1
|
Na JY, Jeon J, Huh KY, Yu K, Lee S, Eom J, Ahn J, You W, Oh J. Population pharmacokinetic model of ABL001/CTX-009 (anti-VEGF/DLL4) in adult cancer patients with solid tumor. Cancer Sci 2024; 115:3943-3951. [PMID: 39375952 PMCID: PMC11611764 DOI: 10.1111/cas.16363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 09/03/2024] [Accepted: 09/19/2024] [Indexed: 10/09/2024] Open
Abstract
ABL001/CTX-009 is a bispecific antibody targeting delta-like ligand-4 and vascular endothelial growth factor A. In this study, we developed a population pharmacokinetic (PK) model of ABL001/CTX-009 in patients with solid tumors. A total of 712 plasma concentrations from 30 patients with relapsed or refractory solid tumors were collected from a phase 1 study (NCT03292783). A population PK model was developed using a nonlinear mixed-effect method and was evaluated by graphical and numerical methods. Using the model, the steady-state concentrations were simulated to compare weight-based and fixed-dose regimens and to find optimal dosing intervals. The PK of ABL001/CTX-009 was well described by a two-compartment model with a parallel first-order and Michaelis-Menten elimination kinetics. Body weight was selected as a significant covariate on V1. Model evaluation results suggested that the model was adequate and robust with good precision. Simulations after administrations of fixed or weight-based doses showed similar plasma concentrations. Additionally, 10 mg/kg for every other week and 15 mg/kg for every three-week administration showed comparable plasma concentrations. In conclusion, the model well described the plasma concentrations of ABL001/CTX-009 in patients with solid tumors. The simulation suggested that weight-based dose and fixed dose can provide equivalent systemic exposure.
Collapse
Affiliation(s)
- Joo Young Na
- Department of Clinical Pharmacology and TherapeuticsSeoul National University College of Medicine and HospitalSeoulRepublic of Korea
- Division of Pharmaceutics and PharmacologyCollege of Pharmacy, The Ohio State UniversityColumbusOhioUSA
| | | | - Ki Young Huh
- Department of Clinical Pharmacology and TherapeuticsSeoul National University College of Medicine and HospitalSeoulRepublic of Korea
| | - Kyung‐Sang Yu
- Department of Clinical Pharmacology and TherapeuticsSeoul National University College of Medicine and HospitalSeoulRepublic of Korea
| | | | | | | | | | - Jaeseong Oh
- Department of Clinical Pharmacology and TherapeuticsSeoul National University College of Medicine and HospitalSeoulRepublic of Korea
- Department of PharmacologyJeju National University College of MedicineJejuRepublic of Korea
- Clinical Research Institute, Jeju National University HospitalJejuRepublic of Korea
| |
Collapse
|
2
|
Yan J, Xie Y, Liu Z, Yang Y, Zhou T. DLL4-targeted CAR-T therapy sensitizes neoadjuvant chemotherapy via eliminating cancer stem cells and reshaping immune microenvironment in HER2 + breast cancer. J Immunother Cancer 2024; 12:e009636. [PMID: 39542653 PMCID: PMC11575309 DOI: 10.1136/jitc-2024-009636] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/18/2024] [Indexed: 11/17/2024] Open
Abstract
BACKGROUND Neoadjuvant therapy with trastuzumab, pertuzumab and paclitaxel (THP) has significantly improved the prognosis of patients with human epidermal growth factor receptor 2 (HER2)+ breast cancer (BC). However, there remains a subset of non-responsive patients. Thus, this study sought to identify key regulators of THP neoadjuvant therapy resistance and potential targets to sensitize sensitivity. METHODS The Cancer Genome Atlas database, Gene Expression Omnibus and membrane protein database were used to identify the key regulator of THP neoadjuvant resistance. The biological functions and mechanisms of delta-like 4 proteins (DLL4) in THP therapy resistance were investigated in vitro and in vivo using the bioinformatic analysis, multiplex immunofluorescence, flow cytometry, sphere formation assays and chromatin immunoprecipitation, etc. Furthermore, DLL4-targeted chimeric antigen receptor (CAR)-T cells were established to sensitize THP therapy. RESULTS DLL4 was identified as a key target in THP neoadjuvant therapy resistance for HER2+ BC. Mechanistically, DLL4+ tumor cells exhibited enhanced stemness and resistance to the THP neoadjuvant chemotherapy. Additionally, soluble DLL4 can split away from tumor cells and diffuse into the stroma, where it can activate the Notch signaling pathway in neutrophils, inducing the formation and release of neutrophil extracellular traps (NETs) by regulating the transcription of MPO, PDIA4 and ELANE. This led to the exclusion of lymphocyte infiltration, thereby enhancing therapy resistance. What is more, we designed a DLL4-targeted CAR-T to eliminate DLL4+ tumor cells and reverse the resistant status. CONCLUSIONS Our study revealed novel functions of DLL4 in cell stemness and immune infiltration, including NET formation and T cell exclusion, which collectively contributed to THP neoadjuvant therapy resistance in HER2+ BC. Furthermore, we provided a CAR-T-based therapy to sensitize the THP neoadjuvant therapy.
Collapse
Affiliation(s)
- Jingrui Yan
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Yongjie Xie
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Ziyun Liu
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Yanfang Yang
- Second Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China
| | - Tianxing Zhou
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| |
Collapse
|
3
|
Zhang X, Xu Z, Chen Q, Zhou Z. Notch signaling regulates pulmonary fibrosis. Front Cell Dev Biol 2024; 12:1450038. [PMID: 39450276 PMCID: PMC11499121 DOI: 10.3389/fcell.2024.1450038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Accepted: 09/23/2024] [Indexed: 10/26/2024] Open
Abstract
Pulmonary fibrosis is a progressive interstitial lung disease associated with aging. The pathogenesis of pulmonary fibrosis remains unclear, however, alveolar epithelial cell injury, myofibroblast activation, and extracellular matrix (ECM) accumulation are recognized as key contributors. Moreover, recent studies have implicated cellular senescence, endothelial-mesenchymal transition (EndMT), and epigenetic modifications in the pathogenesis of fibrotic diseases. Various signaling pathways regulate pulmonary fibrosis, including the TGF-β, Notch, Wnt, Hedgehog, and mTOR pathways. Among these, the TGF-β pathway is extensively studied, while the Notch pathway has emerged as a recent research focus. The Notch pathway influences the fibrotic process by modulating immune cell differentiation (e.g., macrophages, lymphocytes), inhibiting autophagy, and promoting interstitial transformation. Consequently, inhibiting Notch signaling represents a promising approach to mitigating pulmonary fibrosis. In this review, we discuss the role of Notch signaling pathway in pulmonary fibrosis, aiming to offer insights for future therapeutic investigations.
Collapse
Affiliation(s)
| | - Zhihao Xu
- Department of Respiratory and Critical Care Medicine, The Fourth Affiliated Hospital, School of Medicine, Zhejiang University, Yiwu, China
| | | | | |
Collapse
|
4
|
Afzalipour R, Abbasi-Dokht T, Sheikh M, Mohammadlou M, Nili F, Baharlou R. The Prediction of DLL4 as a Prognostic Biomarker in Patients with Gastric Cancer Using Anti-DLL4 Nanobody. J Gastrointest Cancer 2024; 55:1380-1387. [PMID: 39046662 DOI: 10.1007/s12029-024-01093-9] [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] [Accepted: 07/11/2024] [Indexed: 07/25/2024]
Abstract
BACKGROUND Angiogenesis and cancer metastasis depend on the DLL4/Notch signaling pathway. A new approach to treating angiogenesis could inhibit or block this pathway. In the present study, we investigated DLL4 expression as a biomarker capable of predicting survival outcomes in gastric cancer patients using a novel anti-DLL4 Nanobody. PATIENTS AND METHODS By using a recently developed anti-DLL4 Nanobody, the expression of DLL4 was evaluated in tissue samples from 135 gastric cancer patients. It was evaluated whether DLL4 expression is related to clinicopathological factors, overall survival (OS), and recurrence-free survival (RFS). RESULTS Sixty-five (48%) gastric cancer patients had a positive expression of DLL4 within the tumor tissue. Based on both the univariate and multivariate regression analyses, the expression of DLL4 was strongly associated with RFS (HR, 1.94; p = 0.008) and OS (HR, 2.06; p = 0.004). Moreover, the survival analysis demonstrated that DLL4 expression was a significant independent factor of unfavorable OS (HR, 2.7; p = 0.01) and RFS (HR, 2.3; p = 0.02) in gastric cancer patients. CONCLUSION DLL4 expression in gastric cancer patients may predict poor prognosis and survival. Furthermore, the current data demonstrate the potential of Nanobody for detecting DLL4, and it may lead to develop novel therapies and diagnostics for tumors.
Collapse
Affiliation(s)
- Reza Afzalipour
- Molecular Medicine Research Center, Hormozgan Health Institute, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
- Department of Radiology, Faculty of Para-Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Tannaz Abbasi-Dokht
- Cancer Research Center, Semnan University of Medical Sciences, Semnan, Iran
- Department of Immunology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Maryam Sheikh
- Cancer Research Center, Semnan University of Medical Sciences, Semnan, Iran
- Department of Immunology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Maryam Mohammadlou
- Cancer Research Center, Semnan University of Medical Sciences, Semnan, Iran
- Department of Immunology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Fatemeh Nili
- Department of Pathology, Cancer Institute, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
| | - Rasoul Baharlou
- Cancer Research Center, Semnan University of Medical Sciences, Semnan, Iran.
- Department of Immunology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran.
| |
Collapse
|
5
|
Sachan N, Sharma V, Mutsuddi M, Mukherjee A. Notch signalling: multifaceted role in development and disease. FEBS J 2024; 291:3030-3059. [PMID: 37166442 DOI: 10.1111/febs.16815] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 02/08/2023] [Accepted: 05/10/2023] [Indexed: 05/12/2023]
Abstract
Notch pathway is an evolutionarily conserved signalling system that operates to influence an astonishing array of cell fate decisions in different developmental contexts. Notch signalling plays important roles in many developmental processes, making it difficult to name a tissue or a developing organ that does not depend on Notch function at one stage or another. Thus, dysregulation of Notch signalling is associated with many developmental defects and various pathological conditions, including cancer. Although many recent advances have been made to reveal different aspects of the Notch signalling mechanism and its intricate regulation, there are still many unanswered questions related to how the Notch signalling pathway functions in so many developmental events. The same pathway can be deployed in numerous cellular contexts to play varied and critical roles in an organism's development and this is only possible because of the complex regulatory mechanisms of the pathway. In this review, we provide an overview of the mechanism and regulation of the Notch signalling pathway along with its multifaceted functions in different aspects of development and disease.
Collapse
Affiliation(s)
- Nalani Sachan
- Department of Molecular and Human Genetics, Institute of Science, Banaras Hindu University, Varanasi, India
- Department of Cell Biology, NYU Grossman School of Medicine, New York, NY, USA
| | - Vartika Sharma
- Department of Molecular and Human Genetics, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Mousumi Mutsuddi
- Department of Molecular and Human Genetics, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Ashim Mukherjee
- Department of Molecular and Human Genetics, Institute of Science, Banaras Hindu University, Varanasi, India
| |
Collapse
|
6
|
Cheng B, Li Y, Ji YB, Shi W, Li M, Zheng J, Ding L, Liu K, Fang L, Xu Y, Li H, Shao X. Polyethylenimine Triggers Dll4 Degradation to Regulate Angiogenesis In Vitro. ACS OMEGA 2024; 9:7502-7510. [PMID: 38405519 PMCID: PMC10882680 DOI: 10.1021/acsomega.3c06050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 11/28/2023] [Accepted: 01/17/2024] [Indexed: 02/27/2024]
Abstract
The Dll4-Notch signaling pathway plays a crucial role in the regulation of angiogenesis and is a promising therapeutic target for diseases associated with abnormal angiogenesis, such as cancer and ophthalmic diseases. Here, we find that polyethylenimine (PEI), a cationic polymer widely used as nucleic acid transfection reagents, can target the Notch ligand Dll4. By immunostaining and immunoblotting, we demonstrate that PEI significantly induces the clearance of cell-surface Dll4 and facilitates its degradation through the lysosomal pathway. As a result, the activation of Notch signaling in endothelial cells is effectively inhibited by PEI, as evidenced by the observed decrease in the generation of the activated form of Notch and expression of Notch target genes Hes1 and Hey1. Furthermore, through blocking Dll4-mediated Notch signaling, PEI treatment enhances angiogenesis in vitro. Together, our study reveals a novel biological effect of PEI and establishes a foundation for the development of a Dll4-targeted biomaterial for the treatment of angiogenesis-related disease.
Collapse
Affiliation(s)
- Binghua Cheng
- Guangdong
Key Laboratory of Nanomedicine, CAS-HK Joint Lab of Biomaterials,
Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- University
of Chinese Academy of Sciences, Beijing 100049, China
| | - Yanyan Li
- Guangdong
Key Laboratory of Nanomedicine, CAS-HK Joint Lab of Biomaterials,
Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- University
of Chinese Academy of Sciences, Beijing 100049, China
| | - Ya-Bin Ji
- Guangdong
Key Laboratory of Nanomedicine, CAS-HK Joint Lab of Biomaterials,
Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Wenli Shi
- Guangdong
Key Laboratory of Nanomedicine, CAS-HK Joint Lab of Biomaterials,
Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Meiqing Li
- Guangdong
Key Laboratory of Nanomedicine, CAS-HK Joint Lab of Biomaterials,
Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- Sino-Euro
Center of Biomedicine and Health, Shenzhen 518024, China
| | - Jiwei Zheng
- Guangdong
Key Laboratory of Nanomedicine, CAS-HK Joint Lab of Biomaterials,
Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Li Ding
- Guangdong
Key Laboratory of Nanomedicine, CAS-HK Joint Lab of Biomaterials,
Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Ke Liu
- Guangdong
Key Laboratory of Nanomedicine, CAS-HK Joint Lab of Biomaterials,
Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- Sino-Euro
Center of Biomedicine and Health, Shenzhen 518024, China
| | - Lijing Fang
- Guangdong
Key Laboratory of Nanomedicine, CAS-HK Joint Lab of Biomaterials,
Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- Sino-Euro
Center of Biomedicine and Health, Shenzhen 518024, China
| | - Ye Xu
- General
Hospital of Southern Theatre Command, Guangzhou 510010, China
| | - Hongchang Li
- Guangdong
Key Laboratory of Nanomedicine, CAS-HK Joint Lab of Biomaterials,
Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- Sino-Euro
Center of Biomedicine and Health, Shenzhen 518024, China
| | - Ximing Shao
- Guangdong
Key Laboratory of Nanomedicine, CAS-HK Joint Lab of Biomaterials,
Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- Sino-Euro
Center of Biomedicine and Health, Shenzhen 518024, China
| |
Collapse
|
7
|
Domínguez-Cejudo MA, Gil-Torralvo A, Cejuela M, Molina-Pinelo S, Salvador Bofill J. Targeting the Tumor Microenvironment in Breast Cancer: Prognostic and Predictive Significance and Therapeutic Opportunities. Int J Mol Sci 2023; 24:16771. [PMID: 38069096 PMCID: PMC10706312 DOI: 10.3390/ijms242316771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 11/23/2023] [Accepted: 11/24/2023] [Indexed: 12/18/2023] Open
Abstract
Breast cancer is one of the most prevalent tumors among women. Its prognosis and treatment outcomes depend on factors related to tumor cell biology. However, recent studies have revealed the critical role of the tumor microenvironment (TME) in the development, progression, and treatment response of breast cancer. In this review, we explore the different components of the TME and their relevance as prognostic and predictive biomarkers in breast cancer. In addition, techniques for assessing the tumor microenvironment, such as immunohistochemistry or gene expression profiling, and their clinical utility in therapeutic decision-making are examined. Finally, therapeutic strategies targeting the TME are reviewed, highlighting their potential clinical benefits. Overall, this review emphasizes the importance of the TME in breast cancer and its potential as a clinical tool for better patient stratification and the design of personalized therapies.
