1
|
Cheng T, Li F, Zhang Z, Yuan Y, Zhou Y, Zhu X, Xi L, Dong Q, Luo D, Ma X, Fan L. Identification of a Vascular Endothelial Growth Factor Receptor-3 Binding Peptide TMVP1 for Enhancing Drug Delivery Efficiency and Therapeutic Efficacy Against Tumor Lymphangiogenesis. Cancer Biother Radiopharm 2025; 40:196-209. [PMID: 39718836 DOI: 10.1089/cbr.2024.0119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2024] Open
Abstract
Background: Vascular endothelial growth factor receptor-3 (VEGFR-3) plays an indispensable role in lymphangiogenesis. Previous findings suggest that blocking the VEGFR-3 signaling pathway can inhibit lymph node metastasis effectively, thus reducing the incidence of distant metastasis. The development of new VEGFR-3-targeting drugs for early detection and effective treatments is, therefore, urgently required. Methods: In vitro biopanning of a phage-displayed peptide library was used to identify specific peptides binding to the extracellular domain of VEGFR-3. We obtained a novel VEGFR-3-targeting peptide, TMVP1 (LARGR). Our combined immunofluorescence and radiolabeling studies revealed that FITC-TMVP1 and 99mTc-labeled TMVP1 specifically accumulated in VEGFR-3-positive lymphatic vessels of tumors after intravenous administration in tumor xenograft models in vivo. To enhance the therapeutic efficacy of anticancer drugs, TMVP1 was fused to a proapoptotic peptide, D(KLAKLAK)2. Results: The fusion peptide strongly inhibited tumor lymphangiogenesis in vitro and in vivo and specifically suppressed lung metastasis in a 4T1 breast cancer xenograft model. The accumulation of the TMVP1 in lymphatic vessels was specific. Conclusions: Our results suggest that TMVP1 is a potential therapeutic strategy for developing new diagnostic tracers or alternative anticancer agents for tumor lymphangiogenesis and lymphatic metastasis.
Collapse
Affiliation(s)
- Teng Cheng
- National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- Department of Breast and Thyroid Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Fei Li
- National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Zhenzhong Zhang
- National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- Department of Gynecological Oncology, Henan Provincial Tumor Hospital, Affiliated Tumor Hospital of Zhengzhou University, Zhengzhou, People's Republic of China
| | - Yuan Yuan
- National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Ying Zhou
- National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Xiaohua Zhu
- Department of Nuclear Medicine and PET, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Ling Xi
- National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Qingjian Dong
- Department of Nuclear Medicine and PET, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Danfeng Luo
- National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Xiangyi Ma
- National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Liangsheng Fan
- Department of Obstetrics and Gynecology, First Affiliated Hospital, Guangzhou Medical University, Guangzhou, People's Republic of China
| |
Collapse
|
2
|
Cheung SM, Wu WS, Senn N, Sharma R, McGoldrick T, Gagliardi T, Husain E, Masannat Y, He J. Towards detection of early response in neoadjuvant chemotherapy of breast cancer using Bayesian intravoxel incoherent motion. Front Oncol 2023; 13:1277556. [PMID: 38125950 PMCID: PMC10731248 DOI: 10.3389/fonc.2023.1277556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 11/21/2023] [Indexed: 12/23/2023] Open
Abstract
Introduction The early identification of good responders to neoadjuvant chemotherapy (NACT) holds a significant potential in the optimal treatment of breast cancer. A recent Bayesian approach has been postulated to improve the accuracy of the intravoxel incoherent motion (IVIM) model for clinical translation. This study examined the prediction and early sensitivity of Bayesian IVIM to NACT response. Materials and methods Seventeen female patients with breast cancer were scanned at baseline and 16 patients were scanned after Cycle 1. Tissue diffusion and perfusion from Bayesian IVIM were calculated at baseline with percentage change at Cycle 1 computed with reference to baseline. Cellular proliferative activity marker Ki-67 was obtained semi-quantitatively with percentage change at excision computed with reference to core biopsy. Results The perfusion fraction showed a significant difference (p = 0.042) in percentage change between responder groups at Cycle 1, with a decrease in good responders [-7.98% (-19.47-1.73), n = 7] and an increase in poor responders [10.04% (5.09-28.93), n = 9]. There was a significant correlation between percentage change in perfusion fraction and percentage change in Ki-67 (p = 0.042). Tissue diffusion and pseudodiffusion showed no significant difference in percentage change between groups at Cycle 1, nor was there a significant correlation against percentage change in Ki-67. Perfusion fraction, tissue diffusion, and pseudodiffusion showed no significant difference between groups at baseline, nor was there a significant correlation against Ki-67 from core biopsy. Conclusion The alteration in tumour perfusion fraction from the Bayesian IVIM model, in association with cellular proliferation, showed early sensitivity to good responders in NACT. Clinical trial registration https://clinicaltrials.gov/ct2/show/NCT03501394, identifier NCT03501394.
