1
|
Lee YT, Wu SH, Wu CH, Lin YH, Lin CK, Chen ZA, Sun TC, Chen YJ, Chen P, Mou CY, Chen YP. Drug-Free Mesoporous Silica Nanoparticles Enable Suppression of Cancer Metastasis and Confer Survival Advantages to Mice with Tumor Xenografts. ACS APPLIED MATERIALS & INTERFACES 2024. [PMID: 39448366 DOI: 10.1021/acsami.4c16609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/26/2024]
Abstract
Despite advancements in nanomedicine for drug delivery, many drug-loaded nanoparticles reduce tumor sizes but often fail to prevent metastasis. Mesoporous silica nanoparticles (MSNs) have attracted attention as promising nanocarriers. Here, we demonstrated that MSN-PEG/TA 25, with proper surface modifications, exhibited unique antimetastatic properties. In vivo studies showed that overall tumor metastasis decreased in 4T1 xenografts mice treated with MSN-PEG/TA 25 with a notable reduction in lung tumor metastasis. In vitro assays, including wound-healing, Boyden chamber, tube-formation, and real-time cell analyses, showed that MSN-PEG/TA 25 could modulate cell migration of 4T1 breast cancer cells and interrupt tube formation by human umbilical vein endothelial cells (HUVECs), key factors in suppressing cancer metastasis. The synergistic effect of MSN-PEG/TA 25 combined with liposomal-encapsulated doxorubicin (Lipo-Dox) significantly boosted mouse survival rates, outperforming Lipo-Dox monotherapy. We attributed the improved survival to the antimetastatic capabilities of MSN-PEG/TA 25. Moreover, Dox-loaded MSN-PEG/TA 25 suppressed primary tumors while retaining the antimetastatic effect, thereby enhancing therapeutic outcomes and overall survival. Western blot and qPCR analyses revealed that MSN-PEG/TA 25 interfered with the phosphorylation of ERK, FAK, and paxillin, thus impacting focal adhesion turnover and inhibiting cell motility. Our findings suggest that drug-free MSN-PEG/TA 25 is highly efficient for cancer treatment via suppressing metastatic activity and angiogenesis.
Collapse
Affiliation(s)
- Yu-Tse Lee
- Department of Chemistry, National Taiwan University, Taipei 10617, Taiwan
| | - Si-Han Wu
- Graduate Institute of Nanomedicine and Medical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan
- International Ph.D. Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan
| | - Cheng-Hsun Wu
- Nano Targeting & Therapy Biopharma Inc., Taipei 10087, Taiwan
| | - Yu-Han Lin
- Graduate Institute of Nanomedicine and Medical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan
| | - Cong-Kai Lin
- Graduate Institute of Biomedical Materials & Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan
| | - Zih-An Chen
- Graduate Institute of Nanomedicine and Medical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan
- Research Center for Applied Sciences, Academia Sinica, Taipei 11529, Taiwan
| | - Ting-Chung Sun
- Nano Targeting & Therapy Biopharma Inc., Taipei 10087, Taiwan
| | - Yin-Ju Chen
- Graduate Institute of Biomedical Materials & Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan
| | - Peilin Chen
- Research Center for Applied Sciences, Academia Sinica, Taipei 11529, Taiwan
| | - Chung-Yuan Mou
- Department of Chemistry, National Taiwan University, Taipei 10617, Taiwan
- Graduate Institute of Nanomedicine and Medical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan
| | - Yi-Ping Chen
- Graduate Institute of Nanomedicine and Medical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan
- International Ph.D. Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan
| |
Collapse
|
2
|
Zhang Q, Han X, Bi Z, Yang M, Lin J, Li Z, Zhang M, Bu B. Exhausted signature and regulatory network of NK cells in myasthenia gravis. Front Immunol 2024; 15:1397916. [PMID: 39346912 PMCID: PMC11427316 DOI: 10.3389/fimmu.2024.1397916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 08/27/2024] [Indexed: 10/01/2024] Open
Abstract
Introduction NK cells are dysfunctional in myasthenia gravis (MG), but the mechanism is unclear. This study aims to measure associations and underlying mechanisms between the NK cells and the development of MG. Methods Twenty healthy controls (HCs) and 53 MG patients who did not receive glucocorticoids and immunosuppressants were collected. According to the Myasthenia Gravis Foundation of America (MGFA) classification, MG patients were categorized into MGFA I group (n = 18) and MGFA II-IV group (n = 35). Flow cytometry, cell sorting, ELISA, mRNA-sequencing, RT-qPCR, western blot, and cell culture experiments were performed to evaluate the regulatory mechanism of exhausted NK cells. Results Peripheral NK cells in MGFA II-IV patients exhibit exhausted phenotypes than HCs, marked by the dramatic loss of total NK cells, CD56dimCD16- NK cells, elevated PD1 expression, reduced NKG2D expression, impaired cytotoxic activity (perforin, granzyme B, CD107a) and cytokine secretion (IFN-γ). Plasma IL-6 and IL-21 are elevated in MG patients and mainly derived from the aberrant expansion of monocytes and Tfh cells, respectively. IL-6/IL-21 cooperatively induced NK-cell exhausted signature via upregulating SOCS2 and inhibiting the phosphorylation of STAT5. SOCS2 siRNA and IL-2 supplement attenuated the IL-6/IL-21-mediated alteration of NK-cell phenotypes and function. Discussion Inhibition of IL-6/IL-21/SOCS2/STAT5 pathway and recovery of NK-cell ability to inhibit autoimmunity may be a new direction in the treatment of MG.
Collapse
Affiliation(s)
- Qing Zhang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, China
| | - Xingyu Han
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, China
| | - Zhuajin Bi
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, China
| | - Mengge Yang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, China
| | - Jing Lin
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, China
| | - Zhijun Li
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, China
| | - Min Zhang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, China
| | - Bitao Bu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
3
|
Isvoranu G, Chiritoiu-Butnaru M. Therapeutic potential of interleukin-21 in cancer. Front Immunol 2024; 15:1369743. [PMID: 38638431 PMCID: PMC11024325 DOI: 10.3389/fimmu.2024.1369743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 03/12/2024] [Indexed: 04/20/2024] Open
Abstract
Interleukin-21 (IL-21) is an immunostimulatory cytokine which belongs to the common gamma-chain family of cytokines. It plays an import role in the development, differentiation, proliferation, and activation of immune cells, in particular T and natural killer (NK) cells. Since its discovery in 2000, IL-21 has been shown to regulate both adaptive and immune responses associates with key role in antiviral and antitumor responses. Recent advances indicate IL-21 as a promising target for cancer treatment and encouraging results were obtained in preclinical studies which investigated the potency of IL-21 alone or in combination with other therapies, including monoclonal antibodies, checkpoint inhibitory molecules, oncolytic virotherapy, and adoptive cell transfer. Furthermore, IL-21 showed antitumor effects in the treatment of patients with advanced cancer, with minimal side effects in several clinical trials. In the present review, we will outline the recent progress in IL-21 research, highlighting the potential of IL-21 based therapy as single agent or in combination with other drugs to enhance cancer treatment efficiency.
Collapse
Affiliation(s)
- Gheorghita Isvoranu
- Department of Animal Husbandry,” Victor Babeș” National Institute of Pathology, Bucharest, Romania
| | - Marioara Chiritoiu-Butnaru
- Department of Molecular and Cell Biology, Institute of Biochemistry of the Romanian Academy, Bucharest, Romania
| |
Collapse
|
4
|
Pastwińska J, Karwaciak I, Karaś K, Bachorz RA, Ratajewski M. RORγT agonists as immune modulators in anticancer therapy. Biochim Biophys Acta Rev Cancer 2023; 1878:189021. [PMID: 37951483 DOI: 10.1016/j.bbcan.2023.189021] [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: 08/01/2023] [Revised: 10/26/2023] [Accepted: 11/04/2023] [Indexed: 11/14/2023]
Abstract
RORγT is a transcription factor that directs the development of Th17 lymphocytes and other IL-17-expressing cells (e.g., Tc17 and ILC3 cells). These cells are involved in the body's defense against pathogenic bacteria and fungi, but they also participate in maintaining the proinflammatory environment in some autoimmune diseases and play a role in the immune system's response to cancer. Similar to other members of the nuclear receptor superfamily, the activity of RORγT is regulated by low-molecular-weight ligands. Therefore, extensive efforts have been dedicated to identifying inverse agonists that diminish the activity of this receptor and subsequently inhibit the development of autoimmune diseases. Unfortunately, in the pursuit of an ideal inverse agonist, the development of agonists has been overlooked. It is important to remember that these types of compounds, by stimulating lymphocytes expressing RORγT (Th17 and Tc17), can enhance the immune system's response to tumors. In this review, we present recent advancements in the biology of RORγT agonists and their potential application in anticancer therapy.
Collapse
Affiliation(s)
- Joanna Pastwińska
- Laboratory of Epigenetics, Institute of Medical Biology, Polish Academy of Sciences, Lodowa 106, 93-232 Lodz, Poland
| | - Iwona Karwaciak
- Laboratory of Epigenetics, Institute of Medical Biology, Polish Academy of Sciences, Lodowa 106, 93-232 Lodz, Poland
| | - Kaja Karaś
- Laboratory of Epigenetics, Institute of Medical Biology, Polish Academy of Sciences, Lodowa 106, 93-232 Lodz, Poland
| | - Rafał A Bachorz
- Laboratory of Molecular Modeling, Institute of Medical Biology, Polish Academy of Sciences, Lodowa 106, 93-232 Lodz, Poland
| | - Marcin Ratajewski
- Laboratory of Epigenetics, Institute of Medical Biology, Polish Academy of Sciences, Lodowa 106, 93-232 Lodz, Poland.
| |
Collapse
|
5
|
Mieville V, Griffioen AW, Benamran D, Nowak-Sliwinska P. Advanced in vitro models for renal cell carcinoma therapy design. Biochim Biophys Acta Rev Cancer 2023; 1878:188942. [PMID: 37343729 DOI: 10.1016/j.bbcan.2023.188942] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 06/14/2023] [Accepted: 06/15/2023] [Indexed: 06/23/2023]
Abstract
Renal cell carcinoma (RCC) and its principal subtype, clear cell RCC, are the most diagnosed kidney cancer. Despite substantial improvement over the last decades, current pharmacological intervention still fails to achieve long-term therapeutic success. RCC is characterized by a high intra- and inter-tumoral heterogeneity and is heavily influenced by the crosstalk of the cells composing the tumor microenvironment, such as cancer-associated fibroblasts, endothelial cells and immune cells. Moreover, multiple physicochemical properties such as pH, interstitial pressure or oxygenation may also play an important role. These elements are often poorly recapitulated in in vitro models used for drug development. This inadequate recapitulation of the tumor is partially responsible for the current lack of an effective and curative treatment. Therefore, there are needs for more complex in vitro or ex vivo drug screening models. In this review, we discuss the current state-of-the-art of RCC models and suggest strategies for their further development.
Collapse
Affiliation(s)
- Valentin Mieville
- School of Pharmaceutical Sciences, Faculty of Sciences, University of Geneva, Switzerland; Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Geneva, Switzerland; Translational Research Center in Oncohaematology, Geneva, Switzerland
| | - Arjan W Griffioen
- Angiogenesis Laboratory, Department of Medical Oncology, Amsterdam UMC, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Daniel Benamran
- Division of Urology, Geneva University Hospitals, Geneva, Switzerland
| | - Patrycja Nowak-Sliwinska
- School of Pharmaceutical Sciences, Faculty of Sciences, University of Geneva, Switzerland; Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Geneva, Switzerland; Translational Research Center in Oncohaematology, Geneva, Switzerland.
| |
Collapse
|
6
|
Sun Y, Zhang Z, Zhang C, Zhang N, Wang P, Chu Y, Chard Dunmall LS, Lemoine NR, Wang Y. An effective therapeutic regime for treatment of glioma using oncolytic vaccinia virus expressing IL-21 in combination with immune checkpoint inhibition. Mol Ther Oncolytics 2022; 26:105-119. [PMID: 35795092 PMCID: PMC9233193 DOI: 10.1016/j.omto.2022.05.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 05/13/2022] [Indexed: 12/24/2022] Open
Abstract
Glioblastoma (GBM) is the most common primary malignant tumor in the brain, accounting for 51.4% of all primary brain tumors. GBM has a highly immunosuppressive tumor microenvironment (TME) and, as such, responses to immunotherapeutic strategies are poor. Vaccinia virus (VV) is an oncolytic virus that has shown tremendous therapeutic effect in various tumor types. In addition to its directly lytic effect on tumor cells, it has an ability to enhance immune cell infiltration into the TME allowing for improved immune control over the tumor. Here, we used a new generation of VV expressing the therapeutic payload interleukin-21 to treat murine GL261 glioma models. After both intratumoral and intravenous delivery, virus treatment induced remodeling of the TME to promote a robust anti-tumor immune response that resulted in control over tumor growth and long-term survival in both subcutaneous and orthotopic mouse models. Treatment efficacy was significantly improved in combination with systemic α-PD1 therapy, which is ineffective as a standalone treatment but synergizes with oncolytic VV to enhance therapeutic outcomes. Importantly, this study also revealed the upregulation of stem cell memory T cell populations after the virus treatment that exert strong and durable anti-tumor activity.