Collapse
Affiliation(s)
- María A. Domínguez-Cejudo
- Institute of Biomedicine of Seville (IBiS), HUVR, CSIC, Universidad de Sevilla, 41013 Seville, Spain (S.M.-P.)
- Andalusian—Roche Network Mixed Alliance in Precision Medical Oncology, 41092 Sevilla, Spain
| | - Ana Gil-Torralvo
- Institute of Biomedicine of Seville (IBiS), HUVR, CSIC, Universidad de Sevilla, 41013 Seville, Spain (S.M.-P.)
- Andalusian—Roche Network Mixed Alliance in Precision Medical Oncology, 41092 Sevilla, Spain
- Medical Oncology Department, Virgen del Rocio Hospital, 41013 Seville, Spain
| | - Mónica Cejuela
- Medical Oncology Department, Virgen del Rocio Hospital, 41013 Seville, Spain
| | - Sonia Molina-Pinelo
- Institute of Biomedicine of Seville (IBiS), HUVR, CSIC, Universidad de Sevilla, 41013 Seville, Spain (S.M.-P.)
- Andalusian—Roche Network Mixed Alliance in Precision Medical Oncology, 41092 Sevilla, Spain
| | - Javier Salvador Bofill
- Institute of Biomedicine of Seville (IBiS), HUVR, CSIC, Universidad de Sevilla, 41013 Seville, Spain (S.M.-P.)
- Andalusian—Roche Network Mixed Alliance in Precision Medical Oncology, 41092 Sevilla, Spain
- Medical Oncology Department, Virgen del Rocio Hospital, 41013 Seville, Spain
| |
Collapse
|
8
|
Lin M, Dai H, Zheng L, Yu P, Chen Y, Hong R, Zheng C. Tryptanthrin inhibits tumor angiogenesis via Notch/Dll4 signaling pathway in zebrafish. Transl Cancer Res 2023; 12:2660-2672. [PMID: 37969390 PMCID: PMC10643962 DOI: 10.21037/tcr-23-925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 09/19/2023] [Indexed: 11/17/2023]
Abstract
Background Anti-angiogenic pathways are important for inhibiting tumor growth and migration. Tryptanthrin has anticancer properties in vivo but its anti-angiogenesis activities and associated mechanisms remain unclear. Methods The effects of tryptanthrin were investigated in vivo using fluorescent labeling of blood vessels in zebrafish. Fluorescence quantitation was conducted to analyze the level of delta-like ligand 4 (Dll4) gene expression. Transcriptome sequencing and quantitative polymerase chain reaction (qPCR) analyses were performed to explore the molecular mechanisms of anti-tumor angiogenesis. Results Significant anti-tumor effects were observed in all 48-hpf (hours post-fertilization) zebrafish treated with tryptanthrin (P<0.05). The 6-hpf zebrafish were cultured to 48 and 72 hpf following tryptanthrin treatment. It was found that compared with the control groups, the fluorescence area and the number of complete internode vessels reduced significantly following treatment with medium and high concentrations of tryptanthrin (P<0.05). The relative expression of Dll4 in the 48-hpf zebrafish was significantly inhibited only in the high concentration group (P<0.05). qPCR analysis revealed that the levels of Krt18b, desma, Tnnt2c, and Krt4 gene expression were significantly up-regulated in zebrafish following Dll4 overexpression. After Dll4 knockdown, the level of desma and Tnnt2c gene expression was significantly up-regulated. Conclusions Tryptanthrin can inhibit tumor growth in vivo in a concentration-dependent manner by down-regulating Dll4 protein expression, and at the same time up-regulating the level of desma and Tnnt2c gene expression.
Collapse
Affiliation(s)
- Meimei Lin
- Department of Pharmacy, Fuqing City Hospital Affiliated to Fujian Medical University, Fuzhou, China
| | - Hengfen Dai
- Department of Pharmacy, Affiliated Fuzhou First Hospital of Fujian Medical University, Fuzhou, China
| | - Lanlan Zheng
- Department of Endocrinology, Fuqing City Hospital Affiliated to Fujian Medical University, Fuzhou, China
| | - Peiqing Yu
- Department of Endocrinology, Fuqing City Hospital Affiliated to Fujian Medical University, Fuzhou, China
| | - Yan Chen
- Department of Pharmacy, Fuqing City Hospital Affiliated to Fujian Medical University, Fuzhou, China
| | - Ru Hong
- Department of Pharmacy, Fuqing City Hospital Affiliated to Fujian Medical University, Fuzhou, China
| | - Caiyun Zheng
- Department of Pharmacy, Fuqing City Hospital Affiliated to Fujian Medical University, Fuzhou, China
| |
Collapse
|
9
|
Wang W, Chen H, Yin S, Yang Z, Zhang F. Targeting adipocyte-immune cell crosstalk to control breast cancer progression. J Cancer Res Clin Oncol 2023; 149:7969-7979. [PMID: 36914785 DOI: 10.1007/s00432-023-04685-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Accepted: 03/05/2023] [Indexed: 03/16/2023]
Abstract
Adipocytes are crucial components of breast cancer and are involved in regulating the progression, therapeutic efficacy, and prognosis of breast cancer patients. Characterized by storing energy and producing a variety of secretory factors, adipocytes are responsible for inducing obesity and regulating the tumor immune activity. Adipocytes communicate with tumor infiltrating immune cells through the secreted adipokines, cytokines, and exosomes in the breast cancer TIME, which shapes the tumor supporting environment to facilitate the immune escape of tumor cells. In-depth studies of the crosstalk between adipocytes and TIME can not only provide a more comprehensive regulatory landscape of TIME, but also be conducive to screening novel targets for future precision targeted therapy. The aim of this review is to discuss recent studies for understanding the role of crosstalk between adipocytes and immune cells in shaping the breast cancer immune microenvironment.
Collapse
Affiliation(s)
- Weihua Wang
- Department of Breast and Thyroid Surgery, Chongqing General Hospital, 118 Xingguang Avenue, Chongqing, 401147, People's Republic of China
| | - Hongdan Chen
- Department of Breast and Thyroid Surgery, Chongqing General Hospital, 118 Xingguang Avenue, Chongqing, 401147, People's Republic of China
| | - Supeng Yin
- Department of Breast and Thyroid Surgery, Chongqing General Hospital, 118 Xingguang Avenue, Chongqing, 401147, People's Republic of China
| | - Zeyu Yang
- Department of Breast and Thyroid Surgery, Chongqing General Hospital, 118 Xingguang Avenue, Chongqing, 401147, People's Republic of China.
| | - Fan Zhang
- Department of Breast and Thyroid Surgery, Chongqing General Hospital, 118 Xingguang Avenue, Chongqing, 401147, People's Republic of China.
| |
Collapse
|
10
|
Shamis SAK, Edwards J, McMillan DC. The relationship between carbonic anhydrase IX (CAIX) and patient survival in breast cancer: systematic review and meta-analysis. Diagn Pathol 2023; 18:46. [PMID: 37061698 PMCID: PMC10105416 DOI: 10.1186/s13000-023-01325-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 03/14/2023] [Indexed: 04/17/2023] Open
Abstract
PURPOSE Hypoxia is a characteristic of many solid tumours and an adverse prognostic factor for cancer therapy. Hypoxia results in upregulation of carbonic anhydrase IX (CAIX) expression, a pH-regulating enzyme. Many human tissue studies have examined the prognostic value of CAIX expression in breast cancer but have yielded inconsistent results. Therefore, a systematic review and meta-analysis was undertaken to assess the prognostic value of CAIX expression for breast cancer patients. METHODS The electronic databases were systematically searched to identify relevant papers. The clinical outcomes included disease-free survival (DFS), recurrence-free survival (RFS) and overall survival (OS) in breast cancer patients. Review Manager version 5.4 was employed to analysis data from 23 eligible studies (containing 8390 patients). RESULTS High CAIX expression was associated with poorer RFS [HR = 1.42, 95% CI (1.32-1.51), p < 0.00001], DFS [HR = 1.64, 95% CI (1.34-2.00), p < 0.00001], and OS [HR = 1.48, 95% CI (1.22-1.80), p < 0.0001]. Heterogeneity was observed across the studies. There was an effect of the CAIX antibody employed, scoring methods, and tumour localisation on CAIX expression. CONCLUSION CAIX overexpression was significantly associated with poorer RFS, DFS, and OS in breast cancer patients. However, further work in high quantity tissue cohorts is required to define the optimal methodological approach.
Collapse
Affiliation(s)
- Suad A K Shamis
- Academic Unit of Surgery, School of Medicine, University of Glasgow, Royal Infirmary, Alexandria Parade, Glasgow, G31 2ER, UK.
- Unit of Molecular Pathology, School of Cancer Sciences, University of Glasgow, Wolfson Wohl Cancer Research Centre, Garscube Estate, Switchback Road, Glasgow, G61 1QH, UK.
| | - Joanne Edwards
- Unit of Molecular Pathology, School of Cancer Sciences, University of Glasgow, Wolfson Wohl Cancer Research Centre, Garscube Estate, Switchback Road, Glasgow, G61 1QH, UK
| | - Donald C McMillan
- Academic Unit of Surgery, School of Medicine, University of Glasgow, Royal Infirmary, Alexandria Parade, Glasgow, G31 2ER, UK
| |
Collapse
|
11
|
Numprasit W, Yangngam S, Prasopsiri J, Quinn JA, Edwards J, Thuwajit C. Carbonic anhydrase IX-related tumoral hypoxia predicts worse prognosis in breast cancer: A systematic review and meta-analysis. Front Med (Lausanne) 2023; 10:1087270. [PMID: 37007798 PMCID: PMC10063856 DOI: 10.3389/fmed.2023.1087270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 02/17/2023] [Indexed: 03/19/2023] Open
Abstract
BackgroundTumoral hypoxia is associated with aggressiveness in many cancers including breast cancer. However, measuring hypoxia is complicated. Carbonic anhydrase IX (CAIX) is a reliable endogenous marker of hypoxia under the control of the master regulator hypoxia-inducible factor-1α (HIF-1α). The expression of CAIX is associated with poor prognosis in many solid malignancies; however, its role in breast cancer remains controversial.MethodsThe present study performed a meta-analysis to evaluate the correlation between CAIX expression and disease-free survival (DFS) and overall survival (OS) in breast cancer.ResultsA total of 2,120 publications from EMBASE, PubMed, Cochrane, and Scopus were screened. Of these 2,120 publications, 272 full texts were reviewed, and 27 articles were included in the meta-analysis. High CAIX was significantly associated with poor DFS (HR = 1.70, 95% CI = 1.39–2.07, p < 0.00001) and OS (HR = 2.02, 95% CI 1.40–2.91, p = 0.0002) in patients with breast cancer. When stratified by subtype, the high CAIX group was clearly associated with shorter DFS (HR = 2.09, 95% CI =1.11–3.92, p = 0.02) and OS (HR = 2.50, 95% CI =1.53–4.07, p = 0.0002) in TNBC and shorter DFS in ER+ breast cancer (HR = 1.81 95% CI =1.38–2.36, p < 0.0001).ConclusionHigh CAIX expression is a negative prognostic marker of breast cancer regardless of the subtypes.
Collapse
Affiliation(s)
- Warapan Numprasit
- Division of Head Neck and Breast Surgery, Department of Surgery, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- School of Cancer Sciences, Wolfson Wohl Cancer Research Centre, University of Glasgow, Glasgow, United Kingdom
| | - Supaporn Yangngam
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Jaturawitt Prasopsiri
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Jean A. Quinn
- School of Cancer Sciences, Wolfson Wohl Cancer Research Centre, University of Glasgow, Glasgow, United Kingdom
| | - Joanne Edwards
- School of Cancer Sciences, Wolfson Wohl Cancer Research Centre, University of Glasgow, Glasgow, United Kingdom
| | - Chanitra Thuwajit
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- *Correspondence: Chanitra Thuwajit,
| |
Collapse
|
12
|
Wang L, Cao G, Li W, Chen XW, Xiong SS, Mu YN, Jiang JF, Yang L, Zhang DR, Cao YW. Expressions and Prognostic Values of Notch3 and DLL4 in Human Breast Cancer. Technol Cancer Res Treat 2023; 22:15330338221118984. [PMID: 36740988 PMCID: PMC9903027 DOI: 10.1177/15330338221118984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Background: Notch signaling played a critical role in promoting breast tumorigenesis and progression. However, the role and prognostic value of Notch3 combined with DLL4 expression in breast carcinoma had not been explored. Methods: The retrospective study enrolled 90 breast cancer tissues and 60 noncancerous tissues from (conceal). The expression and prognostic value of Notch3 and DLL4 in patients with breast carcinoma were investigated using Oncomine and UALCAN database. Notch3 and DLL4 expression levels were detected by quantitative real-time polymerase chain reaction, western blotting, and immunohistochemistry. We analyzed the correlation between both proteins expression and clinicopathological parameters and survival data, respectively. Results: The expressions of Notch3 and DLL4 were increased, and Notch3 expression was significantly positively associated with DLL4 in breast carcinoma. The 2 proteins dramatically correlated with advanced stage, high grade and negative Her2 status. The overexpressing of single or both Notch3 and DLL4 resulted in shortened survival of breast cancer patients. And Notch3 overexpression was one of independent risk predictors to poor prognosis. Conclusion: The interaction of Notch3 receptor and DLL4 ligand accelerates oncogenesis, progression, and poor prognosis of breast cancer patients. Notch3 protein may serve as one of biomarker to independently predict prognosis of patients.
Collapse
Affiliation(s)
- Lin Wang
- Shihezi University School of Medicine, Shihezi, Xinjiang, China,Changle People's Hospital, Weifang, Shandong, China
| | - Ge Cao
- Shihezi University School of Medicine, Shihezi, Xinjiang, China
| | - Wei Li
- Shandong Provincial Western Hospital, Jinan, , Shandong, China
| | - Xiao-Wen Chen
- Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | | | - Ya-Nan Mu
- Shihezi University School of Medicine, Shihezi, Xinjiang, China
| | - Jin-Fang Jiang
- Shihezi University School of Medicine, Shihezi, Xinjiang, China
| | - Lan Yang
- Shihezi University School of Medicine, Shihezi, Xinjiang, China
| | - De-Rui Zhang
- Shihezi University School of Medicine, Shihezi, Xinjiang, China
| | - Yu-Wen Cao
- Shihezi University School of Medicine, Shihezi, Xinjiang, China,The First Affiliated Hospital of Shihezi University School of Medicine, Shihezi University School of Medicine, Shihezi, Xinjiang, China,Yu-Wen Cao, Department of Pathology, Shihezi University School of Medicine, Shihezi 832000, Xinjiang, China.
| |
Collapse
|
13
|
You WK, Schuetz TJ, Lee SH. Targeting the DLL/Notch Signaling Pathway in Cancer: Challenges and Advances in Clinical Development. Mol Cancer Ther 2023; 22:3-11. [PMID: 36223541 PMCID: PMC9808372 DOI: 10.1158/1535-7163.mct-22-0243] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 08/28/2022] [Accepted: 10/07/2022] [Indexed: 02/03/2023]
Abstract
The DLL/Notch signaling pathway plays an important role in cancer as a key driver in maintaining cancer stemness and inducing tumor angiogenesis. Many different types of DLL/Notch inhibitors have been developed and explored in clinical trials for cancer treatment, including small-molecule compounds to inhibit gamma-secretase and antibodies targeting Notch ligands or receptors. Despite promising efficacy of these inhibitors in preclinical studies, the overall clinical outcomes have been insufficient to advance to the next stage of clinical development primarily due to safety concerns or modest efficacy. To overcome the narrow therapeutic window of DLL/Notch inhibitors, diverse strategies for improving the balance between the safety and efficacy are currently being explored. Here, we review the clinical perspective and potential of DLL/Notch inhibitors as anticancer agents based on recent results from multiple clinical studies. An antibody specifically targeting Notch ligands or receptors may offer a better approach to reduce concerns about toxicity derived from broad-spectrum DLL/Notch blockers. In addition, combination therapy with an angiogenesis inhibitor targeting VEGF could be a better option for increasing anticancer efficacy. Taken together, the results of clinical trials suggest a bispecific antibody blocking the DLL/Notch and VEGF/VEGFR signaling pathways as a promising approach for effective anticancer treatment.