Collapse
Affiliation(s)
- Sai Man Cheung
- Institute of Medical Sciences, School of Medicine, University of Aberdeen, Aberdeen, United Kingdom
| | - Wing-Shan Wu
- Institute of Medical Sciences, School of Medicine, University of Aberdeen, Aberdeen, United Kingdom
| | - Nicholas Senn
- Institute of Medical Sciences, School of Medicine, University of Aberdeen, Aberdeen, United Kingdom
| | - Ravi Sharma
- Department of Oncology, Aberdeen Royal Infirmary, Aberdeen, United Kingdom
| | - Trevor McGoldrick
- Department of Oncology, Aberdeen Royal Infirmary, Aberdeen, United Kingdom
| | - Tanja Gagliardi
- Institute of Medical Sciences, School of Medicine, University of Aberdeen, Aberdeen, United Kingdom
- Department of Radiology, Royal Marsden Hospital, London, United Kingdom
| | - Ehab Husain
- Department of Pathology, Aberdeen Royal Infirmary, Aberdeen, United Kingdom
| | - Yazan Masannat
- Breast Unit, Aberdeen Royal Infirmary, Aberdeen, United Kingdom
| | - Jiabao He
- Institute of Medical Sciences, School of Medicine, University of Aberdeen, Aberdeen, United Kingdom
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| |
Collapse
|
3
|
Kim JH, Jeong JH. Structure-Activity Relationship Studies Based on 3D-QSAR CoMFA/CoMSIA for Thieno-Pyrimidine Derivatives as Triple Negative Breast Cancer Inhibitors. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27227974. [PMID: 36432075 PMCID: PMC9698756 DOI: 10.3390/molecules27227974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 11/04/2022] [Accepted: 11/15/2022] [Indexed: 11/19/2022]
Abstract
Triple-negative breast cancer (TNBC) is defined as a kind of breast cancer that lacks estrogen receptors (ER), progesterone receptors (PR), and human epidermal growth factor receptors (HER2). This cancer accounts for 10-15% of all breast cancers and has the features of high invasiveness and metastatic potential. The treatment regimens are still lacking and need to develop novel inhibitors for therapeutic strategies. Three-dimensional quantitative structure-activity relationship (3D-QSAR) analyses, based on a series of forty-seven thieno-pyrimidine derivatives, were performed to identify the key structural features for the inhibitory biological activities. The established comparative molecular field analysis (CoMFA) presented a leave-one-out cross-validated correlation coefficient q2 of 0.818 and a determination coefficient r2 of 0.917. In comparative molecular similarity indices analysis (CoMSIA), a q2 of 0.801 and an r2 of 0.897 were exhibited. The predictive capability of these models was confirmed by using external validation and was further validated by the progressive scrambling stability test. From these results of validation, the models were determined to be statistically reliable and robust. This study could provide valuable information for further optimization and design of novel inhibitors against metastatic breast cancer.
