1
|
Qiao X, Guo S, Meng Z, Gan H, Wu Z, Sun Y, Liu S, Dou G, Gu R. Advances in the study of death receptor 5. Front Pharmacol 2025; 16:1549808. [PMID: 40144653 PMCID: PMC11936945 DOI: 10.3389/fphar.2025.1549808] [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: 12/22/2024] [Accepted: 02/24/2025] [Indexed: 03/28/2025] Open
Abstract
DR5, a receptor with the highest affinity for TRAIL under physiological conditions, selectively induces apoptosis in specific target cells such as tumor and aberrant immune cells, while minimally affecting normal cells. The TRAIL-DR5 signaling pathway is a crucial regulatory mechanism when the body responds to various exogenous interference factors, including viruses, chemicals, and radiation. This pathway plays a vital role in maintaining physiological homeostasis and in the pathological development of various diseases. Different modulations of DR5, such as upregulation, activation, and antagonism, hold significant potential for therapeutic applications in tumors, cardiovascular diseases, autoimmune diseases, viral infections, and radiation injuries. This article provides an overview of the current research progress on DR5, including the status and prospects of its clinical applications.
Collapse
Affiliation(s)
- Xuan Qiao
- Graduate Collaborative Training Base of Academy of Military Medical Sciences, Hengyang Medical School, University of South China, Hengyang, China
- Beijing Institute of Radiation Medicine, Beijing, China
| | - Shuang Guo
- Beijing Institute of Radiation Medicine, Beijing, China
| | - Zhiyun Meng
- Beijing Institute of Radiation Medicine, Beijing, China
| | - Hui Gan
- Beijing Institute of Radiation Medicine, Beijing, China
| | - Zhuona Wu
- Beijing Institute of Radiation Medicine, Beijing, China
| | - Yunbo Sun
- Beijing Institute of Radiation Medicine, Beijing, China
| | - Shuchen Liu
- Beijing Institute of Radiation Medicine, Beijing, China
| | - Guifang Dou
- Beijing Institute of Radiation Medicine, Beijing, China
| | - Ruolan Gu
- Beijing Institute of Radiation Medicine, Beijing, China
| |
Collapse
|
2
|
Huseynova N, Çetinkaya M, Baran Z, Khalilov R, Mammadova A, Baran Y. Flavonoids as Chemosensitizers in Leukemias. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1479:205-234. [PMID: 39503945 DOI: 10.1007/5584_2024_828] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2025]
Abstract
Flavonoids, a diverse group of natural compounds abundant in plants, fruits, and seeds, are not only responsible for the vibrant colors, fragrances, and flavors found in nature but also possess significant health benefits. Representing a secondary metabolite, these phytonutrients contribute to overall well-being. They have garnered considerable interest due to their diverse biological roles, encompassing antioxidant, anti-inflammatory, and anticancer properties. Flavonoids exert anticancer properties by interfering with different signaling pathways and molecules. Also, they have been demonstrated to exert chemosensitization features, where flavonoids may enhance the effectiveness of chemotherapy, and hold promise for improving cancer treatment outcomes as they have been discovered to make cancer cells more responsive to treatment. Understanding their influence on the regulation of cellular signaling provides a foundation for exploring their potential in combination with different chemotherapy agents and their possible single use for cancer treatment. Besides, they are believed to present a cost-effective approach to cancer therapeutics with possible implications for reducing the side effects of the current chemotherapy regimens, which can be a great therapeutic strategy for treating cancer types, including leukemia. This chapter explores potential approaches for creating anticancer treatments, focusing on leukemia, through integrating flavonoid nutraceuticals with traditional chemotherapy agents.
Collapse
Affiliation(s)
- Nigar Huseynova
- Department of Biophysics and Biochemistry, Baku State University, Baku, Azerbaijan
- Department of Natural Sciences, Western Caspian University, Baku, Azerbaijan
| | - Melisa Çetinkaya
- Department of Molecular Biology and Genetics, Laboratory of Cancer Genetics, İzmir Institute of Technology, İzmir, Turkey
| | - Züleyha Baran
- Department of Pharmacology, Laboratory of Molecular Pharmacology, Anadolu University, Eskişehir, Turkey
| | - Rovshan Khalilov
- Department of Biophysics and Biochemistry, Baku State University, Baku, Azerbaijan
| | - Afat Mammadova
- Department of Botany and Plant Physiology, Baku State University, Baku, Azerbaijan
| | - Yusuf Baran
- Department of Molecular Biology and Genetics, Laboratory of Cancer Genetics, İzmir Institute of Technology, İzmir, Turkey.
| |
Collapse
|
3
|
Wang Y, Qian X, Wang Y, Yu C, Feng L, Zheng X, Wang Y, Gong Q. Turn TRAIL Into Better Anticancer Therapeutic Through TRAIL Fusion Proteins. Cancer Med 2025; 14:e70517. [PMID: 39740038 DOI: 10.1002/cam4.70517] [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: 08/20/2024] [Revised: 11/17/2024] [Accepted: 12/07/2024] [Indexed: 01/02/2025] Open
Abstract
BACKGROUND TNF-related apoptosis-inducing ligand (TRAIL) belongs to the tumor necrosis factor superfamily. TRAIL selectively induces apoptosis in tumor cells while sparing normal cells, which makes it an attractive candidate for cancer therapy. Recombinant soluble TRAIL and agonistic antibodies against TRAIL receptors have demonstrated safety and tolerability in clinical trials. However, they have failed to exhibit expected clinical efficacy. Consequently, extensive research has focused on optimizing TRAIL-based therapies, with one of the most common approaches being the construction of TRAIL fusion proteins. METHODS An extensive literature search was conducted to identify studies published over the past three decades related to TRAIL fusion proteins. These various TRAIL fusion strategies were categorized based on their effects achieved. RESULTS The main fusion strategies for TRAIL include: 1. Construction of stable TRAIL trimers; 2. Enhancing the polymerization capacity of soluble TRAIL; 3. Increasing the accumulation of TRAIL at tumor sites by fusing with antibody fragments or peptides; 4. Decorating immune cells with TRAIL; 5. Prolonging the half-life of TRAIL in vivo; 6. Sensitizing cancer cells to overcome resistance to TRAIL treatment. CONCLUSION This work focuses on the progress in recombinant TRAIL fusion proteins and aims to provide more rational and effective fusion strategies to enhance the efficacy of recombinant soluble TRAIL, facilitating its translation from bench to bedside as an effective anti-cancer therapeutic.
Collapse
Affiliation(s)
- Yan Wang
- College of Agroforestry and Medicine, The Open University of China, Beijing, China
| | - Xin Qian
- Endocrinology Centre, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yubo Wang
- Department of Pharmacy, Beijing Ditan Hospital Capital Medical University, Beijing, China
| | - Caiyuan Yu
- College of Agroforestry and Medicine, The Open University of China, Beijing, China
- State Key Laboratory of Quality Research in Chinese Medicines, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau, China
| | - Li Feng
- College of Agroforestry and Medicine, The Open University of China, Beijing, China
| | - Xiaoyan Zheng
- College of Agroforestry and Medicine, The Open University of China, Beijing, China
| | - Yaya Wang
- College of Agroforestry and Medicine, The Open University of China, Beijing, China
| | - Qiuhong Gong
- Endocrinology Centre, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
4
|
Masum AA, Aoki S, Rahman MM, Hisamatsu Y. Chemical synthetic approaches to mimic the TRAIL: promising cancer therapeutics. RSC Med Chem 2024; 15:d4md00183d. [PMID: 39246747 PMCID: PMC11376135 DOI: 10.1039/d4md00183d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Accepted: 07/29/2024] [Indexed: 09/10/2024] Open
Abstract
Apoptosis is programmed cell death that eliminates undesired cells to maintain homeostasis in metazoan. Aberration of this process may lead to cancer genesis. The tumor necrosis factor related apoptosis inducing ligand (TRAIL) induces apoptosis in cancer cells after ligation with death receptors (DR4/DR5) while sparing most normal cells. Therefore, strategies to induce apoptosis in cancer cells by mimicking the TRAIL emerge as a promising therapeutic tool. Hence, approaches are taken to develop TRAIL/DR-based cancer therapeutics. The recombinant soluble TRAIL (rhTRAIL) and death receptor agonistic antibodies were produced and tested pre-clinically and clinically. Pre-clinical and clinical trial data demonstrate that these therapeutics are safe and relatively well tolerated. But some of these therapeutics failed to exert adequate efficacy in clinical settings. Besides these biotechnologically derived therapeutics, a few chemically synthesized therapeutics are reported. Some of these therapeutics exert considerable efficacy in vitro and in vivo. In this review, we will discuss chemically synthesized TRAIL/DR-based therapeutics, their chemical and biological behaviour, design concepts and strategies that may contribute to further improvement of TRAIL/DR-based therapeutics.
Collapse
Affiliation(s)
- Abdullah-Al Masum
- Department of Pharmaceutical Sciences, North South University Bashundhara R/A Dhaka-1229 Bangladesh
| | - Shin Aoki
- Faculty of Pharmaceutical Sciences, Tokyo University of Science 2641 Yamazaki, Noda-shi Chiba 278-8510 Japan
- Research Institute for Science and Technology, Tokyo University of Science 2641 Yamazaki, Noda-shi Chiba 278-8510 Japan
- Research Institute for Biomedical Sciences, Tokyo University of Science 2641 Yamazaki, Noda-shi Chiba 278-8510 Japan
| | - Md Mahbubur Rahman
- Department of Pharmaceutical Sciences, North South University Bashundhara R/A Dhaka-1229 Bangladesh
| | - Yosuke Hisamatsu
- Graduate School of Pharmaceutical Sciences, Nagoya City University Mizuho-Ku Nagoya 467-8603 Japan
| |
Collapse
|
5
|
Tapadar P, Pal A, Ghosal N, Kumar B, Paul T, Biswas N, Pal R. CDH1 overexpression sensitizes TRAIL resistant breast cancer cells towards rhTRAIL induced apoptosis. Mol Biol Rep 2023; 50:7283-7294. [PMID: 37422537 DOI: 10.1007/s11033-023-08657-1] [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: 04/20/2023] [Accepted: 06/29/2023] [Indexed: 07/10/2023]
Abstract
PURPOSE Tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) is well known for its unique ability to induce apoptosis in cancer cells but not normal cells. However, a subpopulation of cancer cells exist that does not respond to toxic doses of TRAIL. In this study, we aimed to identify key factors regulating TRAIL resistance in breast cancer. METHODS rhTRAIL (recombinant human TRAIL) resistant cells (TR) isolated from TRAIL sensitive MDA-MB-231 parental cells (TS) were confirmed using trypan blue assay, cell viability assay and AO/EtBr (acridine orange/ethidium bromide) staining. Microarray was performed followed by analysis using DAVID and Cytoscape bioinformatics software to identify the candidate hub gene. Gene expression of the candidate gene was confirmed using real-time PCR and western blot. Candidate gene was overexpressed via transient transfection to identify its significance in the context of rhTRAIL. Breast cancer patient data was obtained from The Cancer Genome Atlas (TCGA) database. RESULTS Whole transcriptome analysis identified 4907 differentially expressed genes (DEGs) between TS and TR cells. CDH1 was identified as the candidate hub gene, with 18-degree centrality. We further observed CDH1 protein to be downregulated, overexpression of which increased apoptosis in TR cells after rhTRAIL treatment. TCGA patient data analysis also showed CDH1 mRNA to be low in TRAIL resistant patient group compared to TRAIL sensitive group. CONCLUSION CDH1 overexpression sensitizes TR cells towards rhTRAIL induced apoptosis. Therefore, we can hypothesize that CDH1 expression should be taken into account while performing TRAIL therapy in breast cancer.