Collapse
Affiliation(s)
- Yijie Sun
- National Centre for International Research in Cell and Gene Therapy, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou 450052, China
| | - Zhe Zhang
- National Centre for International Research in Cell and Gene Therapy, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou 450052, China
| | - Chenglin Zhang
- National Centre for International Research in Cell and Gene Therapy, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou 450052, China
| | - Na Zhang
- National Centre for International Research in Cell and Gene Therapy, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou 450052, China
| | - Pengju Wang
- National Centre for International Research in Cell and Gene Therapy, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou 450052, China
| | - Yongchao Chu
- National Centre for International Research in Cell and Gene Therapy, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou 450052, China
| | - Louisa S. Chard Dunmall
- Centre for Cancer Biomarkers & Biotherapeutics, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK
| | - Nicholas R. Lemoine
- National Centre for International Research in Cell and Gene Therapy, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou 450052, China
- Centre for Cancer Biomarkers & Biotherapeutics, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK
| | - Yaohe Wang
- National Centre for International Research in Cell and Gene Therapy, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou 450052, China
- Centre for Cancer Biomarkers & Biotherapeutics, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK
| |
Collapse
|
7
|
Multifaceted Roles of Chemokines and Chemokine Receptors in Tumor Immunity. Cancers (Basel) 2021; 13:cancers13236132. [PMID: 34885241 PMCID: PMC8656932 DOI: 10.3390/cancers13236132] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 11/26/2021] [Accepted: 12/02/2021] [Indexed: 12/22/2022] Open
Abstract
Simple Summary Various immune cells are involved in host immune responses to cancer. T-helper (Th) 1 cells, cytotoxic CD8+ T cells, and natural killer cells are the major effector cells in anti-tumor immunity, whereas cells such as regulatory T cells and myeloid-derived suppressor cells are negatively involved in anti-tumor immunity. Th2 cells and Th17 cells have been shown to have both pro-tumor and anti-tumor activities. The migratory properties of various immune cells are essential for their function and critically regulated by the chemokine superfamily. In this review, we summarize the roles of various immune cells in tumor immunity and their migratory regulation by the chemokine superfamily. We also assess the therapeutic possibilities of targeting chemokines and chemokine receptors in cancer immunotherapy. Abstract Various immune cells are involved in host tumor immune responses. In particular, there are many T cell subsets with different roles in tumor immunity. T-helper (Th) 1 cells are involved in cellular immunity and thus play the major role in host anti-tumor immunity by inducing and activating cytotoxic T lymphocytes (CTLs). On the other hand, Th2 cells are involved in humoral immunity and suppressive to Th1 responses. Regulatory T (Treg) cells negatively regulate immune responses and contribute to immune evasion of tumor cells. Th17 cells are involved in inflammatory responses and may play a role in tumor progression. However, recent studies have also shown that Th17 cells are capable of directly inducting CTLs and thus may promote anti-tumor immunity. Besides these T cell subsets, there are many other innate immune cells such as dendritic cells (DCs), natural killer (NK) cells, and myeloid-derived suppressor cells (MDSCs) that are involved in host immune responses to cancer. The migratory properties of various immune cells are critical for their functions and largely regulated by the chemokine superfamily. Thus, chemokines and chemokine receptors play vital roles in the orchestration of host immune responses to cancer. In this review, we overview the various immune cells involved in host responses to cancer and their migratory properties regulated by the chemokine superfamily. Understanding the roles of chemokines and chemokine receptors in host immune responses to cancer may provide new therapeutic opportunities for cancer immunotherapy.
Collapse
|
8
|
Yang D, Gu X, Li C, Shi J, Chen Y, Dong M, Zhang Z. BCL7B is a potential novel diagnosis and prognosis biomarker for sarcomas using bioinformatics analysis. Medicine (Baltimore) 2021; 100:e26632. [PMID: 34260555 PMCID: PMC8284715 DOI: 10.1097/md.0000000000026632] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 06/23/2021] [Indexed: 01/04/2023] Open
Abstract
BCL7B plays a potential role in the progression of various cancers, while its role in sarcomas is unknown. We aimed to evaluate BCL7B's diagnostic and prognostic value, and potential BCL7B-related mechanisms in sarcomas based on The Cancer Genome Atlas (TCGA) database. We collected patients with sarcoma from TCGA. Wilcoxon rank sum test was used to compare the expression of BCL7B in sarcoma samples with different clinical-pathologic features. Univariate Cox regression analysis and multivariate Cox regression analysis were used to evaluate prognosis factors for sarcoma. Gene set enrichment analysis (GSEA) was conducted to elucidate the significant functions and pathways associated with BCL7B. BCL7B was a potential biomarker for distinguishing normal and tumor tissues with the analysis of ROC curve (AUC = 0.588). Low BCL7B expression was significantly correlated with tumor multifocal (OR = 0.39 for yes vs no), larger residual tumor (OR = 0.40 for R1,R2 vs RO), male gender (OR = 0.48 for male vs female) and White race (OR = 0.29 for White vs Asian, Black or African American). High BCL7B expression was correlated with leiomyosarcoma histological type (OR = 6.08 for leiomyosarcoma vs dedifferentiated liposarcoma, pleomorphic sarcoma). Univariate and multivariate Cox regression analysis showed that low BCL7B expression was independently associated with poor overall survival (P = .008). GSEA showed that GPCR (G protein-coupled receptors) ligand binding, secreted factors, class A1 rhodopsin-like receptors, extracellular matrix organization, core matrisome, Fc epsilon receptor I mediated NF-κB activation, and WNT signaling pathway were differentially enriched in BCL7B low expression phenotype (|NES| > 1, adjusted P value <.05, and FDR value <0.25). BCL7B may play an important role in sarcoma progression and may be a potential biomarker for prognosis and diagnosis in sarcomas.
Collapse
Affiliation(s)
- Dinglong Yang
- Second Clinical Medical College, Shanxi Medical University, China
| | - Xiaodong Gu
- Department of Orthopedics, Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Chunjiang Li
- Department of Orthopedics, Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Junjun Shi
- Department of Orthopedics, Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Yujing Chen
- School of Public Health, Xi’an Jiaotong University, Xian, Shaanxi, China
| | - Mingjie Dong
- Second Clinical Medical College, Shanxi Medical University, China
| | - Zhiqiang Zhang
- Department of Orthopedics, Second Hospital of Shanxi Medical University, Taiyuan, China
| |
Collapse
|
9
|
Abstract
The prevalence of peripheral arterial disease (PAD) in the United States exceeds 10 million people, and PAD is a significant cause of morbidity and mortality across the globe. PAD is typically caused by atherosclerotic obstructions in the large arteries to the leg(s). The most common clinical consequences of PAD include pain on walking (claudication), impaired functional capacity, pain at rest, and loss of tissue integrity in the distal limbs that may lead to lower extremity amputation. Patients with PAD also have higher than expected rates of myocardial infarction, stroke, and cardiovascular death. Despite advances in surgical and endovascular procedures, revascularization procedures may be suboptimal in relieving symptoms, and some patients with PAD cannot be treated because of comorbid conditions. In some cases, relieving obstructive disease in the large conduit arteries does not assure complete limb salvage because of severe microvascular disease. Despite several decades of investigational efforts, medical therapies to improve perfusion to the distal limb are of limited benefit. Whereas recent studies of anticoagulant (eg, rivaroxaban) and intensive lipid lowering (such as PCSK9 [proprotein convertase subtilisin/kexin type 9] inhibitors) have reduced major cardiovascular and limb events in PAD populations, chronic ischemia of the limb remains largely resistant to medical therapy. Experimental approaches to improve limb outcomes have included the administration of angiogenic cytokines (either as recombinant protein or as gene therapy) as well as cell therapy. Although early angiogenesis and cell therapy studies were promising, these studies lacked sufficient control groups and larger randomized clinical trials have yet to achieve significant benefit. This review will focus on what has been learned to advance medical revascularization for PAD and how that information might lead to novel approaches for therapeutic angiogenesis and arteriogenesis for PAD.
Collapse
Affiliation(s)
- Brian H Annex
- Vascular Biology Center, Department of Medicine, Medical College of Georgia, Augusta University (B.H.A.)
| | - John P Cooke
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, TX (J.P.C.)
| |
Collapse
|
10
|
Wang N, Wang J, Zhang Z, Cao H, Yan W, Chu Y, Chard Dunmall LS, Wang Y. A novel vaccinia virus enhances anti-tumor efficacy and promotes a long-term anti-tumor response in a murine model of colorectal cancer. Mol Ther Oncolytics 2021; 20:71-81. [PMID: 33575472 PMCID: PMC7851495 DOI: 10.1016/j.omto.2020.11.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Accepted: 11/11/2020] [Indexed: 02/06/2023] Open
Abstract
Colorectal cancer (CRC) is one of the leading causes of mortality and morbidity in the world, and there remains an urgent need to develop long-lasting therapies to treat CRC and prevent recurrence in patients. Oncolytic virus therapy (OVT) has demonstrated remarkable efficacy in a number of different cancer models. Here, we report a novel vaccinia virus (VV)-based OVT for treatment of CRC. The novel VV, based on the recently reported novel VVLΔTKΔN1L virus, was armed with the pleiotropic cytokine interleukin-21 (IL-21) to enhance anti-tumor immune responses stimulated after viral infection of tumor cells. Compared with an unarmed virus, VVLΔTKΔN1L-mIL-21 had a superior anti-tumor efficacy in murine CMT93 subcutaneous CRC models in vivo, mediated mainly by CD8+ T cells. Treatment resulted in development of long-term immunity against CMT93 tumor cells, as evidenced by prevention of disease recurrence. These results demonstrate that VVLΔTKΔN1L-mIL-21 is a promising therapeutic agent for treatment of CRC.
Collapse
Affiliation(s)
- Na Wang
- National Centre for International Research in Cell and Gene Therapy, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou 450052, China
| | - Jiwei Wang
- National Centre for International Research in Cell and Gene Therapy, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou 450052, China
| | - Zhe Zhang
- National Centre for International Research in Cell and Gene Therapy, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou 450052, China
| | - Hua Cao
- ENT Department, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou 450052, China
| | - Wenli Yan
- National Centre for International Research in Cell and Gene Therapy, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou 450052, China
| | - Yongchao Chu
- National Centre for International Research in Cell and Gene Therapy, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou 450052, China
| | - Louisa S. Chard Dunmall
- Centre for Biomarkers & Biotherapeutics, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK
| | - Yaohe Wang
- National Centre for International Research in Cell and Gene Therapy, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou 450052, China
- Centre for Biomarkers & Biotherapeutics, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK
| |
Collapse
|
11
|
Witek P, Enguita FJ, Grzesiak M, Costa MC, Gabriel A, Koziorowski M, Slomczynska M, Knapczyk-Stwora K. Effects of neonatal exposure to methoxychlor on corpus luteum in gilts: A transcriptomic analysis. Mol Reprod Dev 2021; 88:238-248. [PMID: 33655673 DOI: 10.1002/mrd.23463] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 01/07/2021] [Accepted: 02/20/2021] [Indexed: 11/10/2022]
Abstract
This study investigated the effects of neonatal exposure to methoxychlor (MXC), a synthetic organochlorine used as an insecticide with estrogenic, antiestrogenic, and antiandrogenic activities, on luteal function in pigs. Piglets were injected subcutaneously with MXC (20 μg/kg body weight) or corn oil (control) between postnatal Days 1 and 10 (N = 5/group). Corpora lutea from sexually mature gilts were examined for luteal steroid and prostaglandin concentrations and processed for total RNA isolation and subsequent RNA sequencing. Intra-luteal concentrations of androstenedione and prostaglandin E2 were greater, while that of estrone was lower when compared to control. Fifty-three differentially expressed (DE) microRNAS (miRNAs) (p-adjusted <.05 and log2(fold change) ≥.5) and 359 DE genes (p-adjusted <.05 and log2(fold change) ≥1) were identified in luteal tissue in response to neonatal MXC treatment. MXC was found to affect the expression of genes related to lipogenesis, steroidogenesis, membrane transport, immune response, cell signaling and adhesion. These results suggest an earlier onset of structural luteolysis in pigs caused by MXC actions in neonates. Since negative correlation analysis showed the potential interactions of miRNAs with specific messenger RNAs, we propose that these miRNAs are potential mediators of the long-term MXC effect on the CL function in pigs.