Collapse
Affiliation(s)
- Weon-Kyoo You
- R&D Center, ABL Bio, Inc., Seongnam-si, Republic of Korea.,Corresponding Author: Weon-Kyoo You, R&D, R&D center, ABL Bio, Inc., 2F, 16 Daewangpangyo-ro, 712 beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do 13488, Republic of Korea. Phone: 8231-8018-9803; Fax: 8231-8018-9836; E-mail:
| | | | - Sang Hoon Lee
- R&D Center, ABL Bio, Inc., Seongnam-si, Republic of Korea
| |
Collapse
|
14
|
Cui H, Li H, Zhang M, Li H, Wang X, Wang Z, Zhai W, Chen X, Cheng H, Xu J, Zhao X, Ding Z. Molecular Characterization, Expression, Evolutionary Selection, and Biological Activity Analysis of CD68 Gene from Megalobrama amblycephala. Int J Mol Sci 2022; 23:13133. [PMID: 36361921 PMCID: PMC9656401 DOI: 10.3390/ijms232113133] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 10/21/2022] [Accepted: 10/24/2022] [Indexed: 11/24/2022] Open
Abstract
CD68 is a highly glycosylated transmembrane glycoprotein that belongs to the lysosome-associated membrane glycoprotein family and is involved in various immune processes. In this study, Megalobrama amblycephala CD68 (MaCD68) was cloned and characterized, and its expression patterns and evolutionary characteristics were analyzed. The coding region of MaCD68 was 987 bp, encoding 328 amino acids, and the predicted protein molecular weight was 34.9 kDa. MaCD68 contained two transmembrane helical structures and 18 predicted N-glycosylation sites. Multiple sequence alignments showed that the MaCD68 protein had high homology with other fish, and their functional sites were also highly conserved. Phylogenetic analysis revealed that MaCD68 and other cypriniformes fish clustered into one branch. Adaptive evolution analysis identified several positively selected sites of teleost CD68 using site and branch-site models, indicating that it was under positive selection pressure during evolution. Quantitative real-time reverse transcription polymerase chain reaction analysis showed that MaCD68 was highly expressed in the head kidney, spleen, and heart. After Aeromonas hydrophila infection, MaCD68 was significantly upregulated in all tested tissues, peaking at 12 h post-infection (hpi) in the kidney and head kidney and at 120 hpi in the liver and spleen, suggesting that MaCD68 participated in the innate immune response of the host against bacterial infection. Immunohistochemical and immunofluorescence analyses also showed that positive signals derived from the MaCD68 protein were further enhanced after bacterial and lipopolysaccharide treatment, which suggested that MaCD68 is involved in the immune response and could be used as a macrophage marker. Biological activity analysis indicated that recombinant MaCD68 (rMaCD68) protein had no agglutination or bactericidal effects on A. hydrophila but did have these effects on Escherichia coli. In conclusion, these results suggest that MaCD68 plays a vital role in the immune response against pathogens, which is helpful in understanding the immune responses and mechanisms of M. amblycephala.
Collapse
Affiliation(s)
- Hujun Cui
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Co-Innovation Center of Jiangsu Marine Bio-Industry Technology, Jiangsu Ocean University, Lianyungang 222005, China
- Jiangsu Key Laboratory of Marine Biotechnology, School of Marine Science and Fisheries, Jiangsu Ocean University, Lianyungang 222005, China
| | - Hong Li
- Hunan Fisheries Science Institute, Changsha 410153, China
| | - Minying Zhang
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Co-Innovation Center of Jiangsu Marine Bio-Industry Technology, Jiangsu Ocean University, Lianyungang 222005, China
- Jiangsu Key Laboratory of Marine Biotechnology, School of Marine Science and Fisheries, Jiangsu Ocean University, Lianyungang 222005, China
| | - Hongping Li
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Co-Innovation Center of Jiangsu Marine Bio-Industry Technology, Jiangsu Ocean University, Lianyungang 222005, China
- Jiangsu Key Laboratory of Marine Biotechnology, School of Marine Science and Fisheries, Jiangsu Ocean University, Lianyungang 222005, China
| | - Xu Wang
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Co-Innovation Center of Jiangsu Marine Bio-Industry Technology, Jiangsu Ocean University, Lianyungang 222005, China
- Jiangsu Key Laboratory of Marine Biotechnology, School of Marine Science and Fisheries, Jiangsu Ocean University, Lianyungang 222005, China
| | - Zirui Wang
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Co-Innovation Center of Jiangsu Marine Bio-Industry Technology, Jiangsu Ocean University, Lianyungang 222005, China
- Jiangsu Key Laboratory of Marine Biotechnology, School of Marine Science and Fisheries, Jiangsu Ocean University, Lianyungang 222005, China
| | - Wei Zhai
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Co-Innovation Center of Jiangsu Marine Bio-Industry Technology, Jiangsu Ocean University, Lianyungang 222005, China
- Jiangsu Key Laboratory of Marine Biotechnology, School of Marine Science and Fisheries, Jiangsu Ocean University, Lianyungang 222005, China
| | - Xiangning Chen
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Co-Innovation Center of Jiangsu Marine Bio-Industry Technology, Jiangsu Ocean University, Lianyungang 222005, China
- Jiangsu Key Laboratory of Marine Biotechnology, School of Marine Science and Fisheries, Jiangsu Ocean University, Lianyungang 222005, China
| | - Hanliang Cheng
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Co-Innovation Center of Jiangsu Marine Bio-Industry Technology, Jiangsu Ocean University, Lianyungang 222005, China
- Jiangsu Key Laboratory of Marine Biotechnology, School of Marine Science and Fisheries, Jiangsu Ocean University, Lianyungang 222005, China
| | - Jianhe Xu
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Co-Innovation Center of Jiangsu Marine Bio-Industry Technology, Jiangsu Ocean University, Lianyungang 222005, China
- Jiangsu Key Laboratory of Marine Biotechnology, School of Marine Science and Fisheries, Jiangsu Ocean University, Lianyungang 222005, China
| | - Xiaoheng Zhao
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Co-Innovation Center of Jiangsu Marine Bio-Industry Technology, Jiangsu Ocean University, Lianyungang 222005, China
- Jiangsu Key Laboratory of Marine Biotechnology, School of Marine Science and Fisheries, Jiangsu Ocean University, Lianyungang 222005, China
| | - Zhujin Ding
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Co-Innovation Center of Jiangsu Marine Bio-Industry Technology, Jiangsu Ocean University, Lianyungang 222005, China
- Jiangsu Key Laboratory of Marine Biotechnology, School of Marine Science and Fisheries, Jiangsu Ocean University, Lianyungang 222005, China
| |
Collapse
|
15
|
Prognostic Index for Nonsmall Cell Lung Cancer Based on Immune-Related Genes Expression. BIOMED RESEARCH INTERNATIONAL 2022; 2022:4779811. [PMID: 36193311 PMCID: PMC9526605 DOI: 10.1155/2022/4779811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 08/24/2022] [Indexed: 11/17/2022]
Abstract
Immune system dysregulation is associated with tumor incidence and growth. Here, we established an RNA-based individualized immune signature associated with prognosis for nonsmall cell lung cancer (NSCLC) to guide adjuvant therapy. We downloaded publicly accessible data on RNA expression and clinical characteristics of NSCLC from the Cancer Genome Atlas (TCGA). From immune-related genes (IRGs) retrieved from the immunology database and analysis portal (ImmPort) database, we then screened differentially expressed immune-related genes (DEIRGs). Using overall survival (OS) as a clinical endpoint, we identified 26 prognostic DEIRGs via univariate and multivariate Cox regression analysis, and then developed a risk model based on these 26 IRGs with an area under the curve (AUC) of 0.701, and its predictive ability independent from other clinical factors. We also downloaded tumor immune infiltrate data and analyzed the correlations between lymphocytic infiltration with our risk scores, but found no significant association. Furthermore, we retrieved 86 differentially expressed transcription factors (TFs) to assess their regulatory relationships with the 26 prognostic DEIRGs. In summary, we developed a robust risk model to predict survival in patients with NSCLC, based on the expression of 26 IRGs. It provides novel predictive and therapeutic molecular targets.
Collapse
|
16
|
Rahman MM, Sarker MT, Alam Tumpa MA, Yamin M, Islam T, Park MN, Islam MR, Rauf A, Sharma R, Cavalu S, Kim B. Exploring the recent trends in perturbing the cellular signaling pathways in cancer by natural products. Front Pharmacol 2022; 13:950109. [PMID: 36160435 PMCID: PMC9498834 DOI: 10.3389/fphar.2022.950109] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Accepted: 08/15/2022] [Indexed: 12/12/2022] Open
Abstract
Cancer is commonly thought to be the product of irregular cell division. According to the World Health Organization (WHO), cancer is the major cause of death globally. Nature offers an abundant supply of bioactive compounds with high therapeutic efficacy. Anticancer effects have been studied in a variety of phytochemicals found in nature. When Food and Drug Administration (FDA)-approved anticancer drugs are combined with natural compounds, the effectiveness improves. Several agents have already progressed to clinical trials based on these promising results of natural compounds against various cancer forms. Natural compounds prevent cancer cell proliferation, development, and metastasis by inducing cell cycle arrest, activating intrinsic and extrinsic apoptosis pathways, generating reactive oxygen species (ROS), and down-regulating activated signaling pathways. These natural chemicals are known to affect numerous important cellular signaling pathways, such as NF-B, MAPK, Wnt, Notch, Akt, p53, AR, ER, and many others, to cause cell death signals and induce apoptosis in pre-cancerous or cancer cells without harming normal cells. As a result, non-toxic "natural drugs" taken from nature's bounty could be effective for the prevention of tumor progression and/or therapy of human malignancies, either alone or in combination with conventional treatments. Natural compounds have also been shown in preclinical studies to improve the sensitivity of resistant cancers to currently available chemotherapy agents. To summarize, preclinical and clinical findings against cancer indicate that natural-sourced compounds have promising anticancer efficacy. The vital purpose of these studies is to target cellular signaling pathways in cancer by natural compounds.
Collapse
Affiliation(s)
- Md. Mominur Rahman
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Md. Taslim Sarker
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Mst. Afroza Alam Tumpa
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Md. Yamin
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Tamanna Islam
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Moon Nyeo Park
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul, South Korea
| | - Md. Rezaul Islam
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Abdur Rauf
- Department of Chemistry, University of Swabi, Swabi, Anbar, Pakistan
| | - Rohit Sharma
- Department of Rasa Shastra and Bhaishajya Kalpana, Faculty of Ayurveda, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Simona Cavalu
- Faculty of Medicine and Pharmacy, University of Oradea, Oradea, Romania
| | - Bonglee Kim
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul, South Korea
| |
Collapse
|
17
|
Presence of Dendritic Cell Subsets in Sentinel Nodes of Breast Cancer Patients Is Related to Nodal Burden. Int J Mol Sci 2022; 23:ijms23158461. [PMID: 35955602 PMCID: PMC9369399 DOI: 10.3390/ijms23158461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 07/20/2022] [Accepted: 07/21/2022] [Indexed: 12/04/2022] Open
Abstract
BACKGROUND: Sentinel lymph nodes (SLNs) are both the first site where breast cancer (BC) metastases form and where anti-tumoral immunity develops. Despite being the most potent antigen-presenting cells, dendritic cells (DCs) located in a nodal tissue can both promote or suppress immune response against cancer in SLNs. METHODS: In SLNs excisions obtained from 123 invasive BC patients, we performed immunohistochemistry (IHC) for CD1a, CD1c, DC-LAMP, and DC-SIGN to identify different DCs populations. Then we investigated the numbers of DCs subsets in tumor-free, micrometastatic, and macrometastatic SLNs with the use of a light microscope. RESULTS: We observed that CD1c+ and DC-SIGN+ DCs were more numerous in SLNs with a larger tumor size. More abundant intratumoral DC-LAMP+ population was related to a higher number of metastatic lymph nodes. Conversely, more abundant CD1a+ DCs were associated with a decreasing nodal burden in SLNs and a lower number of involved lymph nodes. Moreover, densities of the investigated DC populations differed with respect to tumor grade, HER2 overexpression, hormone receptor status, and histologic type of BC. CONCLUSIONS: According to their subtype, DCs are associated with either lower or higher nodal burden in SLNs from invasive BC patients. These relationships appear to be dependent not only on the maturation state of DCs but also on the histological and biological characteristics of the tumor.
Collapse
|
18
|
Shi Y, Zhang D, Chen J, Jiang Q, Song S, Mi Y, Wang T, Ye Q. Interaction between BEND5 and RBPJ suppresses breast cancer growth and metastasis via inhibiting Notch signaling. Int J Biol Sci 2022; 18:4233-4244. [PMID: 35844785 PMCID: PMC9274485 DOI: 10.7150/ijbs.70866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Accepted: 06/10/2022] [Indexed: 11/17/2022] Open
Abstract
High frequent metastasis is the major cause of breast cancer (BC) mortality among women. However, the molecular mechanisms underlying BC metastasis remain largely unknown. Here, we identified six hub BC metastasis driver genes (BEND5, HSD11B1, NEDD9, SAA2, SH2D2A and TNFSF4) through bioinformatics analysis, among which BEND5 is the most significant gene. Low BEND5 expression predicted advanced stage and shorter overall survival in BC patients. Functional experiments showed that BEND5 could suppress BC growth and metastasis in vitro and in vivo. Mechanistically, BEND5 inhibits Notch signaling via directly interacting with transcription factor RBPJ/CSL. BEN domain of BEND5 interacts with the N-terminal domain (NTD) domain of RBPJ, thus preventing mastermind like transcriptional coactivator (MAML) from forming a transcription activation complex with RBPJ. Our study provides a novel insight into regulatory mechanisms underlying Notch signaling and suggests that BEND5 may become a promising target for BC therapy.