Collapse
Affiliation(s)
- Poulami Tapadar
- Department of Life Sciences, Presidency University, 86/1 College Street, Kolkata, West Bengal, 700073, India
| | - Ambika Pal
- Department of Life Sciences, Presidency University, 86/1 College Street, Kolkata, West Bengal, 700073, India
| | - Nirajan Ghosal
- Department of Life Sciences, Presidency University, 86/1 College Street, Kolkata, West Bengal, 700073, India
| | - Bhupender Kumar
- Department of Biochemistry, Institute of Home Economics, University of Delhi, New Delhi, 110016, India
| | - Tamalika Paul
- Department of Life Sciences, Presidency University, 86/1 College Street, Kolkata, West Bengal, 700073, India
| | - Nabendu Biswas
- Department of Life Sciences, Presidency University, 86/1 College Street, Kolkata, West Bengal, 700073, India
| | - Ranjana Pal
- Department of Life Sciences, Presidency University, 86/1 College Street, Kolkata, West Bengal, 700073, India.
| |
Collapse
|
6
|
Montinaro A, Areso Zubiaur I, Saggau J, Kretz AL, Ferreira RMM, Hassan O, Kitzig E, Müller I, El-Bahrawy MA, von Karstedt S, Kulms D, Liccardi G, Lemke J, Walczak H. Potent pro-apoptotic combination therapy is highly effective in a broad range of cancers. Cell Death Differ 2022; 29:492-503. [PMID: 34535764 PMCID: PMC8901660 DOI: 10.1038/s41418-021-00869-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 08/31/2021] [Accepted: 09/01/2021] [Indexed: 12/14/2022] Open
Abstract
Primary or acquired therapy resistance is a major obstacle to the effective treatment of cancer. Resistance to apoptosis has long been thought to contribute to therapy resistance. We show here that recombinant TRAIL and CDK9 inhibition cooperate in killing cells derived from a broad range of cancers, importantly without inducing detectable adverse events. Remarkably, the combination of TRAIL with CDK9 inhibition was also highly effective on cancers resistant to both, standard-of-care chemotherapy and various targeted therapeutic approaches. Dynamic BH3 profiling revealed that, mechanistically, combining TRAIL with CDK9 inhibition induced a drastic increase in the mitochondrial priming of cancer cells. Intriguingly, this increase occurred irrespective of whether the cancer cells were sensitive or resistant to chemo- or targeted therapy. We conclude that this pro-apoptotic combination therapy has the potential to serve as a highly effective new treatment option for a variety of different cancers. Notably, this includes cancers that are resistant to currently available treatment modalities.
Collapse
Affiliation(s)
- Antonella Montinaro
- Centre for Cell Death, Cancer, and Inflammation (CCCI), UCL Cancer Institute, University College London, 72 Huntley Street, London, WC1E 6DD, UK
| | - Itziar Areso Zubiaur
- Centre for Cell Death, Cancer, and Inflammation (CCCI), UCL Cancer Institute, University College London, 72 Huntley Street, London, WC1E 6DD, UK
| | - Julia Saggau
- CECAD Cluster of Excellence, University of Cologne, 50931, Cologne, Germany
- Center for Biochemistry, Medical Faculty, Joseph-Stelzmann-Str. 52, University of Cologne, 50931, Cologne, Germany
| | - Anna-Laura Kretz
- Department of General and Visceral Surgery, Ulm University Hospital, Albert-Einstein-Allee 23, 89081, Ulm, Germany
| | - Rute M M Ferreira
- Centre for Cell Death, Cancer, and Inflammation (CCCI), UCL Cancer Institute, University College London, 72 Huntley Street, London, WC1E 6DD, UK
| | - Omar Hassan
- CECAD Cluster of Excellence, University of Cologne, 50931, Cologne, Germany
- Center for Biochemistry, Medical Faculty, Joseph-Stelzmann-Str. 52, University of Cologne, 50931, Cologne, Germany
| | - Ella Kitzig
- Department of General and Visceral Surgery, Ulm University Hospital, Albert-Einstein-Allee 23, 89081, Ulm, Germany
| | - Ines Müller
- Experimental Dermatology, Department of Dermatology, Technical University Dresden, Dresden, Germany
| | - Mona A El-Bahrawy
- Department of Histopathology, Imperial College London, London, W12 0NN, UK
| | - Silvia von Karstedt
- Centre for Cell Death, Cancer, and Inflammation (CCCI), UCL Cancer Institute, University College London, 72 Huntley Street, London, WC1E 6DD, UK
- CECAD Cluster of Excellence, University of Cologne, 50931, Cologne, Germany
- Department of Translational Genomics, Center of Integrated Oncology Cologne-Bonn, Medical Faculty, University of Cologne, 50931, Cologne, Germany
- Center for Molecular Medicine Cologne, Medical Faculty, University Hospital of Cologne, 50931, Cologne, Germany
| | - Dagmar Kulms
- Experimental Dermatology, Department of Dermatology, Technical University Dresden, Dresden, Germany
| | - Gianmaria Liccardi
- Center for Biochemistry, Medical Faculty, Joseph-Stelzmann-Str. 52, University of Cologne, 50931, Cologne, Germany
| | - Johannes Lemke
- Department of General and Visceral Surgery, Ulm University Hospital, Albert-Einstein-Allee 23, 89081, Ulm, Germany
| | - Henning Walczak
- Centre for Cell Death, Cancer, and Inflammation (CCCI), UCL Cancer Institute, University College London, 72 Huntley Street, London, WC1E 6DD, UK.
- CECAD Cluster of Excellence, University of Cologne, 50931, Cologne, Germany.
- Center for Biochemistry, Medical Faculty, Joseph-Stelzmann-Str. 52, University of Cologne, 50931, Cologne, Germany.
| |
Collapse
|
7
|
Soto-Gamez A, Wang Y, Zhou X, Seras L, Quax W, Demaria M. Enhanced extrinsic apoptosis of therapy-induced senescent cancer cells using a death receptor 5 (DR5) selective agonist. Cancer Lett 2022; 525:67-75. [PMID: 34728311 DOI: 10.1016/j.canlet.2021.10.038] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 10/25/2021] [Accepted: 10/26/2021] [Indexed: 01/05/2023]
Abstract
Genotoxic agents are widely used anti-cancer therapies because of their ability to interfere with highly proliferative cells. An important outcome of these interventions is the induction of a state of permanent arrest also known as cellular senescence. However, senescent cancer cells are characterized by genomic instability and are at risk of escaping the growth arrest to eventually facilitate cancer relapse. The tumor necrosis factor related apoptosis inducing ligand (TRAIL) signals extrinsic apoptosis via Death Receptors (DR) 4 and 5, while Decoy Receptors (DcR) 1 and 2, and Osteoprotegerin (OPG) are homologous to death receptors but incapable of transducing an apoptotic signal. The use of recombinant TRAIL as an anti-cancer strategy in combination with chemotherapy is currently in development, and a major question remains whether senescent cancer cells respond to TRAIL. Here, we show variable sensitivity of cancer cells to TRAIL after senescence induction, and upregulation of both pro-apoptotic and anti-apoptotic receptors in therapy-induced senescent cancer cells. A DR5-selective TRAIL variant (DHER), unable to bind to DcR1 or OPG, was more effective in inducing apoptosis of senescent cancer cells compared to wild-type TRAIL. Importantly, no apoptosis induction was observed in non-cancerous cells, even at the highest concentrations tested. Our results suggest that targeting DR5 can serve as a novel therapeutic strategy for the elimination of therapy-induced senescent cancer cells.
Collapse
Affiliation(s)
- Abel Soto-Gamez
- European Institute for the Biology of Aging (ERIBA), University Medical Center Groningen (UMCG), Groningen, Netherlands; University of Groningen, Groningen Research Institute of Pharmacy, Chemical and Pharmaceutical Biology, Groningen, Netherlands
| | - Yizhou Wang
- University of Groningen, Groningen Research Institute of Pharmacy, Chemical and Pharmaceutical Biology, Groningen, Netherlands
| | - Xinyu Zhou
- University of Groningen, Groningen Research Institute of Pharmacy, Chemical and Pharmaceutical Biology, Groningen, Netherlands
| | - Lorina Seras
- University of Groningen, Groningen Research Institute of Pharmacy, Chemical and Pharmaceutical Biology, Groningen, Netherlands
| | - Wim Quax
- University of Groningen, Groningen Research Institute of Pharmacy, Chemical and Pharmaceutical Biology, Groningen, Netherlands.
| | - Marco Demaria
- European Institute for the Biology of Aging (ERIBA), University Medical Center Groningen (UMCG), Groningen, Netherlands.
| |
Collapse
|
8
|
Davoodi Z, Shafiee F. Internalizing RGD, a great motif for targeted peptide and protein delivery: a review article. Drug Deliv Transl Res 2022; 12:2261-2274. [PMID: 35015253 DOI: 10.1007/s13346-022-01116-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/04/2022] [Indexed: 01/10/2023]
Abstract
Understanding that cancer is one of the most important health problems, especially in advanced societies, is not difficult. The term of targeted cancer therapy has also been well known as an ideal treatment strategy in the recent years. Peptides with ability to specifically recognize the cancer cells with suitable penetration properties have been used as the targeting motif in this regard. In the present review article, we focus on an individual RGD-derived peptide with ability to recognize the integrin receptor on the cancer cell surface like its ancestor with an additional outstanding feature to penetrate to extravascular space of tumor and ability to penetrate to cancer cells unlike the original peptide. This peptide which has been named "internalizing RGD" or "iRGD" has been the focus of researches as a new targeting motif since it was discovered. To date, many types of molecules have been associated with this peptide for their targeted delivery to cancer cells. In this review article, we have discussed a summary of penetration mechanisms of iRGD and all introduced peptides and proteins attached to this attractive cell-penetrating peptide and have expressed the results of the studies.
