (R.W.); (C.A.H.)
- National Health Medical Research Council Clinical Trials Centre, University of Sydney, Camperdown, NSW 2050, Australia
| | - Rachel Woodford
- Department of Medical Oncology, St. George Hospital, Kogarah, NSW 2217, Australia; (D.H.W.S.); (R.W.); (C.A.H.)
- Faculty of Medciine and Health, University of Sydney, Camperdown, NSW 2050, Australia
| | - Carole A. Harris
- Department of Medical Oncology, St. George Hospital, Kogarah, NSW 2217, Australia; (D.H.W.S.); (R.W.); (C.A.H.)
- St. George and Sutherland Clinical Schools, University of New South Wales, Sydney, NSW 2217, Australia
| |
Collapse
|
13
|
Ayoub NM, Jaradat SK, Al-Shami KM, Alkhalifa AE. Targeting Angiogenesis in Breast Cancer: Current Evidence and Future Perspectives of Novel Anti-Angiogenic Approaches. Front Pharmacol 2022; 13:838133. [PMID: 35281942 PMCID: PMC8913593 DOI: 10.3389/fphar.2022.838133] [Citation(s) in RCA: 49] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 02/03/2022] [Indexed: 12/12/2022] Open
Abstract
Angiogenesis is a vital process for the growth and dissemination of solid cancers. Numerous molecular pathways are known to drive angiogenic switch in cancer cells promoting the growth of new blood vessels and increased incidence of distant metastasis. Several angiogenesis inhibitors are clinically available for the treatment of different types of advanced solid cancers. These inhibitors mostly belong to monoclonal antibodies or small-molecule tyrosine kinase inhibitors targeting the classical vascular endothelial growth factor (VEGF) and its receptors. Nevertheless, breast cancer is one example of solid tumors that had constantly failed to respond to angiogenesis inhibitors in terms of improved survival outcomes of patients. Accordingly, it is of paramount importance to assess the molecular mechanisms driving angiogenic signaling in breast cancer to explore suitable drug targets that can be further investigated in preclinical and clinical settings. This review summarizes the current evidence for the effect of clinically available anti-angiogenic drugs in breast cancer treatment. Further, major mechanisms associated with intrinsic or acquired resistance to anti-VEGF therapy are discussed. The review also describes evidence from preclinical and clinical studies on targeting novel non-VEGF angiogenic pathways in breast cancer and several approaches to the normalization of tumor vasculature by targeting pericytes, utilization of microRNAs and extracellular tumor-associate vesicles, using immunotherapeutic drugs, and nanotechnology.
Collapse
Affiliation(s)
- Nehad M. Ayoub
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology (JUST), Irbid, Jordan
- *Correspondence: Nehad M. Ayoub,
| | - Sara K. Jaradat
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology (JUST), Irbid, Jordan
| | - Kamal M. Al-Shami
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, AL, United States
| | - Amer E. Alkhalifa
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology (JUST), Irbid, Jordan
| |
Collapse
|
14
|
Baradaran A, Asadzadeh Z, Hemmat N, Baghbanzadeh A, Shadbad MA, Khosravi N, Derakhshani A, Alemohammad H, Afrashteh Nour M, Safarpour H, Silvestris N, Brunetti O, Baradaran B. The cross-talk between tumor-associated macrophages and tumor endothelium: Recent advances in macrophage-based cancer immunotherapy. Biomed Pharmacother 2022; 146:112588. [PMID: 35062062 DOI: 10.1016/j.biopha.2021.112588] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 12/21/2021] [Accepted: 12/23/2021] [Indexed: 11/02/2022] Open
Abstract
Tumor-associated macrophages (TAMs) are among the abundant cell populations of the tumor microenvironment (TME), which have pivotal roles in tumor development, chemoresistance, immune evasion, and metastasis. Growing evidence indicates that TAMs and the cross-talk between TAMs and tumoral endothelial cells can substantially contribute to tumor angiogenesis, which is considered a vital process for cancer development. Besides, tumoral endothelial cells can regulate the leukocyte infiltration to the TME in solid cancers and contribute to immune evasion. Therefore, targeting the immunosuppressive TAMs and the cross-talk between them can be a promising strategy for improving anti-tumoral immune responses. This review aims to summarize the biology of TAMs, their recently identified roles in tumor development/angiogenesis, and recent advances in macrophage-based cancer immunotherapy approaches for treating cancers.
Collapse
Affiliation(s)
- Ali Baradaran
- Diamantina Institute, Translational Research Institute, The University of Queensland, Brisbane, QLD, Australia; Research & Development, BSD Robotics, Queensland, Australia
| | - Zahra Asadzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nima Hemmat
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Baghbanzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahdi Abdoli Shadbad
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Neda Khosravi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Afshin Derakhshani
- Experimental Pharmacology Laboratory, IRCCS Istituto Tumori "Giovanni Paolo II", Bari, Italy
| | - Hajar Alemohammad
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mina Afrashteh Nour
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hossein Safarpour
- Cellular and Molecular Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Nicola Silvestris
- Medical Oncology Unit-IRCCS IstitutoTumori "Giovanni Paolo II" of Bari, Bari, Italy; Department of Biomedical Sciences and Human Oncology DIMO-University of Bari, Bari, Italy
| | - Oronzo Brunetti
- Medical Oncology Unit-IRCCS IstitutoTumori "Giovanni Paolo II" of Bari, Bari, Italy.
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
15
|
Zahedipour F, Zamani P, Jamialahmadi K, Jaafari MR, Sahebkar A. Vaccines targeting angiogenesis in melanoma. Eur J Pharmacol 2021; 912:174565. [PMID: 34656608 DOI: 10.1016/j.ejphar.2021.174565] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 09/28/2021] [Accepted: 10/11/2021] [Indexed: 12/15/2022]
Abstract
Angiogenesis has a significant role in metastasis and progression of melanoma. Even small tumors may be susceptible to metastasis and hence lead to a worse outcome in patients with melanoma. One of the anti-angiogenic treatment approaches that is undergoing comprehensive study is specific immunotherapy. While tumor cells are challenging targets for immunotherapy due to their genetic instability and heterogeneity, endothelial cells (ECs) are genetically stable. Therefore, vaccines targeting angiogenesis in melanoma are appropriate choices that target both tumor cells and ECs while capable of inducing strong, anti-tumor immune responses with limited toxicity. The main targets of angiogenesis are VEGFs and their receptors but other potential targets have also been investigated, especially in preclinical studies. Various types of vaccines that target angiogenesis in melanoma have been studied including DNA, peptide, protein, dendritic cell-based, and endothelial cell vaccines. This review outlines a number of target antigens that are important for potential progress in developing vaccines for targeting angiogenesis in melanoma. We also discuss different types of vaccines that have been investigated, delivery mechanisms and popular adjuvants, and suggest ways to improve future clinical outcomes.
Collapse
Affiliation(s)
- Fatemeh Zahedipour
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Parvin Zamani
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Khadijeh Jamialahmadi
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahmoud Reza Jaafari
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; School of Medicine, The University of Western Australia, Perth, Australia; School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
16
|
Kim MC, Jin Z, Kolb R, Borcherding N, Chatzkel JA, Falzarano SM, Zhang W. Updates on Immunotherapy and Immune Landscape in Renal Clear Cell Carcinoma. Cancers (Basel) 2021; 13:5856. [PMID: 34831009 PMCID: PMC8616149 DOI: 10.3390/cancers13225856] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 11/15/2021] [Accepted: 11/19/2021] [Indexed: 12/24/2022] Open
Abstract
Several clinicopathological features of clear cell renal cell carcinomas (ccRCC) contribute to make an "atypical" cancer, including resistance to chemotherapy, sensitivity to anti-angiogenesis therapy and ICIs despite a low mutational burden, and CD8+ T cell infiltration being the predictor for poor prognosis-normally CD8+ T cell infiltration is a good prognostic factor in cancer patients. These "atypical" features have brought researchers to investigate the molecular and immunological mechanisms that lead to the increased T cell infiltrates despite relatively low molecular burdens, as well as to decipher the immune landscape that leads to better response to ICIs. In the present study, we summarize the past and ongoing pivotal clinical trials of immunotherapies for ccRCC, emphasizing the potential molecular and cellular mechanisms that lead to the success or failure of ICI therapy. Single-cell analysis of ccRCC has provided a more thorough and detailed understanding of the tumor immune microenvironment and has facilitated the discovery of molecular biomarkers from the tumor-infiltrating immune cells. We herein will focus on the discussion of some major immune cells, including T cells and tumor-associated macrophages (TAM) in ccRCC. We will further provide some perspectives of using molecular and cellular biomarkers derived from these immune cell types to potentially improve the response rate to ICIs in ccRCC patients.
Collapse
Affiliation(s)
- Myung-Chul Kim
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL 32610, USA; (M.-C.K.); (Z.J.); (R.K.); (S.M.F.)
- UF Health Cancer Center, University of Florida, Gainesville, FL 32610, USA
| | - Zeng Jin
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL 32610, USA; (M.-C.K.); (Z.J.); (R.K.); (S.M.F.)
- UF Health Cancer Center, University of Florida, Gainesville, FL 32610, USA
| | - Ryan Kolb
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL 32610, USA; (M.-C.K.); (Z.J.); (R.K.); (S.M.F.)