Collapse
Affiliation(s)
- Yanzhu Shi
- Medical College, Guizhou University, Guiyang 550025, P.R. China.,Department of Medical Molecular Biology, Beijing Institute of Biotechnology, Beijing 100850, P.R. China
| | - Deyu Zhang
- Department of Medical Molecular Biology, Beijing Institute of Biotechnology, Beijing 100850, P.R. China
| | - Jingyi Chen
- Department of Medical Molecular Biology, Beijing Institute of Biotechnology, Beijing 100850, P.R. China
| | - Qiwei Jiang
- Department of Medical Molecular Biology, Beijing Institute of Biotechnology, Beijing 100850, P.R. China
| | - Songze Song
- Jinzhou Medical University, Jinzhou, Liaoning 121001, P.R. China
| | - Yue Mi
- Department of Medical Molecular Biology, Beijing Institute of Biotechnology, Beijing 100850, P.R. China
| | - Tao Wang
- Department of Oncology, The fifth Medical Center of Chinese PLA General Hospital, Beijing 100071, P.R. China
| | - Qinong Ye
- Medical College, Guizhou University, Guiyang 550025, P.R. China.,Department of Medical Molecular Biology, Beijing Institute of Biotechnology, Beijing 100850, P.R. China
| |
Collapse
|
19
|
Gordon MS, Nemunaitis J, Barve M, Wainberg ZA, Hamilton EP, Ramanathan RK, Sledge GW, Yue H, Morgan-Lappe SE, Blaney M, Kasichayanula S, Motwani M, Wang L, Naumovski L, Strickler JH. Phase I Open-Label Study Evaluating the Safety, Pharmacokinetics, and Preliminary Efficacy of Dilpacimab in Patients with Advanced Solid Tumors. Mol Cancer Ther 2021; 20:1988-1995. [PMID: 34315767 PMCID: PMC9398147 DOI: 10.1158/1535-7163.mct-20-0985] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 03/30/2021] [Accepted: 07/13/2021] [Indexed: 01/07/2023]
Abstract
Dilpacimab (formerly ABT-165), a novel dual-variable domain immunoglobulin, targets both delta-like ligand 4 (DLL4) and VEGF pathways. Here, we present safety, pharmacokinetic (PK), pharmacodynamic (PD), and preliminary efficacy data from a phase I study (trial registration ID: NCT01946074) of dilpacimab in patients with advanced solid tumors. Eligible patients (≥18 years) received dilpacimab intravenously on days 1 and 15 in 28-day cycles at escalating dose levels (range, 1.25-7.5 mg/kg) until progressive disease or unacceptable toxicity. As of August 2018, 55 patients with solid tumors were enrolled in the dilpacimab monotherapy dose-escalation and dose-expansion cohorts. The most common treatment-related adverse events (TRAE) included hypertension (60.0%), headache (30.9%), and fatigue (21.8%). A TRAE of special interest was gastrointestinal perforation, occurring in 2 patients (3.6%; 1 with ovarian and 1 with prostate cancer) and resulting in 1 death. The PK of dilpacimab showed a half-life ranging from 4.9 to 9.5 days, and biomarker analysis demonstrated that the drug bound to both VEGF and DLL4 targets. The recommended phase II dose for dilpacimab monotherapy was established as 3.75 mg/kg, primarily on the basis of tolerability through multiple cycles. A partial response was achieved in 10.9% of patients (including 4 of 16 patients with ovarian cancer). The remaining patients had either stable disease (52.7%), progressive disease (23.6%), or were deemed unevaluable (12.7%). These results demonstrate that dilpacimab monotherapy has an acceptable safety profile, with clinical activity observed in patients with advanced solid tumors.
Collapse
Affiliation(s)
- Michael S. Gordon
- HonorHealth Research Institute, Scottsdale, Arizona.,Corresponding Author: Michael S. Gordon, HonorHealth Research Institute, 10510 N. 92nd Street, Ste 200, Scottsdale, AZ 85258. Phone: 480-323-1350; Fax: 480-323-1359; E-mail:
| | - John Nemunaitis
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio.,ProMedica Health System, Toledo, Ohio
| | | | - Zev A. Wainberg
- School of Medicine, Ronald Reagan UCLA Medical Center, UCLA Health, University of California Los Angeles, Los Angeles, California
| | - Erika P. Hamilton
- Sarah Cannon Research Institute and Tennessee Oncology, Nashville, Tennessee
| | | | - George W. Sledge
- Stanford Cancer Institute, Stanford Medicine, Stanford, California
| | - Huibin Yue
- Oncology Early Development, AbbVie Inc., Redwood City, California
| | | | - Martha Blaney
- Oncology Early Development, AbbVie Inc., Redwood City, California
| | | | - Monica Motwani
- Translational Oncology, AbbVie Inc., North Chicago, Illinois
| | - Lan Wang
- Oncology Early Development, AbbVie Inc., Redwood City, California
| | - Louie Naumovski
- Oncology Early Development, AbbVie Inc., Redwood City, California
| | - John H. Strickler
- Division of Medical Oncology, Department of Medicine, Duke University School of Medicine, Durham, North Carolina
| |
Collapse
|
20
|
Ribatti D, Annese T, Tamma R. Controversial role of mast cells in breast cancer tumor progression and angiogenesis. Clin Breast Cancer 2021; 21:486-491. [PMID: 34580034 DOI: 10.1016/j.clbc.2021.08.010] [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: 07/08/2021] [Revised: 08/16/2021] [Accepted: 08/26/2021] [Indexed: 10/20/2022]
Abstract
Breast cancer is a neoplastic disease and is a cause of cancer-related mortality for women. Among cellular and molecular regulators of the microenvironment, mast cells and vascular endothelial growth factor (VEGF), are correlated with tumor progression and prognosis in breast cancer. Clinical and experimental studies on breast cancer have revealed a marked correlation between increased angiogenesis, metastasization, and poorer prognosis. After a brief introduction on angiogenesis evidence and angiogenic factors role in different breast cancer subtypes, in this article, we have discerned the relationship between mast cell infiltration, angiogenesis, and tumor progression in human breast cancer with particular reference to the dual role of mast cells, in terms of both pro- or anti-tumoral activity and poor or good biomarker.
Collapse
Affiliation(s)
- Domenico Ribatti
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, Bari, Italy.
| | - Tiziana Annese
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, Bari, Italy
| | - Roberto Tamma
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, Bari, Italy
| |
Collapse
|
21
|
Brzozowa-Zasada M. The role of Notch ligand, Delta-like ligand 4 (DLL4), in cancer angiogenesis—implications for therapy. Eur Surg 2021. [DOI: 10.1007/s10353-021-00707-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Summary
Background
It is generally accepted that angiogenesis is a complex and tightly regulated process characterized by the growth of blood vessels from existing vasculature. Activation of the Notch signalling pathway affects multiple aspects of vascular development. One of the components of the Notch signalling pathway, Delta-like ligand 4 (DLL4), has recently appeared as a critical regulator of tumour angiogenesis and thus as a promising therapeutic target.
Methods
This review article includes available data from peer-reviewed publications associated with the role of DLL4 in cancer angiogenesis. Searches were performed in PubMed, EMBASE, Google Scholar and Web of Science using the terms “tumour angiogenesis”, “DLL4”, “Notch signalling” and “anti-cancer therapy”.
Results
The survival curves of cancer patients revealed that the patients with high DLL4 expression levels had significantly shorter survival times than the patients with low DLL4 expression. Moreover, a positive correlation was also identified between DLL4 and VEGF receptorsʼ expression levels. It seems that inhibition of DLL4 may exert potent growth inhibitory effects on some tumours resistant to anti-VEGF therapies. A great number of blocking agents of DLL4/Notch signalling including anti-DLL4 antibodies, DNA vaccination, Notch antibodies and gamma-secretase inhibitors have been studied in preclinical tumour models.
Conclusion
DLL4 seems to be a promising target in anti-cancer therapy. Nevertheless, the careful evaluation of adverse effects on normal physiological processes in relation to therapeutic doses of anti-DLL4 drugs will be significant for advancement of DLL4 blocking agents in clinical oncology.
Collapse
|
22
|
Prognostic significance and targeting tumor-associated macrophages in cancer: new insights and future perspectives. Breast Cancer 2021; 28:539-555. [PMID: 33661479 DOI: 10.1007/s12282-021-01231-2] [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: 11/17/2020] [Accepted: 02/18/2021] [Indexed: 02/06/2023]
Abstract
Macrophages are phagocytic sentinel cells of the immune system that are central to both innate and adaptive immune responses and serve as the first line of defense against pathogenic insults to tissues. In the tumor microenvironment, tumor-derived factors induce monocyte polarization towards a pro-tumor phenotype. The pro-tumor macrophages regulate key steps in tumorigenicity including tumor growth, angiogenesis, immune suppression, and metastasis. Macrophage infiltration in solid tumors correlates with poor prognosis and resistance to chemotherapy in most cancers. Here in this review, we will shed light on tumor-associated macrophages (TAMs) in regulating tumorigenicity and TAMs as a prognostic biomarker. Also, we will review the recent advances in targeting TAMs to increase the prognosis of cancer patients.
Collapse
|
23
|
Papadopoulou AA, Fluhrer R. Signaling Functions of Intramembrane Aspartyl-Proteases. Front Cardiovasc Med 2020; 7:591787. [PMID: 33381526 PMCID: PMC7768045 DOI: 10.3389/fcvm.2020.591787] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 11/16/2020] [Indexed: 01/18/2023] Open
Abstract
Intramembrane proteolysis is more than a mechanism to "clean" the membranes from proteins no longer needed. By non-reversibly modifying transmembrane proteins, intramembrane cleaving proteases hold key roles in multiple signaling pathways and often distinguish physiological from pathological conditions. Signal peptide peptidase (SPP) and signal peptide peptidase-like proteases (SPPLs) recently have been associated with multiple functions in the field of signal transduction. SPP/SPPLs together with presenilins (PSs) are the only two families of intramembrane cleaving aspartyl proteases known in mammals. PS1 or PS2 comprise the catalytic center of the γ-secretase complex, which is well-studied in the context of Alzheimer's disease. The mammalian SPP/SPPL family of intramembrane cleaving proteases consists of five members: SPP and its homologous proteins SPPL2a, SPPL2b, SPPL2c, and SPPL3. Although these proteases were discovered due to their homology to PSs, it became evident in the past two decades that no physiological functions are shared between these two families. Based on studies in cell culture models various substrates of SPP/SPPL proteases have been identified in the past years and recently-developed mouse lines lacking individual members of this protease family, will help to further clarify the physiological functions of these proteases. In this review we concentrate on signaling roles of mammalian intramembrane cleaving aspartyl proteases. In particular, we will highlight the signaling roles of PS via its substrates NOTCH, VEGF, and others, mainly focusing on its involvement in vasculature. Delineating also signaling pathways that are affected and/or controlled by SPP/SPPL proteases. From SPP's participation in tumor progression and survival, to SPPL3's regulation of protein glycosylation and SPPL2c's control over cellular calcium stores, various crossovers between proteolytic activity of intramembrane proteases and cell signaling will be described.
Collapse
Affiliation(s)
- Alkmini A. Papadopoulou
- Biochemistry and Molecular Biology, Institute of Theoretical Medicine, Medical Faculty, University of Augsburg, Augsburg, Germany
| | - Regina Fluhrer
- Biochemistry and Molecular Biology, Institute of Theoretical Medicine, Medical Faculty, University of Augsburg, Augsburg, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| |
Collapse
|
24
|
López C, Bosch-Príncep R, Orero G, Fontoura Balagueró L, Korzynska A, García-Rojo M, Bueno G, Fernández-Carrobles MDM, Roszkowiak L, Callau Casanova C, Salvadó-Usach MT, Jaén Martínez J, Gibert-Ramos A, Roso-Llorach A, Gras Navarro A, Berenguer-Poblet M, Llobera M, Gil Garcia J, Tomás B, Gestí V, Laine E, Plancoulaine B, Baucells J, Lejeune M. Peritumoral immune infiltrates in primary tumours are not associated with the presence of axillary lymph node metastasis in breast cancer: a retrospective cohort study. PeerJ 2020; 8:e9779. [PMID: 32953267 PMCID: PMC7474517 DOI: 10.7717/peerj.9779] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 07/30/2020] [Indexed: 11/29/2022] Open
Abstract
Background The axillary lymph nodes (ALNs) in breast cancer patients are the body regions to where tumoral cells most often first disseminate. The tumour immune response is important for breast cancer patient outcome, and some studies have evaluated its involvement in ALN metastasis development. Most studies have focused on the intratumoral immune response, but very few have evaluated the peritumoral immune response. The aim of the present article is to evaluate the immune infiltrates of the peritumoral area and their association with the presence of ALN metastases. Methods The concentration of 11 immune markers in the peritumoral areas was studied in 149 patients diagnosed with invasive breast carcinoma of no special type (half of whom had ALN metastasis at diagnosis) using tissue microarrays, immunohistochemistry and digital image analysis procedures. The differences in the concentration of the immune response of peritumoral areas between patients diagnosed with and without metastasis in their ALNs were evaluated. A multivariate logistic regression model was developed to identify the clinical-pathological variables and the peritumoral immune markers independently associated with having or not having ALN metastases at diagnosis. Results No statistically significant differences were found in the concentrations of the 11 immune markers between patients diagnosed with or without ALN metastases. Patients with metastases in their ALNs had a higher histological grade, more lymphovascular and perineural invasion and larger-diameter tumours. The multivariate analysis, after validation by bootstrap simulation, revealed that only tumour diameter (OR = 1.04; 95% CI [1.00–1.07]; p = 0.026), lymphovascular invasion (OR = 25.42; 95% CI [9.57–67.55]; p < 0.001) and histological grades 2 (OR = 3.84; 95% CI [1.11–13.28]; p = 0.033) and 3 (OR = 5.18; 95% CI [1.40–19.17]; p = 0.014) were associated with the presence of ALN metastases at diagnosis. This study is one of the first to study the association of the peritumoral immune response with ALN metastasis. We did not find any association of peritumoral immune infiltrates with the presence of ALN metastasis. Nevertheless, this does not rule out the possibility that other peritumoral immune populations are associated with ALN metastasis. This matter needs to be examined in greater depth, broadening the types of peritumoral immune cells studied, and including new peritumoral areas, such as the germinal centres of the peritumoral tertiary lymphoid structures found in extensively infiltrated neoplastic lesions.