Collapse
Affiliation(s)
- Zeinabosadat Davoodi
- Department of Pharmaceutical Biotechnology, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Hezar Jarib Ave., Isfahan, Iran
| | - Fatemeh Shafiee
- Department of Pharmaceutical Biotechnology, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Hezar Jarib Ave., Isfahan, Iran.
| |
Collapse
|
9
|
Nalawade SA, Shafer P, Bajgain P, McKenna MK, Ali A, Kelly L, Joubert J, Gottschalk S, Watanabe N, Leen A, Parihar R, Vera Valdes JF, Hoyos V. Selectively targeting myeloid-derived suppressor cells through TRAIL receptor 2 to enhance the efficacy of CAR T cell therapy for treatment of breast cancer. J Immunother Cancer 2021; 9:jitc-2021-003237. [PMID: 34815355 PMCID: PMC8611441 DOI: 10.1136/jitc-2021-003237] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/14/2021] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Successful targeting of solid tumors such as breast cancer (BC) using chimeric antigen receptor (CAR) T cells has proven challenging, largely attributed to the immunosuppressive tumor microenvironment (TME). Myeloid-derived suppressor cells (MDSCs) inhibit CAR T cell function and persistence within the breast TME. To overcome this challenge, we have developed CAR T cells targeting tumor-associated mucin 1 (MUC1) with a novel chimeric costimulatory receptor that targets tumor necrosis factor-related apoptosis-inducing ligand receptor 2 (TR2) expressed on MDSCs. METHODS The function of the TR2.41BB costimulatory receptor was assessed by exposing non-transduced (NT) and TR2.41BB transduced T cells to recombinant TR2, after which nuclear translocation of NFκB was measured by ELISA and western blot. The cytolytic activity of CAR.MUC1/TR2.41BB T cells was measured in a 5-hour cytotoxicity assay using MUC1+ tumor cells as targets in the presence or absence of MDSCs. In vivo antitumor activity was assessed using MDSC-enriched tumor-bearing mice treated with CAR T cells with or without TR2.41BB. RESULTS Nuclear translocation of NFκB in response to recombinant TR2 was detected only in TR2.41BB T cells. The presence of MDSCs diminished the cytotoxic potential of CAR.MUC1 T cells against MUC1+ BC cell lines by 25%. However, TR2.41BB expression on CAR.MUC1 T cells induced MDSC apoptosis, thereby restoring the cytotoxic activity of CAR.MUC1 T cells against MUC1+ BC lines. The presence of MDSCs resulted in an approximately twofold increase in tumor growth due to enhanced angiogenesis and fibroblast accumulation compared with mice with tumor alone. Treatment of these MDSC-enriched tumors with CAR.MUC1.TR2.41BB T cells led to superior tumor cell killing and significant reduction in tumor growth (24.54±8.55 mm3) compared with CAR.MUC1 (469.79±81.46 mm3) or TR2.41BB (434.86±64.25 mm3) T cells alone. CAR.MUC1.TR2.41BB T cells also demonstrated improved T cell proliferation and persistence at the tumor site, thereby preventing metastases. We observed similar results using CAR.HER2.TR2.41BB T cells in a HER2+ BC model. CONCLUSIONS Our findings demonstrate that CAR T cells that coexpress the TR2.4-1BB receptor exhibit superior antitumor potential against breast tumors containing immunosuppressive and tumor promoting MDSCs, resulting in TME remodeling and improved T cell proliferation at the tumor site.
Collapse
Affiliation(s)
- Saisha A Nalawade
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas, USA
| | - Paul Shafer
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas, USA
| | - Pradip Bajgain
- Mouse Cancer Genetics Program, National Cancer Institute at Frederick, Frederick, Maryland, USA
| | - Mary K McKenna
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas, USA
| | - Arushana Ali
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas, USA
| | - Lauren Kelly
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas, USA
| | - Jarrett Joubert
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas, USA
| | - Stephen Gottschalk
- Bone Marrow Transplant Department, St Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Norihiro Watanabe
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas, USA
| | - Ann Leen
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas, USA
| | - Robin Parihar
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas, USA
| | | | - Valentina Hoyos
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
10
|
Chemo-Sensitization of CD133+ Cancer Stem Cell Enhances the Effect of Mesenchymal Stem Cell Expressing TRAIL in Non-Small Cell Lung Cancer Cell Lines. BIOLOGY 2021; 10:biology10111103. [PMID: 34827096 PMCID: PMC8614666 DOI: 10.3390/biology10111103] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 10/03/2021] [Accepted: 10/07/2021] [Indexed: 11/17/2022]
Abstract
Simple Summary The anti-tumor properties of mesenchymal stem cell (MSCs) expressing TNF-related apoptosis inducing ligand (TRAIL) or MSC-TRAIL have been well documented by several reports. However, some tumors are resistant to TRAIL due to the existence of cancer stem cells (CSCs). Chemo-sensitization of tumors and their CSCs has been reported to enhance TRAIL-mediated inhibition. In this study, we examined the effect of pre-treatment using first-line chemotherapies on MSC-TRAIL-induced inhibition in non-small cell lung cancers (NSCLCs)–derived CSCs. We found that these chemotherapies were able to induce a chemo-sensitization effect to the CSC, thus improving the MSC-TRAIL-induced inhibition. We also noticed that the effect of chemo-sensitization was cell type specific and selecting chemotherapies for the right NSCLC subtypes might help in inducing a more meaningful combinatory effect. As such, this study has proven that chemo-sensitization of the CSCs was able to enhance the MSC-TRAIL-induced inhibition in NSCLC cell lines. Abstract Pre-clinical studies have demonstrated the efficacy of mesenchymal stem cells (MSCs) expressing tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) or MSC-TRAIL against several tumors. However, due to the existence of cancer stem cells (CSCs), some tumors, including non-small cell lung cancer (NSCLC), exhibit TRAIL resistance. This study was designed to evaluate the capacity of using first-line chemotherapies including cisplatin, 5-fluorouracil (5-FU) and vinorelbine to act as a chemo-sensitizer on CD133+ (prominin-1 positive) CSCs derived from NSCLC cell lines (A549, H460 and H2170) for the purpose of MSC-TRAIL-induced inhibition. We showed that MSC-TRAIL was resistant to all three chemotherapies compared to the NSCLC cell lines, suggesting that the chemotherapies had little effect on MSC-TRAIL viability. Pre-treatment using either cisplatin or 5-FU, but not with vinorelbine, was able to increase the efficacy of MSC-TRAIL to kill the TRAIL-resistant A549-derived CSCs. The study also demonstrated that both 5-FU and vinorelbine were an effective chemo-sensitizer, used to increase the anti-tumor effect of MSC-TRAIL against H460- and H2170-derived CSCs. Furthermore, pre-treatment using cisplatin was noted to enhance the effect of MSC-TRAIL in H460-derived CSCs; however, this effect was not detected in the H2170-derived CSCs. These findings suggest that a pre-treatment using certain chemotherapies in NSCLC could enhance the anti-tumor effect of MSC-TRAIL to target the CSCs, and therefore the combination of chemotherapies and MSC-TRAIL may serve as a novel approach for the treatment of NSCLC.
Collapse
|
11
|
Birtekocak F, Demirbolat GM, Cevik O. TRAIL Conjugated Silver Nanoparticle Synthesis, Characterization and Therapeutic Effects on HT-29 Colon Cancer Cells. IRANIAN JOURNAL OF PHARMACEUTICAL RESEARCH : IJPR 2021; 20:45-56. [PMID: 34567145 PMCID: PMC8457744 DOI: 10.22037/ijpr.2020.112069.13514] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Colon cancer is one of the most prominent causes of cancer-related morbidity and mortality and curable if detected in the early stages. TNF-related apoptosis-inducing ligand (TRAIL) is a therapeutic protein and has a potential anti-cancer activity that is widely used for the treatment of several cancers. In this study, we aimed to develop a silver nanoparticle system conjugated with TRAIL and coated with PEG (AgCTP NPs) to improve the therapeutic effects of colon cancer. AgCTP NPs were characterized by UV spectrum, FTIR and zetasizer. Cytotoxicity, hemolysis assay and apoptotic effects of nanoparticles were investigated using a colon cancer cell line (HT-29) in-vitro. Treatment with AgCTP NPs effectively inhibited proliferation and colony formation of HT-29 cells. The apoptotic effects of nanoparticles on HT-29 cells were determined as Bax, Bcl-2, PARP and clv-PARP protein expression levels using Western blot. Apoptotic proteins were upregulated by AgCTP NPs. In this study, we demonstrated that AgCTP NPs had an anti-cancer effect by activating cell death. Thus, we have confirmed that silver nanoparticles can be selected as a good carrier for TRAIL therapeutic proteins that can be used to treat colon cancer.
Collapse
Affiliation(s)
- Fatih Birtekocak
- Department of Biochemistry, School of Medicine, Aydin Adnan Menderes University, Aydin, Turkey
| | - Gulen Melike Demirbolat
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Biruni University, Istanbul, Turkey
| | - Ozge Cevik
- Department of Biochemistry, School of Medicine, Aydin Adnan Menderes University, Aydin, Turkey
| |
Collapse
|
12
|
Vaughan HJ, Zamboni CG, Radant NP, Bhardwaj P, Revai Lechtich E, Hassan LF, Shah K, Green JJ. Poly(beta-amino ester) nanoparticles enable tumor-specific TRAIL secretion and a bystander effect to treat liver cancer. Mol Ther Oncolytics 2021; 21:377-388. [PMID: 34189258 PMCID: PMC8208964 DOI: 10.1016/j.omto.2021.04.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Accepted: 04/12/2021] [Indexed: 01/23/2023] Open
Abstract
Despite initial promise, tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-based approaches to cancer treatment have yet to yield a clinically approved therapy, due to delivery challenges, a lack of potency, and drug resistance. To address these challenges, we have developed poly(beta-amino ester) (PBAE) nanoparticles (NPs), as well as an engineered cDNA sequence encoding a secretable TRAIL (sTRAIL) protein, to enable reprogramming of liver cancer cells to locally secrete TRAIL protein. We show that sTRAIL initiates apoptosis in transfected cells and has a bystander effect to non-transfected cells. To address TRAIL resistance, NP treatment is combined with histone deacetylase inhibitors, resulting in >80% TRAIL-mediated cell death in target cancer cells and significantly slowed xenograft tumor growth. This anti-cancer effect is specific to liver cancer cells, with up to 40-fold higher cell death in HepG2 cancer cells over human hepatocytes. By combining cancer-specific TRAIL NPs with small-molecule-sensitizing drugs, this strategy addresses multiple challenges associated with TRAIL therapy and offers a new potential approach for cancer treatment.