- UF Health Cancer Center, University of Florida, Gainesville, FL 32610, USA
| | - Nicholas Borcherding
- Department of Pathology and Immunology, Washington University, St. Louis, MO 63110, USA;
| | | | - Sara Moscovita Falzarano
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL 32610, USA; (M.-C.K.); (Z.J.); (R.K.); (S.M.F.)
- UF Health Cancer Center, University of Florida, Gainesville, FL 32610, USA
| | - Weizhou Zhang
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL 32610, USA; (M.-C.K.); (Z.J.); (R.K.); (S.M.F.)
- UF Health Cancer Center, University of Florida, Gainesville, FL 32610, USA
| |
Collapse
|
17
|
王 婧, 彭 文, 江 美, 邬 麟. [Research Progress of Anti-angiogenic Agents Combined with Immunotherapy
in Patients with Advanced Non-small Cell Lung Cancer]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2021; 24:196-203. [PMID: 33819970 PMCID: PMC8143969 DOI: 10.3779/j.issn.1009-3419.2021.101.05] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 01/12/2021] [Accepted: 01/21/2021] [Indexed: 12/05/2022]
Abstract
Lung cancer has the highest incidence rate and mortality in China, even in the world, and non-small cell lung cancer (NSCLC) accounts for about 85%. The growth and metastasis of tumor depend on the generation of blood vessels, and anti-angiogenic therapy is playing an increasingly important role, however, no significant improvement was observed in the underwent anti-angiogenic agents used for patients alone. In recent years, the application of immune checkpoint inhibitor (ICI) has significantly improved the prognosis of some lung cancer patients, however, the objective response rate of patients receiving ICI alone is low. While anti-angiogenic agents and ICI both regulate the tumor immune microenvironment and have a potential synergistic mechanism, showing a bright prospect in the combined application of anti-tumor therapy. In this review, we focused on the research and application of anti-angiogenic agents in combination with ICI in advanced non-small cell lung cancer.
Collapse
Affiliation(s)
- 婧怡 王
- />410013 长沙,中南大学湘雅医学院附属肿瘤医院/湖南省肿瘤医院胸部内二科The Second Department of Thoracic Oncology, the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha 410013, China
| | - 文颖 彭
- />410013 长沙,中南大学湘雅医学院附属肿瘤医院/湖南省肿瘤医院胸部内二科The Second Department of Thoracic Oncology, the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha 410013, China
| | - 美林 江
- />410013 长沙,中南大学湘雅医学院附属肿瘤医院/湖南省肿瘤医院胸部内二科The Second Department of Thoracic Oncology, the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha 410013, China
| | - 麟 邬
- />410013 长沙,中南大学湘雅医学院附属肿瘤医院/湖南省肿瘤医院胸部内二科The Second Department of Thoracic Oncology, the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha 410013, China
| |
Collapse
|
18
|
Wooster AL, Girgis LH, Brazeale H, Anderson TS, Wood LM, Lowe DB. Dendritic cell vaccine therapy for colorectal cancer. Pharmacol Res 2021; 164:105374. [PMID: 33348026 PMCID: PMC7867624 DOI: 10.1016/j.phrs.2020.105374] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 12/03/2020] [Accepted: 12/04/2020] [Indexed: 02/06/2023]
Abstract
Colorectal cancer (CRC) remains a leading cause of cancer-related deaths in the United States despite an array of available treatment options. Current standard-of-care interventions for this malignancy include surgical resection, chemotherapy, and targeted therapies depending on the disease stage. Specifically, infusion of anti-vascular endothelial growth factor agents in combination with chemotherapy was an important development in improving the survival of patients with advanced colorectal cancer, while also helping give rise to other forms of anti-angiogenic therapies. Yet, one approach by which tumor angiogenesis may be further disrupted is through the administration of a dendritic cell (DC) vaccine targeting tumor-derived blood vessels, leading to cytotoxic immune responses that decrease tumor growth and synergize with other systemic therapies. Early generations of such vaccines exhibited protection against various forms of cancer in pre-clinical models, but clinical results have historically been disappointing. Sipuleucel-T (Provenge®) was the first, and to-date, only dendritic cell-based therapy to receive FDA approval after significantly increasing overall survival in prostate cancer patients. The unparalleled success of Sipuleucel-T has helped revitalize the clinical development of dendritic cell vaccines, which will be examined in this review. We also highlight the promise of these vaccines to instill anti-angiogenic immunity for individuals with advanced colorectal cancer.
Collapse
Affiliation(s)
- Amanda L Wooster
- Department of Immunotherotherapeutics and Biotechnology, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX, 79601, United States
| | - Lydia H Girgis
- Department of Immunotherotherapeutics and Biotechnology, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX, 79601, United States
| | - Hayley Brazeale
- Department of Immunotherotherapeutics and Biotechnology, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX, 79601, United States
| | - Trevor S Anderson
- Department of Immunotherotherapeutics and Biotechnology, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX, 79601, United States
| | - Laurence M Wood
- Department of Immunotherotherapeutics and Biotechnology, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX, 79601, United States
| | - Devin B Lowe
- Department of Immunotherotherapeutics and Biotechnology, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX, 79601, United States.
| |
Collapse
|
19
|
Zhao J, Zhang X, Du Y, Zhou L, Dong Z, Zhao J, Lu J. Allogenic mouse cell vaccine inhibits lung cancer progression by inhibiting angiogenesis. Hum Vaccin Immunother 2021; 17:35-50. [PMID: 32460680 DOI: 10.1080/21645515.2020.1759996] [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] [Indexed: 12/26/2022] Open
Abstract
Aim: This research investigated the therapeutic effect of an allogeneic mouse brain microvascular endothelial cell vaccine on lung cancer and further elucidated its potential anti-angiogenic mechanism. Materials & methods: The immune effect of the allogeneic bEnd.3 vaccine and DC vaccine loaded with bEnd.3 antigen on the subcutaneous transplantation of Lewis lung cancer (LLC) was assessed by ELISA, the CCK test and the CTL killing test. The mechanism was preliminarily revealed by immunohistochemistry and immunoblot analysis. Results: This study revealed that tumor volume was decreased (p < .01) and the survival was prolonged significantly (p < .05) by the bEnd.3 vaccine in subcutaneous LLC transplantation in the vaccine prevention group. In contrast, both tumor volume in the serum therapeutic group and survival of bEnd.3 vaccine were not significantly different from those of the control group (p > .05). Importantly, tumor volume and survival of the T lymphocyte therapeutic group were decreased and prolonged (p < .05). In addition, both tumor volume and survival of DC vaccine loaded with bEnd.3 in the vaccine prevention group were decreased and prolonged significantly (p < .01). Furthermore, bEnd.3 vaccine and DC vaccine loaded with bEnd.3 both produced the activity of killing bEnd.3 target cells in vitro.The reason may induce the immune mice to produce anti-VEGFR-II, anti-endoglin and anti-integrin αν antibodies to have an anti-angiogenesis function. Conclusion: The allogeneic mouse bEnd.3 cell vaccine can block angiogenesis and prevent the development of lung cancer transplantation tumors.
Collapse
Affiliation(s)
- Jun Zhao
- Department of Oncology, Changzhi People's Hospital , Changzhi, China.,Department of Pathophysiology, College of Basic Medical Sciences, Zhengzhou University , Zhengzhou, China
| | - Xiaoling Zhang
- Department of Oncology, Changzhi People's Hospital , Changzhi, China
| | - Yunyi Du
- Department of Oncology, Changzhi People's Hospital , Changzhi, China
| | - Lurong Zhou
- Quality Control Department, Changzhi People's Hospital , Changzhi, China
| | - Ziming Dong
- Department of Pathophysiology, College of Basic Medical Sciences, Zhengzhou University , Zhengzhou, China.,Collaborative Innovation Center of Henan Province for Cancer Chemoprevention , Zhengzhou, China.,State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University , Zhengzhou, China
| | - Jimin Zhao
- Department of Pathophysiology, College of Basic Medical Sciences, Zhengzhou University , Zhengzhou, China.,Collaborative Innovation Center of Henan Province for Cancer Chemoprevention , Zhengzhou, China.,State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University , Zhengzhou, China
| | - Jing Lu
- Department of Pathophysiology, College of Basic Medical Sciences, Zhengzhou University , Zhengzhou, China.,Collaborative Innovation Center of Henan Province for Cancer Chemoprevention , Zhengzhou, China.,State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University , Zhengzhou, China
| |
Collapse
|
20
|
Integrating Phenotypic Search and Phosphoproteomic Profiling of Active Kinases for Optimization of Drug Mixtures for RCC Treatment. Cancers (Basel) 2020; 12:cancers12092697. [PMID: 32967224 PMCID: PMC7564658 DOI: 10.3390/cancers12092697] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 09/10/2020] [Accepted: 09/15/2020] [Indexed: 12/22/2022] Open
Abstract
Combined application of multiple therapeutic agents presents the possibility of enhanced efficacy and reduced development of resistance. Definition of the most appropriate combination for any given disease phenotype is challenged by the vast number of theoretically possible combinations of drugs and doses, making extensive empirical testing a virtually impossible task. We have used the streamlined-feedback system control (s-FSC) technique, a phenotypic approach, which converges to optimized drug combinations (ODC) within a few experimental steps. Phosphoproteomics analysis coupled to kinase activity analysis using the novel INKA (integrative inferred kinase activity) pipeline was performed to evaluate ODC mechanisms in a panel of renal cell carcinoma (RCC) cell lines. We identified different ODC with up to 95% effectivity for each RCC cell line, with low doses (ED5-25) of individual drugs. Global phosphoproteomics analysis demonstrated inhibition of relevant kinases, and targeting remaining active kinases with additional compounds improved efficacy. In addition, we identified a common RCC ODC, based on kinase activity data, to be effective in all RCC cell lines under study. Combining s-FSC with a phosphoproteomic profiling approach provides valuable insight in targetable kinase activity and allows for the identification of superior drug combinations for the treatment of RCC.