Collapse
Affiliation(s)
- Carlos López
- Department of Pathology, Hospital de Tortosa Verge de la Cinta, Tortosa, Spain.,Campus Terres de l'Ebre, Universitat Rovira Virgili Tarragona, Tortosa, Spain
| | - Ramón Bosch-Príncep
- Department of Pathology, Hospital de Tortosa Verge de la Cinta, Tortosa, Spain
| | - Guifré Orero
- Department of Pathology, Hospital de Tortosa Verge de la Cinta, Tortosa, Spain
| | | | - Anna Korzynska
- Laboratory of Processing and Analysis of Microscopic Images, Nałęcz Institute of Biocybernetics and Biomedical Engineering, Warsaw, Poland
| | - Marcial García-Rojo
- Department of Pathology, Hospital Universitario Puerta del Mar, Cádiz, Spain
| | - Gloria Bueno
- VISILAB, Universidad de Castilla-La Mancha, Ciudad Real, Spain
| | | | - Lukasz Roszkowiak
- Laboratory of Processing and Analysis of Microscopic Images, Nałęcz Institute of Biocybernetics and Biomedical Engineering, Warsaw, Poland
| | | | - M Teresa Salvadó-Usach
- Department of Pathology, Hospital de Tortosa Verge de la Cinta, Tortosa, Spain.,Campus Terres de l'Ebre, Universitat Rovira Virgili Tarragona, Tortosa, Spain
| | | | - Albert Gibert-Ramos
- Department of Pathology, Hospital de Tortosa Verge de la Cinta, Tortosa, Spain
| | - Albert Roso-Llorach
- Institut Universitari d'Investigació en Atenció Primària Jordi Gol, Barcelona, Spain
| | - Andrea Gras Navarro
- Department of Pathology, Hospital de Tortosa Verge de la Cinta, Tortosa, Spain
| | - Marta Berenguer-Poblet
- Campus Terres de l'Ebre, Universitat Rovira Virgili Tarragona, Tortosa, Spain.,Department of Knowledge Management, Hospital de Tortosa Verge de la Cinta, Tortosa, Spain
| | - Montse Llobera
- Department of Oncology, Hospital de Tortosa Verge de la Cinta, Tortosa, Spain
| | - Júlia Gil Garcia
- Department of Surgery, Hospital Universitari de Girona Dr Josep Trueta, Girona, Spain
| | - Bárbara Tomás
- Department of Pathology, Hospital de Tortosa Verge de la Cinta, Tortosa, Spain
| | - Vanessa Gestí
- Department of Pathology, Hospital de Tortosa Verge de la Cinta, Tortosa, Spain
| | - Eeva Laine
- Department of Knowledge Management, Hospital de Tortosa Verge de la Cinta, Tortosa, Spain
| | | | - Jordi Baucells
- Department of Informatics, Hospital de Tortosa Verge de la Cinta, Tortosa, Spain
| | - Maryléne Lejeune
- Department of Pathology, Hospital de Tortosa Verge de la Cinta, Tortosa, Spain.,Campus Terres de l'Ebre, Universitat Rovira Virgili Tarragona, Tortosa, Spain
| |
Collapse
|
25
|
Wang S, Wen H, Fei W, Zhao Y, Feng Y, Kuang L, Wang M, Wu M. Two engineered site-specific antibody-drug conjugates, HLmD4 and HLvM4, have potent therapeutic activity in two DLL4-positive tumour xenograft models. Am J Cancer Res 2020; 10:2387-2408. [PMID: 32905508 PMCID: PMC7471348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 07/01/2020] [Indexed: 06/11/2023] Open
Abstract
The humanized Delta-like 4 (DLL4) monoclonal antibody H3L2 with a quite high affinity for hrDLL4 inhibits the DLL4-mediated human umbilical vein endothelial cell (HUVEC) phenotype, inducing dysfunctional angiogenesis and tumour cell apoptosis, which effectively arrests breast cancer cell growth in vivo. To develop a more effective therapy, an engineered cysteine residue at alanine 121 (Kabat numbering) on each H3L2 heavy chain or at valine 207 (Kabat numbering) on each H3L2 light chain was established by site-directed mutagenesis. Three engineered antibodies, THL4, TH2 and TL2, were identified, and the specific-site antibody-drug conjugates (ADCs) THL4-mpeoDM1 (named HLmD4), TH2-mpeoDM1 (named HmD2), TL2-mpeoDM1 (named LmD2) and THL4-vcMMAE (named HLvM4), were produced, which exhibit much more potent antitumour activity than the naked antibody. The engineered ADCs can be directed against DLL4 and effectively internalized, followed by the release of small molecule cytotoxic agents, e.g., DM1 or MMAE, into the cytosol, which inhibit the synthesis of microtubules and induce G2/M phase growth arrest and cell death through the induction of apoptosis. ADC-conjugated DM1 was highly potent against DLL4-expressing cells in vitro. We systematically compared the in vitro potency and the in vivo preclinical efficacy and safety profiles of the heterogeneous conventional ADC, H3L2-mpeoDM1 (named JmD4) with that of the homogeneous engineered conjugate HLmD4. The engineered anti-DLL4 ADCs, particularly HLmD4, showed more potent antitumour activity than Docetaxel and superior safety compared with JmD4 in two xenograft tumour models. Our findings indicate that engineered ADCs have promising potential as effective preclinical therapies for cancers.
Collapse
Affiliation(s)
- Shijing Wang
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University Nanjing 210009, China
| | - Hui Wen
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University Nanjing 210009, China
| | - Wenyi Fei
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University Nanjing 210009, China
| | - Yuhong Zhao
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University Nanjing 210009, China
| | - Yuqi Feng
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University Nanjing 210009, China
| | - Lu Kuang
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University Nanjing 210009, China
| | - Min Wang
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University Nanjing 210009, China
| | - Min Wu
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University Nanjing 210009, China
| |
Collapse
|
26
|
Leach A, Smyth P, Ferguson L, Steven J, Greene MK, Branco CM, McCann AP, Porter A, Barelle CJ, Scott CJ. Anti-DLL4 VNAR targeted nanoparticles for targeting of both tumour and tumour associated vasculature. NANOSCALE 2020; 12:14751-14763. [PMID: 32626858 DOI: 10.1039/d0nr02962a] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Whilst there is an extensive body of preclinical nanomedicine research, translation to clinical settings has been slow. Here we present a novel approach to the targeted nanoparticle (NP) concept: utilizing both a novel targeting ligand, VNAR (Variable New Antigen Receptor), a shark-derived single chain binding domain, and an under-investigated target in delta-like ligand 4 (DLL4). We describe the development of an anti-DLL4 VNAR and the site-specific conjugation of this to poly(lactic-co-glycolic) acid PEGylated NPs using surface maleimide functional groups. These nanoconjugates were shown to specifically bind DLL4 with high affinity and were preferentially internalized by DLL4-expressing pancreatic cancer cell lines and endothelial cells. Furthermore, a distinct anti-angiogenic effect endowed by the anti-DLL4 VNAR was evident in in vitro tubulogenic assays. Taken together these findings highlight the potential of anti-DLL4 targeted polymeric NPs as a novel therapeutic approach in pancreatic cancer.
Collapse
Affiliation(s)
- Adam Leach
- The Patrick G. Johnston Centre for Cancer Research, School of Medicine, Queen's University Belfast, Belfast, UK.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Jamiyan T, Kuroda H, Yamaguchi R, Abe A, Hayashi M. CD68- and CD163-positive tumor-associated macrophages in triple negative cancer of the breast. Virchows Arch 2020; 477:767-775. [PMID: 32607685 PMCID: PMC7683466 DOI: 10.1007/s00428-020-02855-z] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 05/24/2020] [Accepted: 05/28/2020] [Indexed: 12/13/2022]
Abstract
Tumor-associated macrophages (TAMs) have recently been reported as an important factor in tumor growth and the progression of cancer. The prognostic significance of localizations and densities of TAMs in triple negative cancer (TNC) of the breast is not well understood. The aim of this study was to assess the localizations and densities of the TAMs subtype in TNC and examine their clinicopathological features. The study was based on 107 TNC cases operated on at Dokkyo Medical University Hospital using the pan-macrophage marker CD68 and the M2 macrophage marker CD163 in the tumor stroma (TS) and tumor nest (TN), respectively, and examined the clinicopathological significance. Multivariate Cox regression analyses revealed that age and CD163+ TAMs in both the TS and TN were independent prognostic factors for relapse-free survival and overall survival. No correlation was found between the number of CD68+ cells or the CD163/CD68 ratio either in TS or TN, or clinicopathological features. Our study found that infiltration of CD163+ TAMs, rather than CD68+, in both TS and TN was associated with poor prognosis in TNC patients by multivariate analysis. This suggests that CD163+ TAMs may affect the prognosis of TNC by not only regulating the immune reaction by TAMs in TS, but also because of their direct influence on TN.
Collapse
Affiliation(s)
- Tsengelmaa Jamiyan
- Department of Diagnostic Pathology, Dokkyo Medical University, 880 Kitakobayashi, Mibu, Shimotsuga District, Tochigi, 321-0293, Japan.,Department of Pathology and Forensic medicine, Mongolian National University of Medical Sciences, Jamyan St 3, Ulaanbaatar, 14210, Mongolia
| | - Hajime Kuroda
- Department of Diagnostic Pathology, Dokkyo Medical University, 880 Kitakobayashi, Mibu, Shimotsuga District, Tochigi, 321-0293, Japan. .,Department of Diagnostic Pathology, Tokyo Women's Medical University, Medical Center East, 2-1-10 Nishiogu, Arakawa-ku, Tokyo 116-8567, Japan.
| | - Rin Yamaguchi
- Department of Pathology and Laboratory Medicine, Kurume University Medical Center, 155-1 Kokubumachi, Kurume, Fukuoka, 839-0863, Japan
| | - Akihito Abe
- Breast center, Dokkyo Medical University, 880 Kitakobayashi, Mibu, Shimotsuga District, Tochigi 321-0293, Japan.,Department of Surgery II, Dokkyo Medical University, 880 Kitakobayashi, Mibu, Shimotsuga District, Tochigi, 321-0293, Japan
| | - Mitsuhiro Hayashi
- Breast center, Dokkyo Medical University, 880 Kitakobayashi, Mibu, Shimotsuga District, Tochigi 321-0293, Japan
| |
Collapse
|
28
|
Xiu MX, Liu YM, Kuang BH. The Role of DLLs in Cancer: A Novel Therapeutic Target. Onco Targets Ther 2020; 13:3881-3901. [PMID: 32440154 PMCID: PMC7213894 DOI: 10.2147/ott.s244860] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2020] [Accepted: 04/06/2020] [Indexed: 12/18/2022] Open
Abstract
Delta-like ligands (DLLs) control Notch signaling. DLL1, DLL3 and DLL4 are frequently deregulated in cancer and influence tumor growth, the tumor vasculature and tumor immunity, which play different roles in cancer progression. DLLs have attracted intense research interest as anti-cancer therapeutics. In this review, we discuss the role of DLLs in cancer and summarize the emerging DLL-relevant targeting methods to aid future studies.
Collapse
Affiliation(s)
- Meng-Xi Xiu
- Medical School of Nanchang University, Nanchang, People's Republic of China
| | - Yuan-Meng Liu
- Medical School of Nanchang University, Nanchang, People's Republic of China
| | - Bo-Hai Kuang
- Medical School of Nanchang University, Nanchang, People's Republic of China
| |
Collapse
|
29
|
Sharma G, Jagtap JM, Parchur AK, Gogineni VR, Ran S, Bergom C, White SB, Flister MJ, Joshi A. Heritable modifiers of the tumor microenvironment influence nanoparticle uptake, distribution and response to photothermal therapy. Theranostics 2020; 10:5368-5383. [PMID: 32373218 PMCID: PMC7196309 DOI: 10.7150/thno.41171] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 03/16/2020] [Indexed: 12/14/2022] Open
Abstract
We report the impact of notch-DLL4-based hereditary vascular heterogeneities on the enhanced permeation and retention (EPR) effect and plasmonic photothermal therapy response in tumors. Methods: We generated two consomic rat strains with differing DLL4 expression on 3rd chromosome. These strains were based on immunocompromised Salt-sensitive or SSIL2Rγ- (DLL4-high) and SS.BN3IL2Rγ- (DLL4-low) rats with 3rd chromosome substituted from Brown Norway rat. We further constructed three novel SS.BN3IL2Rγ- congenic strains by introgressing varying segments of BN chromosome 3 into the parental SSIL2Rγ- strain to localize the role of SSIL2Rγ- DLL4 on tumor EPR effect with precision. We synthesized multimodal theranostic nanoparticles (TNPs) based on Au-nanorods which provide magnetic resonance imaging (MRI), X-ray, and optical contrasts to assess image guided PTT response and quantify host specific therapy response differences in tumors orthotopically xenografted in DLL4-high and -low strains. We tested recovery of therapy sensitivity of PTT resistant strains by employing anti-DLL4 conjugated TNPs in two triple negative breast cancer tumor xenografts. Results: Host strains with high DLL4 allele demonstrated slightly increased tumor nanoparticle uptake but consistently developed photothermal therapy resistance compared to tumors in host strains with low DLL4 allele. Tumor micro-environment with low DLL4 expression altered the geographic distribution of nanoparticles towards closer proximity with vasculature which improved efficacy of PTT in spite of lower overall TNP uptake. Targeting TNPs to tumor endothelium via anti-DLL4 antibody conjugation improved therapy sensitivity in high DLL4 allele hosts for two triple negative human breast cancer xenografts. Conclusions: Inherited DLL4 expression modulates EPR effects in tumors, and molecular targeting of endothelial DLL4 via nanoparticles is an effective personalized nanomedicine strategy.
Collapse
Affiliation(s)
- Gayatri Sharma
- Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Jaidip M. Jagtap
- Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Abdul K. Parchur
- Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WI, USA
| | | | - Sophia Ran
- Simmons Cancer Institute, Southern Illinois University School of Medicine, Springfield, IL, USA
- Department of Medical Microbiology, Immunology, and Cell Biology, Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Carmen Bergom
- Department of Radiation Oncology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Sarah B. White
- Department of Radiology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Michael J. Flister
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA
- Genomic Sciences and Precision Medicine Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Amit Joshi
- Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WI, USA
- Department of Radiology, Medical College of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
30
|
Zohny SF, Zamzami MA, Al-Malki AL, Trabulsi NH. Highly Expressed DLL4 and JAG1: Their Role in Incidence of Breast Cancer Metastasis. Arch Med Res 2020; 51:145-152. [PMID: 32111499 DOI: 10.1016/j.arcmed.2019.12.019] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 12/12/2019] [Accepted: 12/31/2019] [Indexed: 01/02/2023]
Abstract
BACKGROUND The role of Notch signaling dysregulation in causing metastatic breast cancer is not yet elucidated, therefore, this study aimed to investigate the expression of DLL4 and JAG1 in metastatic breast cancer. Moreover, we examined the possible association between clinicopathological features and studied parameters. DESIGN AND METHODS A total of 90 patients with invasive ductal breast carcinomas (52 non-metastatic and 38 metastatic) were enrolled in the current study. Furthermore, there were 42 patients with benign breast diseases. The mRNA and protein expression of DLL4 and JAG1 were analyzed by RT-PCR and ELISA, respectively in breast cell lysates. RESULTS The mRNA and protein expression of DLL4 and JAG1 were obviously higher in patients with breast cancer compared to patients with benign breast diseases and in metastatic versus non-metastatic breast cancer. A significant positive correlation was declared between DLL4 and JAG1 at both mRNA and protein levels in metastatic and localized breast cancer patients. Highly expressed mRNA and protein of DLL4 and JAG1 were associated with late tumor stages; moreover, upregulation of mRNA and protein of JAG1 was correlated with poorly differentiated tumors. CONCLUSION Our data emphasize that overexpression of DLL4 and JAG1 could predict the development of distant metastasis in breast cancer patients.