Collapse
Affiliation(s)
- Hannah J. Vaughan
- Department of Biomedical Engineering, Institute for NanoBioTechnology, and the Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Camila G. Zamboni
- Department of Biomedical Engineering, Institute for NanoBioTechnology, and the Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Nicholas P. Radant
- Department of Biomedical Engineering, Institute for NanoBioTechnology, and the Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Pranshu Bhardwaj
- Department of Biomedical Engineering, Institute for NanoBioTechnology, and the Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Esther Revai Lechtich
- Center for Stem Cell Therapeutics and Imaging, Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Laboni F. Hassan
- Department of Biomedical Engineering, Institute for NanoBioTechnology, and the Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Khalid Shah
- Center for Stem Cell Therapeutics and Imaging, Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Jordan J. Green
- Department of Biomedical Engineering, Institute for NanoBioTechnology, and the Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
- Departments of Ophthalmology, Oncology, Neurosurgery, Materials Science & Engineering, and Chemical & Biomolecular Engineering, and the Bloomberg∼Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| |
Collapse
|
13
|
NCTR 25 fusion facilitates the formation of TRAIL polymers that selectively activate TRAIL receptors with higher potency and efficacy than TRAIL. Cancer Chemother Pharmacol 2021; 88:289-306. [PMID: 33942150 DOI: 10.1007/s00280-021-04283-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 04/17/2021] [Indexed: 10/21/2022]
Abstract
PURPOSE Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) binds to death receptor (DR) 4 and DR5 and induces tumor-selective apoptosis. The fusion proteins NCTR25-TRAIL and NCTR25-TGF3L-TRAIL self-assembled into polymers and triggered super-active cancer cell killing. The role of TGF3L in self-assembly and super-activity was unclear. These multivalent TRAILs elicited apoptosis with great potency, but their specificity towards receptors and subsequent efficacy in signal activation were unclear. METHODS NCTR25-TRAIL fusion was constructed and prokaryotically expressed. The size of fusion protein polymers was estimated. Their cytotoxicity was assessed in eight cancer cell lines and two noncancerous cell lines. Receptor binding and activation specificity were determined by antibody blockade. Apoptosis was evaluated, and the associated pathway was verified by quantifying caspase activity. The NF-κB signaling pathway was assessed by dual-luciferase assay. The in vivo antitumor activity was also evaluated in nude mice. RESULTS NCTR25 fusion to TRAIL promoted its self-assembly into polymers and showed similar super-cytotoxicity to NCTR25-TGF3L-TRAIL in vitro. The multivalent TRAILs exclusively activated both DR4 and DR5 and showed a bias towards DR4 in mediating cytotoxicity in NCI-H460 cells. They activated caspase pathway and induced apoptosis with higher potency but in similar efficacy than TRAIL. A higher potency and a greater efficacy were observed in activating NF-κB pathway by NCTR25-TRAIL comparing to TRAIL. Both the polymers showed better in vivo antitumor activity than TRAIL. CONCLUSIONS NCTR25 fusion alone facilitates the formation of TRAIL polymers. Multivalent TRAIL polymers bind and activate DR4 and DR5 specifically and exclusively, triggering the signaling pathways with higher potency, and greater efficacy than TRAIL.
Collapse
|
14
|
Cytotoxic Efficacy and Resistance Mechanism of a TRAIL and VEGFA-Peptide Fusion Protein in Colorectal Cancer Models. Int J Mol Sci 2021; 22:ijms22063160. [PMID: 33808900 PMCID: PMC8003782 DOI: 10.3390/ijms22063160] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 03/16/2021] [Accepted: 03/17/2021] [Indexed: 11/16/2022] Open
Abstract
TNF-related apoptosis-inducing ligand (TRAIL) is a type II transmembrane protein capable of selectively inducing apoptosis in cancer cells by binding to its cognate receptors. Here, we examined the anticancer efficacy of a recently developed chimeric AD-O51.4 protein, a TRAIL fused to the VEGFA-originating peptide. We tested AD-O51.4 protein activity against human colorectal cancer (CRC) models and investigated the resistance mechanism in the non-responsive CRC models. The quantitative comparison of apoptotic activity between AD-O51.4 and the native TRAIL in nine human colorectal cancer cell lines revealed dose-dependent toxicity in seven of them; the immunofluorescence-captured receptor abundance correlated with the extent of apoptosis. AD-O51.4 reduced the growth of CRC patient-derived xenografts (PDXs) with good efficacy. Cell lines that acquired AD-O51.4 resistance showed a significant decrease in surface TRAIL receptor expression and apoptosis-related proteins, including Caspase-8, HSP60, and p53. These results demonstrate the effectiveness of AD-O51.4 protein in CRC preclinical models and identify the potential mechanism underlying acquired resistance. Progression of AD-O51.4 to clinical trials is expected.
Collapse
|
15
|
Singh D, Tewari M, Singh S, Narayan G. Revisiting the role of TRAIL/TRAIL-R in cancer biology and therapy. Future Oncol 2021; 17:581-596. [PMID: 33401962 DOI: 10.2217/fon-2020-0727] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
TNF-related apoptosis-inducing ligand (TRAIL), a member of the TNF superfamily, can induce apoptosis in cancer cells, sparing normal cells when bound to its associated death receptors (DR4/DR5). This unique mechanism makes TRAIL a potential anticancer therapeutic agent. However, clinical trials of recombinant TRAIL protein and TRAIL receptor agonist monoclonal antibodies have shown disappointing results due to its short half-life, poor pharmacokinetics and the resistance of the cancer cells. This review summarizes TRAIL-induced apoptotic and survival pathways as well as mechanisms leading to apoptotic resistance. Recent development of methods to overcome cancer cell resistance to TRAIL-induced apoptosis, such as protein modification, combination therapy and TRAIL-based gene therapy, appear promising. We also discuss the challenges and opportunities in the development of TRAIL-based therapies for the treatment of human cancers.
Collapse
Affiliation(s)
- Deepika Singh
- Department of Molecular & Human Genetics, Cancer Genetics Laboratory, Institute of Science, Banaras Hindu University, Varanasi, 221005, India
| | - Mallika Tewari
- Department of Surgical Oncology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221005, India
| | - Sunita Singh
- Department of Zoology, Mahila Mahavidyalaya, Banaras Hindu University, Varanasi, 221005, India
| | - Gopeshwar Narayan
- Department of Molecular & Human Genetics, Cancer Genetics Laboratory, Institute of Science, Banaras Hindu University, Varanasi, 221005, India
| |
Collapse
|
16
|
Hydroxychavicol sensitizes imatinib-resistant chronic myelogenous leukemia cells to TRAIL-induced apoptosis by ROS-mediated IAP downregulation. Anticancer Drugs 2020; 30:167-178. [PMID: 30418193 DOI: 10.1097/cad.0000000000000710] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
The tumor necrosis factor-related apoptosis-inducing ligand (TRAIL), a member of cytokine superfamily, induces apoptosis in a number of tumor cells through the activation of extrinsic apoptotic pathway but shows little or no cytotoxicity toward normal cells. However some tumor cells are inherently resistant to TRAIL-mediated apoptosis, which needs to be addressed to establish TRAIL as a potential chemotherapeutic drug. In this study, our aim was to manipulate TRAIL-apoptosis pathway by hydroxychavicol (HCH), a polyphenol from Piper betel leaf, for the induction of apoptosis in TRAIL resistant chronic myeloid leukemia cell. When imatinib-resistant K562 cells were treated with HCH, it made these K562 cells sensitive to TRAIL. It was observed that HCH downregulated antiapoptotic proteins XIAP and FLIP, whereas the expression of TRAIL receptors, DR4 and DR5, remains unchanged. Moreover, we observed that reactive oxygen species or ROS played a crucial role in the downregulation of FLIP and XIAP because ROS scavenger significantly reversed the decrease of XIAP, and FLIP. Ubiquitin-proteasome pathway was observed to play a crucial role in the downregulation of XIAP and FLIP, as proteasomal inhibitor MG132 significantly reversed the downregulation of XIAP and FLIP. In conclusion, this study demonstrates the combinatorial treatment of TRAIL and HCH as promising alternative therapeutic approach to treat the imatinib-resistant leukemia, which are also resistant to TRAIL.
Collapse
|
17
|
Liu M, Hu Y, Chen G. The Antitumor Effect of Gene-Engineered Exosomes in the Treatment of Brain Metastasis of Breast Cancer. Front Oncol 2020; 10:1453. [PMID: 32850457 PMCID: PMC7406780 DOI: 10.3389/fonc.2020.01453] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 07/08/2020] [Indexed: 12/16/2022] Open
Abstract
Strategies for treating brain metastases of breast cancer have demonstrated limited efficacy due to the blood–brain barrier (BBB). Gene therapy could improve the efficacy of chemotherapeutic drugs. Exosomes derived from the mesenchymal stem cells (MSCs) are small membrane-based gene vectors that can pass through the BBB. CXCR4 is the most commonly found chemokine receptor in human cancer cells. Furthermore, the SDF-1/CXCR4 axis plays an important role in the homing of MSCs for tumor cell diffusion and metastasis. TRAIL can selectively induce apoptosis in transformed cells without significant toxic side effects in normal tissues. In this study, exosomes were isolated from MSCCXCR4+TRAIL transduced with CXCR4 and TRAIL using a lentiviral vector. Synergistic antitumor study showed that exosomeCXCR4+TRAIL exerted significant activity as a cooperative agent with carboplatin in an MDA-MB-231Br SCID mouse model, potentially engendering a novel strategy for advancing the treatment of brain metastases of breast cancer. Based on this study, further investigation of the effect of the vector on BBB and inducing apoptosis of brain tumors is warranted. In addition, the safety of the vector in animals during the treatment needs to be evaluated.
Collapse
Affiliation(s)
- Minchen Liu
- Engineering Research Center of Modern Preparation Technology of TCM, Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yulan Hu
- Department of Cultural Heritage and Museology, Zhejiang University, Hangzhou, China
| | - Guiqian Chen
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| |
Collapse
|
18
|
Thapa B, Kc R, Uludağ H. TRAIL therapy and prospective developments for cancer treatment. J Control Release 2020; 326:335-349. [PMID: 32682900 DOI: 10.1016/j.jconrel.2020.07.013] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 06/01/2020] [Accepted: 07/11/2020] [Indexed: 12/22/2022]
Abstract
Tumor Necrosis Factor (TNF) Related Apoptosis-Inducing Ligand (TRAIL), an immune cytokine of TNF-family, has received much attention in late 1990s as a potential cancer therapeutics due to its selective ability to induce apoptosis in cancer cells. TRAIL binds to cell surface death receptors, TRAIL-R1 (DR4) and TRAIL-R2 (DR5) and facilitates formation of death-inducing signaling complex (DISC), eventually activating the p53-independent apoptotic cascade. This unique mechanism makes the TRAIL a potential anticancer therapeutic especially for p53-mutated tumors. However, recombinant human TRAIL protein (rhTRAIL) and TRAIL-R agonist monoclonal antibodies (mAb) failed to exert robust anticancer activities due to inherent and/or acquired resistance, poor pharmacokinetics and weak potencies for apoptosis induction. To get TRAIL back on track as a cancer therapeutic, multiple strategies including protein modification, combinatorial approach and TRAIL gene therapy are being extensively explored. These strategies aim to enhance the half-life and bioavailability of TRAIL and synergize with TRAIL action ultimately sensitizing the resistant and non-responsive cells. We summarize emerging strategies for enhanced TRAIL therapy in this review and cover a wide range of recent technologies that will provide impetus to rejuvenate the TRAIL therapeutics in the clinical realm.