Collapse
|
21
|
Jo Y, Ali LA, Shim JA, Lee BH, Hong C. Innovative CAR-T Cell Therapy for Solid Tumor; Current Duel between CAR-T Spear and Tumor Shield. Cancers (Basel) 2020; 12:cancers12082087. [PMID: 32731404 PMCID: PMC7464778 DOI: 10.3390/cancers12082087] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 07/21/2020] [Accepted: 07/23/2020] [Indexed: 12/12/2022] Open
Abstract
Novel engineered T cells containing chimeric antigen receptors (CAR-T cells) that combine the benefits of antigen recognition and T cell response have been developed, and their effect in the anti-tumor immunotherapy of patients with relapsed/refractory leukemia has been dramatic. Thus, CAR-T cell immunotherapy is rapidly emerging as a new therapy. However, it has limitations that prevent consistency in therapeutic effects in solid tumors, which accounts for over 90% of all cancer patients. Here, we review the literature regarding various obstacles to CAR-T cell immunotherapy for solid tumors, including those that cause CAR-T cell dysfunction in the immunosuppressive tumor microenvironment, such as reactive oxygen species, pH, O2, immunosuppressive cells, cytokines, and metabolites, as well as those that impair cell trafficking into the tumor microenvironment. Next-generation CAR-T cell therapy is currently undergoing clinical trials to overcome these challenges. Therefore, novel approaches to address the challenges faced by CAR-T cell immunotherapy in solid tumors are also discussed here.
Collapse
Affiliation(s)
- Yuna Jo
- Department of Anatomy, Pusan National University School of Medicine, Yangsan 50612, Korea; (Y.J.); (L.A.A.); (J.A.S.)
| | - Laraib Amir Ali
- Department of Anatomy, Pusan National University School of Medicine, Yangsan 50612, Korea; (Y.J.); (L.A.A.); (J.A.S.)
| | - Ju A Shim
- Department of Anatomy, Pusan National University School of Medicine, Yangsan 50612, Korea; (Y.J.); (L.A.A.); (J.A.S.)
| | - Byung Ha Lee
- NeoImmuneTech, Inc., 2400 Research Blvd., Suite 250, Rockville, MD 20850, USA;
| | - Changwan Hong
- Department of Anatomy, Pusan National University School of Medicine, Yangsan 50612, Korea; (Y.J.); (L.A.A.); (J.A.S.)
- Correspondence: ; Tel.: +82-51-510-8041
| |
Collapse
|
22
|
Taguchi K, Onoe T, Yoshida T, Yamashita Y, Tanaka Y, Ohdan H. Tumor Endothelial Cell–Mediated Antigen-Specific T-cell Suppression via the PD-1/PD-L1 Pathway. Mol Cancer Res 2020; 18:1427-1440. [DOI: 10.1158/1541-7786.mcr-19-0897] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 04/01/2020] [Accepted: 06/05/2020] [Indexed: 11/16/2022]
|
23
|
Mercurio L, Failla CM, Capriotti L, Scarponi C, Facchiano F, Morelli M, Rossi S, Pagnanelli G, Albanesi C, Cavani A, Madonna S. Interleukin (IL)-17/IL-36 axis participates to the crosstalk between endothelial cells and keratinocytes during inflammatory skin responses. PLoS One 2020; 15:e0222969. [PMID: 32352958 PMCID: PMC7192413 DOI: 10.1371/journal.pone.0222969] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 04/15/2020] [Indexed: 12/22/2022] Open
Abstract
In inflammatory skin conditions, such as psoriasis, vascular enlargement is associated with endothelial cell proliferation, release of cytokines and adhesion molecule expression. Interleukin (IL)-17A is a pro-inflammatory cytokine mainly secreted by T helper-17 cells that is critically involved in psoriasis pathogenesis. IL-36α, IL-36β and IL-36γ are also inflammatory cytokines up-regulated in psoriasis and induced by various stimuli, including IL-17A. In this study, we found that human keratinocytes are the main source of IL-36, in particular of IL-36γ. This cytokine was strongly induced by IL-17A and, together with IL-17A, efficiently activated human dermal microvascular endothelial cells (HDMECs), which expressed both IL-17 and IL-36 receptors. Both IL-36γ and IL-17A induced cell proliferation through specific molecular cascades involving ERK1/2 only or ERK1/2, STAT3 and NF-κB, respectively. We highlighted the intense IL-17A- and IL-36γ -dependent interplay between keratinocytes and HDMECs, likely active in the psoriatic lesions and leading to the establishment of a cytokine network responsible for the development and maintenance of the inflamed state. IL-17A or IL-36γ showed in HDMECs a synergic activity with TNF-α by potently inducing inflammatory cytokine/chemokine release and ICAM-1 expression. We also investigated the involvement of IL-36γ and VEGF-A, substantially reduced in lesional skin of psoriatic patients pharmacologically treated with the anti-IL-17A antibody Secukinumab. Importantly, keratinocyte-derived IL-36γ represented an additional pro-angiogenic mediator of IL-17A. We observed that keratinocyte-derived VEGF-A influenced proliferation but did not act on expression of adhesion molecules in HDMECs. On the other hand, inhibition of IL-36γ released by IL-17A-treated keratinocytes impaired either proliferation or ICAM-1 expression both in HDMECs and in an in vivo murine model of psoriasis. Taken together, our data demonstrated that IL-17A and IL-36γ are highly involved in endothelial cells/keratinocytes crosstalk in inflammatory skin conditions.
Collapse
Affiliation(s)
- Laura Mercurio
- Laboratory of Experimental Immunology, IDI-IRCCS, Rome, Italy
| | | | | | | | - Francesco Facchiano
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità (ISS), Rome, Italy
| | - Martina Morelli
- Laboratory of Experimental Immunology, IDI-IRCCS, Rome, Italy
| | - Stefania Rossi
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità (ISS), Rome, Italy
| | | | | | - Andrea Cavani
- National Institute for Health, Migration and Poverty (NIHMP), Rome, Italy
| | | |
Collapse
|
24
|
Zhang J, Endres S, Kobold S. Enhancing tumor T cell infiltration to enable cancer immunotherapy. Immunotherapy 2020; 11:201-213. [PMID: 30730277 DOI: 10.2217/imt-2018-0111] [Citation(s) in RCA: 88] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Cancer immunotherapy has changed the treatment landscape for cancer patients, especially for those with metastatic spread. While the immunotherapeutic armamentarium is constantly growing, as exemplified by approved compounds, clinical outcome remains variable both within and across entities. A sufficient infiltration into the tumor microenvironment and successful activation of effector T lymphocytes against tumor cells have been identified as predictors for responses to T cell-based immunotherapies. However, tumor cells have developed a variety of mechanisms to reduce T cell homing and access to the tumor tissue to prevent activity of anticancer immunity. As a consequence, investigations have interrogated strategies to improve the efficacy of cancer immunotherapies by enhancing T cell infiltration into tumor tissues. In this review, we summarize mechanisms of how tumor tissue shapes immune suppressive microenvironment to prevent T cell access to the tumor site. We focus on current strategies to improve cancer immunotherapies through enhancing T cell infiltration.
Collapse
Affiliation(s)
- Jin Zhang
- Center of Integrated Protein Science Munich (CIPS-M) & Division of Clinical Pharmacology, Klinikum der Universität München, Lindwurmstrasse 2a, 80337 Munich, Germany, Member of the German Center of Lung Research
| | - Stefan Endres
- Center of Integrated Protein Science Munich (CIPS-M) & Division of Clinical Pharmacology, Klinikum der Universität München, Lindwurmstrasse 2a, 80337 Munich, Germany, Member of the German Center of Lung Research
| | - Sebastian Kobold
- Center of Integrated Protein Science Munich (CIPS-M) & Division of Clinical Pharmacology, Klinikum der Universität München, Lindwurmstrasse 2a, 80337 Munich, Germany, Member of the German Center of Lung Research
| |
Collapse
|
25
|
Ribatti D, Annese T, Ruggieri S, Tamma R, Crivellato E. Limitations of Anti-Angiogenic Treatment of Tumors. Transl Oncol 2019; 12:981-986. [PMID: 31121490 PMCID: PMC6529826 DOI: 10.1016/j.tranon.2019.04.022] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 04/11/2019] [Indexed: 01/26/2023] Open
Abstract
Clinical trials using anti-vascular endothelial growth factor /(VEGF) molecules induce a modest improvement in overall survival, measurable in weeks to just a few months, and tumors respond differently to these agents. In this review article, we have exposed some tumor characteristics and processes that may impair the effectiveness of anti-angiogenic approaches, including genotypic changes on endothelial cells, the vascular normalization phenomenon, and the vasculogenic mimicry. The usage of anti-angiogenic molecules leads to hypoxic tumor microenvironment which enhances tumor invasiveness. The role of tumor-infiltrating cells, including tumor associated macrophages and fibroblasts (TAMs and TAFs) in the therapeutic response to anti-angiogenic settings was also highlighted. Finally, among the new therapeutic approaches to target tumor vasculature, anti-PD-1 or anti-PD-L1 therapy sensitizing and prolonging the efficacy of anti-angiogenic therapy, have been discussed.