Collapse
Affiliation(s)
- Patrycja Witek
- Department of Endocrinology, Jagiellonian University in Krakow, Krakow, Poland
| | - Francisco J Enguita
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Malgorzata Grzesiak
- Department of Endocrinology, Jagiellonian University in Krakow, Krakow, Poland
| | - Marina C Costa
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - André Gabriel
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Marek Koziorowski
- Department of Physiology and Reproduction of Animals, Institute of Biotechnology, University of Rzeszow, Kolbuszowa, Poland
| | - Maria Slomczynska
- Department of Endocrinology, Jagiellonian University in Krakow, Krakow, Poland
| | | |
Collapse
|
12
|
Marelli G, Chard Dunmall LS, Yuan M, Di Gioia C, Miao J, Cheng Z, Zhang Z, Liu P, Ahmed J, Gangeswaran R, Lemoine N, Wang Y. A systemically deliverable Vaccinia virus with increased capacity for intertumoral and intratumoral spread effectively treats pancreatic cancer. J Immunother Cancer 2021; 9:e001624. [PMID: 33500259 PMCID: PMC7839893 DOI: 10.1136/jitc-2020-001624] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/09/2020] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Pancreatic cancer remains one of the most lethal cancers and is refractory to immunotherapeutic interventions. Oncolytic viruses are a promising new treatment option, but current platforms demonstrate limited efficacy, especially for inaccessible and metastatic cancers that require systemically deliverable therapies. We recently described an oncolytic vaccinia virus (VV), VVLΔTKΔN1L, which has potent antitumor activity, and a regime to enhance intravenous delivery of VV by pharmacological inhibition of pharmacological inhibition of PI3 Kinase δ (PI3Kδ) to prevent virus uptake by macrophages. While these platforms improve the clinical prospects of VV, antitumor efficacy must be improved. METHODS VVLΔTKΔN1L was modified to improve viral spread within and between tumors via viral B5R protein modification, which enhanced production of the extracellular enveloped virus form of VV. Antitumor immunity evoked by viral treatment was improved by arming the virus with interleukin-21, creating VVL-21. Efficacy, functional activity and synergy with α-programmed cell death protein 1 (α-PD1) were assessed after systemic delivery to murine and Syrian hamster models of pancreatic cancer. RESULTS VVL-21 could reach tumors after systemic delivery and demonstrated antitumor efficacy in subcutaneous, orthotopic and disseminated models of pancreatic cancer. The incorporation of modified B5R improved intratumoural accumulation of VV. VVL-21 treatment increased the numbers of effector CD8+ T cells within the tumor, increased circulating natural killer cells and was able to polarize macrophages to an M1 phenotype in vivo and in vitro. Importantly, treatment with VVL-21 sensitized tumors to the immune checkpoint inhibitor α-PD1. CONCLUSIONS Intravenously administered VVL-21 successfully remodeled the suppressive tumor-microenvironment to promote antitumor immune responses and improve long-term survival in animal models of pancreatic cancer. Importantly, treatment with VVL-21 sensitized tumors to the immune checkpoint inhibitor α-PD1. Combination of PI3Kδ inhibition, VVL-21 and α-PD1 creates an effective platform for treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Giulia Marelli
- Centre for Cancer Biomarkers and Biotherapeutics, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Louisa S Chard Dunmall
- Centre for Cancer Biomarkers and Biotherapeutics, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Ming Yuan
- Centre for Cancer Biomarkers and Biotherapeutics, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Carmela Di Gioia
- Centre for Cancer Biomarkers and Biotherapeutics, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Jinxin Miao
- National Centre for International Research in Cell and Gene Therapy, Zhengzhou University, Zhengzhou, Henan, China
- Academy of Chinese Medicine Science, Henan University of Chinese Medicine, Zhengzhou 450000, Henan Province, People's Republic of China
| | - Zhenguo Cheng
- National Centre for International Research in Cell and Gene Therapy, Zhengzhou University, Zhengzhou, Henan, China
| | - Zhongxian Zhang
- National Centre for International Research in Cell and Gene Therapy, Zhengzhou University, Zhengzhou, Henan, China
| | - Peng Liu
- Centre for Cancer Biomarkers and Biotherapeutics, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Jahangir Ahmed
- Centre for Cancer Biomarkers and Biotherapeutics, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Rathi Gangeswaran
- Centre for Cancer Biomarkers and Biotherapeutics, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Nicholas Lemoine
- Centre for Cancer Biomarkers and Biotherapeutics, Barts Cancer Institute, Queen Mary University of London, London, UK
- National Centre for International Research in Cell and Gene Therapy, Zhengzhou University, Zhengzhou, Henan, China
| | - Yaohe Wang
- Centre for Cancer Biomarkers and Biotherapeutics, Barts Cancer Institute, Queen Mary University of London, London, UK
- National Centre for International Research in Cell and Gene Therapy, Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
13
|
Picado C, Roca-Ferrer J. Role of the Cyclooxygenase Pathway in the Association of Obstructive Sleep Apnea and Cancer. J Clin Med 2020; 9:E3237. [PMID: 33050416 PMCID: PMC7601393 DOI: 10.3390/jcm9103237] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Revised: 09/30/2020] [Accepted: 10/02/2020] [Indexed: 11/16/2022] Open
Abstract
The objective of this review is to examine the findings that link obstructive sleep apnea (OSA) with cancer and the role played by the cyclooxygenase (COX) pathway in this association. Epidemiological studies in humans suggest a link between OSA and increased cancer incidence and mortality. Studies carried out in animal models have shown that intermittent hypoxia (IH) induces changes in several signaling pathways involved in the regulation of host immunological surveillance that results in tumor establishment and invasion. IH induces the expression of cyclooxygenase 2 (COX-2) that results in an increased synthesis of prostaglandin E2 (PGE2). PGE2 modulates the function of multiple cells involved in immune responses including T lymphocytes, NK cells, dendritic cells, macrophages, and myeloid-derived suppressor cells. In a mouse model blockage of COX-2/PGE2 abrogated the pro-oncogenic effects of IH. Despite the fact that aspirin inhibits PGE2 production and prevents the development of cancer, none of the epidemiological studies that investigated the association of OSA and cancer included aspirin use in the analysis. Studies are needed to investigate the regulation of the COX-2/PGE2 pathway and PGE2 production in patients with OSA, to better define the role of this axis in the physiopathology of OSA and the potential role of aspirin in preventing the development of cancer.
Collapse
Affiliation(s)
- César Picado
- Hospital Clinic, Department of Medicine, Universitat de Barcelona, 08036 Barcelona, Spain
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain;
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto Carlos III, 28029 Madrid, Spain
| | - Jordi Roca-Ferrer
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain;
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto Carlos III, 28029 Madrid, Spain
| |
Collapse
|
14
|
Rada M, Lazaris A, Kapelanski-Lamoureux A, Mayer TZ, Metrakos P. Tumor microenvironment conditions that favor vessel co-option in colorectal cancer liver metastases: A theoretical model. Semin Cancer Biol 2020; 71:52-64. [PMID: 32920126 DOI: 10.1016/j.semcancer.2020.09.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 09/03/2020] [Accepted: 09/03/2020] [Indexed: 02/07/2023]
Abstract
Vessel co-option is an alternative strategy by which tumour cells vascularize and gain access to nutrients to support tumour growth, survival and metastasis. In vessel co-option, the cancer cells move towards the pre-existing vasculature and hijack them. Vessel co-option is adopted by a wide range of human tumours including colorectal cancer liver metastases (CRCLM) and is responsible for the effectiveness of treatment in CRCLM. Furthermore, vessel co-option is an intrinsic feature and an acquired mechanism of resistance to anti-angiogenic treatment. In this review, we describe the microenvironment, the molecular players, discovered thus far of co-opting CRCLM lesions and propose a theoretical model. We also highlight key unanswered questions that are critical to improving our understanding of CRCLM vessel co-option and for the development of effective approaches for the treatment of co-opting tumours.
Collapse
Affiliation(s)
- Miran Rada
- Cancer Research Program, McGill University Health Centre Research Institute, Montreal, Quebec, H4A3J1, Canada
| | - Anthoula Lazaris
- Cancer Research Program, McGill University Health Centre Research Institute, Montreal, Quebec, H4A3J1, Canada
| | - Audrey Kapelanski-Lamoureux
- Cancer Research Program, McGill University Health Centre Research Institute, Montreal, Quebec, H4A3J1, Canada
| | - Thomas Z Mayer
- Cancer Research Program, McGill University Health Centre Research Institute, Montreal, Quebec, H4A3J1, Canada
| | - Peter Metrakos
- Cancer Research Program, McGill University Health Centre Research Institute, Montreal, Quebec, H4A3J1, Canada.
| |
Collapse
|
15
|
Harmsen MJ, Wong CFC, Mijatovic V, Griffioen AW, Groenman F, Hehenkamp WJK, Huirne JAF. Role of angiogenesis in adenomyosis-associated abnormal uterine bleeding and subfertility: a systematic review. Hum Reprod Update 2020; 25:647-671. [PMID: 31504506 PMCID: PMC6737562 DOI: 10.1093/humupd/dmz024] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 06/11/2019] [Accepted: 06/19/2019] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Adenomyosis commonly occurs with abnormal uterine bleeding (AUB) and is associated with subfertility and a higher miscarriage rate. Recent evidence showed abnormal vascularization in the endometrium in patients with adenomyosis, suggesting a role of angiogenesis in the pathophysiology of AUB and subfertility in adenomyosis and providing a possible treatment target. OBJECTIVE AND RATIONALE We hypothesized that the level of abnormal vascularization and expression of angiogenic markers is increased in the ectopic and eutopic endometrium of adenomyosis patients in comparison with the endometrium of control patients. This was investigated through a search of the literature. SEARCH METHODS A systematic search was performed in PubMed and Embase until February 2019. Combinations of terms for angiogenesis and adenomyosis were applied as well as AUB, subfertility or anti-angiogenic therapy. The main search was limited to clinical studies carried out on premenopausal women. Original research articles focusing on markers of angiogenesis in the endometrium of patients with adenomyosis were included. Studies in which no comparison was made to control patients or which were not published in a peer-reviewed journal were excluded. A second search was performed to explore the therapeutic potential of targeting angiogenesis in adenomyosis. This search also included preclinical studies. OUTCOMES A total of 20 articles out of 1669 hits met our selection criteria. The mean vascular density (MVD) was studied by quantification of CD31, CD34, von Willebrand Factor (vWF) or factor-VIII-antibody-stained microvessels in seven studies. All these studies reported a significantly increased MVD in ectopic endometrium, and out of the six articles that took it into account, four studies reported a significantly increased MVD in eutopic endometrium compared with control endometrium. Five articles showed a significantly higher vascular endothelial growth factor expression in ectopic endometrium and three articles in eutopic endometrium compared with control endometrium. The vascular and pro-angiogenic markers α-smooth muscle actin, endoglin, S100A13, vimentin, matrix metalloproteinases (MMPs), nuclear factor (NF)-kB, tissue factor (TF), DJ-1, phosphorylated mammalian target of rapamycin, activin A, folli- and myostatin, CD41, SLIT, roundabout 1 (ROBO1), cyclooxygenase-2, lysophosphatidic acid (LPA) 1,4-5, phospho signal transducer and activator of transcription 3 (pSTAT3), interleukin (IL)-6, IL-22 and transforming growth factor-β1 were increased in ectopic endometrium, and the markers S100A13, MMP-2 and -9, TF, follistatin, myostatin, ROBO1, LPA1 and 4-5, pSTAT3, IL-6 and IL-22 were increased in eutopic endometrium, compared with control endometrium. The anti-angiogenic markers E-cadherin, eukaryotic translation initiation factor 3 subunit and gene associated with retinoic-interferon-induced mortality 19 were decreased in ectopic endometrium and IL-10 in eutopic endometrium, compared with control endometrium. The staining level of vWF and two pro-angiogenic markers (NF-κB nuclear p65 and TF) correlated with AUB in patients with adenomyosis. We found no studies that investigated the possible relationship between markers of angiogenesis and subfertility in adenomyosis patients. Nine articles reported on direct or indirect targeting of angiogenesis in adenomyosis-either by testing hormonal therapy or herbal compounds in clinical studies or by testing angiogenesis inhibitors in preclinical studies. However, there are no clinical studies on the effectiveness of such therapy for adenomyosis-related AUB or subfertility. WIDER IMPLICATIONS The results are in agreement with our hypothesis that increased angiogenesis is present in the endometrium of patients with adenomyosis compared with the endometrium of control patients. It is likely that increased angiogenesis leads to fragile and more permeable vessels resulting in adenomyosis-related AUB and possibly subfertility. While this association has not sufficiently been studied yet, our results encourage future studies to investigate the exact role of angiogenesis in the etiology of adenomyosis and related AUB or subfertility in women with adenomyosis in order to design curative or preventive therapeutic strategies.
Collapse
Affiliation(s)
- Marissa J Harmsen
- Department of Obstetrics and Gynecology, Amsterdam UMC, location VUMC, Amsterdam, The Netherlands.,Angiogenesis Laboratory, Amsterdam UMC, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Caroline F C Wong
- Department of Obstetrics and Gynecology, Amsterdam UMC, location VUMC, Amsterdam, The Netherlands.,Angiogenesis Laboratory, Amsterdam UMC, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Velja Mijatovic
- Department of Obstetrics and Gynecology, Amsterdam UMC, location VUMC, Amsterdam, The Netherlands
| | - Arjan W Griffioen
- Angiogenesis Laboratory, Amsterdam UMC, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Freek Groenman
- Department of Obstetrics and Gynecology, Amsterdam UMC, location VUMC, Amsterdam, The Netherlands
| | - Wouter J K Hehenkamp
- Department of Obstetrics and Gynecology, Amsterdam UMC, location VUMC, Amsterdam, The Netherlands
| | - Judith A F Huirne
- Department of Obstetrics and Gynecology, Amsterdam UMC, location VUMC, Amsterdam, The Netherlands
| |
Collapse
|
16
|
Pokuri VK, Tomaszewski GM, Ait-Oudhia S, Groman A, Khushalani NI, Lugade AA, Thanavala Y, Ashton EA, Grande C, Fetterly GJ, Iyer R. Efficacy, Safety, and Potential Biomarkers of Sunitinib and Transarterial Chemoembolization (TACE) Combination in Advanced Hepatocellular Carcinoma (HCC): Phase II Trial. Am J Clin Oncol 2019; 41:332-338. [PMID: 27014931 DOI: 10.1097/coc.0000000000000286] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
OBJECTIVES To evaluate the safety/efficacy and explore biomarkers for a rationally designed combination of sunitinib and transarterial chemoembolization (TACE) in a prospective phase 2 study of advanced hepatocellular carcinoma (HCC). METHODS Inoperable HCC patients with Child-Pugh A disease received 37.5 mg sunitinib from days 1 to 7 followed by TACE on day 8. Sunitinib was resumed from days 15 to 36 followed by 2 weeks off. Patients received subsequent sunitinib cycles of 4 weeks on and 2 weeks off. Dynamic contrast-enhanced magnetic resonance imaging and circulating soluble biomarkers were assessed at baseline, day 8, day 10, and day 36. RESULTS Sixteen patients with liver only (n=10) and extrahepatic disease (n=6) were enrolled. After a median follow-up of 12.8 months, 2 partial responses, 11 stable disease, and 3 clinical deteriorations were seen for a clinical benefit rate of 81%. Median progression-free survival (PFS) was 8 months (95% CI, 4.3-9.3) and overall survival was 14.9 months (95% CI, 6.3-27.1). Eleven of 16 patients (69%) had grade 3/4 toxicities attributable to sunitinib, the most frequent being thrombocytopenia, amylase/lipase elevations, lymphopenia, and fatigue. Mean K (volume transfer constant) and viable tumor percent in consented patients decreased by 27% and 14.8%, respectively, with combination therapy. Soluble vascular endothelial growth factor receptor-2 (sVEGFR2) levels, cytokines (interleukin-8, interleukin-21), and monocytes decreased with combination therapy. Estimated sunitinib IC50 values of 15 and 10 ng/mL modulated K and AUC90. sVEGFR2 levels decreased with K and AUC90. CONCLUSIONS Encouraging progression-free survival and overall survival were seen with acceptable toxicity in our study of sunitinib and TACE combination in advanced HCC. Potential imaging and serum biomarkers showed increased benefit with combination therapy.