Collapse
Affiliation(s)
- Bindu Thapa
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada.
| | - Remant Kc
- Department of Chemical & Material Engineering, Faculty of Engineering, University of Alberta, Edmonton, AB, Canada.
| | - Hasan Uludağ
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada; Department of Chemical & Material Engineering, Faculty of Engineering, University of Alberta, Edmonton, AB, Canada; Department of Biomedical Engineering, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
19
|
Sordo-Bahamonde C, Lorenzo-Herrero S, Payer ÁR, Gonzalez S, López-Soto A. Mechanisms of Apoptosis Resistance to NK Cell-Mediated Cytotoxicity in Cancer. Int J Mol Sci 2020; 21:ijms21103726. [PMID: 32466293 PMCID: PMC7279491 DOI: 10.3390/ijms21103726] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 05/20/2020] [Accepted: 05/22/2020] [Indexed: 12/14/2022] Open
Abstract
Natural killer (NK) cells are major contributors to immunosurveillance and control of tumor development by inducing apoptosis of malignant cells. Among the main mechanisms involved in NK cell-mediated cytotoxicity, the death receptor pathway and the release of granules containing perforin/granzymes stand out due to their efficacy in eliminating tumor cells. However, accumulated evidence suggest a profound immune suppression in the context of tumor progression affecting effector cells, such as NK cells, leading to decreased cytotoxicity. This diminished capability, together with the development of resistance to apoptosis by cancer cells, favor the loss of immunogenicity and promote immunosuppression, thus partially inducing NK cell-mediated killing resistance. Altered expression patterns of pro- and anti-apoptotic proteins along with genetic background comprise the main mechanisms of resistance to NK cell-related apoptosis. Herein, we summarize the main effector cytotoxic mechanisms against tumor cells, as well as the major resistance strategies acquired by tumor cells that hamper the extrinsic and intrinsic apoptotic pathways related to NK cell-mediated killing.
Collapse
Affiliation(s)
- Christian Sordo-Bahamonde
- Department of Functional Biology, Immunology, University of Oviedo, 33006 Oviedo, Spain; (S.L.-H.); (S.G.)
- Instituto Universitario de Oncología del Principado de Asturias, IUOPA, 33006 Oviedo, Spain;
- Instituto de Investigación Biosanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
- Correspondence: (C.S.-B.); (A.L.-S.)
| | - Seila Lorenzo-Herrero
- Department of Functional Biology, Immunology, University of Oviedo, 33006 Oviedo, Spain; (S.L.-H.); (S.G.)
- Instituto Universitario de Oncología del Principado de Asturias, IUOPA, 33006 Oviedo, Spain;
- Instituto de Investigación Biosanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
| | - Ángel R. Payer
- Instituto Universitario de Oncología del Principado de Asturias, IUOPA, 33006 Oviedo, Spain;
- Instituto de Investigación Biosanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
- Department of Hematology, Hospital Universitario Central de Asturias (HUCA), 33011 Oviedo, Spain
| | - Segundo Gonzalez
- Department of Functional Biology, Immunology, University of Oviedo, 33006 Oviedo, Spain; (S.L.-H.); (S.G.)
- Instituto Universitario de Oncología del Principado de Asturias, IUOPA, 33006 Oviedo, Spain;
- Instituto de Investigación Biosanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
| | - Alejandro López-Soto
- Instituto Universitario de Oncología del Principado de Asturias, IUOPA, 33006 Oviedo, Spain;
- Instituto de Investigación Biosanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
- Department of Biochemistry and Molecular Biology, University of Oviedo, 33006 Oviedo, Spain
- Correspondence: (C.S.-B.); (A.L.-S.)
| |
Collapse
|
20
|
Stöhr D, Jeltsch A, Rehm M. TRAIL receptor signaling: From the basics of canonical signal transduction toward its entanglement with ER stress and the unfolded protein response. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2020; 351:57-99. [PMID: 32247582 DOI: 10.1016/bs.ircmb.2020.02.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The cytokine tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) is a member of the large TNF superfamily that can trigger apoptosis in transformed or infected cells by binding and activating two receptors, TRAIL receptor 1 (TRAILR1) and TRAIL receptor 2 (TRAILR2). Compared to other death ligands of the same family, TRAIL induces apoptosis preferentially in malignant cells while sparing normal tissue and has therefore been extensively investigated for its suitability as an anti-cancer agent. Recently, it was noticed that TRAIL receptor signaling is also linked to endoplasmic reticulum (ER) stress and the unfolded protein response (UPR). The role of TRAIL receptors in regulating cellular apoptosis susceptibility therefore is broader than previously thought. Here, we provide an overview of TRAIL-induced signaling, covering the core signal transduction during extrinsic apoptosis as well as its link to alternative outcomes, such as necroptosis or NF-κB activation. We discuss how environmental factors, transcriptional regulators, and genetic or epigenetic alterations regulate TRAIL receptors and thus alter cellular TRAIL susceptibility. Finally, we provide insight into the role of TRAIL receptors in signaling scenarios that engage the unfolded protein response and discuss how these findings might be translated into new combination therapies for cancer treatment.
Collapse
Affiliation(s)
- Daniela Stöhr
- University of Stuttgart, Institute of Cell Biology and Immunology, Stuttgart, Germany; University of Stuttgart, Stuttgart Research Center Systems Biology, Stuttgart, Germany.
| | - Albert Jeltsch
- Department of Biochemistry, University of Stuttgart, Institute of Biochemistry and Technical Biochemistry, Stuttgart, Germany
| | - Markus Rehm
- University of Stuttgart, Institute of Cell Biology and Immunology, Stuttgart, Germany; University of Stuttgart, Stuttgart Research Center Systems Biology, Stuttgart, Germany; University of Stuttgart, Stuttgart Centre for Simulation Science, Stuttgart, Germany
| |
Collapse
|
21
|
Double-Edged Lipid Nanoparticles Combining Liposome-Bound TRAIL and Encapsulated Doxorubicin Showing an Extraordinary Synergistic Pro-Apoptotic Potential. Cancers (Basel) 2019; 11:cancers11121948. [PMID: 31817469 PMCID: PMC6966652 DOI: 10.3390/cancers11121948] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 11/27/2019] [Accepted: 11/28/2019] [Indexed: 12/29/2022] Open
Abstract
Although TRAIL (TNF-related apoptosis-inducing ligand, also known as Apo2L) was described as capable of inducing apoptosis in transformed cells while sparing normal cells, limited results obtained in clinical trials has limited its use as an anti-tumor agent. Consequently, novel TRAIL formulations with enhanced bioactivity are necessary for overcoming resistance to conventional soluble TRAIL (sTRAIL) exhibited by many primary tumors. Our group has generated artificial liposomes with sTRAIL anchored on their surface (large unilamellar vesicle (LUV)-TRAIL), which have shown a greater cytotoxic activity both in vitro and in vivo when compared to sTRAIL against distinct hematologic and epithelial carcinoma cells. In this study, we have improved LUV-TRAIL by loading doxorubicin (DOX) in its liposomal lumen (LUVDOX-TRAIL) in order to improve their cytotoxic potential. LUVDOX-TRAIL killed not only to a higher extent, but also with a much faster kinetic than LUV-TRAIL. In addition, the concerted action of the liposomal DOX and TRAIL was specific of the liposomal DOX and was not observed when with soluble DOX. The cytotoxicity induced by LUVDOX-TRAIL was proven to rely on two processes due to different molecular mechanisms: a dynamin-mediated internalization of the doxorubicin-loaded particle, and the strong activation of caspase-8 exerted by the liposomal TRAIL. Finally, greater cytotoxic activity of LUVDOX-TRAIL was also observed in vivo in a tumor xenograft model. Therefore, we developed a novel double-edged nanoparticle combining the cytotoxic potential of DOX and TRAIL, showing an exceptional and remarkable synergistic effect between both agents.
Collapse
|
22
|
Thapa B, KC R, Bahniuk M, Schmitke J, Hitt M, Lavasanifar A, Kutsch O, Seol DW, Uludag H. Breathing New Life into TRAIL for Breast Cancer Therapy: Co-Delivery of pTRAIL and Complementary siRNAs Using Lipopolymers. Hum Gene Ther 2019; 30:1531-1546. [DOI: 10.1089/hum.2019.096] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Affiliation(s)
- Bindu Thapa
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Canada
| | - Remant KC
- Department of Chemical and Materials Engineering, Faculty of Engineering, University of Alberta, Edmonton, Canada
| | - Markian Bahniuk
- Department of Chemical and Materials Engineering, Faculty of Engineering, University of Alberta, Edmonton, Canada
| | - Janine Schmitke
- Department of Chemical and Materials Engineering, Faculty of Engineering, University of Alberta, Edmonton, Canada
| | - Mary Hitt
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
| | - Afsaneh Lavasanifar
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Canada
| | - Olaf Kutsch
- Department of Medicine, University of Alabama, Birmingham, Alabama
| | - Dai-Wu Seol
- College of Pharmacy, Chung-Ang University, Seoul, South Korea
| | - Hasan Uludag
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Canada
- Department of Chemical and Materials Engineering, Faculty of Engineering, University of Alberta, Edmonton, Canada
- Department of Biomedical Engineering, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
| |
Collapse
|
23
|
Dhuriya YK, Sharma D, Naik AA. Cellular demolition: Proteins as molecular players of programmed cell death. Int J Biol Macromol 2019; 138:492-503. [PMID: 31330212 DOI: 10.1016/j.ijbiomac.2019.07.113] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 06/25/2019] [Accepted: 07/19/2019] [Indexed: 12/11/2022]
Abstract
Apoptosis, a well-characterized and regulated cell death programme in eukaryotes plays a fundamental role in developing or later-life periods to dispose of unwanted cells to maintain typical tissue architecture, homeostasis in a spatiotemporal manner. This silent cellular death occurs without affecting any neighboring cells/tissue and avoids triggering of immunological response. Furthermore, diminished forms of apoptosis result in cancer and autoimmune diseases, whereas unregulated apoptosis may also lead to the development of a myriad of neurodegenerative diseases. Unraveling the mechanistic events in depth will provide new insights into understanding physiological control of apoptosis, pathological consequences of abnormal apoptosis and development of novel therapeutics for diseases. Here we provide a brief overview of molecular players of programmed cell death with discussion on the role of caspases, modifications, ubiquitylation in apoptosis, removal of the apoptotic body and its relevance to diseases.