Collapse
|
4
|
Chen JM, Luo B, Ma R, Luo XX, Chen YS, Li Y. Lymphatic Endothelial Markers and Tumor Lymphangiogenesis Assessment in Human Breast Cancer. Diagnostics (Basel) 2021; 12:diagnostics12010004. [PMID: 35054174 PMCID: PMC8774380 DOI: 10.3390/diagnostics12010004] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 12/09/2021] [Accepted: 12/17/2021] [Indexed: 12/11/2022] Open
Abstract
Metastasis via lymphatic vessels or blood vessels is the leading cause of death for breast cancer, and lymphangiogenesis and angiogenesis are critical prerequisites for the tumor invasion–metastasis cascade. The research progress for tumor lymphangiogenesis has tended to lag behind that for angiogenesis due to the lack of specific markers. With the discovery of lymphatic endothelial cell (LEC) markers, growing evidence demonstrates that the LEC plays an active role in lymphatic formation and remodeling, tumor cell growth, invasion and intravasation, tumor–microenvironment remodeling, and antitumor immunity. However, some studies have drawn controversial conclusions due to the variation in the LEC markers and lymphangiogenesis assessments used. In this study, we review recent findings on tumor lymphangiogenesis, the most commonly used LEC markers, and parameters for lymphangiogenesis assessments, such as the lymphatic vessel density and lymphatic vessel invasion in human breast cancer. An in-depth understanding of tumor lymphangiogenesis and LEC markers can help to illustrate the mechanisms and distinct roles of lymphangiogenesis in breast cancer progression, which will help in exploring novel potential predictive biomarkers and therapeutic targets for breast cancer.
Collapse
Affiliation(s)
- Jia-Mei Chen
- Center of Oncology, Renmin Hospital of Wuhan University, Wuhan 430060, China; (J.-M.C.); (X.-X.L.)
| | - Bo Luo
- Department of Pathology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430014, China;
| | - Ru Ma
- Department of Peritoneal Cancer Surgery, Beijing Shijitan Hospital of Capital Medical University, Beijing 100038, China;
| | - Xi-Xi Luo
- Center of Oncology, Renmin Hospital of Wuhan University, Wuhan 430060, China; (J.-M.C.); (X.-X.L.)
| | - Yong-Shun Chen
- Center of Oncology, Renmin Hospital of Wuhan University, Wuhan 430060, China; (J.-M.C.); (X.-X.L.)
- Correspondence: (Y.-S.C.); (Y.L.); Tel.: +86-027-88048911 (Y.-S.C.); +86-010-63926525 (Y.L.)
| | - Yan Li
- Department of Peritoneal Cancer Surgery, Beijing Shijitan Hospital of Capital Medical University, Beijing 100038, China;
- Department of Pathology, Beijing Shijitan Hospital, Capital Medical University, Beijing 100038, China
- Correspondence: (Y.-S.C.); (Y.L.); Tel.: +86-027-88048911 (Y.-S.C.); +86-010-63926525 (Y.L.)
| |
Collapse
|
5
|
Wang S, Yang T, He Z. Investigations on the Role of the MicroRNA-338-5p/Wnt Family Member 2B (WNT2B) Axis in Regulating the Pathogenesis of Nasopharyngeal Carcinoma (NPC). Front Oncol 2021; 11:684462. [PMID: 34268117 PMCID: PMC8276634 DOI: 10.3389/fonc.2021.684462] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 06/03/2021] [Indexed: 11/13/2022] Open
Abstract
Background The involvement of microRNA-338-5p in modulating NPC pathogenesis is still largely unknown, and this study aimed to investigate this issue. Methods The expressions of cancer associated genes were determined by Real-Time qPCR and Western Blot, and cell apoptosis was determined by flow cytometer (FCM). CCK-8 assay and colony formation assay were respectively used to determine cell proliferation and colony formation abilities. Transwell assay was used to evaluate cell migration. The expression levels of Ki67 protein in mice tissues were measured by Immunohistochemistry (IHC) assay. Results The present study found that microRNA-338-5p suppressed NPC progression by degrading its downstream target, Wnt family member 2B (WNT2B). Specifically, microRNA-338-5p tended to be low-expressed in NPC tissues and cell lines, compared to the non-tumor nasopharyngeal mucosa tissues and normal nasopharyngeal cell line (NP69). Upregulation of microRNA-338-5p inhibited proliferation, mobility, and epithelial-mesenchymal transition (EMT) in NPC cells in vitro, while silencing of microRNA-338-5p had opposite effects. Consistently, microRNA-338-5p suppressed tumorigenesis of NPC cells in vivo. In addition, microRNA-338-5p targeted WNT2B for degradation and inhibition, and the inhibiting effects of microRNA-338-5p overexpression on NPC development were reversed by upregulating WNT2B. Conclusions Taken together, we concluded that microRNA-338-5p targeted WNT2B to hinder NPC development.