Collapse
Affiliation(s)
- Samir F Zohny
- Biochemistry Department, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia; Biochemistry Department, Faculty of Science, Ain Shams University, Abbassia, Cairo, Egypt.
| | - Mazin A Zamzami
- Biochemistry Department, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Abdulrahman L Al-Malki
- Biochemistry Department, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Nora H Trabulsi
- Department of Surgery, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
31
|
Colaprico A, Olsen C, Bailey MH, Odom GJ, Terkelsen T, Silva TC, Olsen AV, Cantini L, Zinovyev A, Barillot E, Noushmehr H, Bertoli G, Castiglioni I, Cava C, Bontempi G, Chen XS, Papaleo E. Interpreting pathways to discover cancer driver genes with Moonlight. Nat Commun 2020; 11:69. [PMID: 31900418 PMCID: PMC6941958 DOI: 10.1038/s41467-019-13803-0] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 11/22/2019] [Indexed: 12/28/2022] Open
Abstract
Cancer driver gene alterations influence cancer development, occurring in oncogenes, tumor suppressors, and dual role genes. Discovering dual role cancer genes is difficult because of their elusive context-dependent behavior. We define oncogenic mediators as genes controlling biological processes. With them, we classify cancer driver genes, unveiling their roles in cancer mechanisms. To this end, we present Moonlight, a tool that incorporates multiple -omics data to identify critical cancer driver genes. With Moonlight, we analyze 8000+ tumor samples from 18 cancer types, discovering 3310 oncogenic mediators, 151 having dual roles. By incorporating additional data (amplification, mutation, DNA methylation, chromatin accessibility), we reveal 1000+ cancer driver genes, corroborating known molecular mechanisms. Additionally, we confirm critical cancer driver genes by analysing cell-line datasets. We discover inactivation of tumor suppressors in intron regions and that tissue type and subtype indicate dual role status. These findings help explain tumor heterogeneity and could guide therapeutic decisions.
Collapse
Affiliation(s)
- Antonio Colaprico
- Interuniversity Institute of Bioinformatics in Brussels (IB)2, Brussels, Belgium.
- Machine Learning Group, Université Libre de Bruxelles (ULB), Brussels, Belgium.
- Department of Public Health Sciences, University of Miami, Miller School of Medicine, Miami, FL, 33136, USA.
| | - Catharina Olsen
- Interuniversity Institute of Bioinformatics in Brussels (IB)2, Brussels, Belgium
- Machine Learning Group, Université Libre de Bruxelles (ULB), Brussels, Belgium
- Center for Medical Genetics, Reproduction and Genetics, Reproduction Genetics and Regenerative Medicine, Vrije Universiteit Brussel, UZ Brussel, Laarbeeklaan 101, 1090, Brussels, Belgium
- Brussels Interuniversity Genomics High Throughput core (BRIGHTcore), VUB-ULB, Laarbeeklaan 101, 1090, Brussels, Belgium
| | - Matthew H Bailey
- Division of Oncology, Department of Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA
- McDonnell Genome Institute, Washington University, St. Louis, MO, 63108, USA
| | - Gabriel J Odom
- Department of Public Health Sciences, University of Miami, Miller School of Medicine, Miami, FL, 33136, USA
- Department of Biostatistics, Stempel College of Public Health, Florida International University, Miami, FL, 33199, USA
| | - Thilde Terkelsen
- Computational Biology Laboratory, and Center for Autophagy, Recycling and Disease, Danish Cancer Society Research Center, Strandboulevarden 49, 2100, Copenhagen, Denmark
| | - Tiago C Silva
- Department of Public Health Sciences, University of Miami, Miller School of Medicine, Miami, FL, 33136, USA
- Department of Genetics, Ribeirão Preto Medical School, University of Sao Paulo, Ribeirão Preto, Brazil
| | - André V Olsen
- Computational Biology Laboratory, and Center for Autophagy, Recycling and Disease, Danish Cancer Society Research Center, Strandboulevarden 49, 2100, Copenhagen, Denmark
| | - Laura Cantini
- Institut Curie, 26 rue d'Ulm, F-75248, Paris, France
- INSERM, U900, Paris, F-75248, France
- Mines ParisTech, Fontainebleau, F-77300, France
- Computational Systems Biology Team, Institut de Biologie de l'Ecole Normale Supérieure, CNRS UMR8197, INSERM U1024, Ecole Normale Supérieure, Paris Sciences et Lettres Research University, 75005, Paris, France
| | - Andrei Zinovyev
- Institut Curie, 26 rue d'Ulm, F-75248, Paris, France
- INSERM, U900, Paris, F-75248, France
- Mines ParisTech, Fontainebleau, F-77300, France
| | - Emmanuel Barillot
- Institut Curie, 26 rue d'Ulm, F-75248, Paris, France
- INSERM, U900, Paris, F-75248, France
- Mines ParisTech, Fontainebleau, F-77300, France
| | - Houtan Noushmehr
- Department of Genetics, Ribeirão Preto Medical School, University of Sao Paulo, Ribeirão Preto, Brazil
- Department of Neurosurgery, Brain Tumor Center, Henry Ford Health System, Detroit, MI, USA
| | - Gloria Bertoli
- Institute of Molecular Bioimaging and Physiology of the National Research Council (IBFM-CNR), Milan, Italy
| | - Isabella Castiglioni
- Institute of Molecular Bioimaging and Physiology of the National Research Council (IBFM-CNR), Milan, Italy
| | - Claudia Cava
- Institute of Molecular Bioimaging and Physiology of the National Research Council (IBFM-CNR), Milan, Italy
| | - Gianluca Bontempi
- Interuniversity Institute of Bioinformatics in Brussels (IB)2, Brussels, Belgium
- Machine Learning Group, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Xi Steven Chen
- Department of Public Health Sciences, University of Miami, Miller School of Medicine, Miami, FL, 33136, USA.
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, 33136, USA.
| | - Elena Papaleo
- Computational Biology Laboratory, and Center for Autophagy, Recycling and Disease, Danish Cancer Society Research Center, Strandboulevarden 49, 2100, Copenhagen, Denmark.
- Translational Disease System Biology, Faculty of Health and Medical Science, Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
32
|
Gibert-Ramos A, López C, Bosch R, Fontoura L, Bueno G, García-Rojo M, Berenguer M, Lejeune M. Immune response profile of primary tumour, sentinel and non-sentinel axillary lymph nodes related to metastasis in breast cancer: an immunohistochemical point of view. Histochem Cell Biol 2019; 152:177-193. [DOI: 10.1007/s00418-019-01802-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/01/2019] [Indexed: 12/24/2022]
|
33
|
Zhou R, Wang S, Wen H, Wang M, Wu M. The bispecific antibody HB-32, blockade of both VEGF and DLL4 shows potent anti-angiogenic activity in vitro and anti-tumor activity in breast cancer xenograft models. Exp Cell Res 2019; 380:141-148. [DOI: 10.1016/j.yexcr.2019.04.025] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 04/19/2019] [Accepted: 04/22/2019] [Indexed: 10/26/2022]
|
34
|
Li S, Chen C, Xiong X, Huang Y, Hu J, Fan Z, Ling K. Type Iγ phosphatidylinositol phosphate kinase dependent cell migration and invasion are dispensable for tumor metastasis. Am J Cancer Res 2019; 9:959-974. [PMID: 31218104 PMCID: PMC6556613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 04/01/2019] [Indexed: 06/09/2023] Open
Abstract
Type Iγ phosphatidylinositol phosphate kinase (PIPKIγ) has been associated with poor prognosis in breast cancer patients by promoting metastasis. Among the six alternative-splicing isoforms of PIPKIγ, PIPKIγ_i2 specifically targets to focal adhesions and regulates focal adhesion turnover, thus was proposed responsible for tumor metastasis. In the present study, we specifically depleted PIPKIγ_i2 from mouse triple negative breast cancer (TNBC) 4T1 cells and analyzed their behaviors. As expected, PIPKIγ_i2-depleted 4T1 cells exhibited reduced proliferation, migration, and invasion in vitro at a comparable level as pan-PIPKIγ depleted cells. However, PIPKIγ_i2 depletion had no effect on metastasis and progression of 4T1 tumors in vivo. PIPKIγ_i2-depleted tumors showed similar levels of growth, hypoxia state, macrophage infiltration, and angiogenesis as parental tumors, although the pan-PIPKIγ depletion led to substantial inhibition on these aspects. Further investigation revealed that depleting PIPKIγ_i2 alone, unlike depleting all PIPKIγ isoforms, had no effect on PD-L1 expression, the status of the epithelial-to-mesenchymal transition, and the anchorage-independent growth of 4T1 cells. In human TNBC MDA-MB-231 cells, we obtained similar results. Thus, while PIPKIγ_i2 indeed is required for cell migration and invasion, these characteristics are not decisive for metastasis. Other PIPKIγ isoform(s) that regulate the expression of key factors to support cell survival under stresses is more critical for the malignant progression of TNBCs.
Collapse
Affiliation(s)
- Shiheng Li
- Department of Breast Surgery, The First Hospital of Jilin University71 Xinmin Street, Changchun, Jilin Province, China
- Department of Biochemistry and Molecular Biology, Mayo Clinic200 First Street SW, Rochester, MN 55902, USA
| | - Chunhua Chen
- Department of Biochemistry and Molecular Biology, Mayo Clinic200 First Street SW, Rochester, MN 55902, USA
| | - Xunhao Xiong
- Department of Pathology, The University of Oklahoma Health Sciences Center940 Stanton L Young Blvd, Oklahoma City, Oklahoma, USA
| | - Yan Huang
- Department of Biochemistry and Molecular Biology, Mayo Clinic200 First Street SW, Rochester, MN 55902, USA
- Department of Nephrology and Hypertension, Mayo Clinic200 First Street SW, Rochester, MN 55902, USA
| | - Jinghua Hu
- Department of Biochemistry and Molecular Biology, Mayo Clinic200 First Street SW, Rochester, MN 55902, USA
- Department of Nephrology and Hypertension, Mayo Clinic200 First Street SW, Rochester, MN 55902, USA
| | - Zhimin Fan
- Department of Breast Surgery, The First Hospital of Jilin University71 Xinmin Street, Changchun, Jilin Province, China
| | - Kun Ling
- Department of Biochemistry and Molecular Biology, Mayo Clinic200 First Street SW, Rochester, MN 55902, USA
| |
Collapse
|
35
|
Baharlou R, Tajik N, Behdani M, Shokrgozar MA, Tavana V, Kazemi-Lomedasht F, Faraji F, Habibi-Anbouhi M. An antibody fragment against human delta-like ligand-4 for inhibition of cell proliferation and neovascularization. Immunopharmacol Immunotoxicol 2018; 40:368-374. [DOI: 10.1080/08923973.2018.1505907] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Rasoul Baharlou
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Immunology Research Center (IRC), Iran University of Medical Sciences, Tehran, Iran
| | - Nader Tajik
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Immunology Research Center (IRC), Iran University of Medical Sciences, Tehran, Iran
| | - Mahdi Behdani
- Biotechnology Research Center, Venom & Biotherapeutics Molecules Laboratory, Pasteur Institute of Iran, Tehran, Iran
| | | | - Vajiheh Tavana
- Biotechnology Research Center, Venom & Biotherapeutics Molecules Laboratory, Pasteur Institute of Iran, Tehran, Iran
| | - Fatemeh Kazemi-Lomedasht
- Biotechnology Research Center, Venom & Biotherapeutics Molecules Laboratory, Pasteur Institute of Iran, Tehran, Iran
| | - Fatemeh Faraji
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Immunology Research Center (IRC), Iran University of Medical Sciences, Tehran, Iran
| | | |
Collapse
|
36
|
Propranolol inhibits proliferation and invasion of hemangioma-derived endothelial cells by suppressing the DLL4/Notch1/Akt pathway. Chem Biol Interact 2018; 294:28-33. [PMID: 30130526 DOI: 10.1016/j.cbi.2018.08.018] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 07/29/2018] [Accepted: 08/17/2018] [Indexed: 11/21/2022]
Abstract
Infantile hemangioma (IH) is one of the most common benign vascular tumors of infancy. Propranolol has been recently introduced for the treatment of IH. However, the mechanism of protective effect has not been fully understood. In this study, hemangioma-derived endothelial cells (HemECs) were isolated and treated with propranolol. The cell viability was measured by MTT assay, and the cell cycle arrest was detected using flow cytometry. Cell invasion was determined using transwell assay. The expressions of matrix metalloproteinase (MMP)-2, MMP-9, Delta-like 4 (DLL4), Notch1, Akt, p-Akt, and vascular endothelial growth factor (VEGF) were detected using western blot. HemECs were incubated with recombinant human DLL4 (rhDLL4) to investigate the role of DLL4/Notch1 in the effect of propranolol. The results showed that propranolol inhibited cell viability of HemECs in a time-dependent manner. Propranolol suppressed cell proliferation of HemECs by arresting cell progression at G0/G1 phase. Propranolol inhibited the invasion ability of HemECs and reduced the expression levels of MMP-2 and MMP-9 in HemECs. Besides, propranolol treatment blocked the DLL4/Notch1 and Akt signaling and inhibited VEGF expression in HemECs. Treatment with rhDLL4 activated the Akt signaling and attenuated the effect of propranolol on HemECs. Our data indicated that propranolol inhibited the cell proliferation and invasion of HemECs. The effect was possibly involved in the DLL4/Notch1/Akt signaling pathway.
Collapse
|
37
|
The Notch Pathway in Breast Cancer Progression. ScientificWorldJournal 2018; 2018:2415489. [PMID: 30111989 PMCID: PMC6077551 DOI: 10.1155/2018/2415489] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 05/10/2018] [Accepted: 06/12/2018] [Indexed: 12/29/2022] Open
Abstract
Objective Notch signaling pathway is a vital parameter of the mammalian vascular system. In this review, the authors summarize the current knowledge about the impact of the Notch signaling pathway in breast cancer progression and the therapeutic role of Notch's inhibition. Methods The available literature in MEDLINE, PubMed, and Scopus, regarding the role of the Notch pathway in breast cancer progression was searched for related articles from about 1973 to 2017 including terms such as “Notch,” “Breast Cancer,” and “Angiogenesis.” Results. Notch signaling controls the differentiation of breast epithelial cells during normal development. Studies confirm that the Notch pathway has a major participation in breast cancer progression through overexpression and/or abnormal genetic type expression of the notch receptors and ligands that determine angiogenesis. The cross-talk of Notch and estrogens, the effect of Notch in breast cancer stem cells formation, and the dependable Notch overexpression during breast tumorigenesis have been studied enough and undoubtedly linked to breast cancer development. The already applied therapeutic inhibition of Notch for breast cancer can drastically change the course of the disease. Conclusion Current data prove that Notch pathway has a major participation and multiple roles during breast tumor progression. Inhibition of Notch receptors and ligands provides innovative therapeutic results and could become the therapy of choice in the next few years, even though further research is needed to reach safe conclusions.
Collapse
|
38
|
Yang M, Li Z, Ren M, Li S, Zhang L, Zhang X, Liu F. Stromal Infiltration of Tumor-Associated Macrophages Conferring Poor Prognosis of Patients with Basal-Like Breast Carcinoma. J Cancer 2018; 9:2308-2316. [PMID: 30026826 PMCID: PMC6036715 DOI: 10.7150/jca.25155] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 03/30/2018] [Indexed: 12/12/2022] Open
Abstract
Aims: Tumor associated macrophages (TAMs) play a critical role in the initiation and progression of breast cancer. However, their prognostic significance in the molecular subtype of basal-like breast cancer (BLBC) is poorly understood. The aim of this study was to investigate the extent and patterns of TAMs in BLBC and their associations with clinicopathological features and patient survival. Methods and Results: We evaluated TAMs in 200 cases of BLBC by immunohistochemistry using the M2 macrophage marker CD163 and the pan-macrophage marker CD68 in tumor nest and stroma, and assessed their prognostic significance. The study demonstrated that infiltration of CD163+ and CD68+ macrophages in tumor stroma was of clinical relevance in BLBC, but not those in tumor nest. Increased stromal infiltration of CD68+ or CD163+ macrophages correlated with larger tumor size, higher histological grade, higher 5-year recurrence and 5-year breast cancer mortality. Although both of CD68+ and CD163+ macrophages in tumor stroma were associated with poor recurrence-free survival (RFS) and overall survival (OS), multivariate analysis demonstrated that only CD163+ macrophage was an independent predictor of RFS and OS. Conclusions: Our results highlight the prognostic importance of TAMs' location in BLBC. CD163, a highly specific biomarker for M2 macrophages, is an independent prognostic marker for BLBC patients, and may serve as an indicator or potential target of macrophage-centred therapeutic strategies.