Collapse
Affiliation(s)
- Yogesh Kumar Dhuriya
- Developmental Toxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow 226 001, India
| | - Divakar Sharma
- Department of Biochemistry, National JALMA Institute for Leprosy and Other Mycobacterial Diseases, Tajganj, Agra, India; Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh 202002, India.
| | - Aijaz A Naik
- Neurology, School of Medicine, University of Virginia, Charlottesville 22908, United States of America
| |
Collapse
|
24
|
Evstratova YV, Kobyakova MI, Novikova VV, Senotov AS, Akatov VS, Fadeev RS. Monocyte-Macrophage Differentiation Suppresses the Expression of Proapoptotic Receptors to Apo2L/TRAIL and Increases Resistance to TRAIL-Induced Apoptosis. Biophysics (Nagoya-shi) 2019. [DOI: 10.1134/s0006350919050038] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
25
|
Zhang S, Zheng C, Zhu W, Xiong P, Zhou D, Huang C, Zheng D. A novel anti-DR5 antibody-drug conjugate possesses a high-potential therapeutic efficacy for leukemia and solid tumors. Am J Cancer Res 2019; 9:5412-5423. [PMID: 31410224 PMCID: PMC6691585 DOI: 10.7150/thno.33598] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 06/21/2019] [Indexed: 12/17/2022] Open
Abstract
It is well known that tumor necrosis factor-related apoptosis inducing ligand receptor 1 or 2 (DR4/DR5) is specifically expressed in various tumor cells, but less or no expression in most normal cells. Many first generations of TRAIL agonists including recombinant preparations of TRAIL, agonistic antibodies against DR4/DR5 have been developed in phase I/II clinical trials for cancer therapy. However, the outcomes of clinical trials by using DR4/DR5 agonist mono-therapy were disappointed even though the safety profile was well tolerance. In the present study, we report an anti-DR5 antibody-drug conjugate (ADC, named as Zapadcine-1) possesses a higher potential for the therapy of lymphocyte leukemia and solid cancers. Methods: Zapadcine-1 was made by a fully humanized DR5-specific monoclonal antibody (Zaptuzumab) coupled via a cleavable linker to a highly toxic inhibitor of tubulin, monomethyl auristatin D (MMAD), by using ThioBridge technology. Cytotoxicity of the ADC in various tumor cells was identified by luminescent cell viability assay and the efficacy in vivo was determined in cells derived xenografts (CDX) of Jurkat E6-1, BALL-1, Reh, and patient derived xenografts (PDX) of human acute leukemia. Preliminary safety evaluation was carried out in rat and monkey. Results: Zapadcine-1 possesses a similar binding ability to the death receptor DR5 as the naked monoclonal antibody Zaptuzumab, and can be rapidly endocytosed into the lysosome of cancer cells. Zapadcine-1 specifically kills human lymphocyte leukemia cells and solid tumor cells, but not normal cells tested. More importantly, Zapadcine-1 drastically eliminates the xenografts in both CDX and PDX models of human acute leukemia. The excellent and comparable therapeutic efficacy is also observed in lung cancer NCI-H1975 CDX mouse model. The maximum-tolerated dose (MTD) of single injected Zapadcine-1 in rat and cynomolgus monkey shows an acceptable safety profile. Conclusion: These data demonstrate a promising anti-cancer activity, meriting further exploration of its potential as a novel cancer therapeutic agent, especially for the acute lymphocyte leukemia.
Collapse
|
26
|
Yu B, Zhang X, Yan J, Liu D, Li L, Pei R, Yu X, You T. Improved Stability, Antitumor Effect, and Controlled Release of Recombinant Soluble TRAIL by Combining Genetic Engineering with Coaxial Electrospinning. ACS APPLIED BIO MATERIALS 2019; 2:2414-2420. [DOI: 10.1021/acsabm.9b00119] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Affiliation(s)
- Bin Yu
- School of Agricultural Equipment Engineering Institute of Agricultural Engineering, Jiangsu University, Zhenjiang, Jiangsu 212013, China
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Xueping Zhang
- Department of Materials Science and Engineering, National University of Singapore, Singapore 117575, Singapore
| | - Jingyi Yan
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Dong Liu
- School of Agricultural Equipment Engineering Institute of Agricultural Engineering, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Libo Li
- School of Agricultural Equipment Engineering Institute of Agricultural Engineering, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Renjun Pei
- Suzhou Institute of Nano-tech and Nano-bionics, Chinese Academy of Sciences, Suzhou, Jiangsu 215123, China
| | - Xianghui Yu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Tianyan You
- School of Agricultural Equipment Engineering Institute of Agricultural Engineering, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| |
Collapse
|
27
|
Lee YR, Hwang E, Jang YJ. Involvement of p38 Activation and Mitochondria in Death of Human Leukemia Cells Induced by an Agonistic Human Monoclonal Antibody Fab Specific to TRAIL Receptor 1. Int J Mol Sci 2019; 20:ijms20081967. [PMID: 31013630 PMCID: PMC6515105 DOI: 10.3390/ijms20081967] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 04/05/2019] [Accepted: 04/18/2019] [Indexed: 11/16/2022] Open
Abstract
The tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) induces cancer cell death with minimal damage to normal cells; however, some cancer cells are resistant to TRAIL. TRAIL resistance may be overcome by agonistic antibodies to TRAIL receptors. In this study, we report the toxic effects of a novel recombinant agonistic human anti-TRAIL receptor 1 (DR4) monoclonal antibody Fab fragment, DR4-4, on various TRAIL-resistant and -sensitive cancer cell lines. The mechanisms of DR4-4 Fab-induced cell death in a human T cell leukemia cell line (Jurkat) were investigated using cell viability testing, immunoblotting, immunoassays, flow cytometry, and morphological observation. DR4-4 Fab-induced caspase-independent necrosis was observed to occur in Jurkat cells in association with p38 mitogen-activated protein kinase activation, cellular FLICE (FADD-like IL-1β-converting enzyme)-inhibitory protein degradation, decreased mitochondrial membrane potential, and increased mitochondrial reactive oxygen species production. Increased cytotoxic effects of DR4-4 Fab were observed in combination with TRAIL or γ-irradiation. Our results indicate that the novel DR4-4 Fab might overcome TRAIL-resistance and induce death in leukemia cells via cellular mechanisms different from those activated by TRAIL. DR4-4 Fab may have application as a potential therapeutic antibody fragment in single or combination therapy for cancer.
Collapse
Affiliation(s)
- You-Ri Lee
- Department of Microbiology, Ajou University School of Medicine, Suwon 16499, Korea.
| | - Eunjoo Hwang
- Department of Microbiology, Ajou University School of Medicine, Suwon 16499, Korea.
| | - Young-Ju Jang
- Department of Microbiology, Ajou University School of Medicine, Suwon 16499, Korea.
| |
Collapse
|
28
|
Zhang F, Chen D, Yang W, Duan S, Chen Y. Combined effects of XAF1 and TRAIL on the apoptosis of lung adenocarcinoma cells. Exp Ther Med 2019; 17:4663-4669. [PMID: 31086598 DOI: 10.3892/etm.2019.7491] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Accepted: 04/10/2017] [Indexed: 12/29/2022] Open
Abstract
This study aimed to investigate the effects and mechanisms of X-linked inhibitor of apoptosis protein (XIAP)-associated factor 1 (XAF1) and tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) on the apoptosis of A549 lung adenocarcinoma cell lines. Recombinant lentiviral vector of Ad5/F35-XAF1 and controlled lentiviral vector of Ad5/F35-Null were transfected into A549 cells at same multiplicity of infection (MOI), respectively. Based on whether recombinant human TRAIL (rhTRAIL) was added or not, cells were divided into different groups as follows: XAF1 group, XAF1 + TRAIL group, XAF1-Null group, and XAF1-Null + TRAIL group. Following culturing for 48 h, the mRNA and protein expression levels of related genes were determined by reverse transcription-quantitative polymerase chain reaction and western blotting analyses, respectively. Cell proliferationand cell apoptosis were detected by MTT assay and Annexin V-FITC/PI double staining, respectively. Xenograft mice models were established with A549 lung adenocarcinoma cells and treated with recombinant virus Ad5/F35-XAF1 and controlled virus Ad5/F35-Null for immunohistochemical analysis. Expression levels of XAFl at the mRNA and protein levels were significantly higher in the XAF1 group and XAF1 + TRAIL groups when compared with the levels in the other groups (P<0.05). Cleavage of apoptosis-associated proteins, poly ADP-ribose polymerase and caspase-3, was noted in the XAF1 + TRAIL group, whereas they were not detected in other groups. Apoptosis rates of A549 cells in the XAF1, Null + TRAIL and XAFl + TRAIL groups were significantly higher than those in the NOR and Null groups (P<0.05). Apoptotic rates were highest in the XAF1 + TRAIL group. In conclusion, these findings suggest that combined use of XAF1 and TRAIL may synergistically induce the apoptosis of A549 lung adenocarcinoma cells.
Collapse
Affiliation(s)
- Fuquan Zhang
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, P.R. China
| | - Donglai Chen
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, P.R. China
| | - Wentao Yang
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, P.R. China
| | - Shanzhou Duan
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, P.R. China
| | - Yongbing Chen
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, P.R. China
| |
Collapse
|
29
|
Kretz AL, Trauzold A, Hillenbrand A, Knippschild U, Henne-Bruns D, von Karstedt S, Lemke J. TRAILblazing Strategies for Cancer Treatment. Cancers (Basel) 2019; 11:cancers11040456. [PMID: 30935038 PMCID: PMC6521007 DOI: 10.3390/cancers11040456] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 03/25/2019] [Accepted: 03/26/2019] [Indexed: 01/07/2023] Open
Abstract
In the late 1990s, tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL), a member of the TNF-family, started receiving much attention for its potential in cancer therapy, due to its capacity to induce apoptosis selectively in tumour cells in vivo. TRAIL binds to its membrane-bound death receptors TRAIL-R1 (DR4) and TRAIL-R2 (DR5) inducing the formation of a death-inducing signalling complex (DISC) thereby activating the apoptotic cascade. The ability of TRAIL to also induce apoptosis independently of p53 makes TRAIL a promising anticancer agent, especially in p53-mutated tumour entities. Thus, several so-called TRAIL receptor agonists (TRAs) were developed. Unfortunately, clinical testing of these TRAs did not reveal any significant anticancer activity, presumably due to inherent or acquired TRAIL resistance of most primary tumour cells. Since the potential power of TRAIL-based therapies still lies in TRAIL's explicit cancer cell-selectivity, a desirable approach going forward for TRAIL-based cancer therapy is the identification of substances that sensitise tumour cells for TRAIL-induced apoptosis while sparing normal cells. Numerous of such TRAIL-sensitising strategies have been identified within the last decades. However, many of these approaches have not been verified in animal models, and therefore potential toxicity of these approaches has not been taken into consideration. Here, we critically summarise and discuss the status quo of TRAIL signalling in cancer cells and strategies to force tumour cells into undergoing apoptosis triggered by TRAIL as a cancer therapeutic approach. Moreover, we provide an overview and outlook on innovative and promising future TRAIL-based therapeutic strategies.