Collapse
Affiliation(s)
- Suzhen Wang
- Department of Otolaryngology, Wuwei People's Hospital, Wuwei, China
| | - Tianning Yang
- Department of Otolaryngology, The First Hospital of Lanzhou University, Lanzhou, China
| | - Zhengxiang He
- Department of Otolaryngology, Wuwei People's Hospital, Wuwei, China
| |
Collapse
|
6
|
Chan KH, Lee CH, Sharif SZ, Hayati F, Sallapan S. Diagnostic challenge in diagnosing bilateral breast metastases from mediastinal neuroendocrine tumor: A case report. Ann Med Surg (Lond) 2020; 60:438-441. [PMID: 33251002 PMCID: PMC7683178 DOI: 10.1016/j.amsu.2020.11.035] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 11/08/2020] [Accepted: 11/08/2020] [Indexed: 12/03/2022] Open
Abstract
Background Metastatic neuroendocrine tumours (NETs) to the breast are very rare entities. Case presentation A 26-year-old lady presented with anterior neck swelling with symptoms of superior vena cava syndrome for 6 months. Imaging study revealed a mediastinal mass which was preceded with core biopsy which was consistent with high-grade small cell NETs. Despite second-line adjuvant chemotherapy and radiotherapy, her disease became advanced which was confirmed via restaging scan. There were bilateral breast lesions discovered during the scan which was deemed to be metastatic NETs histologically. Despite prompt initiation of treatment, she succumbed 1 year after the radiotherapy due to disease progression. Conclusion High suspicion of an index is needed for diagnosis when patients with known primary NETs present with suspicious breast lesions. Triple assessment is mandatory, however histopathology assessment and immunohistochemistry staining are the mainstay of diagnosis. Metastatic breast neuroendocrine tumours are very rare entities especially when they arise from the mediastinum. High suspicion of an index is needed for diagnosis when patients with known primary neuroendocrine tumours presented with suspicious breast lesions. Any breast pathology requires a triple assessment including secondary breast neuroendocrine tumours.
Collapse
Affiliation(s)
- Kheng Hooi Chan
- Department of Surgery, Hospital University Kebangsaan Malaysia Medical Centre, Kuala Lumpur, Malaysia
| | - Chang Haur Lee
- Department of Breast and Endocrine Surgery, Queen Elizabeth Hospital 2, Ministry of Health Malaysia, Kota Kinabalu, Sabah, Malaysia
| | - Siti Zubaidah Sharif
- Department of Breast and Endocrine Surgery, Queen Elizabeth Hospital 2, Ministry of Health Malaysia, Kota Kinabalu, Sabah, Malaysia
| | - Firdaus Hayati
- Department of Surgery, Faculty of Medicine and Health Sciences, Universiti Malaysia Sabah, Kota Kinabalu, Sabah, Malaysia
- Corresponding author.