Collapse
Affiliation(s)
- Domenico Ribatti
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, Bari, Italy.
| | - Tiziana Annese
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, Bari, Italy
| | - Simona Ruggieri
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, Bari, Italy
| | - Roberto Tamma
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, Bari, Italy
| | - Enrico Crivellato
- Department of Medicine, Section of Human Anatomy, University of Udine, Italy
| |
Collapse
|
26
|
Goedegebuure RSA, de Klerk LK, Bass AJ, Derks S, Thijssen VLJL. Combining Radiotherapy With Anti-angiogenic Therapy and Immunotherapy; A Therapeutic Triad for Cancer? Front Immunol 2019; 9:3107. [PMID: 30692993 PMCID: PMC6339950 DOI: 10.3389/fimmu.2018.03107] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 12/17/2018] [Indexed: 12/19/2022] Open
Abstract
Radiotherapy has been used for the treatment of cancer for over a century. Throughout this period, the therapeutic benefit of radiotherapy has continuously progressed due to technical developments and increased insight in the biological mechanisms underlying the cellular responses to irradiation. In order to further improve radiotherapy efficacy, there is a mounting interest in combining radiotherapy with other forms of therapy such as anti-angiogenic therapy or immunotherapy. These strategies provide different opportunities and challenges, especially with regard to dose scheduling and timing. Addressing these issues requires insight in the interaction between the different treatment modalities. In the current review, we describe the basic principles of the effects of radiotherapy on tumor vascularization and tumor immunity and vice versa. We discuss the main strategies to combine these treatment modalities and the hurdles that have to be overcome in order to maximize therapeutic effectivity. Finally, we evaluate the outstanding questions and present future prospects of a therapeutic triad for cancer.
Collapse
Affiliation(s)
- Ruben S A Goedegebuure
- Amsterdam UMC, Location VUmc, Medical Oncology, Cancer Center Amsterdam, Amsterdam, Netherlands
| | - Leonie K de Klerk
- Amsterdam UMC, Location VUmc, Medical Oncology, Cancer Center Amsterdam, Amsterdam, Netherlands.,Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States
| | - Adam J Bass
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States.,Cancer Program, The Broad Institute of MIT and Harvard, Cambridge, MA, United States
| | - Sarah Derks
- Amsterdam UMC, Location VUmc, Medical Oncology, Cancer Center Amsterdam, Amsterdam, Netherlands
| | - Victor L J L Thijssen
- Amsterdam UMC, Location VUmc, Medical Oncology, Cancer Center Amsterdam, Amsterdam, Netherlands.,Amsterdam UMC, Location VUmc, Radiation Oncology, Cancer Center Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
27
|
Caruana I, Simula L, Locatelli F, Campello S. T lymphocytes against solid malignancies: winning ways to defeat tumours. Cell Stress 2018; 2:200-212. [PMID: 31225487 PMCID: PMC6551626 DOI: 10.15698/cst2018.07.148] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
In the last decades, a novel field has emerged in the cure of cancer, by boosting the ability of the patient’s immune system to recognize and kill tumour cells. Although excellent and encouraging results, exploiting the effect of genetically modified T cells, have been obtained, it is now evident that tumour malignancies can evolve several mechanisms to escape such immune responses, thus continuing their growth in the body. These mechanisms are in part due to tumour cell metabolic or genetic alterations, which can render the target invisible to the immune system or can favour the generation of an extracellular milieu preventing immune cell infiltration or cytotoxicity. Such mechanisms may also involve the accumulation inside the tumour microenvironment of different immune-suppressive cell types, which further down-regulate the activity of cytotoxic immune cells either directly by interacting with them or indirectly by releasing suppressive molecules. In this review, we will first focus on describing several mechanisms by which tumour cells may dampen or abrogate the immune response inside the tumour microenvironment and, second, on current strategies that are adopted to cope with and possibly overcome such alterations, thus ameliorating the efficacy of the current-in-use anti-cancer immuno-therapies.
Collapse
Affiliation(s)
- Ignazio Caruana
- Dept. of Pediatric Onco-Hematology and cell and gene therapy, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
| | - Luca Simula
- Dept. of Pediatric Onco-Hematology and cell and gene therapy, IRCCS Bambino Gesù Children's Hospital, Rome, Italy.,Dept. of Biology, University of Rome Tor Vergata, Rome, Italy
| | - Franco Locatelli
- Dept. of Pediatric Onco-Hematology and cell and gene therapy, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
| | - Silvia Campello
- Dept. of Biology, University of Rome Tor Vergata, Rome, Italy.,IRCCS, Santa Lucia Foundation, Rome, Italy
| |
Collapse
|
28
|
Weishaupt C, Emmert S. Connecting basic cold plasma technology to dermato-oncology. CLINICAL PLASMA MEDICINE 2018. [DOI: 10.1016/j.cpme.2018.03.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
29
|
Abstract
Immunotherapies have revolutionized medical oncology following the remarkable and, in some cases, unprecedented outcomes observed in certain groups of patients with cancer. Combination with other therapeutic modalities, including anti-angiogenic agents, is one of the many strategies currently under investigation to improve the response rates and duration of immunotherapies. Such a strategy might seem counterintuitive given that anti-angiogenic agents can increase tumour hypoxia and reduce the number of blood vessels within tumours. Herein, we review the additional effects mediated by drugs targeting VEGF-dependent signalling and other pathways, such as those mediated by angiopoietin 2 or HGF, which might increase the efficacy of immunotherapies. In addition, we discuss the seldom considered possibility that immunotherapies, and immune-checkpoint inhibitors in particular, might increase the efficacy of anti-angiogenic or other types of antivascular therapies and/or promote changes in the tumour vasculature. In short, we propose that interactions between both therapeutic modalities could be considered a 'two-way street'.
Collapse
|
30
|
Abstract
PURPOSE OF REVIEW Most cancer patients experience pain and many will require opioids. However, the effects of opioids on cancer progression, metastasis, and recurrence is increasingly being questioned. There is evidence that opioids affect immune system function, angiogenesis, apoptosis, and invasion in a potentially deleterious manner. This review will examine the preclinical and clinical evidence. RECENT FINDINGS Recent clinical data have struggled to find robust evidence that opioids promote cancer progression. Although most study has involved morphine, differential effects of other opioids on immune function and cancer are revealing a more complex picture. SUMMARY Although there is a biologically plausible story, evidence for the action of opioids on cancer is mixed. Indeed, it may even be that in the chronic setting morphine has a beneficial effect on outcome in certain cancer types. This review critically examines and evaluates the evidence for the action of opioids on the processes involved in cancer progression. In the light of the uncertainty of opioid effect on cancer, any decision making should be tempered by knowing that stress and pain undoubtedly contribute to cancer progression.
Collapse
|
31
|
Xu Q, Gu J, Lv Y, Yuan J, Yang N, Chen J, Wang C, Hou X, Jia X, Feng L, Yin G. Angiogenesis for tumor vascular normalization of Endostar on hepatoma 22 tumor-bearing mice is involved in the immune response. Oncol Lett 2018; 15:3437-3446. [PMID: 29467868 PMCID: PMC5795950 DOI: 10.3892/ol.2018.7734] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2015] [Accepted: 05/18/2017] [Indexed: 12/11/2022] Open
Abstract
Tumor vascular normalization involved in immune response is beneficial to the chemotherapy of tumors. Recombinant human endostatin (Endostar), an angiogenesis inhibitor, has been demonstrated to be effective in hepatocellular cancer (HCC). However, its vascular normalization in HCC and the role of the immune response in angiogenesis were unclear. In the present study, effects of Endostar on tumor vascular normalization were evaluated in hepatoma 22 (H22) tumor-bearing mice. Endostar was able to inhibit the proliferation and infiltration of tumor cells and improve α-fetoprotein, tumor necrosis factor-α and cyclic adenosine 5′-phosphate levels in the serum of H22-bearing mice, as well as the protein expression levels of the immune factors interferon-γ and cluster of differentiation (CD)86 in liver tissue. Endostar also exhibited more marked downregulation of the levels of vascular endothelial growth factor, CD31, matrix metalloproteinase (MMP)-2, MMP-9 and interleukin-17 during day 3–9 treatment, resulting in short-term normalization of tumor blood vessels. The period of vascular normalization was 3–9 days. The results of the present study demonstrated that Endostar was able to induce the period of vascular normalization, contributing to a more efficacious means of HCC treatment combined with other chemotherapy, and this effect was associated with the immune response. It may be concluded that Endostar inhibited immunity-associated angiogenesis behaviors of vascular endothelial cells in response to HCC. The results of the present study provided more reasonable possibility for the combination therapy of Endostar for the treatment of HCC.