Collapse
Affiliation(s)
- Anna-Laura Kretz
- Department of General and Visceral Surgery, Ulm University Hospital, Albert-Einstein-Allee 23, 89081 Ulm, Germany.
| | - Anna Trauzold
- Institute for Experimental Cancer Research, University of Kiel, 24105 Kiel, Germany.
- Clinic for General Surgery, Visceral, Thoracic, Transplantation and Pediatric Surgery, University Hospital Schleswig-Holstein, 24105 Kiel, Germany.
| | - Andreas Hillenbrand
- Department of General and Visceral Surgery, Ulm University Hospital, Albert-Einstein-Allee 23, 89081 Ulm, Germany.
| | - Uwe Knippschild
- Department of General and Visceral Surgery, Ulm University Hospital, Albert-Einstein-Allee 23, 89081 Ulm, Germany.
| | - Doris Henne-Bruns
- Department of General and Visceral Surgery, Ulm University Hospital, Albert-Einstein-Allee 23, 89081 Ulm, Germany.
| | - Silvia von Karstedt
- Department of Translational Genomics, University Hospital Cologne, Weyertal 115b, 50931 Cologne, Germany.
- Cologne Excellence Cluster on Cellular Stress Response in Aging-Associated Diseases (CECAD), University of Cologne, Joseph-Stelzmann Straße 26, 50931 Cologne, Germany.
| | - Johannes Lemke
- Department of General and Visceral Surgery, Ulm University Hospital, Albert-Einstein-Allee 23, 89081 Ulm, Germany.
| |
Collapse
|
30
|
Le DHT, Commandeur U, Steinmetz NF. Presentation and Delivery of Tumor Necrosis Factor-Related Apoptosis-Inducing Ligand via Elongated Plant Viral Nanoparticle Enhances Antitumor Efficacy. ACS NANO 2019; 13:2501-2510. [PMID: 30668110 DOI: 10.1021/acsnano.8b09462] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Potato virus X (PVX) is a flexuous plant virus-based nanotechnology with promise in cancer therapy. As a high aspect ratio biologic (13 × 515 nm), PVX has excellent spatial control in structures and functions, offering high-precision nanoengineering for multivalent display of functional moieties. Herein, we demonstrate the preparation of the PVX-based nanocarrier for delivery of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL), a promising protein drug that induces apoptosis in cancer cells but not healthy cells. TRAIL bound to PVX by coordination bonds between nickel-coordinated nitrilotriacetic acid on PVX and His-tag on the protein could mimic the bioactive "membrane-bound" state in native TRAIL, resulting in an elongated nanoparticle displaying up 490 therapeutic protein molecules. Our data show that PVX-delivered TRAIL activates caspase-mediated apoptosis more efficiently compared to soluble TRAIL; also in vivo the therapeutic nanoparticle outperforms in delaying tumor growth in an athymic nude mouse model bearing human triple-negative breast cancer xenografts. This proof-of-concept work highlights the potential of filamentous plant virus nanotechnologies, particularly for targeting protein drug delivery for cancer therapy.
Collapse
Affiliation(s)
- Duc H T Le
- Department of Biomedical Engineering , Case Western Reserve University School of Medicine , Cleveland , Ohio 44106 , United States
| | - Ulrich Commandeur
- Department of Molecular Biology , RWTH-Aachen University , Aachen 52064 , Germany
| | - Nicole F Steinmetz
- Department of NanoEngineering, Moores Cancer Center, Department of Radiology, Department of Bioengineering , University of California, San Diego , La Jolla , California 92093 , United States
- Department of Biomedical Engineering , Case Western Reserve University School of Medicine , Cleveland , Ohio 44106 , United States
| |
Collapse
|
31
|
Improved Anticancer Effect of Recombinant Protein izTRAIL Combined with Sorafenib and Peptide iRGD. Int J Mol Sci 2019; 20:ijms20030525. [PMID: 30691192 PMCID: PMC6387460 DOI: 10.3390/ijms20030525] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 01/19/2019] [Accepted: 01/24/2019] [Indexed: 01/10/2023] Open
Abstract
One of the main problems in oncology is the development of drugs that cause the death of cancer cells without damaging normal cells. Another key problem to be solved is to suppress the drug resistance of cancer cells. The third important issue is to provide effective penetration of drug molecules to cancer cells. TRAIL (TNFα-related apoptosis inducing ligand)/Apo2L is a highly selective anticancer agent. However, the recombinant TRAIL protein having high efficiency against cancer cells in vitro was not effective in clinical trials. Recently we have discovered an acquisition of TRAIL resistance by cancer cells in confluent cultures, which is apparently a manifestation of the general phenomenon of multicellular resistance. The aim of this study was to evaluate whether the anticancer effect of the recombinant protein TRAIL in vivo can be improved by the suppression of multicellular TRAIL-resistance using sorafenib and a tumor-penetrating peptide iRGD, c(CRGDKGPDC). The results testified a great increase in the resistance of human fibrosarcoma HT-1080 cells to izTRAIL both in confluent cultures and in spheroids. Sorafenib administered at nontoxic concentration effectively suppressed confluent- or spheroid-mediated TRAIL-resistance of HT-1080 cells in vitro. Sorafenib combined with iRGD significantly improved the anticancer effect of the recombinant protein izTRAIL in HT-1080 human fibrosarcoma grafts in BALB/c nude mice. Consistent with this finding, multicellular TRAIL-resistance may be a reason of inefficacy of izTRAIL alone in vivo. The anticancer effect of the recombinant protein izTRAIL in vivo may be improved in combination with sorafenib, an inhibitor of multicellular TRAIL resistance and iRGD, the tumor-penetrating peptide.
Collapse
|
32
|
Zhang B, van Roosmalen IAM, Reis CR, Setroikromo R, Quax WJ. Death receptor 5 is activated by fucosylation in colon cancer cells. FEBS J 2019; 286:555-571. [PMID: 30589515 PMCID: PMC6849799 DOI: 10.1111/febs.14742] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Revised: 11/01/2018] [Accepted: 12/24/2018] [Indexed: 12/24/2022]
Abstract
The remarkable pro‐apoptotic properties of tumour necrosis factor (TNF)‐related apoptosis‐inducing ligand (TRAIL) have led to considerable interest in this protein as a potential anticancer therapeutic. However, TRAIL is largely ineffective in inducing apoptosis in certain cancer cells, and the mechanisms underlying this selectivity are unknown. In colon adenocarcinomas, posttranslational modifications including O‐ and N‐ glycosylation of death receptors were found to correlate with TRAIL‐induced apoptosis. Additionally, mRNA levels of fucosyltransferase 3 (FUT3) and 6 (FUT6) were found to be high in the TRAIL‐sensitive colon adenocarcinoma cell line COLO 205. In this study, we use agonistic receptor‐specific TRAIL variants to dissect the contribution of FUT3 and FUT6‐mediated fucosylation to TRAIL‐induced apoptosis via its two death receptors, DR4 and DR5. Triggering of apoptosis by TRAIL revealed that the low FUT3/6‐expressing cells DLD‐1 and HCT 116 are insensitive to DR5 but not to DR4‐mediated apoptosis. By contrast, efficient apoptosis is mediated via both receptors in high FUT3/6‐expressing COLO 205 cells. The reconstitution of FUT3/6 expression in DR5‐resistant cells completely restored TRAIL sensitivity via this receptor, while only marginally enhancing apoptosis via DR4 at lower TRAIL concentrations. Interestingly, we observed that induction of the salvage pathway by external administration of l‐fucose restores DR5‐mediated apoptosis in both DLD‐1 and HCT 116 cells. Finally, we show that fucosylation influences the ligand‐independent receptor association that leads to increased death inducing signalling complex (DISC) formation and caspase‐8 activation. Taken together, these results provide evidence for the differential impact of fucosylation on signalling via DR4 or DR5. These findings provide novel opportunities to enhance TRAIL sensitivity in colon adenocarcinoma cells that are highly resistant to DR5‐mediated apoptosis.
Collapse
Affiliation(s)
- Baojie Zhang
- Department of Chemical and Pharmaceutical Biology, Groningen Research Institute of Pharmacy, University of Groningen, The Netherlands
| | - Ingrid A M van Roosmalen
- Department of Chemical and Pharmaceutical Biology, Groningen Research Institute of Pharmacy, University of Groningen, The Netherlands
| | - Carlos R Reis
- Department of Chemical and Pharmaceutical Biology, Groningen Research Institute of Pharmacy, University of Groningen, The Netherlands
| | - Rita Setroikromo
- Department of Chemical and Pharmaceutical Biology, Groningen Research Institute of Pharmacy, University of Groningen, The Netherlands
| | - Wim J Quax
- Department of Chemical and Pharmaceutical Biology, Groningen Research Institute of Pharmacy, University of Groningen, The Netherlands
| |
Collapse
|
33
|
Zhou X, Liu Z, Wang H, Liu X, Zhou Z, Tang J, Liu X, Zheng M, Shen Y. SAHA (vorinostat) facilitates functional polymer-based gene transfection via upregulation of ROS and synergizes with TRAIL gene delivery for cancer therapy. J Drug Target 2018; 27:306-314. [PMID: 30188217 DOI: 10.1080/1061186x.2018.1519028] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Non-viral gene delivery is an attractive approach for the treatment of many diseases including cancer, benefiting from its safety and large-scale production concerns. However, the relatively low transfection efficacy compared with viral vectors restricts the clinical applications of non-viral gene vectors. Reactive oxygen species (ROS) triggered charge reversal polymers (named B-PDEAEA) presented improved transfection efficacy, because of fast release of plasmid DNA responding to enhanced oxidative stress in cancer cells. But inadequate dissociation can still occur owing to the insufficient intracellular ROS generation. Here, we report SAHA (vorinostat), which is a clinical histone deacetylase inhibitor and anticancer drug, induces the ROS accumulation in cancer cells, and facilitates the charge reversal process of B-PDEAEA and the cellular dissociation of the delivered gene from the vectors. As a result, SAHA remarkably increases the gene transfection efficacy in an ROS-dependent manner. Importantly, SAHA synergizes with B-PDEAEA mediated therapeutic gene TNF-related apoptosis-inducing ligand (TRAIL) delivery in inducing apoptosis of cancer cells. These findings support the first concept of improving the gene delivery efficacy of stimuli-responsive vectors through upregulating the cellular ROS via an FDA approved anticancer agent. Additionally, combination of SAHA and TRAIL gene therapy could be a potential strategy for cancer treatment.