Collapse
Affiliation(s)
| | | | - Sihem Ait-Oudhia
- Pharmacometrics and Systems Pharmacology at Lake Nona, University of Florida, Orlando, FL
| | | | | | - Amit A Lugade
- Center for Immunotherapy, Roswell Park Cancer Institute (RPCI), Buffalo
| | | | | | | | | | | |
Collapse
|
17
|
Najafi S, Mirshafiey A. The role of T helper 17 and regulatory T cells in tumor microenvironment. Immunopharmacol Immunotoxicol 2019; 41:16-24. [PMID: 30714422 DOI: 10.1080/08923973.2019.1566925] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
T helper 17 (Th17) cells were first described as a novel T helper cell lineage independent from Th1 and Th2 subsets. Th17 cells play vital roles in inflammation and tumor immunity. It causes the dissipation of antitumor immunity and contribution to the survival of tumor cells, worsening tumor growth and metastasis. Tumor-infiltrating Th17 cells were seen innumerous cancers in mice and humans. There has been an association between intratumoral Th17 cell infiltration and both good and bad prognoses. Besides the protumoral roles defined for IL-17 andTh17 cells, several reports have shown that Th17 cells also drive antitumoral immunity. Various mechanisms by which Th17 cells control tumor growth are as following: recruitment of several immune cells including DCs, CD4+ T cells, and CD8+ T cells within tumors, activation of CD8+ T cells, and probably plasticity toward Th1 phenotype, related to IFN-γ and TNF-α production. Regulatory T cells (Tregs) have been exhibited to infiltrate human tumors and are believed to restrict antitumor immunity. The effect of Treg cells has been more controversial. Whereas some studies have proposed that a high density of Treg cells within the tumor associated with a poor clinical prognosis, other studies have presented a positive clinical prognosis, underlining the importance of elucidating the clinical significance of Treg cells further. Treg and Th17 cells play both positive and negative roles in regulating antitumor immune responses. In spite of the presence of these cells, yet some tumors develop and grow. These T cells by themselves are not adequate to efficiently mount antitumor immune responses.
Collapse
Affiliation(s)
- Soheil Najafi
- a Department of Immunology , School of Public Health, International Campus, Tehran University of Medical Sciences , Tehran , Iran
| | - Abbas Mirshafiey
- b Department of Immunology , School of Public Health, Tehran University of Medical Sciences , Tehran , Iran
| |
Collapse
|
18
|
The roles of interleukins in perfusion recovery after peripheral arterial disease. Biosci Rep 2018; 38:BSR20171455. [PMID: 29358309 PMCID: PMC5809615 DOI: 10.1042/bsr20171455] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2017] [Revised: 01/02/2018] [Accepted: 01/20/2018] [Indexed: 11/22/2022] Open
Abstract
In peripheral arterial disease (PAD) patients, occlusions in the major arteries that supply the leg makes blood flow dependent on the capacity of neovascularization. There is no current medication that is able to increase neovascularization to the ischemic limb and directly treat the primary problem of PAD. An increasing body of evidence supports the notion that inflammation plays an important role in the vascular remodeling and perfusion recovery after PAD. Interleukins (ILs), a group of proteins produced during inflammation, have been considered to be important for angiogenesis and arteriogenesis after tissue ischemia. This review summarizes the latest clinical and experimental developments of the role of ILs in blood perfusion recovery after PAD.
Collapse
|
19
|
Gonçalves RSG, Pereira MC, Dantas AT, Almeida ARD, Marques CDL, Rego MJBM, Pitta IR, Duarte ALBP, Pitta MGR. IL-17 and related cytokines involved in systemic sclerosis: Perspectives. Autoimmunity 2017; 51:1-9. [PMID: 29256263 DOI: 10.1080/08916934.2017.1416467] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Systemic sclerosis (SSc) is a multisystemic, complex, and rare disease of connective tissue, with high morbidity and mortality, and without specific treatment. The disease is characterized by three main principles: vascular disease, autoantibody production and inflammation, and fibrosis. Since it is well defined that SSc is characterized by elevated production of TGF-β, IL-6, and IL-1, all of them cytokines related to Th17 differentiation, the hypothesis is that this disease may be strongly related to a polarization of the immune response towards the Th17 pathway. Considering the importance of a better understanding of the pathophysiology of Th17 pathway in SSc, this article aims to propose an update for a better understanding of current knowledge on main cytokines secreted by the Th17 cells (IL-17 A, IL-21, and IL-22) and the future prospects in the current disease.
Collapse
Affiliation(s)
- Rafaela Silva Guimarães Gonçalves
- a Hospital das Clínicas de Pernambuco , Universidade Federal de Pernambuco , Recife , Brazil.,b Laboratório de Imunomodulação e Novas Abordagens Terapêuticas Suely Galdino , Universidade Federal de Pernambuco , Recife , Brazil
| | - Michelly C Pereira
- b Laboratório de Imunomodulação e Novas Abordagens Terapêuticas Suely Galdino , Universidade Federal de Pernambuco , Recife , Brazil
| | - Andréa Tavares Dantas
- a Hospital das Clínicas de Pernambuco , Universidade Federal de Pernambuco , Recife , Brazil.,b Laboratório de Imunomodulação e Novas Abordagens Terapêuticas Suely Galdino , Universidade Federal de Pernambuco , Recife , Brazil
| | - Anderson Rodrigues de Almeida
- b Laboratório de Imunomodulação e Novas Abordagens Terapêuticas Suely Galdino , Universidade Federal de Pernambuco , Recife , Brazil
| | | | - Moacyr J B M Rego
- b Laboratório de Imunomodulação e Novas Abordagens Terapêuticas Suely Galdino , Universidade Federal de Pernambuco , Recife , Brazil
| | - Ivan R Pitta
- b Laboratório de Imunomodulação e Novas Abordagens Terapêuticas Suely Galdino , Universidade Federal de Pernambuco , Recife , Brazil
| | - Angela Luzia Branco Pinto Duarte
- a Hospital das Clínicas de Pernambuco , Universidade Federal de Pernambuco , Recife , Brazil.,b Laboratório de Imunomodulação e Novas Abordagens Terapêuticas Suely Galdino , Universidade Federal de Pernambuco , Recife , Brazil
| | - Maira Galdino R Pitta
- b Laboratório de Imunomodulação e Novas Abordagens Terapêuticas Suely Galdino , Universidade Federal de Pernambuco , Recife , Brazil
| |
Collapse
|
20
|
Lewis KE, Selby MJ, Masters G, Valle J, Dito G, Curtis WR, Garcia R, Mink KA, Waggie KS, Holdren MS, Grosso JF, Korman AJ, Jure-Kunkel M, Dillon SR. Interleukin-21 combined with PD-1 or CTLA-4 blockade enhances antitumor immunity in mouse tumor models. Oncoimmunology 2017; 7:e1377873. [PMID: 29296539 PMCID: PMC5739581 DOI: 10.1080/2162402x.2017.1377873] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Revised: 08/17/2017] [Accepted: 09/04/2017] [Indexed: 01/19/2023] Open
Abstract
Recent advances in cancer treatment with checkpoint blockade of receptors such as CTLA-4 and PD-1 have demonstrated that combinations of agents with complementary immunomodulatory effects have the potential to enhance antitumor activity as compared to single agents. We investigated the efficacy of immune-modulatory interleukin-21 (IL-21) combined with checkpoint blockade in several syngeneic mouse tumor models. After tumor establishment, mice were administered recombinant mouse IL-21 (mIL-21) alone or in combination with blocking monoclonal antibodies against mouse PD-1 or CTLA-4. In contrast to monotherapy, IL-21 enhanced antitumor activity of mCTLA-4 mAb in four models and anti-PD-1 mAb in two models, with evidence of synergy for one or both of the combination treatments in the EMT-6 and MC38 models. The enhanced efficacy was associated with increased intratumoral CD8+ T cell infiltrates, CD8+ T cell proliferation, and increased effector memory T cells, along with decreased frequency of central memory CD8+ T cells. In vivo depletion of CD8+ T cells abolished the antitumor activities observed for both combination and monotherapy treatments, further supporting a beneficial role for CD8+ T cells. In all studies, the combination therapies were well tolerated. These results support the hypothesis that the combination of recombinant human IL-21 with CTLA-4 or PD-1 monoclonal antibodies could lead to improved outcomes in cancer patients.
Collapse
Affiliation(s)
| | - Mark J Selby
- Oncology Discovery Research, Bristol-Myers Squibb, Redwood City, CA
| | - Gregg Masters
- Oncology Translational Research, Bristol-Myers Squibb, Princeton, NJ
| | - Jose Valle
- Oncology Discovery Research, Bristol-Myers Squibb, Redwood City, CA
| | - Gennaro Dito
- Oncology Translational Research, Bristol-Myers Squibb, Princeton, NJ
| | - Wendy R Curtis
- Oncology Discovery Research, Bristol-Myers Squibb, Seattle, WA
| | - Richard Garcia
- Oncology Discovery Research, Bristol-Myers Squibb, Seattle, WA
| | - Kathy A Mink
- Oncology Discovery Research, Bristol-Myers Squibb, Seattle, WA
| | | | | | - Joseph F Grosso
- Early Clinical Development, Bristol-Myers Squibb, Princeton, NJ
| | - Alan J Korman
- Oncology Discovery Research, Bristol-Myers Squibb, Redwood City, CA
| | - Maria Jure-Kunkel
- Oncology Translational Research, Bristol-Myers Squibb, Princeton, NJ
| | - Stacey R Dillon
- Oncology Discovery Research, Bristol-Myers Squibb, Seattle, WA
| |
Collapse
|
21
|
Sharma J, Bhar S, Devi CS. A review on interleukins: The key manipulators in rheumatoid arthritis. Mod Rheumatol 2017; 27:723-746. [DOI: 10.1080/14397595.2016.1266071] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Jatin Sharma
- School of Biosciences and Technology, VIT University, Vellore, India
| | - Sutonuka Bhar
- School of Biosciences and Technology, VIT University, Vellore, India
| | - C. Subathra Devi
- School of Biosciences and Technology, VIT University, Vellore, India
| |
Collapse
|
22
|
Yuan MJ, Wang T. Advances of the interleukin-21 signaling pathway in immunity and angiogenesis. Biomed Rep 2016; 5:3-6. [PMID: 27330746 DOI: 10.3892/br.2016.665] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 04/25/2016] [Indexed: 12/23/2022] Open
Abstract
Interleukin-21 (IL-21) and its receptor (IL-21R) are broadly expressed on human B cells, activated T cells and other myeloid cells. IL-21 cooperates with IL-6 and transforming growth factor-β to regulate T-cell differentiation. IL-21-mediated human B cell and dendritic cells differentiation requires signal transducer and activator of transcription 3 (STAT3), and also induces B-cell apoptosis dependents on the Toll-like receptor signal. Recently, in vitro and in vivo experiments showed that IL-21/IL-21R regulate angiogenesis through STAT3. IL-21 signaling pathways are complex due to its cooperation with other transcriptional factors, such as interferon regulatory factor 4 and granulocyte-macrophage colony-stimulating factor. The Janus kinase-STAT pathway has been the most extensively studied. With the increase in the understanding of IL-21 biology in the context of each specific disease or pathological condition, IL-21 could be a new therapeutic target for immune-related disease.
Collapse
Affiliation(s)
- Ming-Jie Yuan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Tao Wang
- Cardiovascular Research Center, University of Virginia, Charlottesville, VA 22908, USA
| |
Collapse
|
23
|
De Simone V, Ronchetti G, Franzè E, Colantoni A, Ortenzi A, Fantini MC, Rizzo A, Sica GS, Sileri P, Rossi P, MacDonald TT, Pallone F, Monteleone G, Stolfi C. Interleukin-21 sustains inflammatory signals that contribute to sporadic colon tumorigenesis. Oncotarget 2016; 6:9908-23. [PMID: 25839161 PMCID: PMC4496406 DOI: 10.18632/oncotarget.3532] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Accepted: 02/17/2015] [Indexed: 02/07/2023] Open
Abstract
Interleukin (IL)-21 triggers inflammatory signals that contribute to the growth of neoplastic cells in mouse models of colitis-associated colorectal cancer (CRC). Because most CRCs are sporadic and arise in the absence of overt inflammation we have investigated the role of IL-21 in these tumors in mouse and man. IL-21 was highly expressed in human sporadic CRC and produced mostly by IFN-γ-expressing T-bet/RORγt double-positive CD3+CD8− cells. Stimulation of human CRC cell lines with IL-21 did not directly activate the oncogenic transcription factors STAT3 and NF-kB and did not affect CRC cell proliferation and survival. In contrast, IL-21 modulated the production of protumorigenic factors by human tumor infiltrating T cells. IL-21 was upregulated in the neoplastic areas, as compared with non-tumor mucosa, of Apcmin/+ mice, and genetic ablation of IL-21 in such mice resulted in a marked decrease of both tumor incidence and size. IL-21 deficiency was associated with reduced STAT3/NF-kB activation in both immune cells and neoplastic cells, diminished synthesis of protumorigenic cytokines (that is, IL-17A, IL-22, TNF-α and IL-6), downregulation of COX-2/PGE2 pathway and decreased angiogenesis in the lesions of Apcmin/+ mice. Altogether, data suggest that IL-21 promotes a protumorigenic inflammatory circuit that ultimately sustains the development of sporadic CRC.