Collapse
Affiliation(s)
- Mu Yang
- Department of Breast Pathology and Research Laboratory, Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education), National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China.,Department of Pathology, University Medical Center of Princeton, Plainsboro, NJ 08854, USA
| | - Zhenhua Li
- Department of Breast Pathology and Research Laboratory, Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education), National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China
| | - Meijing Ren
- Department of Breast Pathology and Research Laboratory, Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education), National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China
| | - Shuai Li
- Department of Breast Pathology and Research Laboratory, Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education), National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China
| | - Lanjing Zhang
- Department of Pathology, University Medical Center of Princeton, Plainsboro, NJ 08854, USA.,Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey, USA.,Department of Biological Sciences, Rutgers University, Newark, New Jersey, USA.,Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey, USA
| | - Xinmin Zhang
- Department of Pathology, Cooper University Hospital, Cooper Medical School of Rowan University, Camden, New Jersey 08103, USA
| | - Fangfang Liu
- Department of Breast Pathology and Research Laboratory, Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education), National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China
| |
Collapse
|
39
|
Li Y, Hickson JA, Ambrosi DJ, Haasch DL, Foster-Duke KD, Eaton LJ, DiGiammarino EL, Panchal SC, Jiang F, Mudd SR, Zhang C, Akella SS, Gao W, Ralston SL, Naumovski L, Gu J, Morgan-Lappe SE. ABT-165, a Dual Variable Domain Immunoglobulin (DVD-Ig) Targeting DLL4 and VEGF, Demonstrates Superior Efficacy and Favorable Safety Profiles in Preclinical Models. Mol Cancer Ther 2018; 17:1039-1050. [PMID: 29592882 DOI: 10.1158/1535-7163.mct-17-0800] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Revised: 12/28/2017] [Accepted: 03/08/2018] [Indexed: 11/16/2022]
Abstract
Antiangiogenic therapy is a clinically validated modality in cancer treatment. To date, all approved antiangiogenic drugs primarily inhibit the VEGF pathway. Delta-like ligand 4 (DLL4) has been identified as a potential drug target in VEGF-independent angiogenesis and tumor-initiating cell (TIC) survival. A dual-specific biologic targeting both VEGF and DLL4 could be an attractive strategy to improve the effectiveness of anti-VEGF therapy. ABT-165 was uniquely engineered using a proprietary dual-variable domain immunoglobulin (DVD-Ig) technology based on its ability to bind and inhibit both DLL4 and VEGF. In vivo, ABT-165 induced significant tumor growth inhibition compared with either parental antibody treatment alone, due, in part, to the disruption of functional tumor vasculature. In combination with chemotherapy agents, ABT-165 also induced greater antitumor response and outperformed anti-VEGF treatment. ABT-165 displayed nonlinear pharmacokinetic profiles in cynomolgus monkeys, with an apparent terminal half-life > 5 days at a target saturation dose. In a GLP monkey toxicity study, ABT-165 was well-tolerated at doses up to 200 mg/kg with non-adverse treatment-related histopathology findings limited to the liver and thymus. In summary, ABT-165 represents a novel antiangiogenic strategy that potently inhibits both DLL4 and VEGF, demonstrating favorable in vivo efficacy, pharmacokinetic, and safety profiles in preclinical models. Given these preclinical attributes, ABT-165 has progressed to a phase I study. Mol Cancer Ther; 17(5); 1039-50. ©2018 AACR.
Collapse
Affiliation(s)
- Yingchun Li
- Oncology Discovery, AbbVie Inc., North Chicago, Illinois
| | | | | | | | | | | | | | | | - Fang Jiang
- Oncology Discovery, AbbVie Inc., North Chicago, Illinois
| | - Sarah R Mudd
- Translational Imaging, AbbVie Inc., North Chicago, Illinois
| | - Catherine Zhang
- Drug Metabolism and Pharmacokinetics - Bioanalysis, AbbVie Biotherapeutics, Redwood City, California
| | - Surekha S Akella
- Preclinical Safety, AbbVie Biotherapeutics, Redwood City, California
| | - Wenqing Gao
- Drug Metabolism and Pharmacokinetics, AbbVie Inc., North Chicago, Illinois
| | | | - Louie Naumovski
- Oncology Early Development, AbbVie Inc., Redwood City, California
| | - Jijie Gu
- AbbVie Bioresearch Center, Worcester, Massachusetts
| | | |
Collapse
|
40
|
Baharlou R, Tajik N, Habibi-Anbouhi M, Shokrgozar MA, Zarnani AH, Shahhosseini F, Behdani M. Generation and characterization of an anti-delta like ligand-4 Nanobody to induce non-productive angiogenesis. Anal Biochem 2018; 544:34-41. [DOI: 10.1016/j.ab.2017.12.014] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Revised: 10/23/2017] [Accepted: 12/11/2017] [Indexed: 12/25/2022]
|
41
|
Wang S, Zhou R, Sun F, Li R, Wang M, Wu M. The two novel DLL4-targeting antibody-drug conjugates MvM03 and MGD03 show potent anti-tumour activity in breast cancer xenograft models. Cancer Lett 2017; 409:125-136. [PMID: 28923397 DOI: 10.1016/j.canlet.2017.09.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 08/28/2017] [Accepted: 09/10/2017] [Indexed: 11/18/2022]
Abstract
The anti-human Delta-like 4 (DLL4) monoclonal antibody MMGZ01 has a high affinity to hrDLL4 and arrests the DLL4-mediated human umbilical vein endothelial cell (HUVEC) phenotype, promotes immature vessels, and effectively reduces breast cancer cell growth in vivo. To develop a much more effective therapy, we conjugated MMGZ01 with two small-molecule cytotoxic agents, i.e., monomethyl auristatin E (MMAE) and doxorubicin (DOX), with different linkers to generate antibody-drug conjugates (ADCs), i.e., MMGZ01-vc-MMAE (named MvM03) and MMGZ01-GMBS-DOX (named MGD03), that are more potent therapeutic agents than naked antibody therapeutic agents. The produced anti-DLL4 ADCs can be effectively directed against DLL4 and internalized. Then, the release of MMAE or DOX into the cytosol can induce G2/M or G0/G1 phase growth arrest and cell death through the induction of apoptosis. In vitro, MvM03 was highly potent and selective against DLL4 cell lines. The anti-DLL4 ADCs, particularly MvM03, showed more potent anti-tumour activity than Docetaxel, which is an inhibitor of the depolymerisation of microtubules, in two xenograft breast cancer tumour models. Our findings indicate that anti-DLL4 ADCs have promising potential as an effective therapy for breast cancer.
Collapse
Affiliation(s)
- Shijing Wang
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China
| | - Rihong Zhou
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China
| | - Fumou Sun
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China
| | - Renjie Li
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China
| | - Min Wang
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China.
| | - Min Wu
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
42
|
Oon CE, Bridges E, Sheldon H, Sainson RC, Jubb A, Turley H, Leek R, Buffa F, Harris AL, Li JL. Role of Delta-like 4 in Jagged1-induced tumour angiogenesis and tumour growth. Oncotarget 2017; 8:40115-40131. [PMID: 28445154 PMCID: PMC5522274 DOI: 10.18632/oncotarget.16969] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Accepted: 03/11/2017] [Indexed: 12/20/2022] Open
Abstract
Delta-like 4 (DLL4) and Jagged1 (JAG1) are two key Notch ligands implicated in tumour angiogenesis. They were shown to have opposite effects on mouse retinal and adult regenerative angiogenesis. In tumours, both ligands are upregulated but their relative effects and interactions in tumour biology, particularly in tumour response to therapeutic intervention are unclear. Here we demonstrate that DLL4 and JAG1 displayed equal potency in stimulating Notch target genes in HMEC-1 endothelial cells but had opposing effects on sprouting angiogenesis in vitro. Mouse DLL4 or JAG1 expressed in glioblastoma cells decreased tumour cell proliferation in vitro but promoted tumour growth in vivo. mDLL4-expressing tumours showed fewer but larger vessels whereas mJAG1-tumours produced more vessels. In both tumour types pericyte coverage was decreased but the vessels were more perfused. Both ligands increased tumour resistance towards anti-VEGF therapy but the resistance was higher in mDLL4-tumours versus mJAG1-tumours. However, their sensitivity to the therapy was restored by blocking Notch signalling with dibenzazepine. Importantly, anti-DLL4 antibody blocked the effect of JAG1 on tumour growth and increased vessel branching in vivo. The mechanism behind the differential responsiveness was due to a positive feedback loop for DLL4-Notch signalling, rendering DLL4 more dominant in activating Notch signalling in the tumour microenvironment. We concluded that DLL4 and JAG1 promote tumour growth by modulating tumour angiogenesis via different mechanisms. JAG1 is not antagonistic but utilises DLL4 in tumour angiogenesis. The results suggest that anti-JAG1 therapy should be explored in conjunction with anti-DLL4 treatment in developing anti-Notch therapies in clinics.
Collapse
Affiliation(s)
- Chern Ein Oon
- Molecular Oncology Laboratories, Department of Oncology, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, Penang, Malaysia
| | - Esther Bridges
- Molecular Oncology Laboratories, Department of Oncology, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Helen Sheldon
- Molecular Oncology Laboratories, Department of Oncology, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Richard C.A. Sainson
- Molecular Oncology Laboratories, Department of Oncology, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Adrian Jubb
- Molecular Oncology Laboratories, Department of Oncology, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Helen Turley
- Molecular Oncology Laboratories, Department of Oncology, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Russell Leek
- Molecular Oncology Laboratories, Department of Oncology, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Francesca Buffa
- Molecular Oncology Laboratories, Department of Oncology, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Adrian L. Harris
- Molecular Oncology Laboratories, Department of Oncology, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Ji-Liang Li
- Molecular Oncology Laboratories, Department of Oncology, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
- Institute of Translational and Stratified Medicine, Plymouth University Peninsula Schools of Medicine and Dentistry, Plymouth, UK
| |
Collapse
|
43
|
Trindade A, Djokovic D, Gigante J, Mendonça L, Duarte A. Endothelial Dll4 overexpression reduces vascular response and inhibits tumor growth and metastasization in vivo. BMC Cancer 2017; 17:189. [PMID: 28288569 PMCID: PMC5348880 DOI: 10.1186/s12885-017-3171-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Accepted: 03/04/2017] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND The inhibition of Delta-like 4 (Dll4)/Notch signaling has been shown to result in excessive, nonfunctional vessel proliferation and significant tumor growth suppression. However, safety concerns emerged with the identification of side effects resulting from chronic Dll4/Notch blockade. Alternatively, we explored the endothelial Dll4 overexpression using different mouse tumor models. METHODS We used a transgenic mouse model of endothelial-specific Dll4 overexpression, previously produced. Growth kinetics and vascular histopathology of several types of solid tumors was evaluated, namely Lewis Lung Carcinoma xenografts, chemically-induced skin papillomas and RIP1-Tag2 insulinomas. RESULTS We found that increased Dll4/Notch signaling reduces tumor growth by reducing vascular endothelial growth factor (VEGF)-induced endothelial proliferation, tumor vessel density and overall tumor blood supply. In addition, Dll4 overexpression consistently improved tumor vascular maturation and functionality, as indicated by increased vessel calibers, enhanced mural cell recruitment and increased network perfusion. Importantly, the tumor vessel normalization is not more effective than restricted vessel proliferation, but was found to prevent metastasis formation and allow for increased delivery to the tumor of concomitant chemotherapy, improving its efficacy. CONCLUSIONS By reducing endothelial sensitivity to VEGF, these results imply that Dll4/Notch stimulation in tumor microenvironment could be beneficial to solid cancer patient treatment by reducing primary tumor size, improving tumor drug delivery and reducing metastization. Endothelial specific Dll4 overexpression thus appears as a promising anti-angiogenic modality that might improve cancer control.
Collapse
Affiliation(s)
- Alexandre Trindade
- Centro Interdisciplinar de Investigação em Sanidade Animal (CIISA), Faculdade de Medicina Veterinária, University of Lisbon, Lisbon, Portugal
| | - Dusan Djokovic
- Centro Interdisciplinar de Investigação em Sanidade Animal (CIISA), Faculdade de Medicina Veterinária, University of Lisbon, Lisbon, Portugal.,Faculdade de Ciências Médicas, Nova Medical School, Nova University of Lisbon, Lisbon, Portugal.,Serviço de Obstetrícia e Ginecologia, Centro Hospitalar de Lisboa Ocidental, Hospital de São Francisco Xavier, Lisbon, Portugal
| | - Joana Gigante
- Centro Interdisciplinar de Investigação em Sanidade Animal (CIISA), Faculdade de Medicina Veterinária, University of Lisbon, Lisbon, Portugal
| | - Liliana Mendonça
- Centro Interdisciplinar de Investigação em Sanidade Animal (CIISA), Faculdade de Medicina Veterinária, University of Lisbon, Lisbon, Portugal
| | - António Duarte
- Centro Interdisciplinar de Investigação em Sanidade Animal (CIISA), Faculdade de Medicina Veterinária, University of Lisbon, Lisbon, Portugal.
| |
Collapse
|
44
|
Merk H, Zhang S, Lehr T, Müller C, Ulrich M, Bibb JA, Adams RH, Bracher F, Zahler S, Vollmar AM, Liebl J. Inhibition of endothelial Cdk5 reduces tumor growth by promoting non-productive angiogenesis. Oncotarget 2017; 7:6088-104. [PMID: 26755662 PMCID: PMC4868742 DOI: 10.18632/oncotarget.6842] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Accepted: 12/29/2015] [Indexed: 11/25/2022] Open
Abstract
Therapeutic success of VEGF-based anti-angiogenic tumor therapy is limited due to resistance. Thus, new strategies for anti-angiogenic cancer therapy based on novel targets are urgently required. Our previous in vitro work suggested that small molecule Cdk5 inhibitors affect angiogenic processes such as endothelial migration and proliferation. Moreover, we recently uncovered a substantial role of Cdk5 in the development of lymphatic vessels. Here we pin down the in vivo impact of endothelial Cdk5 inhibition in angiogenesis and elucidate the underlying mechanism in order to judge the potential of Cdk5 as a novel anti-angiogenic and anti-cancer target. By the use of endothelial-specific Cdk5 knockout mouse models and various endothelial and tumor cell based assays including human tumor xenograft models, we show that endothelial-specific knockdown of Cdk5 results in excessive but non-productive angiogenesis during development but also in tumors, which subsequently leads to inhibition of tumor growth. As Cdk5 inhibition disrupted Notch function by reducing the generation of the active Notch intracellular domain (NICD) and Cdk5 modulates Notch-dependent endothelial cell proliferation and sprouting, we propose that the Dll4/Notch driven angiogenic signaling hub is an important and promising mechanistic target of Cdk5. In fact, Cdk5 inhibition can sensitize tumors to conventional anti-angiogenic treatment as shown in tumor xenograft models. In summary our data set the stage for Cdk5 as a drugable target to inhibit Notch-driven angiogenesis condensing the view that Cdk5 is a promising target for cancer therapy.