Collapse
Affiliation(s)
- Qingyu Xu
- Department of Intervention, Cancer Hospital of Jiangsu, Nanjing, Jiangsu 210009, P.R. China
| | - Junfei Gu
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, P.R. China
| | - You Lv
- Department of Intervention, Cancer Hospital of Jiangsu, Nanjing, Jiangsu 210009, P.R. China
| | - Jiarui Yuan
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, P.R. China
| | - Nan Yang
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, P.R. China
| | - Juan Chen
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, P.R. China
| | - Chunfei Wang
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, P.R. China
| | - Xuefeng Hou
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, P.R. China
| | - Xiaobin Jia
- Key Laboratory of Delivery Systems of Chinese Materia Medica, Jiangsu Provincial Academy of Chinese Medicine, Nanjing, Jiangsu 210028, P.R. China
| | - Liang Feng
- Key Laboratory of Delivery Systems of Chinese Materia Medica, Jiangsu Provincial Academy of Chinese Medicine, Nanjing, Jiangsu 210028, P.R. China
| | - Guowen Yin
- Department of Intervention, Cancer Hospital of Jiangsu, Nanjing, Jiangsu 210009, P.R. China
| |
Collapse
|
32
|
Impact of Blood Vessel Quantity and Vascular Expression of CD133 and ICAM-1 on Survival of Glioblastoma Patients. NEUROSCIENCE JOURNAL 2017; 2017:5629563. [PMID: 29250531 PMCID: PMC5698821 DOI: 10.1155/2017/5629563] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/12/2017] [Revised: 08/07/2017] [Accepted: 10/04/2017] [Indexed: 11/18/2022]
Abstract
Glioblastoma (GB) is the most angiogenic tumor. Nevertheless, antiangiogenic therapy has not shown significant clinical efficacy. The aim of this study was to assess blood vessel characteristics on survival of GB patients. Surgically excised GB tissues were histologically examined for overall proportion of glomeruloid microvascular proliferation (MP) and the total number of blood vessels. Also, immunohistochemical vascular staining intensities of CD133 and ICAM-1 were determined. Vessel parameters were correlated with patients' overall survival. The survival time depended on the number of blood vessels (p = 0.03) but not on the proportion of MP. Median survival times for patients with low (<median) and high (≥median) number of blood vessels were 9.0 months (95% CI: 7.5–10.5) and 12.0 months (95% CI: 9.3–14.7). Also, median survival times for patients with low (<median) and high (≥median) vascular expression level of CD133 were 9.0 months (95% CI: 8.0–10.1) and 12.0 months (95% CI: 10.3–13.7). In contrast, the staining intensity of vascular ICAM-1 did not affect survival. In multivariate analysis, the number of blood vessels emerged as an independent predictor for longer overall survival (HR: 2.4, 95% CI: 1.2–5.0, p = 0.02). For success in antiangiogenic therapy, better understanding about tumor vasculature biology is needed.
Collapse
|
33
|
Reply to Marchiò et al.: Antitumor immune regulation by angiostatic therapy. Proc Natl Acad Sci U S A 2017; 114:E3166-E3167. [DOI: 10.1073/pnas.1703074114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
34
|
Berndsen RH, Abdul UK, Weiss A, Zoetemelk M, te Winkel MT, Dyson PJ, Griffioen AW, Nowak-Sliwinska P. Epigenetic approach for angiostatic therapy: promising combinations for cancer treatment. Angiogenesis 2017; 20:245-267. [DOI: 10.1007/s10456-017-9551-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2017] [Accepted: 03/10/2017] [Indexed: 12/15/2022]
|
35
|
Hu C, Jiang X. The effect of anti-angiogenic drugs on regulatory T cells in the tumor microenvironment. Biomed Pharmacother 2017; 88:134-137. [DOI: 10.1016/j.biopha.2017.01.051] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 01/06/2017] [Accepted: 01/09/2017] [Indexed: 11/30/2022] Open
|
36
|
Fabian KPL, Chi-Sabins N, Taylor JL, Fecek R, Weinstein A, Storkus WJ. Therapeutic efficacy of combined vaccination against tumor pericyte-associated antigens DLK1 and DLK2 in mice. Oncoimmunology 2017; 6:e1290035. [PMID: 28405524 DOI: 10.1080/2162402x.2017.1290035] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Revised: 01/25/2017] [Accepted: 01/26/2017] [Indexed: 12/28/2022] Open
Abstract
When compared with vascular cells in normal tissues, pericytes and vascular endothelial cells (VEC) in tumor blood vessels exhibit altered morphology and epigenetic programming that leads to the expression of unique antigens that allow for differential recognition by CD8+ T cells. We have previously shown that the Notch antagonist delta-like homolog 1 (DLK1) is a tumor pericyte-associated antigen expressed in setting of melanoma and a range of carcinomas. In this report, we show that therapeutic vaccination against DLK1 in murine models results in slowed tumor growth, but also to the compensatory expression of the DLK1 homolog, DLK2, by tumor-associated pericytes. Vaccines targeting both DLK1 and DLK2 resulted in superior antitumor benefits in association with improved activation and recruitment of antigen-specific Type 1 CD8+ T cells, reduced presence of myeloid-derived suppressive cells, T regulatory cell and tumor vascular normalization. The antitumor efficacy of vaccines coordinately targeting DLK1 and DLK2 was further improved by inclusion of PD-L1 blockade, thus defining a combination immunotherapy theoretically suitable for the treatment of a broad range of solid (vascularized) cancers.
Collapse
Affiliation(s)
- Kellsye Paula L Fabian
- Department of Immunology, University of Pittsburgh School of Medicine , Pittsburgh, PA, USA
| | - Nina Chi-Sabins
- Department of Immunology, University of Pittsburgh School of Medicine , Pittsburgh, PA, USA
| | - Jennifer L Taylor
- Department of Dermatology, University of Pittsburgh School of Medicine , Pittsburgh, PA, USA
| | - Ronald Fecek
- Department of Dermatology, University of Pittsburgh School of Medicine , Pittsburgh, PA, USA
| | - Aliyah Weinstein
- Department of Immunology, University of Pittsburgh School of Medicine , Pittsburgh, PA, USA
| | - Walter J Storkus
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; The University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
| |
Collapse
|
37
|
Xu A, Zhang L, Chen Y, Lin Z, Li R. Immunogenicity and efficacy of a rationally designed vaccine against vascular endothelial growth factor in mouse solid tumor models. Cancer Immunol Immunother 2017; 66:181-192. [PMID: 27872951 PMCID: PMC11028649 DOI: 10.1007/s00262-016-1928-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2016] [Accepted: 11/07/2016] [Indexed: 11/27/2022]
Abstract
Vascular endothelial growth factor (VEGF) plays an important role in the progression of various cancers. The VEGF-specific antibody bevacizumab combined with chemotherapy was shown to significantly improve progression-free survival in certain cancers. However, repeated administration is necessary for effective suppression of VEGF, thereby making the therapy expensive and cumbersome. Thus, it is urgent to develop alternative reagents such as VEGF vaccines. Here we report that DTT-VEGF, a VEGF-based antigen consisting of the receptor-binding domain of VEGF and diphtheria toxin T domain (DTT), not only stimulated neutralizing antibody response, but also induced type 1 immune response as well as anti-tumor cytotoxic T lymphocytes in mice when administered with aluminum hydroxide adjuvant. The antibodies triggered by DTT-VEGF immunization inhibited the binding of VEGF to VEGF receptor and downregulated the serum VEGF levels in tumor-bearing mice. VEGF-specific IgG2a and IgG2b antibodies as well as type 1 cytokines were stimulated by DTT-VEGF vaccination. The splenocytes from DTT-VEGF-immunized mice showed cytotoxic activity against B16-F10 cells expressing VEGF. Extensive necrosis with severe hemorrhage and enhanced CD8+ T cell infiltration were observed in tumors from DTT-VEGF-immunized mice. The percentages of CD31+ vascular areas in the tumor sections from DTT-VEGF-immunized mice were significantly lower than those of control mice. DTT-VEGF significantly inhibited tumor growth in preventive and therapeutic vaccination settings in mouse models. Our data suggest that DTT is an effective antigen carrier to break immune self-tolerance and our vaccine design has potential to be used for human cancer therapy.
Collapse
Affiliation(s)
- Aizhang Xu
- State Key Laboratory of Microbial Metabolism, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Li Zhang
- State Key Laboratory of Microbial Metabolism, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Yangyang Chen
- State Key Laboratory of Microbial Metabolism, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Zhibing Lin
- State Key Laboratory of Microbial Metabolism, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Rongxiu Li
- State Key Laboratory of Microbial Metabolism, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China.
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China.
- Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
38
|
Church SE, Galon J. Regulation of CTL Infiltration Within the Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1036:33-49. [PMID: 29275463 DOI: 10.1007/978-3-319-67577-0_3] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The tumor microenvironment consists of a complex milieu of cells and factors that maintain equilibrium between tumor progression and destruction. Characterization of the immune contexture in primary tumors has consistently shown that T lymphocytes are an integral predictor of improved clinical outcome. This is notably true in colorectal carcinoma where high densities of cytotoxic or memory T lymphocytes in the invasive margin and the center of the primary tumor predict better patient survival, a measure termed Immunoscore. Since a high Immunoscore and pre-existing adaptive immune response are significantly correlated with improved clinical outcome, it is essential to understand the mechanisms underlying functional T lymphocyte infiltration into the tumor. The ability of cytolytic and memory T lymphocytes to migrate into tumors is regulated by multiple strategies including T lymphocyte help, homing factors, cytokines, tumor genotype, angiogenesis, lymphangiogenesis, and neurological signals. This chapter will discuss the predominant factors that mediate T-lymphocyte infiltration into tumors and how analysis of these biomarkers determine patients' disease-related survival and predicts response to cancer therapy.