Collapse
Affiliation(s)
- Veronica De Simone
- Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Giulia Ronchetti
- Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Eleonora Franzè
- Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Alfredo Colantoni
- Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Angela Ortenzi
- Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Massimo C Fantini
- Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Angelamaria Rizzo
- Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Giuseppe S Sica
- Department of Surgery, University of Rome "Tor Vergata", Rome, Italy
| | - Pierpaolo Sileri
- Department of Surgery, University of Rome "Tor Vergata", Rome, Italy
| | - Piero Rossi
- Department of Surgery, University of Rome "Tor Vergata", Rome, Italy
| | - Thomas T MacDonald
- Centre for Immunology and Infectious Disease, Blizard Institute of Cell and Molecular Science, Barts and the London School of Medicine and Dentistry, London, UK
| | - Francesco Pallone
- Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Giovanni Monteleone
- Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Carmine Stolfi
- Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
| |
Collapse
|
24
|
ANNEX BRIANH, BELLER GEORGEA. TOWARDS THE DEVELOPMENT OF NOVEL THERAPEUTICS FOR PERIPHERAL ARTERY DISEASE. TRANSACTIONS OF THE AMERICAN CLINICAL AND CLIMATOLOGICAL ASSOCIATION 2016; 127:224-234. [PMID: 28066055 PMCID: PMC5216482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Atherosclerosis is the leading cause of morbidity and mortality in the Western world. Peripheral artery disease (PAD) has been less studied then coronary artery disease but is nearly as common. PAD impairs blood flow to the leg(s) and causes functional impairment, leg pain, and amputation. The last drug approved for PAD was in 1999. Blood flow to leg proceeds through one major artery and in PAD total occlusions in the course of that vessel are common. Thus, the extent of new blood vessel growth determines a patients' clinical course. Promoting the growth of new blood vessels (therapeutic angiogenesis) was a major goal of therapy. Results from studies using cytokine growth factors have shown disappointing results. Using clinical and preclinical studies, our laboratory has identified several novel therapeutic approaches. One, a modulator of innate immunity, will be reviewed as an approach that has the potential to create new therapies for PAD.
Collapse
Affiliation(s)
- BRIAN H. ANNEX
- Correspondence and reprint requests: Brian H. Annex, MD,
Department of Medicine, University of Virginia, 1215 Lee Street, PO Box 800158, Charlottesville, VA 22908434-982-0853434-982-1998
| | | |
Collapse
|
25
|
Wang T, Cunningham A, Houston K, Sharma AM, Chen L, Dokun AO, Lye RJ, Spolski R, Leonard WJ, Annex BH. Endothelial interleukin-21 receptor up-regulation in peripheral artery disease. Vasc Med 2015; 21:99-104. [PMID: 26705256 DOI: 10.1177/1358863x15621798] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
In most patients with symptomatic peripheral artery disease (PAD), severe stenosis in or occlusion of the major blood vessels that supply the legs make the amount of distal blood flow dependent on the capacity to induce angiogenesis and collateral vessel formation. Currently, there are no medications that improve perfusion to the ischemic limb, and thus directly treat the primary problem of PAD. A recent report from our group in a pre-clinical mouse PAD model showed that interleukin-21 receptor (IL-21R) is up-regulated in the endothelial cells from ischemic hindlimb muscle. We further showed that loss of IL-21R resulted in impaired perfusion recovery in this model. In our study, we sought to determine whether IL-21R is present in the endothelium from ischemic muscle of patients with PAD. Using human gastrocnemius muscle biopsies, we found increased levels of IL-21R in the skeletal muscle endothelial cells of patients with PAD compared to control individuals. Interestingly, PAD patients had approximately 1.7-fold higher levels of circulating IL-21. These data provide direct evidence that the IL-21R pathway is indeed up-regulated in patients with PAD. This pathway may serve as a therapeutic target for modulation.
Collapse
Affiliation(s)
- Tao Wang
- Robert M Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, USA
| | - Alexis Cunningham
- Robert M Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, USA
| | - Kevin Houston
- Robert M Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, USA
| | - Aditya M Sharma
- Division of Cardiovascular Medicine, Department of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Lingdan Chen
- Robert M Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, USA
| | - Ayotunde O Dokun
- Robert M Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, USA
| | - R John Lye
- Robert M Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, USA
| | - Rosanne Spolski
- Laboratory of Molecular Immunology and the Immunology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Warren J Leonard
- Laboratory of Molecular Immunology and the Immunology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Brian H Annex
- Robert M Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, USA Division of Cardiovascular Medicine, Department of Medicine, University of Virginia, Charlottesville, VA, USA
| |
Collapse
|
26
|
Tudoran O, Virtic O, Balacescu L, Lisencu C, Fetica B, Gherman C, Balacescu O, Berindan-Neagoe I. Baseline blood immunological profiling differentiates between Her2-breast cancer molecular subtypes: implications for immunomediated mechanisms of treatment response. Onco Targets Ther 2015; 8:3415-23. [PMID: 26604799 PMCID: PMC4655955 DOI: 10.2147/ott.s91720] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Purpose Breast cancer patients’ response to treatment is highly dependent on the primary tumor molecular features, with triple-negative breast tumors having the worst prognosis of all subtypes. According to the molecular features, tumors stimulate the microenvironment to induce distinct immune responses, baseline immune activation being associated with higher likelihood of pathologic response. In this study, we investigated the deconvolution of the immunological status of triple-negative tumors in comparison with luminal tumors and the association with patients’ clinicopathological characteristics. Patients and methods Gene expression of 84 inflammatory molecules and their receptors were analyzed in 40 peripheral blood samples from patients with Her2− primary breast cancer tumors. We studied the association of triple-negative phenotype with age, clinical stage, tumor size, lymph nodes, and menopausal status. Results We observed that more patients with estrogen (ER)/progesterone (PR)-negative tumors had grade III, while more patients with ER/PR-positive tumors had grade II tumors. Gene expression analysis revealed a panel of 14 genes to have differential expression between the two groups: several interleukins: IL13, IL16, IL17C and IL17F, IL1A, IL3; interleukin receptors: IL10RB, IL5RA; chemokines: CXCL13 and CCL26; and cytokines: CSF2, IFNA2, OSM, TNSF13. Conclusion The expression levels of these genes have been previously shown to be associated with reduced immunological status; indeed, the triple-negative breast cancer patients presented with lower counts of lymphocytes and eosinophils than the ER/PR-positive ones. These results contribute to a better understanding of the possible role of antitumor immune responses in mediating the clinical outcome.
Collapse
Affiliation(s)
- Oana Tudoran
- Department of Functional Genomics and Experimental Pathology, The Oncology Institute "Prof Dr Ion Chiricuţă", Cluj-Napoca, Romania ; Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Oana Virtic
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Loredana Balacescu
- Department of Functional Genomics and Experimental Pathology, The Oncology Institute "Prof Dr Ion Chiricuţă", Cluj-Napoca, Romania ; Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | | | - Bogdan Fetica
- Department of Pathology, The Oncology Institute "Prof Dr Ion Chiricuţă", Cluj-Napoca, Romania
| | - Claudia Gherman
- Department of Functional Genomics and Experimental Pathology, The Oncology Institute "Prof Dr Ion Chiricuţă", Cluj-Napoca, Romania
| | - Ovidiu Balacescu
- Department of Functional Genomics and Experimental Pathology, The Oncology Institute "Prof Dr Ion Chiricuţă", Cluj-Napoca, Romania
| | - Ioana Berindan-Neagoe
- Department of Functional Genomics and Experimental Pathology, The Oncology Institute "Prof Dr Ion Chiricuţă", Cluj-Napoca, Romania ; Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania ; Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| |
Collapse
|
27
|
Th17 Cell Plasticity and Functions in Cancer Immunity. BIOMED RESEARCH INTERNATIONAL 2015; 2015:314620. [PMID: 26583099 PMCID: PMC4637016 DOI: 10.1155/2015/314620] [Citation(s) in RCA: 168] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/03/2015] [Accepted: 06/01/2015] [Indexed: 12/12/2022]
Abstract
Th17 cells represent a particular subset of T helper lymphocytes characterized by high production of IL-17 and other inflammatory cytokines. Th17 cells participate in antimicrobial immunity at mucosal and epithelial barriers and particularly fight against extracellular bacteria and fungi. While a role for Th17 cells in promoting inflammation and autoimmune disorders has been extensively and elegantly demonstrated, it is still controversial whether and how Th17 cells influence tumor immunity. Although Th17 cells specifically accumulate in many different types of tumors compared to healthy tissues, the outcome might however differ from a tumor type to another. Th17 cells were consequently associated with both good and bad prognoses. The high plasticity of those cells toward cells exhibiting either anti-inflammatory or in contrast pathogenic functions might contribute to Th17 versatile functions in the tumor context. On one hand, Th17 cells promote tumor growth by inducing angiogenesis (via IL-17) and by exerting themselves immunosuppressive functions. On the other hand, Th17 cells drive antitumor immune responses by recruiting immune cells into tumors, activating effector CD8(+) T cells, or even directly by converting toward Th1 phenotype and producing IFN-γ. In this review, we are discussing the impact of the tumor microenvironment on Th17 cell plasticity and function and its implications in cancer immunity.
Collapse
|
28
|
T Helper Lymphocyte Subsets and Plasticity in Autoimmunity and Cancer: An Overview. BIOMED RESEARCH INTERNATIONAL 2015; 2015:327470. [PMID: 26583100 PMCID: PMC4637008 DOI: 10.1155/2015/327470] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Revised: 05/16/2015] [Accepted: 05/24/2015] [Indexed: 12/20/2022]
Abstract
In response to cytokine signalling and other factors, CD4-positive T lymphocytes differentiate into distinct populations that are characterized by the production of certain cytokines and are controlled by different master transcription factors. The spectrum of such populations, which was initially limited to Th1 and Th2 subsets, is currently broadened to include Th17 and Treg subsets, as well as a number of less studied subtypes, such as Tfh, Th9, and Th22. Although these subsets appear to be relatively stable, certain plasticity exists that allows for transition between the subsets and formation of hybrid transition forms. This provides the immune system flexibility needed for adequate response to pathogens but, at the same time, can play a role in the pathogenic processes in cases of deregulation. In this review, we will discuss the properties of T lymphocyte subsets and their plasticity, as well as its implications for cancer and autoimmune diseases.
Collapse
|
29
|
Boland S, Bourin A, Alen J, Geraets J, Schroeders P, Castermans K, Kindt N, Boumans N, Panitti L, Fransen S, Vanormelingen J, Stassen JM, Leysen D, Defert O. Design, Synthesis, and Biological Evaluation of Novel, Highly Active Soft ROCK Inhibitors. J Med Chem 2015; 58:4309-24. [DOI: 10.1021/acs.jmedchem.5b00308] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Sandro Boland
- Amakem Therapeutics, Agoralaan
Abis, 3590 Diepenbeek, Belgium
| | - Arnaud Bourin
- Amakem Therapeutics, Agoralaan
Abis, 3590 Diepenbeek, Belgium
| | - Jo Alen
- Amakem Therapeutics, Agoralaan
Abis, 3590 Diepenbeek, Belgium
| | - Jacques Geraets
- Amakem Therapeutics, Agoralaan
Abis, 3590 Diepenbeek, Belgium
| | | | | | - Nele Kindt
- Amakem Therapeutics, Agoralaan
Abis, 3590 Diepenbeek, Belgium
| | - Nicki Boumans
- Amakem Therapeutics, Agoralaan
Abis, 3590 Diepenbeek, Belgium
| | - Laura Panitti
- Amakem Therapeutics, Agoralaan
Abis, 3590 Diepenbeek, Belgium
| | - Silke Fransen
- Amakem Therapeutics, Agoralaan
Abis, 3590 Diepenbeek, Belgium
| | | | | | - Dirk Leysen
- CSD Farmakem, Elvire Boelensstraat
7, 9160 Lokeren, Belgium
| | - Olivier Defert
- Amakem Therapeutics, Agoralaan
Abis, 3590 Diepenbeek, Belgium
| |
Collapse
|
30
|
Wang T, Cunningham A, Dokun AO, Hazarika S, Houston K, Chen L, Lye RJ, Spolski R, Leonard WJ, Annex BH. Loss of interleukin-21 receptor activation in hypoxic endothelial cells impairs perfusion recovery after hindlimb ischemia. Arterioscler Thromb Vasc Biol 2015; 35:1218-25. [PMID: 25838422 PMCID: PMC4865891 DOI: 10.1161/atvbaha.115.305476] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Accepted: 03/10/2015] [Indexed: 01/19/2023]
Abstract
OBJECTIVE Surgical hindlimb ischemia (HLI) in mice has become a valuable preclinical model to study peripheral arterial disease. We previously identified that the different phenotypic outcomes after HLI across inbred mouse strains is related to a region on the short arm of mouse chromosome 7. The gene coding the interleukin-21 receptor (IL-21R) lies at the peak of association in this region. APPROACH AND RESULTS With quantitative real-time polymerase chain reaction, we found that a mouse strain with a greater ability to upregulate IL-21R after HLI had better perfusion recovery than a strain with no upregulation after HLI. Immunofluorescent staining of ischemic hindlimb tissue showed IL-21R expression on endothelial cells (ECs) from C57BL/6 mice. An EC-enriched fraction isolated from ischemic hindlimb muscle showed higher Il-21R levels than an EC-enriched fraction from nonischemic limbs. In vitro, human umbilical vein ECs showed elevated IL-21R expression after hypoxia and serum starvation. Under these conditions, IL-21 treatment increased cell viability, decreased cell apoptosis, and augmented tube formation. In vivo, either knockout Il21r or blocking IL-21 signaling by treating with IL-21R-Fc (fusion protein that blocks IL-21 binding to its receptor) in C57BL/6 mice resulted in less perfusion recovery after HLI. Both in vitro and in vivo modulation of the IL-21/IL-21R axis under hypoxic conditions resulted in increased signal transducer and activator of transcription 3 phosphorylation and a subsequent increase in the B-cell lymphoma leukemia-2/BCL-2-associated X protein ratio. CONCLUSION Our data indicate that IL-21R upregulation and ligand activation in hypoxic ECs may help perfusion recovery by limiting/preventing apoptosis and favoring cell survival and angiogenesis through the signal transducer and activator of transcription 3 pathway.