| | - Sugunah Sallapan
- Department of Pathology, Queen Elizabeth Hospital, Ministry of Health Malaysia, Kota Kinabalu, Sabah, Malaysia
| |
Collapse
|
7
|
Zhang Y, Lun X, Guo W. Expression of TRPC1 and SBEM protein in breast cancer tissue and its relationship with clinicopathological features and prognosis of patients. Oncol Lett 2020; 20:392. [PMID: 33193852 DOI: 10.3892/ol.2020.12255] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Accepted: 07/22/2020] [Indexed: 12/23/2022] Open
Abstract
This study investigated the relationship of the expression of transient receptor potential channel 1 (TRPC1), small breast epithelial mucin (SBEM) in breast cancer tissues with clinical pathological features and prognosis of patients. Altogether 50 patients with breast cancer who were treated in Weifang People's hospital from April 2017 to November 2018 were selected, and the mRNA and protein differences of TRPC1 and SBEM in breast cancer patients and normal breast cancer tissues were detected by qRT-PCR and Western blot. Spearman test was used for correlation analysis. Logistic univariate and multivariate analysis were performed on the risk factors related to breast cancer metastasis in breast cancer patients. The expression of TRPC1 and SBEM in breast cancer tissues was significantly higher than that in normal breast tissues (P<0.001). The mRNA expression of TRPC1, SBEM and protein was not related to age, tumor size and tissue grade of breast cancer patients, but related to TNM stage, clinical stage and lymph node metastasis (P<0.001). The relative expression of TRPC1 was positively correlated with clinical stage of breast cancer (r=0.992, P<0.001). The relative expression of SBEM was positively correlated with the clinical stage of breast cancer (r=0.853, P<0.001). The relative expression of TRPC1 was positively correlated with TNM staging of breast cancer (r=0.860, P<0.001). The relative expression of SBEM was positively correlated with TNM staging of breast cancer (r=0.880, P<0.001). Multivariate conditional Logistic regression analysis showed that TNM staging, TRPC1, SBEM were independent risk factors for malignant breast cancer metastasis. On the contrary, expression of TRPC1 and SBEM in breast cancer tissues was up-regulated. TRPC1 and SBEM may be involved in the process of breast cancer occurrence, development and metastasis, and can be used as potential tissue biomarkers in diagnosis of breast cancer metastasis and disease assessment.
Collapse
Affiliation(s)
- Yongqing Zhang
- Department of General Surgery, Weifang People's Hospital, Weifang, Shandong 261041, P.R. China
| | - Xiaoqin Lun
- Department of General Surgery, Weifang People's Hospital, Weifang, Shandong 261041, P.R. China
| | - Weiling Guo
- Department of General Surgery, Weifang People's Hospital, Weifang, Shandong 261041, P.R. China
| |
Collapse
|
8
|
van Pul KM, Vuylsteke RJCLM, de Beijer MTA, van de Ven R, van den Tol MP, Stockmann HBAC, de Gruijl TD. Breast cancer-induced immune suppression in the sentinel lymph node is effectively countered by CpG-B in conjunction with inhibition of the JAK2/STAT3 pathway. J Immunother Cancer 2020; 8:jitc-2020-000761. [PMID: 33046620 PMCID: PMC7552844 DOI: 10.1136/jitc-2020-000761] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/07/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND We previously showed selectively hampered activation of lymph node-resident (LNR) dendritic cell (DC) subsets in the breast cancer (BrC) sentinel lymph node (SLN) to precede a state of profound T cell anergy. Reactivating these DC subsets by intratumoral delivery of the Toll-like receptor-9 (TLR9) agonist CpG-B could potentially offer a promising immune therapeutic strategy to combat this immune suppression and prevent disease spread. Unfortunately, CpG-B can limit its own immune stimulatory activity through direct TLR9-mediated activation of signal transducer and activator of transcription 3 (STAT3), pinpointed as a key regulator of immune suppression in the tumor microenvironment. Here, we have investigated whether in vitro exposure to CpG-B, with or without simultaneous inhibition of STAT3 signaling, could overcome immune suppression in BrC SLN. METHODS Immune modulatory effects of CpG-B (CPG7909) with or without the JAK2/STAT3 inhibitor (STAT3i) AG490 were assessed in ex vivo cultured BrC SLN-derived single-cell suspensions (N=29). Multiparameter flow cytometric analyses were conducted for DC and T cell subset characterization and assessment of (intracellular) cytokine profiles. T cell reactivity against the BrC-associated antigen Mammaglobin-A was determined by means of interferon-γ ELISPOT assay. RESULTS Although CpG-B alone induced activation of all DC subsets, combined inhibition of the JAK2/STAT3 pathway resulted in superior DC maturation (ie, increased CD83 expression), with most profound activation and maturation of LNR DC subsets. Furthermore, combined CpG-B and JAK2/STAT3 inhibition promoted Th1 skewing by counterbalancing the CpG-induced Th2/regulatory T cell response and significantly enhanced Mammaglobin-A specific T cell reactivity. CONCLUSION Ex vivo immune modulation of the SLN by CpG-B and simultaneous JAK2/STAT3 inhibition can effectively overcome BrC-induced immune suppression by preferential activation of LNR DC, ultimately restoring type 1-mediated antitumor immunity, thereby securing a BrC-specific T cell response. These findings provide a clear rationale for clinical exploration of SLN-immune potentiation through local CpG/STAT3i administration in patients with BrC.