Collapse
Affiliation(s)
- Sarah E Church
- Laboratory of Integrative Cancer Immunology, INSERM, UMRS1138, 15 Rue de l'Ecole de Medecine, Paris, France.
- Université Paris Descartes, Paris, France.
- Cordeliers Research Centre, Université Pierre et Marie Curie Paris 6, Paris, France.
| | - Jérôme Galon
- Laboratory of Integrative Cancer Immunology, INSERM, UMRS1138, 15 Rue de l'Ecole de Medecine, Paris, France
- Université Paris Descartes, Paris, France
- Cordeliers Research Centre, Université Pierre et Marie Curie Paris 6, Paris, France
| |
Collapse
|
39
|
Hendry SA, Farnsworth RH, Solomon B, Achen MG, Stacker SA, Fox SB. The Role of the Tumor Vasculature in the Host Immune Response: Implications for Therapeutic Strategies Targeting the Tumor Microenvironment. Front Immunol 2016; 7:621. [PMID: 28066431 PMCID: PMC5168440 DOI: 10.3389/fimmu.2016.00621] [Citation(s) in RCA: 131] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Accepted: 12/07/2016] [Indexed: 12/22/2022] Open
Abstract
Recently developed cancer immunotherapy approaches including immune checkpoint inhibitors and chimeric antigen receptor T cell transfer are showing promising results both in trials and in clinical practice. These approaches reflect increasing recognition of the crucial role of the tumor microenvironment in cancer development and progression. Cancer cells do not act alone, but develop a complex relationship with the environment in which they reside. The host immune response to tumors is critical to the success of immunotherapy; however, the determinants of this response are incompletely understood. The immune cell infiltrate in tumors varies widely in density, composition, and clinical significance. The tumor vasculature is a key component of the microenvironment that can influence tumor behavior and treatment response and can be targeted through the use of antiangiogenic drugs. Blood vascular and lymphatic endothelial cells have important roles in the trafficking of immune cells, controlling the microenvironment, and modulating the immune response. Improving access to the tumor through vascular alteration with antiangiogenic drugs may prove an effective combinatorial strategy with immunotherapy approaches and might be applicable to many tumor types. In this review, we briefly discuss the host's immune response to cancer and the treatment strategies utilizing this response, before focusing on the pathological features of tumor blood and lymphatic vessels and the contribution these might make to tumor immune evasion.
Collapse
Affiliation(s)
- Shona A Hendry
- Department of Pathology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia; The Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, Australia
| | - Rae H Farnsworth
- Tumour Angiogenesis and Microenvironment Program, Peter MacCallum Cancer Centre , Melbourne, VIC , Australia
| | - Benjamin Solomon
- Department of Medical Oncology, Peter MacCallum Cancer Centre , Melbourne, VIC , Australia
| | - Marc G Achen
- The Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, Australia; Tumour Angiogenesis and Microenvironment Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Steven A Stacker
- The Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, Australia; Tumour Angiogenesis and Microenvironment Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Stephen B Fox
- Department of Pathology, Peter MacCallum Cancer Centre , Melbourne, VIC , Australia
| |
Collapse
|
40
|
Liang J, Liu X, Xie Q, Chen G, Li X, Jia Y, Yin B, Qu X, Li Y. Endostatin enhances antitumor effect of tumor antigen-pulsed dendritic cell therapy in mouse xenograft model of lung carcinoma. Chin J Cancer Res 2016; 28:452-60. [PMID: 27647974 PMCID: PMC5018541 DOI: 10.21147/j.issn.1000-9604.2016.04.09] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Objective To investigate the antitumor effect of endostatin combined with tumor antigen-pulsed dendritic cell (DC)-T cell therapy on lung cancer. Methods Transplanted Lewis lung cancer (LLC) models of C57BL/6 mice were established by subcutaneous injection of LLC cells in left extremity axillary. Tumor antigen-pulsed DC-T cells from spleen cells and bone of mice were cultured in vitro. Tumor-bearing mice were randomly divided into three groups, including DC-T+endostatin group, DC-T group, and phosphate-buffered saline (PBS) control group. Microvessel density (MVD) of tumor tissue in tumor-bearing mice was determined by immunohistochemistry (IHC). The expressions of vascular endothelial growth factor (VEGF) and hypoxia-inducible factor-1α (HIF-1α) were determined by Western blotting and IHC staining. The proportions of CD8+ T cells, mature dendritic cells (mDC), tumor-associated macrophages [TAM (M1/M2)], and myeloid-derived suppressor cells (MDSC) in suspended cells of tumor tissue were determined by flow cytometry. The expressions of interleukin (IL)-6, IL-10, IL-17, transforming growth factor-β (TGF-β) and interferon-γ (IFN-γ) in suspended cells of tumor tissue were detected by enzyme-linked immune sorbent assay (ELISA). Results DC-T cells combined with endostatin remarkably suppressed tumor growth. MVD of mice in DC-T+endostatin group was significantly lower than that of the control group and DC-T monotherapy group. The expressions of VEGF, IL-6 and IL-17 in tumors were markedly decreased, but IFN-γ and HIF-1α increased after treating with DC-T cells combined with endostatin, compared to control group and DC-T group. In the DC-T+endostatin group, the proportions of MDSC and TAM (M2 type) were significantly decreased, mDC and TAM (M1 type) were up-regulated, and CD8+ T cells were recruited to infiltrate tumors, in contrast to PBS control and DC-T monotherapy. DC-T cells combined with endostatin potently reduced the expressions of IL-6, IL-10, TGF-β and IL-17 in tumor tissue, and enhanced the expression of IFN-γ. Conclusions The study indicated the synergic antitumor effects between endostatin and tumor antigen-pulsed DC-T cells, which may be a prospective therapy strategy to achieve potent antitumor effects on lung cancer.
Collapse
Affiliation(s)
| | | | - Qi Xie
- Central Laboratory, Qianfoshan Hospital, Shandong University, Jinan 250014, China
| | - Guoling Chen
- Islet Cell Lab, MedStar Georgetown University Hospital, Washington DC 20007, USA
| | | | | | - Beibei Yin
- Central Laboratory, Qianfoshan Hospital, Shandong University, Jinan 250014, China
| | - Xun Qu
- Institute of Basic Medical Sciences and Key Laboratory of Cardiovascular Proteomics of Shandong Province, Qilu Hospital, Shandong University, Jinan 250012, China
| | | |
Collapse
|
41
|
Reardon DA, Gilbert MR, Wick W, Liau L. Immunotherapy for neuro-oncology: the critical rationale for combinatorial therapy. Neuro Oncol 2016; 17 Suppl 7:vii32-vii40. [PMID: 26516225 DOI: 10.1093/neuonc/nov178] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
A successful therapeutic paradigm established historically in oncology involves combining agents with potentially complementary mechanisms of antitumor activity into rationally designed regimens. For example, cocktails of cytotoxic agents, which were carefully designed based on mechanisms of action, dose, and scheduling considerations, have led to dramatic improvements in survival including cures for childhood leukemia, Hodgkin's lymphoma, and several other complex cancers. Outcome for glioblastoma, the most common primary malignant CNS cancer, has been more modest, but nonetheless our current standard of care derives from confirmation that combination therapy surpasses single modality therapy. Immunotherapy has recently come of age for medical oncology with exciting therapeutic benefits achieved by several types of agents including vaccines, adoptive T cells, and immune checkpoint inhibitors against several types of cancers. Nonetheless, most benefits are relatively short, while others are durable but are limited to a minority of treated patients. Critical factors limiting efficacy of immunotherapeutics include insufficient immunogenicity and/or inadequate ability to overcome immunosuppressive factors exploited by tumors. The paradigm of rationally designed combinatorial regimens, originally established by cytotoxic therapy for oncology, may also prove relevant for immunotherapy. Realization of the true therapeutic potential of immunotherapy for medical oncology and neuro-oncology patients may require development of combinatorial regimens that optimize immunogenicity and target tumor adaptive immunosuppressive factors.
Collapse
Affiliation(s)
- David A Reardon
- Center of Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R.); Department of Medical Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R.); Neurology Clinic and National Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (M.R.G.); Neurology Clinic and National Center for Tumor Diseases, University of Heidelberg and German Consortium for Translational Cancer Research (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany (W.W); Brain Tumor Program, Department of Neurosurgery, University of California Los Angeles, David Geffen School of Medicine at UCLA, Los Angeles, California (L.L.)
| | - Mark R Gilbert
- Center of Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R.); Department of Medical Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R.); Neurology Clinic and National Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (M.R.G.); Neurology Clinic and National Center for Tumor Diseases, University of Heidelberg and German Consortium for Translational Cancer Research (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany (W.W); Brain Tumor Program, Department of Neurosurgery, University of California Los Angeles, David Geffen School of Medicine at UCLA, Los Angeles, California (L.L.)
| | - Wolfgang Wick
- Center of Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R.); Department of Medical Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R.); Neurology Clinic and National Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (M.R.G.); Neurology Clinic and National Center for Tumor Diseases, University of Heidelberg and German Consortium for Translational Cancer Research (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany (W.W); Brain Tumor Program, Department of Neurosurgery, University of California Los Angeles, David Geffen School of Medicine at UCLA, Los Angeles, California (L.L.)
| | - Linda Liau
- Center of Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R.); Department of Medical Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R.); Neurology Clinic and National Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (M.R.G.); Neurology Clinic and National Center for Tumor Diseases, University of Heidelberg and German Consortium for Translational Cancer Research (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany (W.W); Brain Tumor Program, Department of Neurosurgery, University of California Los Angeles, David Geffen School of Medicine at UCLA, Los Angeles, California (L.L.)