Collapse
Affiliation(s)
- Tao Wang
- From the Robert M. Berne Cardiovascular Research Center (T.W., A.C., A.O.D., S.H., K.H., L.C., R.J.L., B.H.A.) and Division of Cardiovascular Medicine, Department of Medicine (S.H., B.H.A.), University of Virginia, Charlottesville; and Laboratory of Molecular Immunology and the Immunology Center, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD (R.S., W.J.L.)
| | - Alexis Cunningham
- From the Robert M. Berne Cardiovascular Research Center (T.W., A.C., A.O.D., S.H., K.H., L.C., R.J.L., B.H.A.) and Division of Cardiovascular Medicine, Department of Medicine (S.H., B.H.A.), University of Virginia, Charlottesville; and Laboratory of Molecular Immunology and the Immunology Center, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD (R.S., W.J.L.)
| | - Ayotunde O Dokun
- From the Robert M. Berne Cardiovascular Research Center (T.W., A.C., A.O.D., S.H., K.H., L.C., R.J.L., B.H.A.) and Division of Cardiovascular Medicine, Department of Medicine (S.H., B.H.A.), University of Virginia, Charlottesville; and Laboratory of Molecular Immunology and the Immunology Center, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD (R.S., W.J.L.)
| | - Surovi Hazarika
- From the Robert M. Berne Cardiovascular Research Center (T.W., A.C., A.O.D., S.H., K.H., L.C., R.J.L., B.H.A.) and Division of Cardiovascular Medicine, Department of Medicine (S.H., B.H.A.), University of Virginia, Charlottesville; and Laboratory of Molecular Immunology and the Immunology Center, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD (R.S., W.J.L.)
| | - Kevin Houston
- From the Robert M. Berne Cardiovascular Research Center (T.W., A.C., A.O.D., S.H., K.H., L.C., R.J.L., B.H.A.) and Division of Cardiovascular Medicine, Department of Medicine (S.H., B.H.A.), University of Virginia, Charlottesville; and Laboratory of Molecular Immunology and the Immunology Center, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD (R.S., W.J.L.)
| | - Lingdan Chen
- From the Robert M. Berne Cardiovascular Research Center (T.W., A.C., A.O.D., S.H., K.H., L.C., R.J.L., B.H.A.) and Division of Cardiovascular Medicine, Department of Medicine (S.H., B.H.A.), University of Virginia, Charlottesville; and Laboratory of Molecular Immunology and the Immunology Center, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD (R.S., W.J.L.)
| | - R John Lye
- From the Robert M. Berne Cardiovascular Research Center (T.W., A.C., A.O.D., S.H., K.H., L.C., R.J.L., B.H.A.) and Division of Cardiovascular Medicine, Department of Medicine (S.H., B.H.A.), University of Virginia, Charlottesville; and Laboratory of Molecular Immunology and the Immunology Center, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD (R.S., W.J.L.)
| | - Rosanne Spolski
- From the Robert M. Berne Cardiovascular Research Center (T.W., A.C., A.O.D., S.H., K.H., L.C., R.J.L., B.H.A.) and Division of Cardiovascular Medicine, Department of Medicine (S.H., B.H.A.), University of Virginia, Charlottesville; and Laboratory of Molecular Immunology and the Immunology Center, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD (R.S., W.J.L.)
| | - Warren J Leonard
- From the Robert M. Berne Cardiovascular Research Center (T.W., A.C., A.O.D., S.H., K.H., L.C., R.J.L., B.H.A.) and Division of Cardiovascular Medicine, Department of Medicine (S.H., B.H.A.), University of Virginia, Charlottesville; and Laboratory of Molecular Immunology and the Immunology Center, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD (R.S., W.J.L.)
| | - Brian H Annex
- From the Robert M. Berne Cardiovascular Research Center (T.W., A.C., A.O.D., S.H., K.H., L.C., R.J.L., B.H.A.) and Division of Cardiovascular Medicine, Department of Medicine (S.H., B.H.A.), University of Virginia, Charlottesville; and Laboratory of Molecular Immunology and the Immunology Center, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD (R.S., W.J.L.).
| |
Collapse
|
31
|
Xuan W, Yan Y, Wan M, Wu X, Ji D, Wang L, Lin C, Chen Y, Yu Y, Zhang X. Antitumor activity of mHSP65-TTL enhanced by administration of low dose cyclophosphamide in pancreatic cancer-bearing mice. Int Immunopharmacol 2015; 27:95-103. [PMID: 25907246 DOI: 10.1016/j.intimp.2015.04.014] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2014] [Revised: 04/07/2015] [Accepted: 04/07/2015] [Indexed: 12/13/2022]
Abstract
Pancreatic cancer remains a lethal malignancy. Despite chemotherapy or/and radiotherapy after the surgery, the improvement on the overall survival of the patients has still been minimal. To develop novel therapeutic approaches, we tried to prepare mHSP65-TTL, a candidate vaccine prepared by mixing the recombinant mycobacterial heat shock protein 65 (mHSP65) with tumor tissue lysate (TTL) of Panc02 pancreatic cancer tissue. The mHSP65-TTL were used to immune the C57BL/6 mice implanted with the Panc02 cancer cells, in combination with or without low dose cyclophosphamide (CY). The results showed that mHSP65-TTL significantly prolonged the survival of the pancreatic cancer bearing mice and low dose CY enhanced the efficacy of the mHSP65-TTL. In addition, we detected mRNA expression of RORγt and IL-17A in spleen cells of mice received mHSP65-TTL or mHSP65-TTL plus CY, and found that mHSP65-TTL up-regulated mRNA expressions of RORγt and IL-17A, CY alone or mHSP65-TTL plus CY up-regulated mRNA expressions of RORγt. The work could provide an insight into a combinational approach for the immunotherapy of pancreatic cancer.
Collapse
Affiliation(s)
- Wei Xuan
- Department of Hepatobiliary-Pancreatic Surgery, Third Hospital (China-Japan Union Hospital) of Jilin University, Changchun 130021, China
| | - Youyou Yan
- Department of Immunology, Norman Bethune College of Medicine, Jilin University, Changchun 130021, China
| | - Min Wan
- Department of Molecular Biology, Norman Bethune College of Medicine, Jilin University, Changchun 130021, China
| | - Xiuli Wu
- Department of Molecular Biology, Norman Bethune College of Medicine, Jilin University, Changchun 130021, China
| | - Degang Ji
- Department of Hepatobiliary-Pancreatic Surgery, Third Hospital (China-Japan Union Hospital) of Jilin University, Changchun 130021, China
| | - Liying Wang
- Department of Molecular Biology, Norman Bethune College of Medicine, Jilin University, Changchun 130021, China
| | - Chao Lin
- Department of Hepatobiliary-Pancreatic Surgery, Third Hospital (China-Japan Union Hospital) of Jilin University, Changchun 130021, China
| | - Yang Chen
- Department of Hepatobiliary-Pancreatic Surgery, Third Hospital (China-Japan Union Hospital) of Jilin University, Changchun 130021, China
| | - Yongli Yu
- Department of Immunology, Norman Bethune College of Medicine, Jilin University, Changchun 130021, China.
| | - Xuewen Zhang
- Department of Hepatobiliary-Pancreatic Surgery, Third Hospital (China-Japan Union Hospital) of Jilin University, Changchun 130021, China.
| |
Collapse
|
32
|
Davis MR, Zhu Z, Hansen DM, Bai Q, Fang Y. The role of IL-21 in immunity and cancer. Cancer Lett 2015; 358:107-114. [DOI: 10.1016/j.canlet.2014.12.047] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Revised: 12/17/2014] [Accepted: 12/19/2014] [Indexed: 01/05/2023]
|
33
|
Abstract
Angiostatic therapies are now routinely embedded in the daily clinical management of cancer. Although these agents clearly benefit patient survival rates, the effect is only moderate with sometimes considerable side effects. A major cause of failure in this respect is the induction of resistance and tolerability against these drugs. Most angiostatic drugs are tyrosine kinase inhibitors that aim to inhibit or neutralize the activity of tumour-produced growth factors. Frustrating the tumour cells in this way results in genetic adaptations in the cells, turning them into mutants that are dependent on other growth mechanisms. It may therefore be necessary to shift to another class of drugs that directly target the tumour vasculature. It is evident that improvement of future angiogenesis inhibitors can only arise from two efforts. First, through the identification of better targets, preferably specifically expressed in the tumour vasculature. Secondly, through the development of combination therapies. The present review highlights the current efforts and challenges in trying to develop effective angiostatic combination therapies.
Collapse
|
34
|
Yang B, Kang H, Fung A, Zhao H, Wang T, Ma D. The role of interleukin 17 in tumour proliferation, angiogenesis, and metastasis. Mediators Inflamm 2014; 2014:623759. [PMID: 25110397 PMCID: PMC4119694 DOI: 10.1155/2014/623759] [Citation(s) in RCA: 93] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2014] [Accepted: 06/25/2014] [Indexed: 02/08/2023] Open
Abstract
With 7.6 million deaths globally, cancer according to the World Health Organisation is still one of the leading causes of death worldwide. Interleukin 17 (IL-17) is a cytokine produced by Th17 cells, a T helper cell subset developed from an activated CD4+ T-cell. Whilst the importance of IL-17 in human autoimmune disease, inflammation, and pathogen defence reactions has already been established, its potential role in cancer progression still needs to be updated. Interestingly studies have demonstrated that IL-17 plays an intricate role in the pathophysiology of cancer, from tumorigenesis, proliferation, angiogenesis, and metastasis, to adapting the tumour in its ability to confer upon itself both immune, and chemotherapy resistance. This review will look into IL-17 and summarise the current information and data on its role in the pathophysiology of cancer as well as its potential application in the overall management of the disease.
Collapse
Affiliation(s)
- Bob Yang
- Department of Anesthesiology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
- Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea & Westminster Hospital, London SW10 9NH, UK
| | - Heechan Kang
- Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea & Westminster Hospital, London SW10 9NH, UK
| | - Anthony Fung
- Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea & Westminster Hospital, London SW10 9NH, UK
| | - Hailin Zhao
- Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea & Westminster Hospital, London SW10 9NH, UK
| | - Tianlong Wang
- Department of Anesthesiology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Daqing Ma
- Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea & Westminster Hospital, London SW10 9NH, UK
| |
Collapse
|
35
|
Bailey SR, Nelson MH, Himes RA, Li Z, Mehrotra S, Paulos CM. Th17 cells in cancer: the ultimate identity crisis. Front Immunol 2014; 5:276. [PMID: 24987392 PMCID: PMC4060300 DOI: 10.3389/fimmu.2014.00276] [Citation(s) in RCA: 225] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2014] [Accepted: 05/27/2014] [Indexed: 12/12/2022] Open
Abstract
T helper 17 (Th17) cells play a complex and controversial role in tumor immunity and have been found to exhibit a fluctuating identity within the context of cancer. The recent, expanding literature on these cells attests to their puzzling nature, either promoting or suppressing tumor growth depending on the malignancy and course of therapeutic intervention investigated. This review addresses several newly appreciated factors that may help delineate Th17 cells' immunological properties in the context of cancer. Several reports suggest that inflammatory signals induced in the tumor milieu regulate the functional fate and antitumor activity of Th17 cells. Recent findings also point to significant alterations in Th17 cells due to their interplay with regulatory T lymphocytes and cytotoxic CD8(+) T cells within the tumor microenvironment. Finally, an appreciation for the stem cell-like properties of Th17 cells that augment their persistence and activity emerges from recent reports. The impact of these factors on Th17 cells' antitumor efficacy and how these factors may be exploited to improve cancer therapies will be discussed.