Collapse
Affiliation(s)
- Kim M van Pul
- Medical Oncology-Cancer Center Amsterdam, Amsterdam UMC-VUMC location, Amsterdam, The Netherlands.,Surgical Oncology, Amsterdam UMC-VUMC location, Amsterdam, The Netherlands
| | | | - Monique T A de Beijer
- Medical Oncology-Cancer Center Amsterdam, Amsterdam UMC-VUMC location, Amsterdam, The Netherlands
| | - Rieneke van de Ven
- Medical Oncology and Otolaryngology-Head and Neck Surgery-Cancer Center Amsterdam, Amsterdam UMC-VUMC location, Amsterdam, The Netherlands
| | | | | | - Tanja D de Gruijl
- Medical Oncology-Cancer Center Amsterdam, Amsterdam UMC-VUMC location, Amsterdam, The Netherlands
| |
Collapse
|
9
|
Shibata MA, Shibata E, Tanaka Y, Shiraoka C, Kondo Y. Soluble Vegfr3 gene therapy suppresses multi-organ metastasis in a mouse mammary cancer model. Cancer Sci 2020; 111:2837-2849. [PMID: 32539229 PMCID: PMC7419054 DOI: 10.1111/cas.14531] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 05/28/2020] [Accepted: 06/08/2020] [Indexed: 01/09/2023] Open
Abstract
Accumulating evidence on the association of VEGF-C with lymphangiogenesis and lymph node metastasis implicates lymphatic vessels as a potential target in anti-cancer therapy. To evaluate whether blocking VEGF-C and VEGFR-3 signaling can inhibit multi-organ metastases, a mouse metastatic mammary cancer model was subjected to gene therapy using a soluble VEGFR-3 expression vector (psVEGFR-3). We showed that psVEGFR-3 significantly diminished cell growth in vitro with or without added VEGF-C, and significantly reduced primary tumor growth and tumor metastases to wide-spectrum organs in vivo. Although apoptotic cell death and angiogenesis levels did not differ between the control and psVEGFR-3 groups, cell proliferation and lymphangiogenesis in the mammary tumors were significantly decreased in the psVEGFR-3 group. Furthermore, lymphatic vessel invasion was significantly inhibited in this group. Real-time RT-PCR analysis revealed significantly high expression of the Vegfr3 gene due to gene therapy, and the transcriptional levels of Pcna and Lyve1 tended to decrease in the psVEGFR-3 group. Immunofluorescence staining indicated that phospho-tyrosine expression was considerably lower in tumor cells of psVEGFR-3-treated mammary carcinomas than those of control tumors. Double immunofluorescence staining indicated that phospho-tyrosine+ /LYVE-1+ (a lymphatic vessel marker) tended to decrease in psVEGFR-3-treated mammary carcinomas compared with control mice, indicating a decline in the activity of the VEGF-C/VEGFR-3 axis. These findings showed that a blockade of VEGF-C/VEGFR-3 signaling caused by sVEGFR-3 sequestered VEGF-C and prevented the side-effects of anti-angiogenesis and suppressed overall metastases, suggesting their high clinical significance.
Collapse
Affiliation(s)
- Masa-Aki Shibata
- Department of Anatomy and Cell Biology, Osaka Medical College, Takatsuki, Japan
| | - Eiko Shibata
- Department of Molecular Innovation in Lipidology, National Cerebral & Cardiovascular Center Research Institute, Suita, Japan
| | - Yoshihisa Tanaka
- Department of Anatomy and Cell Biology, Osaka Medical College, Takatsuki, Japan
| | - Chinatsu Shiraoka
- Department of Anatomy and Cell Biology, Osaka Medical College, Takatsuki, Japan
| | - Yoichi Kondo
- Department of Anatomy and Cell Biology, Osaka Medical College, Takatsuki, Japan
| |
Collapse
|