| |
Collapse
|
42
|
Mao X, Xu J, Cui H. Functional nanoparticles for magnetic resonance imaging. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2016; 8:814-841. [PMID: 27040463 DOI: 10.1002/wnan.1400] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Revised: 02/01/2016] [Accepted: 02/15/2016] [Indexed: 12/16/2022]
Abstract
Nanoparticle-based magnetic resonance imaging (MRI) contrast agents have received much attention over the past decade. By virtue of a high payload of magnetic moieties, enhanced accumulation at disease sites, and a large surface area for additional modification with targeting ligands, nanoparticle-based contrast agents offer promising new platforms to further enhance the high resolution and sensitivity of MRI for various biomedical applications. T 2 * superparamagnetic iron oxide nanoparticles (SPIONs) first demonstrated superior improvement on MRI sensitivity. The prevailing SPION attracted growing interest in the development of refined nanoscale versions of MRI contrast agents. Afterwards, T 1 -based contrast agents were developed, and became the most studied subject in MRI due to the positive contrast they provide that avoids the susceptibility associated with MRI signal reduction. Recently, chemical exchange saturation transfer (CEST) contrast agents have emerged and rapidly gained popularity. The unique aspect of CEST contrast agents is that their contrast can be selectively turned 'on' and 'off' by radiofrequency saturation. Their performance can be further enhanced by incorporating a large number of exchangeable protons into well-defined nanostructures. Besides activatable CEST contrast agents, there is growing interest in developing nanoparticle-based activatable MRI contrast agents responsive to stimuli (pH, enzyme, etc.), which improves sensitivity and specificity. In this review, we summarize the recent development of various types of nanoparticle-based MRI contrast agents, and have focused our discussions on the key advantages of introducing nanoparticles in MRI. WIREs Nanomed Nanobiotechnol 2016, 8:814-841. doi: 10.1002/wnan.1400 For further resources related to this article, please visit the WIREs website.
Collapse
Affiliation(s)
- Xinpei Mao
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD, USA.,Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, MD, USA
| | - Jiadi Xu
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,F. M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Research Institute, Baltimore, MD, USA
| | - Honggang Cui
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD, USA. .,Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, MD, USA. .,Department of Oncology and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA. .,Center for Nanomedicine, The Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
43
|
Wagner SC, Ichim TE, Ma H, Szymanski J, Perez JA, Lopez J, Bogin V, Patel AN, Marincola FM, Kesari S. Cancer anti-angiogenesis vaccines: Is the tumor vasculature antigenically unique? J Transl Med 2015; 13:340. [PMID: 26510973 PMCID: PMC4625691 DOI: 10.1186/s12967-015-0688-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2015] [Accepted: 10/03/2015] [Indexed: 12/19/2022] Open
Abstract
Angiogenesis is essential for the growth and metastasis of solid tumors. The tumor endothelium exists in a state of chronic activation and proliferation, fueled by the tumor milieu where angiogenic mediators are aberrantly over-expressed. Uncontrolled tumor growth, immune evasion, and therapeutic resistance are all driven by the dysregulated and constitutive angiogenesis occurring in the vasculature. Accordingly, great efforts have been dedicated toward identifying molecular signatures of this pathological angiogenesis in order to devise selective tumor endothelium targeting therapies while minimizing potential autoimmunity against physiologically normal endothelium. Vaccination with angiogenic antigens to generate cellular and/or humoral immunity against the tumor endothelium has proven to be a promising strategy for inhibiting or normalizing tumor angiogenesis and reducing cancer growth. Here we review tumor endothelium vaccines developed to date including active immunization strategies using specific tumor endothelium-associated antigens and whole endothelial cell-based vaccines designed to elicit immune responses against diverse target antigens. Among the novel therapeutic options, we describe a placenta-derived endothelial cell vaccine, ValloVax™, a polyvalent vaccine that is antigenically similar to proliferating tumor endothelium and is supported by pre-clinical studies to be safe and efficacious against several tumor types.
Collapse
Affiliation(s)
- Samuel C Wagner
- Batu Biologics Inc., Towne Center Drive, San Diego, CA, 92121, USA.
| | - Thomas E Ichim
- Batu Biologics Inc., Towne Center Drive, San Diego, CA, 92121, USA.
| | - Hong Ma
- Batu Biologics Inc., Towne Center Drive, San Diego, CA, 92121, USA.
| | - Julia Szymanski
- Batu Biologics Inc., Towne Center Drive, San Diego, CA, 92121, USA.
| | | | - Javier Lopez
- Pan Am Cancer Treatment Center, Tijuana, Mexico.
| | - Vladimir Bogin
- Batu Biologics Inc., Towne Center Drive, San Diego, CA, 92121, USA.
| | - Amit N Patel
- Department of Surgery, University of Utah, Salt Lake City, UT, USA.
| | | | | |
Collapse
|
44
|
Shi S, Tao L, Song H, Chen L, Huang G. Synergistic antitumor effect of combining metronomic chemotherapy with adoptive cell immunotherapy in nude mice. APMIS 2014; 122:380-91. [PMID: 24628659 DOI: 10.1111/apm.12235] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Accepted: 11/18/2013] [Indexed: 12/30/2022]
Abstract
Adoptive cell immunotherapy with cytokine-induced killer cell (CIK cell) represents a promising non-toxic anticancer therapy. However, the clinical efficacy of CIK cells is limited because of abnormal tumor vasculature. Metronomic chemotherapy shows promising anticancer activity by its potential antiangiogenic effect and reduced toxicity. We hypothesized that metronomic chemotherapy with paclitaxel could improve the antitumor effect of adoptive CIK cell immunotherapy. Mice health status was analyzed by measuring mice weight and observing mice behavior. Immunohistochemistry was used to investigate the recruitment of CIK cells, the expression of endothelial cell molecules, as well as the hypoxic tumor area. Metronomic paclitaxel synergized with adoptive CIK cell immunotherapy to inhibit the growth of non-small cell lung cancer (NSCLC). Metronomic paclitaxel reduced hypoxic tumor area and increased CIK cell infiltration. Hypoxia impeded the adhesion of CIK cells and reduced the expression of endothelial cell adhesion molecules. In vivo studies demonstrated that more CIK cells were found in endothelial cell adhesion molecules high expressed area. Our study provides a new rationale for combining metronomic chemotherapy with adoptive cell immunotherapy in the treatment of xenograft NSCLC tumors in immunodeficient mice. Further clinical trials integrating translational research are necessary to better evaluate the clinical benefit of this promising approach.
Collapse
Affiliation(s)
- Shujing Shi
- Medical Oncology Department of Jinling Hospital, Medical school of Nanjing University, Nanjing, China
| | | | | | | | | |
Collapse
|
45
|
Reardon DA, Wucherpfennig KW, Freeman G, Wu CJ, Chiocca EA, Wen PY, Curry WT, Mitchell DA, Fecci PE, Sampson JH, Dranoff G. An update on vaccine therapy and other immunotherapeutic approaches for glioblastoma. Expert Rev Vaccines 2013; 12:597-615. [PMID: 23750791 DOI: 10.1586/erv.13.41] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Outcome for glioblastoma (GBM), the most common primary CNS malignancy, remains poor. The overall survival benefit recently achieved with immunotherapeutics for melanoma and prostate cancer support evaluation of immunotherapies for other challenging cancers, including GBM. Much historical dogma depicting the CNS as immunoprivileged has been replaced by data demonstrating CNS immunocompetence and active interaction with the peripheral immune system. Several glioma antigens have been identified for potential immunotherapeutic exploitation. Active immunotherapy studies for GBM, supported by preclinical data, have focused on tumor lysate and synthetic antigen vaccination strategies. Results to date confirm consistent safety, including a lack of autoimmune reactivity; however, modest efficacy and variable immunogenicity have been observed. These findings underscore the need to optimize vaccination variables and to address challenges posed by systemic and local immunosuppression inherent to GBM tumors. Additional immunotherapy strategies are also in development for GBM. Future studies may consider combinatorial immunotherapy strategies with complimentary actions.
Collapse
Affiliation(s)
- David A Reardon
- Center for Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, MA, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Vascular remodeling in cancer. Oncogene 2013; 33:3496-505. [PMID: 23912450 DOI: 10.1038/onc.2013.304] [Citation(s) in RCA: 121] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2013] [Revised: 05/31/2013] [Accepted: 06/10/2013] [Indexed: 12/17/2022]
Abstract
The growth and dissemination of tumors rely on an altered vascular network, which supports their survival and expansion and provides accessibility to the vasculature and a route of transport for metastasizing tumor cells. The remodeling of vascular structures through generation of new vessels (for example, via tumor angiogenesis) is a well studied, even if still quite poorly understood, process in human cancer. Antiangiogenic therapies have provided insight into the contribution of angiogenesis to the biology of human tumors, yet have also revealed the ease with which resistance to antiangiogenic drugs can develop, presumably involving alterations to vascular signaling mechanisms. Furthermore, cellular and/or molecular changes to pre-existing vessels could represent subtle pre-metastatic alterations to the vasculature, which are important for cancer progression. These changes, and associated molecular markers, may forecast the behavior of individual tumors and contribute to the early detection, diagnosis and prognosis of cancer. This review, which primarily focuses on the blood vasculature, explores current knowledge of how tumor vessels can be remodeled, and the cellular and molecular events responsible for this process.