Collapse
Affiliation(s)
- Stefanie R Bailey
- Department of Microbiology and Immunology, Medical University of South Carolina , Charleston, SC , USA ; Department of Surgery, Medical University of South Carolina , Charleston, SC , USA
| | - Michelle H Nelson
- Department of Microbiology and Immunology, Medical University of South Carolina , Charleston, SC , USA ; Department of Surgery, Medical University of South Carolina , Charleston, SC , USA
| | - Richard A Himes
- Department of Chemistry, College of Charleston , Charleston, SC , USA
| | - Zihai Li
- Department of Microbiology and Immunology, Medical University of South Carolina , Charleston, SC , USA
| | - Shikhar Mehrotra
- Department of Surgery, Medical University of South Carolina , Charleston, SC , USA
| | - Chrystal M Paulos
- Department of Microbiology and Immunology, Medical University of South Carolina , Charleston, SC , USA ; Department of Surgery, Medical University of South Carolina , Charleston, SC , USA
| |
Collapse
|
36
|
Vivanco-Cid H, Maldonado-Rentería MJ, Sánchez-Vargas LA, Izaguirre-Hernández IY, Hernández-Flores KG, Remes-Ruiz R. Dynamics of interleukin-21 production during the clinical course of primary and secondary dengue virus infections. Immunol Lett 2014; 161:89-95. [PMID: 24858204 DOI: 10.1016/j.imlet.2014.05.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2013] [Revised: 04/22/2014] [Accepted: 05/07/2014] [Indexed: 12/12/2022]
Abstract
Previous studies have revealed the clinical relevance of pro-inflammatory cytokine production during dengue virus (DENV) infections. In this study, we evaluated the production of interleukin-21 (IL-21), a key soluble mediator mainly produced by CD4+ T cells. The aim of this study was to investigate the role of IL-21 production during the clinical course of primary and secondary DENV infections and the potential association of IL-21 serum levels with the disease pathogenesis. Blood samples from DENV-infected patients were collected on different days after the onset of symptoms. Patients were classified according to their phase of disease (acute vs. convalescent phases), the type of infection (primary vs. secondary), and the clinical severity of their disease (dengue fever (DF) vs. dengue hemorrhagic fever (DHF)). IL-21 levels were measured using a quantitative capture ELISA assay. The levels of IL-21 were significantly elevated in the disease group compared with the control group. IL-21 was detected in primary and secondary DENV infections, with a significantly higher concentration in the convalescent phase of primary infections. IL-21 levels were significantly higher in patients with secondary acute DHF infections when compared with those with secondary acute DF infection. There was a relationship between the elevated serum levels of IL-21 and the production of DENV-specific IgM and IgG antibodies. Taking together, our results show for the first time the involvement of IL-21 during the clinical course of DENV infections. We speculate that IL-21 may play a protective role in the context of the convalescent phase of primary infections and the acute phase of secondary infections.
Collapse
Affiliation(s)
- H Vivanco-Cid
- Instituto de Investigaciones Medico-Biológicas, Universidad Veracruzana, Veracruz, México; Universidad del Valle de México, campus Villa Rica, Facultad de Medicina "Dr. Porfirio Sosa Zárate", México.
| | - M J Maldonado-Rentería
- Instituto de Investigaciones Medico-Biológicas, Universidad Veracruzana, Veracruz, México
| | - L A Sánchez-Vargas
- Instituto de Investigaciones Medico-Biológicas, Universidad Veracruzana, Veracruz, México
| | | | - K G Hernández-Flores
- Instituto de Investigaciones Medico-Biológicas, Universidad Veracruzana, Veracruz, México
| | - R Remes-Ruiz
- Hospital Regional de Alta Especialidad de Veracruz, Servicios de Salud de Veracruz, México
| |
Collapse
|
37
|
Olewicz-Gawlik A, Danczak-Pazdrowska A, Kuznar-Kaminska B, Gornowicz-Porowska J, Katulska K, Trzybulska D, Batura-Gabryel H, Silny W, Poplawski D, Hrycaj P. Interleukin-17 and interleukin-23: importance in the pathogenesis of lung impairment in patients with systemic sclerosis. Int J Rheum Dis 2014; 17:664-70. [DOI: 10.1111/1756-185x.12290] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Anna Olewicz-Gawlik
- Department of Rheumatology and Clinical Immunology; Poznan University of Medical Sciences; Poznan Poland
| | | | - Barbara Kuznar-Kaminska
- Department of Pulmonology, Allergology and Pulmonary Oncology; Poznan University of Medical Sciences; Poznan Poland
| | | | - Katarzyna Katulska
- Department of General Radiology and Neuroradiology; Poznan University of Medical Sciences; Poznan Poland
| | - Dorota Trzybulska
- Department of Rheumatology and Clinical Immunology; Poznan University of Medical Sciences; Poznan Poland
| | - Halina Batura-Gabryel
- Department of Pulmonology, Allergology and Pulmonary Oncology; Poznan University of Medical Sciences; Poznan Poland
| | - Wojciech Silny
- Department of Dermatology; Poznan University of Medical Sciences; Poznan Poland
| | - Dariusz Poplawski
- Department of Pulmonology, Allergology and Pulmonary Oncology; Poznan University of Medical Sciences; Poznan Poland
| | - Pawel Hrycaj
- Department of Rheumatology and Clinical Immunology; Poznan University of Medical Sciences; Poznan Poland
| |
Collapse
|
38
|
Bhatia S, Curti B, Ernstoff MS, Gordon M, Heath EI, Miller WH, Puzanov I, Quinn DI, Flaig TW, VanVeldhuizen P, Byrnes-Blake K, Freeman JA, Bittner R, Hunder N, Souza S, Thompson JA. Recombinant interleukin-21 plus sorafenib for metastatic renal cell carcinoma: a phase 1/2 study. J Immunother Cancer 2014; 2:2. [PMID: 24829759 PMCID: PMC4019894 DOI: 10.1186/2051-1426-2-2] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2013] [Accepted: 01/20/2014] [Indexed: 01/27/2023] Open
Abstract
Background Despite the positive impact of targeted therapies on metastatic renal cell carcinoma (mRCC), durable responses are infrequent and an unmet need exists for novel therapies with distinct mechanisms of action. We investigated the combination of recombinant Interleukin 21 (IL-21), a cytokine with unique immunostimulatory properties, plus sorafenib, a VEGFR tyrosine kinase inhibitor. Methods In this phase 1/2 study, 52 mRCC patients received outpatient treatment with oral sorafenib 400 mg twice daily plus intravenous IL-21 (10–50 mcg/kg) on days 1–5 and 15–19 of each 7-week treatment course. The safety, antitumor activity, pharmacokinetic and pharmacodynamic effects of the combination were evaluated. Results In phase 1 (n = 19), the maximum tolerated dose for IL-21 with the standard dose of sorafenib was determined to be 30 mcg/kg/day; grade 3 skin rash was the only dose-limiting toxicity. In phase 2, 33 previously-treated patients tolerated the combination therapy well with appropriate dose reductions; toxicities were mostly grade 1 or 2. The objective response rate was 21% and disease control rate was 82%. Two patients have durable responses that are ongoing, despite cessation of both IL-21 and sorafenib, at 41+ and 30+ months, respectively. The median progression-free survival in phase 2 was 5.6 months. The pharmacokinetic and pharmacodynamic properties of IL-21 appeared to be preserved in the presence of sorafenib. Conclusion IL-21 plus sorafenib has antitumor activity and acceptable safety in previously treated mRCC patients. IL-21 may represent a suitable immunotherapy in further exploration of combination strategies in mRCC. Trial registration ClinicalTrials.gov Identifier: NCT00389285
Collapse
Affiliation(s)
- Shailender Bhatia
- University of Washington, Seattle, WA, USA ; Division of Medical Oncology, Department of Medicine, University of Washington, Seattle Cancer Care Alliance, 825 Eastlake Ave East, Mailstop G4-830, Seattle, WA 98109-1023, USA
| | - Brendan Curti
- Providence Portland Medical Center, Portland, OR, USA
| | - Marc S Ernstoff
- Norris Cotton Cancer Center, Dartmouth Hitchcock Medical Center, Lebanon, NH, USA
| | | | | | - Wilson H Miller
- Lady Davis Institute and Segal Cancer Centre, Jewish General Hospital, McGill University, Montreal, Quebec
| | - Igor Puzanov
- Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, TN, USA
| | - David I Quinn
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
| | | | | | | | - Jeremy A Freeman
- Formerly of ZymoGenetics (Bristol-Myers Squibb), Seattle, WA, USA
| | | | - Naomi Hunder
- Formerly of ZymoGenetics (Bristol-Myers Squibb), Seattle, WA, USA
| | - Sonia Souza
- Formerly of ZymoGenetics (Bristol-Myers Squibb), Seattle, WA, USA
| | | |
Collapse
|
39
|
Martin F, Apetoh L, Ghiringhelli F. Controversies on the role of Th17 in cancer: a TGF-β-dependent immunosuppressive activity? Trends Mol Med 2012; 18:742-9. [PMID: 23083809 DOI: 10.1016/j.molmed.2012.09.007] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2012] [Revised: 09/13/2012] [Accepted: 09/22/2012] [Indexed: 01/29/2023]
Abstract
The immune system has important roles in limiting the spread of cancer and shaping the tumor microenvironment. Although the contributions of T helper 17 (Th17) cells (a subtype of CD4(+) T lymphocytes) to autoimmunity and allergy response are well known, their roles in cancer remain ambiguous. Despite adoptive transfer studies indicating that mouse Th17 cells support anticancer immunity, the Th17 cells that naturally infiltrate experimental tumors appear to have a tumor-promoting effect. These contradictory properties can be related to the high degree of plasticity inherent in Th17 cells and their capacity to differentiate into tumoricidal Th1-like cells. Mouse Th17 cells induced by transforming growth factor-β (TGF-β) express CD39 and CD73 ectonucleotidases on their surfaces, which leads to adenosine release and suppression of T cell immunity. Here, we discuss how TGF-β acts as a molecular switch controlling the immunoregulatory properties of Th17 cells.
Collapse
|
40
|
Elishmereni M, Kheifetz Y, Søndergaard H, Overgaard RV, Agur Z. An integrated disease/pharmacokinetic/pharmacodynamic model suggests improved interleukin-21 regimens validated prospectively for mouse solid cancers. PLoS Comput Biol 2011; 7:e1002206. [PMID: 22022259 PMCID: PMC3182868 DOI: 10.1371/journal.pcbi.1002206] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2011] [Accepted: 08/08/2011] [Indexed: 11/20/2022] Open
Abstract
Interleukin (IL)-21 is an attractive antitumor agent with potent immunomodulatory functions. Yet thus far, the cytokine has yielded only partial responses in solid cancer patients, and conditions for beneficial IL-21 immunotherapy remain elusive. The current work aims to identify clinically-relevant IL-21 regimens with enhanced efficacy, based on mathematical modeling of long-term antitumor responses. For this purpose, pharmacokinetic (PK) and pharmacodynamic (PD) data were acquired from a preclinical study applying systemic IL-21 therapy in murine solid cancers. We developed an integrated disease/PK/PD model for the IL-21 anticancer response, and calibrated it using selected “training” data. The accuracy of the model was verified retrospectively under diverse IL-21 treatment settings, by comparing its predictions to independent “validation” data in melanoma and renal cell carcinoma-challenged mice (R2>0.90). Simulations of the verified model surfaced important therapeutic insights: (1) Fractionating the standard daily regimen (50 µg/dose) into a twice daily schedule (25 µg/dose) is advantageous, yielding a significantly lower tumor mass (45% decrease); (2) A low-dose (12 µg/day) regimen exerts a response similar to that obtained under the 50 µg/day treatment, suggestive of an equally efficacious dose with potentially reduced toxicity. Subsequent experiments in melanoma-bearing mice corroborated both of these predictions with high precision (R2>0.89), thus validating the model also prospectively in vivo. Thus, the confirmed PK/PD model rationalizes IL-21 therapy, and pinpoints improved clinically-feasible treatment schedules. Our analysis demonstrates the value of employing mathematical modeling and in silico-guided design of solid tumor immunotherapy in the clinic. Among the many potential drugs explored within the scope of cancer immunotherapy are selected cytokines which possess promising immune-boosting properties. Yet, the natural involvement of these proteins in multiple, often contradicting biological processes can complicate their use in the clinic. The cytokine interleukin (IL)-21 is no exception: while its strength as an anticancer agent has been established in several animal studies, response rates in melanoma and renal cell carcinoma patients remain low. To help guide the design of effective IL-21 therapy, we have developed a mathematical model that bridges between the complex biology of IL-21 and its optimal clinical use. Our model integrates data from preclinical studies under diverse IL-21 treatment settings, and was validated by extensive experiments in tumor-bearing mice. Model simulations predicted that beneficial, clinically practical IL-21 therapy should be composed of low-dose schedules, and/or schedules in which several partial doses are administered rather than a single complete dose. These findings were subsequently confirmed in mice with melanoma. Thus, future testing of these strategies in solid cancer patients can be a promising starting point for improving IL-21 therapy. Our model can thus provide a computational platform for rationalizing IL-21 regimens and streamlining its clinical development.