Collapse
Affiliation(s)
- Henriette Merk
- Department of Pharmacy, Pharmaceutical Biology, Ludwig-Maximilians-University, 81377 Munich, Germany
| | - Siwei Zhang
- Department of Pharmacy, Pharmaceutical Biology, Ludwig-Maximilians-University, 81377 Munich, Germany
| | - Thorsten Lehr
- Clinical Pharmacy, Saarland University, 66123 Saarbrücken, Germany
| | - Christoph Müller
- Department of Pharmacy, Pharmaceutical Chemistry, Ludwig-Maximilians-University, 81377 Munich, Germany
| | - Melanie Ulrich
- Department of Pharmacy, Pharmaceutical Biology, Ludwig-Maximilians-University, 81377 Munich, Germany
| | - James A Bibb
- Department of Psychiatry and Neurology and Neurotherapeutics, The University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Ralf H Adams
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, 48149 Münster, Germany.,University of Münster, Faculty of Medicine, 48149 Münster, Germany
| | - Franz Bracher
- Department of Pharmacy, Pharmaceutical Chemistry, Ludwig-Maximilians-University, 81377 Munich, Germany
| | - Stefan Zahler
- Department of Pharmacy, Pharmaceutical Biology, Ludwig-Maximilians-University, 81377 Munich, Germany
| | - Angelika M Vollmar
- Department of Pharmacy, Pharmaceutical Biology, Ludwig-Maximilians-University, 81377 Munich, Germany
| | - Johanna Liebl
- Department of Pharmacy, Pharmaceutical Biology, Ludwig-Maximilians-University, 81377 Munich, Germany
| |
Collapse
|
45
|
A pilot study on plasma levels of micro-RNAs involved in angiogenesis and vascular maturation in patients with breast cancer. Med Oncol 2017; 34:20. [DOI: 10.1007/s12032-016-0881-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Accepted: 12/29/2016] [Indexed: 01/10/2023]
|
46
|
Ibrahim SA, Katara GK, Kulshrestha A, Jaiswal MK, Amin MA, Beaman KD. Breast cancer associated a2 isoform vacuolar ATPase immunomodulates neutrophils: potential role in tumor progression. Oncotarget 2016; 6:33033-45. [PMID: 26460736 PMCID: PMC4741747 DOI: 10.18632/oncotarget.5439] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Accepted: 09/29/2015] [Indexed: 12/22/2022] Open
Abstract
In invasive breast cancer, tumor associated neutrophils (TAN) represent a significant portion of the tumor mass and are associated with increased angiogenesis and metastasis. Identifying the regulatory factors that control TAN behavior will help in developing ideal immunotherapies. Vacuolar ATPases (V-ATPases), multi-subunit proton pumps, are highly expressed in metastatic breast cancer cells. A cleaved peptide from a2 isoform V-ATPase (a2NTD) has immunomodulatory role in tumor microenvironment. Here, we report for the first time the role of V-ATPase in neutrophils modulation. In invasive breast cancer cells, a2NTD was detected and a2V was highly expressed on the surface. Immunohistochemical analysis of invasive breast cancer tissues revealed that increased neutrophil recruitment and blood vessel density correlated with increased a2NTD levels. In order to determine the direct regulatory role of a2NTD on neutrophils, recombinant a2NTD was used for the treatment of neutrophils isolated from the peripheral blood of healthy volunteers. Neutrophils treated with a2NTD (a2Neuɸ) showed increased secretion of IL-1RA, IL-10, CCL-2 and IL-6 that are important mediators in cancer related inflammation. Moreover, a2Neuɸ exhibited an increased production of protumorigenic factors including IL-8, matrix metaloprotinase-9 and vascular endothelial growth factor. Further, functional characterization of a2Neuɸ revealed that a2Neuɸ derived products induce in vitro angiogenesis as well as increase the invasiveness of breast cancer cells. This study establishes the modulatory effect of breast cancer associated a2V on neutrophils, by the action of a2NTD, which has a positive impact on tumor progression, supporting that a2V can be a potential selective target for breast cancer therapy.
Collapse
Affiliation(s)
- Safaa A Ibrahim
- Department of Microbiology and Immunology, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA.,Department of Microbiology and Immunology, Faculty of Pharmacy, Cairo University, Giza, Egypt
| | - Gajendra K Katara
- Department of Microbiology and Immunology, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Arpita Kulshrestha
- Department of Microbiology and Immunology, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Mukesh K Jaiswal
- Department of Microbiology and Immunology, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Magdy A Amin
- Department of Microbiology and Immunology, Faculty of Pharmacy, Cairo University, Giza, Egypt
| | - Kenneth D Beaman
- Department of Microbiology and Immunology, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| |
Collapse
|
47
|
Lee JH, Zhao XM, Yoon I, Lee JY, Kwon NH, Wang YY, Lee KM, Lee MJ, Kim J, Moon HG, In Y, Hao JK, Park KM, Noh DY, Han W, Kim S. Integrative analysis of mutational and transcriptional profiles reveals driver mutations of metastatic breast cancers. Cell Discov 2016; 2:16025. [PMID: 27625789 PMCID: PMC5004232 DOI: 10.1038/celldisc.2016.25] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 06/21/2016] [Indexed: 12/11/2022] Open
Abstract
Despite the explosion in the numbers of cancer genomic studies, metastasis is still the major cause of cancer mortality. In breast cancer, approximately one-fifth of metastatic patients survive 5 years. Therefore, detecting the patients at a high risk of developing distant metastasis at first diagnosis is critical for effective treatment strategy. We hereby present a novel systems biology approach to identify driver mutations escalating the risk of metastasis based on both exome and RNA sequencing of our collected 78 normal-paired breast cancers. Unlike driver mutations occurring commonly in cancers as reported in the literature, the mutations detected here are relatively rare mutations occurring in less than half metastatic samples. By supposing that the driver mutations should affect the metastasis gene signatures, we develop a novel computational pipeline to identify the driver mutations that affect transcription factors regulating metastasis gene signatures. We identify driver mutations in ADPGK, NUP93, PCGF6, PKP2 and SLC22A5, which are verified to enhance cancer cell migration and prompt metastasis with in vitro experiments. The discovered somatic mutations may be helpful for identifying patients who are likely to develop distant metastasis.
Collapse
Affiliation(s)
- Ji-Hyun Lee
- Medicinal Bioconvergence Research Center, College of Pharmacy, Seoul National University, Seoul, Republic of Korea; Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Xing-Ming Zhao
- Department of Computer Science and Technology, Tongji University , Shanghai, China
| | - Ina Yoon
- Medicinal Bioconvergence Research Center, College of Pharmacy, Seoul National University , Seoul, Republic of Korea
| | - Jin Young Lee
- Medicinal Bioconvergence Research Center, College of Pharmacy, Seoul National University , Seoul, Republic of Korea
| | - Nam Hoon Kwon
- Medicinal Bioconvergence Research Center, College of Pharmacy, Seoul National University , Seoul, Republic of Korea
| | - Yin-Ying Wang
- Department of Computer Science and Technology, Tongji University , Shanghai, China
| | - Kyung-Min Lee
- Department of Surgery, Seoul National University College of Medicine , Seoul, Republic of Korea
| | - Min-Joo Lee
- Department of Surgery, Seoul National University College of Medicine , Seoul, Republic of Korea
| | - Jisun Kim
- Department of Surgery, Seoul National University College of Medicine , Seoul, Republic of Korea
| | - Hyeong-Gon Moon
- Department of Surgery, Seoul National University College of Medicine , Seoul, Republic of Korea
| | - Yongho In
- Medicinal Bioconvergence Research Center, College of Pharmacy, Seoul National University , Seoul, Republic of Korea
| | - Jin-Kao Hao
- LERIA, University of Angers , Angers, France
| | - Kyung-Mii Park
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University , Seoul, Republic of Korea
| | - Dong-Young Noh
- Department of Surgery, Seoul National University College of Medicine , Seoul, Republic of Korea
| | - Wonshik Han
- Department of Surgery, Seoul National University College of Medicine, Seoul, Republic of Korea; Cancer Research Institute, Seoul National University, Seoul, Republic of Korea
| | - Sunghoon Kim
- Medicinal Bioconvergence Research Center, College of Pharmacy, Seoul National University, Seoul, Republic of Korea; Department of Molecular Medicine and Biopharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
48
|
Wang Q, Shi Y, Butler HJ, Xue J, Wang G, Duan P, Zheng H. Role of delta-like ligand-4 in chemoresistance against docetaxel in MCF-7 cells. Hum Exp Toxicol 2016; 36:328-338. [PMID: 27334972 DOI: 10.1177/0960327116650006] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
As Notch receptors have been shown to induce chemoresistance, we hypothesized that delta-like ligand-4 (DLL4), a central Notch signalling ligand, might also participate in chemoresistance in breast cancer. To investigate this issue, overexpression of DLL4 was induced by transfection with expression vectors for DLL4 in the human breast cancer cell line Michigan cancer foundation-7 (MCF-7). It was found that DLL4 could be adaptively upregulated by docetaxel (DOC) treatment in a dose-dependent manner, but Notch1 was unaffected. Overexpression of DLL4 could significantly attenuate the cytotoxic effects of DOC by increasing Bcl-2 expression, while decreasing Bax expression, apoptosis rate and DNA damage. The protective effects of DLL4 made cells acquire chemoresistance against DOC and resulted in cancer cell survival. DLL4 is normally regarded as a regulator of vascular development. Our results expanded the understanding of DLL4. Since DLL4 may play an important role in the process of acquiring chemoresistance, it may be a promising target in overcoming chemoresistance in breast cancer.
Collapse
Affiliation(s)
- Q Wang
- 1 Shiyan Taihe Hospital (affiliated Hospital of Hubei University of Medicine), Hubei, Shiyan, China.,2 HuBei University of Medicine, Hubei Shiyan, China
| | - Y Shi
- 1 Shiyan Taihe Hospital (affiliated Hospital of Hubei University of Medicine), Hubei, Shiyan, China
| | - H J Butler
- 3 Centre for Biophotonics, Lancaster Environment Centre, Lancaster University, Bailrigg, Lancaster, UK
| | - J Xue
- 1 Shiyan Taihe Hospital (affiliated Hospital of Hubei University of Medicine), Hubei, Shiyan, China
| | - G Wang
- 1 Shiyan Taihe Hospital (affiliated Hospital of Hubei University of Medicine), Hubei, Shiyan, China
| | - P Duan
- 4 Ministry of Education, Key Lab of Environment and Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - H Zheng
- 2 HuBei University of Medicine, Hubei Shiyan, China
| |
Collapse
|
49
|
Jia X, Wang W, Xu Z, Wang S, Wang T, Wang M, Wu M. A humanized anti-DLL4 antibody promotes dysfunctional angiogenesis and inhibits breast tumor growth. Sci Rep 2016; 6:27985. [PMID: 27301650 PMCID: PMC4908374 DOI: 10.1038/srep27985] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 05/27/2016] [Indexed: 01/24/2023] Open
Abstract
Blockage of Delta-like 4 (DLL4)-directed Notch signaling induces excessive tip cell formation and endothelial proliferation resulting in dysfunctional angiogenesis in tumors. MMGZ01, as a murine anti-human DLL4 monoclonal antibody, specifically binds to human DLL4 and blocks Notch pathway. Here, the structure of MMGZ01 variable fragment (Fv) was established and framework region (FR) residues which supported complementarily determining region (CDR) loop conformation were identified. Important residues interactions were also identified through docking MMGZ01 Fv with antigen epitope in DLL4. To humanize the murine antibody, we modified MMGZ01 Fv through CDR grafting and the reconstructed antibody (H3L2) maintained similar structure and binding affinity to parental MMGZ01 after back mutation of 12 canonical murine residues in the FRs. Meanwhile, H3L2 promoted human umbilical vein endothelial cell (HUVEC) proliferation through inhibiting DLL4-directed Notch pathway. Moreover, in MDA-MB-231-bearing nude mice, H3L2 induced dysfunctional angiogenesis and tumor cell apoptosis and showed superior anti-tumor activity. In conclusion, H3L2 is an ideal humanized antibody that inhibits tumor growth through targeting DLL4-Notch pathway and has attracting potentials for clinical applications.
Collapse
Affiliation(s)
- Xuelian Jia
- State Key Laboratory of Natural Medicines, School of Life Science &Technology, China Pharmaceutical University, Nanjing, Jiangsu 210009, China
| | - Wenyi Wang
- State Key Laboratory of Natural Medicines, School of Life Science &Technology, China Pharmaceutical University, Nanjing, Jiangsu 210009, China.,The Rutgers Center for Computational and Integrative Biology, Rutgers University, Camden, NJ 08102, United States
| | - Zhuobin Xu
- State Key Laboratory of Natural Medicines, School of Life Science &Technology, China Pharmaceutical University, Nanjing, Jiangsu 210009, China
| | - Shijing Wang
- State Key Laboratory of Natural Medicines, School of Life Science &Technology, China Pharmaceutical University, Nanjing, Jiangsu 210009, China
| | - Tong Wang
- State Key Laboratory of Natural Medicines, School of Life Science &Technology, China Pharmaceutical University, Nanjing, Jiangsu 210009, China
| | - Min Wang
- State Key Laboratory of Natural Medicines, School of Life Science &Technology, China Pharmaceutical University, Nanjing, Jiangsu 210009, China
| | - Min Wu
- State Key Laboratory of Natural Medicines, School of Life Science &Technology, China Pharmaceutical University, Nanjing, Jiangsu 210009, China
| |
Collapse
|
50
|
Xu Z, Wang Z, Jia X, Wang L, Chen Z, Wang S, Wang M, Zhang J, Wu M. MMGZ01, an anti-DLL4 monoclonal antibody, promotes nonfunctional vessels and inhibits breast tumor growth. Cancer Lett 2015; 372:118-27. [PMID: 26739060 DOI: 10.1016/j.canlet.2015.12.025] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Revised: 12/15/2015] [Accepted: 12/16/2015] [Indexed: 12/21/2022]
Abstract
Increasing evidence suggests that DLL4 (Delta-like 4)-Notch signaling plays a critical role in cell fate determination and differentiation in tissues. Blocking DLL4-Notch signaling results in inhibition of tumor growth, which is associated with increased nonfunctional vessels and poor perfusion in the tumor. We successfully generated a human DLL4 monoclonal antibody MMGZ01 that binds specifically to DLL4 to disrupt the interaction between DLL4 and Notch1. MMGZ01 showed high affinity to DLL4 to inhibit the DLL4-mediated human umbilical vein endothelial cell (HUVEC) phenotype. Furthermore, MMGZ01 stimulated HUVEC vessel sprouting and tubule formation in vitro. In addition, MMGZ01 had a pronounced effect in promoting immature vessels and reduced breast cancer cell growth in vivo. Finally, MMGZ01 treatment inhibited the proliferation of breast cancer cells, induced tumor cell apoptosis, suppressed mammosphere formation, decreased CD44(+)/CD24(-) cell population, and reduced epithelial mesenchymal transition (EMT). These findings suggest that antagonism of the DLL4-Notch signaling pathway might provide a potential therapeutic approach for breast cancer treatment.
Collapse
Affiliation(s)
- Zhuobin Xu
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China
| | - Zegen Wang
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China
| | - Xuelian Jia
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China
| | - Luxuan Wang
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China
| | - Zhiguo Chen
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China
| | - Shijing Wang
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China
| | - Min Wang
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China.
| | - Juan Zhang
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China.
| | - Min Wu
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|