Collapse
|
47
|
Mikucki ME, Fisher DT, Ku AW, Appenheimer MM, Muhitch JB, Evans SS. Preconditioning thermal therapy: flipping the switch on IL-6 for anti-tumour immunity. Int J Hyperthermia 2013; 29:464-73. [PMID: 23862980 DOI: 10.3109/02656736.2013.807440] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Cancer immunotherapy aims to generate long-lived, tumour-specific adaptive immunity to limit dysregulated tumour progression and metastasis. Tumour vasculature has emerged as a critical checkpoint controlling the efficacy of immunotherapy since it is the main access point for cytotoxic T cells to reach tumour cell targets. Therapeutic success has been particularly challenging to achieve because of the local, cytokine-rich inflammatory milieu that drives a pro-tumourigenic programme supporting the growth and survival of malignant cells. Here, we focus on recent evidence that systemic thermal therapy can switch the activities of the inflammatory cytokine, interleukin-6 (IL-6), to a predominantly anti-tumourigenic function that promotes anti-tumour immunity by mobilising T cell trafficking in the recalcitrant tumour microenvironment.
Collapse
Affiliation(s)
- Maryann E Mikucki
- Department of Immunology, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| | | | | | | | | | | |
Collapse
|
48
|
Shi S, Wang R, Chen Y, Song H, Chen L, Huang G. Combining antiangiogenic therapy with adoptive cell immunotherapy exerts better antitumor effects in non-small cell lung cancer models. PLoS One 2013; 8:e65757. [PMID: 23799045 PMCID: PMC3683034 DOI: 10.1371/journal.pone.0065757] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2012] [Accepted: 04/29/2013] [Indexed: 12/14/2022] Open
Abstract
Introduction Cytokine-induced killer cells (CIK cells) are a heterogeneous subset of ex-vivo expanded T lymphocytes which are characterized with a MHC-unrestricted tumor-killing activity and a mixed T-NK phenotype. Adoptive CIK cells transfer, one of the adoptive immunotherapy represents a promising nontoxic anticancer therapy. However, in clinical studies, the therapeutic activity of adoptive CIK cells transfer is not as efficient as anticipated. Possible explanations are that abnormal tumor vasculature and hypoxic tumor microenvironment could impede the infiltration and efficacy of lymphocytes. We hypothesized that antiangiogenesis therapy could improve the antitumor activity of CIK cells by normalizing tumor vasculature and modulating hypoxic tumor microenvironment. Methods We combined recombinant human endostatin (rh-endostatin) and CIK cells in the treatment of lung carcinoma murine models. Intravital microscopy, dynamic contrast enhanced magnetic resonance imaging, immunohistochemistry, and flow cytometry were used to investigate the tumor vasculature and hypoxic microenvironment as well as the infiltration of immune cells. Results Our results indicated that rh-endostatin synergized with adoptive CIK cells transfer to inhibit the growth of lung carcinoma. We found that rh-endostatin normalized tumor vasculature and reduced hypoxic area in the tumor microenvironment. Hypoxia significantly inhibited the proliferation, cytotoxicity and migration of CIK cells in vitro and impeded the homing of CIK cells into tumor parenchyma ex vivo. Furthermore, we found that treatment with rh-endostatin significantly increased the homing of CIK cells and decreased the accumulation of suppressive immune cells in the tumor tissue. In addition, combination therapy produced higher level of tumor-infiltration lymphocytes compared with other treatments. Conclusions Our results demonstrate that rh-endostatin improves the therapeutic effect of adoptive CIK cells therapy against lung carcinomas and unmask the mechanisms of the synergistic antitumor efficacy, providing a new rationale for combining antiangiogenesis therapy with immunotherapy in the treatment of lung cancer.
Collapse
Affiliation(s)
- Shujing Shi
- Medical Oncology Department of Jinling Hospital, Medical school of Nanjing University, Nanjing, People’s Republic of China
| | - Rui Wang
- Medical Oncology Department of Jinling Hospital, Medical school of Nanjing University, Nanjing, People’s Republic of China
| | - Yitian Chen
- Medical Oncology Department of Jinling Hospital, Medical school of Nanjing University, Nanjing, People’s Republic of China
| | - Haizhu Song
- Medical Oncology Department of Jinling Hospital, Medical school of Nanjing University, Nanjing, People’s Republic of China
| | - Longbang Chen
- Medical Oncology Department of Jinling Hospital, Medical school of Nanjing University, Nanjing, People’s Republic of China
- * E-mail: (GCH); (LBC)
| | - Guichun Huang
- Medical Oncology Department of Jinling Hospital, Medical school of Nanjing University, Nanjing, People’s Republic of China
- * E-mail: (GCH); (LBC)
| |
Collapse
|
49
|
Salnikova O, Breuhahn K, Hartmann N, Schmidt J, Ryschich E. Endothelial plasticity governs the site-specific leukocyte recruitment in hepatocellular cancer. Int J Cancer 2013; 133:2372-82. [PMID: 23661267 DOI: 10.1002/ijc.28268] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2013] [Accepted: 04/23/2013] [Indexed: 12/12/2022]
Abstract
The correct programming of the endothelial cell phenotype is crucial for efficient leukocyte recruitment to tumor tissue. It has been previously described that T cells infiltrated hepatocellular cancer (HCC) tissue mainly in peritumoral, stromal and tumor border areas. In the current study, phenotype features of tumor endothelial cells and their potential impact on leukocyte recruitment were analyzed in murine tissue of HCC. In the murine model, proinflammatory stimulation with IL-1β induced leukocyte recruitment in the blood vessels of peripheral tumor areas and in nonmalignant liver tissue, but not in deeper tumor blood vessels. Furthermore, peripheral tumor endothelium, but not deeper tumor blood vessels exhibited a "normalized" hepatic sinusoidal endothelial cell (HSEC)-like phenotype with regard to the expression of adhesion molecules and liver sinusoidal endothelial markers. When tumor endothelial cells were isolated and incubated in vitro, their phenotype rapidly changed and became almost identical to normal hepatic endothelial cells. Interestingly, cytokine production in HCC was strongly dysregulated as compared to normal liver, with IL-1RN exhibiting the most prominent elevation. Experiments with isolated hepatic endothelial cells showed that IL-1RN effectively antagonized the activating action of IL-1β on the expression of adhesion molecules and T cell attachment. These novel insights indicate that tumor endothelium of HCC represents a plastic system that is susceptible to microenvironmental changes. The peritumoral and tumor border areas have distinct endothelial cell phenotype, which promotes leukocyte recruitment to HCC tissue.
Collapse
Affiliation(s)
- Olga Salnikova
- Department of Surgery, University Hospital of Heidelberg, Heidelberg, Germany
| | | | | | | | | |
Collapse
|
50
|
Chinnasamy D, Tran E, Yu Z, Morgan RA, Restifo NP, Rosenberg SA. Simultaneous targeting of tumor antigens and the tumor vasculature using T lymphocyte transfer synergize to induce regression of established tumors in mice. Cancer Res 2013; 73:3371-80. [PMID: 23633494 PMCID: PMC3686092 DOI: 10.1158/0008-5472.can-12-3913] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Most systemic cancer therapies target tumor cells directly, although there is increasing interest in targeting the tumor stroma that can comprise a substantial portion of the tumor mass. We report here a synergy between two T-cell therapies, one directed against the stromal tumor vasculature and the other directed against antigens expressed on the tumor cell. Simultaneous transfer of genetically engineered syngeneic T cells expressing a chimeric antigen receptor targeting the VEGF receptor-2 (VEGFR2; KDR) that is overexpressed on tumor vasculature and T-cells specific for the tumor antigens gp100 (PMEL), TRP-1 (TYRP1), or TRP-2 (DCT) synergistically eradicated established B16 melanoma tumors in mice and dramatically increased the tumor-free survival of mice compared with treatment with either cell type alone or T cells coexpressing these two targeting molecules. Host lymphodepletion before cell transfer was required to mediate the antitumor effect. The synergistic antitumor response was accompanied by a significant increase in the infiltration and expansion and/or persistence of the adoptively transferred tumor antigen-specific T cells in the tumor microenvironment and thus enhanced their antitumor potency. The data presented here emphasize the possible beneficial effects of combining antiangiogenic with tumor-specific immunotherapeutic approaches for the treatment of patients with cancer.
Collapse
MESH Headings
- Animals
- Antigens, Neoplasm/immunology
- Disease Models, Animal
- Immunotherapy, Adoptive/methods
- Lymphocyte Activation/immunology
- Melanoma, Experimental/blood supply
- Melanoma, Experimental/immunology
- Melanoma, Experimental/therapy
- Mice
- Mice, Inbred C57BL
- Neovascularization, Pathologic/immunology
- Neovascularization, Pathologic/therapy
- Receptors, Antigen/genetics
- Receptors, Antigen/immunology
- Recombinant Fusion Proteins/genetics
- Recombinant Fusion Proteins/immunology
- T-Lymphocytes/immunology
- Vascular Endothelial Growth Factor Receptor-2/immunology
Collapse
Affiliation(s)
- Dhanalakshmi Chinnasamy
- Surgery Branch, National Cancer Institute, Clinical Research Center, Bethesda, MD 20892, USA
| | | | | | | | | | | |
Collapse
|