Collapse
Affiliation(s)
| | - Yuri Kheifetz
- Institute for Medical Biomathematics (IMBM), Bene-Ataroth, Israel
| | | | | | - Zvia Agur
- Institute for Medical Biomathematics (IMBM), Bene-Ataroth, Israel
- Optimata Ltd., Ramat-Gan, Israel
- * E-mail:
| |
Collapse
|
41
|
Angiogenesis and immunity: a bidirectional link potentially relevant for the monitoring of antiangiogenic therapy and the development of novel therapeutic combination with immunotherapy. Cancer Metastasis Rev 2011; 30:83-95. [PMID: 21249423 DOI: 10.1007/s10555-011-9281-4] [Citation(s) in RCA: 201] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The immune system regulates angiogenesis in cancer with both pro- and antiangiogenic activities. The induction of angiogenesis is mediated by tumor-associated macrophages and myeloid-derived suppressor cells (MDSC) which produce proinflammatory cytokines, endothelial growth factors (VEGF, bFGF…), and protease (MMP9) implicated in neoangiogenesis. Some cytokines (IL-6, IL-17…) activated Stat3 which also led to the production of VEGF and bFGF. In contrast, other cytokines (IFN, IL-12, IL-21, and IL-27) display an antiangiogenic activity. Recently, it has been shown that some antiangiogenic molecules alleviates immunosuppression associated with cancer by decreasing immunosuppressive cells (MDSC, regulatory T cells), immunosuppressive cytokines (IL-10, TGFβ), and inhibitory molecules on T cells (PD-1). Some of these broad effects may result from the ability of some antiangiogenic molecules, especially cytokines to inhibit the Stat3 transcription factor. The association often observed between angiogenesis and immunosuppression may be related to hypoxia which induces both neoangiogenesis via activation of HIF-1 and VEGF and favors the intratumor recruitment and differentiation of regulatory T cells and MDSC. Preliminary studies suggest that modulation of immune markers (intratumoral MDSC and IL-8, peripheral regulatory T cells…) may predict clinical response to antiangiogenic therapy. In preclinical models, a synergy has been observed between antiangiogenic molecules and immunotherapy which may be explained by an improvement of immune status in tumor-bearing mice after antiangiogenic therapy. In preclinical models, antiangiogenic molecules promoted intratumor trafficking of effector cells, enhance endogenous anti-tumor response, and synergyzed with immunotherapy protocols to cure established murine tumors. All these results warrant the development of clinical trials combining antiangiogenic drugs and immunotherapy.
Collapse
|
42
|
Larochelle C, Alvarez JI, Prat A. How do immune cells overcome the blood-brain barrier in multiple sclerosis? FEBS Lett 2011; 585:3770-80. [PMID: 21550344 DOI: 10.1016/j.febslet.2011.04.066] [Citation(s) in RCA: 271] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2011] [Revised: 04/26/2011] [Accepted: 04/27/2011] [Indexed: 11/17/2022]
Abstract
The presence of the blood-brain barrier (BBB) restricts the movement of soluble mediators and leukocytes from the periphery to the central nervous system (CNS). Leukocyte entry into the CNS is nonetheless an early event in multiple sclerosis (MS), an inflammatory disorder of the CNS. Whether BBB dysfunction precedes immune cell infiltration or is the consequence of perivascular leukocyte accumulation remains enigmatic, but leukocyte migration modifies BBB permeability. Immune cells of MS subjects express inflammatory cytokines, reactive oxygen species (ROS) and enzymes that can facilitate their migration to the CNS by influencing BBB function, either directly or indirectly. In this review, we describe how immune cells from the peripheral blood overcome the BBB and promote CNS inflammation in MS through BBB disruption.
Collapse
Affiliation(s)
- Catherine Larochelle
- Neuroimmunology Research Laboratory, Center of Excellence in Neuromics, CRCHUM, Notre-Dame Hospital, Faculty of Medicine, Université de Montréal, Montréal, Québec, Canada
| | | | | |
Collapse
|
43
|
Kesselring R, Thiel A, Pries R, Trenkle T, Wollenberg B. Human Th17 cells can be induced through head and neck cancer and have a functional impact on HNSCC development. Br J Cancer 2010; 103:1245-54. [PMID: 20877351 PMCID: PMC2967064 DOI: 10.1038/sj.bjc.6605891] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Background: The T helper 17 (Th17) cells recently identified as distinct T helper cell lineage are characterised by their production of the proinflammatory cytokine interleukin 17. Although much effort has been made in understanding the function of Th17 cells in the pathogenesis of different diseases, their influence in carcinogenesis remain largely unknown. Methods: We studied the prevalence and induction of Th17 cells in head and neck squamous cell carcinoma (HNSCC) patients by flow cytometry. To determine the migration mechanism of Th17 cells into primary tumours and metastasis of HNSCC, we performed chemotaxis assays. We analysed the proliferation and the angiogenesis-related proteins of HNSCCs in the presence of Th17 cells with MTT-based proliferation assay and an angiogenesis protein array. Results: In this study, we showed that the prevalence of Th17 cells is elevated in peripheral blood of HNSCC patients. In addition, tumour tissue and tumour-draining lymph nodes are infiltrated by a huge number of Th17 cells representing an important fraction of the tumour-infiltrating lymphocytes (TILs). We further showed that Th17 cells can be induced and expanded in tumour microenvironment through cytokines produced by tumour cells and TILs, and in addition can be recruited to the tumour milieu through a CCR6/CCL20-dependent mechanism. Furthermore, we showed that the proliferation and angiogenesis of HNSCC are impaired in the presence of Th17 cells. Conclusion: We conclude that Th17 cells have a substantial impact on the carcinogenesis of HNSCCs and on their metastasis and could serve as a potential therapeutic target to modulate anti-tumour response in HNSCC.
Collapse
Affiliation(s)
- R Kesselring
- Department for Otorhinolaryngology, University of Luebeck, Ratzeburger Allee 160, Luebeck 23538, Germany
| | | | | | | | | |
Collapse
|
44
|
Riaz W, Hernandez-Ilizaliturri FJ, Czuczman MS. Strategies to enhance rituximab anti-tumor activity in the treatment of CD20-positive B-cell neoplasms. Immunol Res 2010; 46:192-205. [PMID: 19763890 DOI: 10.1007/s12026-009-8121-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Rituximab is a chimeric monoclonal anti-CD20 antibody and was the first monoclonal antibody (mAb) therapy approved by FDA (Food and Drug Administration) for the treatment of B-cell lymphoma. It has revolutionized the treatment of patients with CD20-positive non-Hodgkin's lymphoma and CLL. Rituximab is currently being used in virtually all patients with B-cell lymphomas either alone or in combination with chemotherapy. Despite its excellent safety and efficacy profile, only a small portion of B-cell lymphoma patients treated with rituximab as a single agent have sustained complete remissions. Combining rituximab with standard chemotherapy regimens is associated with higher response rates, and improved survival in a subset of patients. Unfortunately, a significant percentage of patients who initially respond to rituximab eventually relapse, and there are patients that demonstrate intrinsic resistance to initial therapy. In the last decade, ongoing scientific research has led to a better understanding of rituximab-associated cytotoxic mechanisms against lymphoma target cells. Scientific efforts are increasingly being focused in developing new strategies to improve mAb activity. Various strategies include the following: combining rituximab with different biologic agents (e.g. cytokines, immunomodulatory drugs); developing novel antibody constructs (including bi-specific antibodies); and/or inhibiting signaling pathways associated with lymphomagenesis and immuno-chemotherapy resistance. In this review article, we will provide an overview of various rituximab-associated cytotoxic mechanisms and novel strategies to improve mAb activity against B-cell lymphoma.
Collapse
Affiliation(s)
- Wasif Riaz
- Department of Medicine, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| | | | | |
Collapse
|
45
|
Kim H, Kang JS, Lee WJ. The Production IL-21 and VEGF in UVB-irradiated Human Keratinocyte Cell Line, HaCaT. Immune Netw 2010; 10:75-80. [PMID: 20532127 PMCID: PMC2881424 DOI: 10.4110/in.2010.10.2.75] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2010] [Revised: 04/08/2010] [Accepted: 04/13/2010] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Ultraviolet B (UVB) induces multiple inflammatory and carcinogenic reactions. In skin, UVB induces to secrete several kinds of inflammatory cytokines from keratinocytes and also increases angiogenic process via the modulation of vascular endothelial growth factor (VEGF) production. Interleukin-21 (IL-21) is an inflammatory cytokine and produced by activated T cells. The biologic functions of IL-21 have not yet extensively studied. METHODS In the present study, we investigate the production of IL-21 from human keratinocyte cell line, HaCaT and its biological effect after exposure to UVB. RESULTS First, we confirmed the IL-21 production and its receptor expression in HaCaT. And then, the change of IL-21 and VEGF production in HaCaT by UVB irradiation was examined. Not only IL-21 but also VEGF production was enhanced by UVB irradiation. Next, to determine relationship of enhanced production of IL-21 and VEGF, we detected VEGF production after neutralization of IL-21. VEGF production was reduced by IL-21 neutralization, which indicates that the IL-21 is involved in the VEGF production. CONCLUSION Taken together, our results suggest that IL-21 and VEGF production is enhanced by UVB irradiation in HaCaT. In addition, it seems that IL-21 plays a role in the angiogenic process in skin via the modulation of VEGF production.
Collapse
Affiliation(s)
- Hyemin Kim
- Department of Anatomy and Tumor Immunity Medical Research Center, Seoul National University College of Medicine, Seoul 110-799, Korea
| | | | | |
Collapse
|
46
|
The Chick Embryo Chorioallantoic Membrane as an In Vivo Assay to Study Antiangiogenesis. Pharmaceuticals (Basel) 2010; 3:482-513. [PMID: 27713265 PMCID: PMC4033966 DOI: 10.3390/ph3030482] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2010] [Revised: 01/29/2010] [Accepted: 03/02/2010] [Indexed: 12/15/2022] Open
Abstract
Antiangiogenesis, e.g., inhibition of blood vessel growth, is being investigated as a way to prevent the growth of tumors and other angiogenesis-dependent diseases. Pharmacological inhibition interferes with the angiogenic cascade or the immature neovasculature with synthetic or semi-synthetic substances, endogenous inhibitors or biological antagonists.The chick embryo chorioallantoic membrane (CAM) is an extraembryonic membrane, which serves as a gas exchange surface and its function is supported by a dense capillary network. Because its extensive vascularization and easy accessibility, CAM has been used to study morphofunctional aspects of the angiogenesis process in vivo and to study the efficacy and mechanism of action of pro- and anti-angiogenic molecules. The fields of application of CAM in the study of antiangiogenesis, including our personal experience, are illustrated in this review article.
Collapse
|
47
|
Abstract
Cytokines are secreted signalling molecules with decisive effects on haematopoiesis, innate and adaptive immunity, and immunopathology. Interleukin (IL)-21 is a novel cytokine produced by activated CD4(+) T cells and natural killer T (NKT) cells. IL-21 is part of a family of cytokines which include IL-2, -4, -7, -9 and -15 that all share the common IL-2 receptor gamma chain (gamma(c)) in their individual receptor complexes. IL-21 receptor (IL-21R) is widely expressed on both myeloid and lymphoid cell lineages and IL-21 actions include co-stimulation of B cell differentiation and immunoglobulin (Ig) production, co-mitogen of T cells, and stimulation of NK and CD8(+) T cell cytotoxic function. Initially, IL-21 was recognized for its anti-tumour effects in several preclinical tumour models, warranting its currently ongoing clinical development as a cancer immunotherapeutic. More recently, IL-21 has been associated with the development of a panel of autoimmune and inflammatory diseases, where neutralization of IL-21 has been suggested as a potential new therapy. In this review, we will cover the latest discoveries of IL-21 as a cancer therapy and its implications in immunopathologies.
Collapse
Affiliation(s)
- H Søndergaard
- Department of Immunopharmacology, Novo Nordisk A/S, Måløv, Denmark.
| | | |
Collapse
|
48
|
Davis ID, Brady B, Kefford RF, Millward M, Cebon J, Skrumsager BK, Mouritzen U, Hansen LT, Skak K, Lundsgaard D, Frederiksen KS, Kristjansen PEG, McArthur G. Clinical and biological efficacy of recombinant human interleukin-21 in patients with stage IV malignant melanoma without prior treatment: a phase IIa trial. Clin Cancer Res 2009; 15:2123-9. [PMID: 19276257 DOI: 10.1158/1078-0432.ccr-08-2663] [Citation(s) in RCA: 113] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Human interleukin-21 (IL-21) is a class I cytokine that mediates activation of CD8(+) T cells, natural killer (NK) cells, and other cell types. We report final clinical and biological results of a phase II study of recombinant human IL-21 (rIL-21) in patients with metastatic melanoma. EXPERIMENTAL DESIGN Open-label, single-arm, two-stage trial. ELIGIBILITY CRITERIA unresectable metastatic melanoma, measurable disease by Response Evaluation Criteria in Solid Tumors, no prior systemic therapy (adjuvant IFN permitted), adequate major organ function, good performance status, no significant autoimmune disease, and life expectancy at least 4 months. PRIMARY OBJECTIVE antitumor efficacy (response rate). SECONDARY OBJECTIVES safety, blood biomarkers, and generation of anti-rIL-21 antibodies. rIL-21 (30 microg/kg/dose) was administered by intravenous bolus injection in 8-week cycles (5 dosing days followed by 9 days of rest for 6 weeks and then 2 weeks off treatment). RESULTS Stage I of the study comprised 14 patients. One confirmed complete response (CR) was observed, and as per protocol, 10 more patients were accrued to stage II (total n = 24: 10 female and 14 male). Best tumor response included one confirmed CR and one confirmed partial response, both with lung metastases. Treatment was overall well tolerated. Biomarker analyses showed increases in serum soluble CD25, frequencies of CD25(+) NK and CD8(+) T cells, and mRNA for IFN-gamma, perforin, and granzyme B in CD8(+) T and NK cells. CONCLUSIONS rIL-21 administered at 30 microg/kg/d in 5-day cycles every second week is biologically active and well tolerated in patients with metastatic melanoma. Confirmed responses, including one CR, were observed.
Collapse
Affiliation(s)
- Ian D Davis
- Ludwig Oncology Unit, Austin Health, Melbourne, Victoria, Australia
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|