Collapse
Affiliation(s)
- Xuefei Zhou
- a Key Laboratory of Biomass Chemical Engineering of Ministry of Education and Center for Bionanoengineering, College of Chemical and Biological Engineering , Zhejiang University , Hangzhou , China
| | - Zimo Liu
- a Key Laboratory of Biomass Chemical Engineering of Ministry of Education and Center for Bionanoengineering, College of Chemical and Biological Engineering , Zhejiang University , Hangzhou , China
| | - Huifang Wang
- a Key Laboratory of Biomass Chemical Engineering of Ministry of Education and Center for Bionanoengineering, College of Chemical and Biological Engineering , Zhejiang University , Hangzhou , China
| | - Xin Liu
- a Key Laboratory of Biomass Chemical Engineering of Ministry of Education and Center for Bionanoengineering, College of Chemical and Biological Engineering , Zhejiang University , Hangzhou , China
| | - Zhuxian Zhou
- a Key Laboratory of Biomass Chemical Engineering of Ministry of Education and Center for Bionanoengineering, College of Chemical and Biological Engineering , Zhejiang University , Hangzhou , China
| | - Jianbin Tang
- a Key Laboratory of Biomass Chemical Engineering of Ministry of Education and Center for Bionanoengineering, College of Chemical and Biological Engineering , Zhejiang University , Hangzhou , China
| | - Xiangrui Liu
- a Key Laboratory of Biomass Chemical Engineering of Ministry of Education and Center for Bionanoengineering, College of Chemical and Biological Engineering , Zhejiang University , Hangzhou , China
| | - Min Zheng
- b State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Medical School , Zhejiang University , Hangzhou , China
| | - Youqing Shen
- a Key Laboratory of Biomass Chemical Engineering of Ministry of Education and Center for Bionanoengineering, College of Chemical and Biological Engineering , Zhejiang University , Hangzhou , China
| |
Collapse
|
34
|
Nihira K, Nan-ya KI, Kakuni M, Ono Y, Yoshikawa Y, Ota T, Hiura M, Yoshinari K. Chimeric Mice With Humanized Livers Demonstrate Human-Specific Hepatotoxicity Caused by a Therapeutic Antibody Against TRAIL-Receptor 2/Death Receptor 5. Toxicol Sci 2018; 167:190-201. [DOI: 10.1093/toxsci/kfy228] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Affiliation(s)
- Kaito Nihira
- Translational Research Unit, Kyowa Hakko Kirin Co., Ltd., Nagaizumi-cho, Sunto-gun, Shizuoka 411-8731, Japan
- Laboratory of Molecular Toxicology, School of Pharmaceutical Sciences, University of Shizuoka, Suruga-ku, Shizuoka 422-8526, Japan
| | - Ken-ichiro Nan-ya
- Translational Research Unit, Kyowa Hakko Kirin Co., Ltd., Nagaizumi-cho, Sunto-gun, Shizuoka 411-8731, Japan
| | - Masakazu Kakuni
- PhoenixBio Co., Ltd., Higashihiroshima, Hiroshima 739-0046, Japan
| | - Yoko Ono
- Translational Research Unit, Kyowa Hakko Kirin Co., Ltd., Nagaizumi-cho, Sunto-gun, Shizuoka 411-8731, Japan
| | - Yukitaka Yoshikawa
- Translational Research Unit, Kyowa Hakko Kirin Co., Ltd., Nagaizumi-cho, Sunto-gun, Shizuoka 411-8731, Japan
| | - Toshio Ota
- Translational Research Unit, Kyowa Hakko Kirin Co., Ltd., Nagaizumi-cho, Sunto-gun, Shizuoka 411-8731, Japan
| | - Masanori Hiura
- Translational Research Unit, Kyowa Hakko Kirin Co., Ltd., Nagaizumi-cho, Sunto-gun, Shizuoka 411-8731, Japan
| | - Kouichi Yoshinari
- Laboratory of Molecular Toxicology, School of Pharmaceutical Sciences, University of Shizuoka, Suruga-ku, Shizuoka 422-8526, Japan
| |
Collapse
|
35
|
Jiang H, Wang S, Zhou X, Wang L, Ye L, Zhou Z, Tang J, Liu X, Teng L, Shen Y. New path to treating pancreatic cancer: TRAIL gene delivery targeting the fibroblast-enriched tumor microenvironment. J Control Release 2018; 286:254-263. [PMID: 30075209 DOI: 10.1016/j.jconrel.2018.07.047] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 07/12/2018] [Accepted: 07/30/2018] [Indexed: 12/18/2022]
Abstract
Gene therapy has shown promise in antitumor strategies for advanced cancer. However, efficient and safe delivery of potent therapeutic gene expressing in specific tumor tissues remains elusive, especially when there exist stromal obstacles. Here we report a non-viral gene delivery approach targeting pancreatic stellate cells (PSCs) as the transfection host in the fibroblast-enriched tumor microenvironment of pancreatic cancer. Plasmid DNA (pDNA) encapsulated in branched polyethylenemine (BPEI) was found to selectively transfect PSCs rather than pancreatic cancer cells and other fibroblast cell lines. Mechanism investigations reveal that the highly expressed fibroblast growth factor receptors (FGFRs) in PSCs facilitated the cellular uptake of polyplexes in PSCs. This delivery platform carrying gene encoding of TNF-related apoptosis-inducing ligand (TRAIL) displayed effective by-stander effect and tumor cell-selective cytotoxicity. More importantly, the therapeutic efficacy was proved in a PSC-enriched orthotopic pancreatic tumor model. Thus, this gene delivery strategy smartly converts PSCs as the microenvironment obstacle for drug delivery into the producer and reservoir of selective tumor-killing proteins.
Collapse
Affiliation(s)
- Haiping Jiang
- Department of Medical Oncology, the First Affiliated Hospital of Medical School of Zhejiang University, Hangzhou, People's Republic of China
| | - Sujuan Wang
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, China
| | - Xuefei Zhou
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, China
| | - Liying Wang
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, China
| | - Lidan Ye
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, China
| | - Zhuxian Zhou
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, China
| | - Jianbin Tang
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, China
| | - Xiangrui Liu
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, China.
| | - Lisong Teng
- Department of Surgical Oncology, the First Affiliated Hospital of Medical School of Zhejiang University, Hangzhou, People's Republic of China
| | - Youqing Shen
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, China
| |
Collapse
|
36
|
TGF3L fusion enhances the antitumor activity of TRAIL by promoting assembly into polymers. Biochem Pharmacol 2018; 155:510-523. [PMID: 30059675 DOI: 10.1016/j.bcp.2018.07.035] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Accepted: 07/25/2018] [Indexed: 01/21/2023]
Abstract
TRAIL, a promising antitumor immuno-agent, exerted limited efficacy in clinical trials. The third disulfide loop of TGF-α (TGF3L peptide) with a very low affinity for EGFR has been reported to enhance the activity of fused antigens or cytokines. We wondered whether fusion of this peptide could enhance TRAIL activity and what the underlying mechanism for this enhancement would be. The TGF3L-TRAIL showed greatly enhanced cytotoxicity in a variety of cancer cell lines while spared normal cells unharmed. Typical apoptosis and cellular caspase activation were potently induced by TGF3L-TRAIL at the concentration levels corresponding to its cytotoxicity. TGF3L-TRAIL was able to activate both DR4 and DR5 the same as TRAIL did. It induced complete cell death in Colo205 through only one receptor when the other one was blocked, different from TRAIL-induced cell death (through DR4 dominantly). TGF3L-TRAIL cytotoxicity was not reduced in some cell lines even if both receptors are blocked simultaneously. Surprisingly, TGF3L-TRAIL self-assembled into stable polymers, which was responsible for its enhanced cytotoxicity. In human tumor xenograft mouse models, TGF3L-TRAIL showed anti-tumor activity similar to or better than TRAIL in different cancer cell types, consistent with its differing enhancement of cytotoxicity in vitro. Taken together, TGF3L fusion of TRAIL obviously enhances the anticancer activity of TRAIL by promoting assembly into polymers, which presents a novel fusion strategy for improving TRAIL function.
Collapse
|
37
|
Fakiruddin KS, Ghazalli N, Lim MN, Zakaria Z, Abdullah S. Mesenchymal Stem Cell Expressing TRAIL as Targeted Therapy against Sensitised Tumour. Int J Mol Sci 2018; 19:ijms19082188. [PMID: 30060445 PMCID: PMC6121609 DOI: 10.3390/ijms19082188] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 06/30/2018] [Accepted: 07/02/2018] [Indexed: 02/06/2023] Open
Abstract
Tapping into the ability of engineered mesenchymal stem cells (MSCs) to mobilise into the tumour has expanded the scope of cancer treatment. Engineered MSCs expressing tumour necrosis factor (TNF)-related apoptosis inducing ligand (MSC-TRAIL) could serve as a platform for an efficient and targeted form of therapy. However, the presence of cancer stem cells (CSCs) that are resistant to TRAIL and apoptosis may represent a challenge for effective treatment. Nonetheless, with the discovery of small molecular inhibitors that could target CSCs and tumour signalling pathways, a higher efficacy of MSC-TRAIL mediated tumour inhibition can be achieved. This might pave the way for a more effective form of combined therapy, which leads to a better treatment outcome. In this review, we first discuss the tumour-homing capacity of MSCs, its effect in tumour tropism, the different approach behind genetically-engineered MSCs, and the efficacy and safety of each agent delivered by these MSCs. Then, we focus on how sensitisation of CSCs and tumours using small molecular inhibitors can increase the effect of these cells to either TRAIL or MSC-TRAIL mediated inhibition. In the conclusion, we address a few questions and safety concerns regarding the utilization of engineered MSCs for future treatment in patients.
Collapse
Affiliation(s)
- Kamal Shaik Fakiruddin
- Stem Cell Laboratory, Haematology Unit, Cancer Research Centre, Institute for Medical Research, Kuala Lumpur 50588, Malaysia.
- UPM-MAKNA Cancer Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia.
| | - Nadiah Ghazalli
- Medical Genetics Laboratory, Department of Biomedical Sciences, Faculty of Medicine & Health Sciences, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia.
| | - Moon Nian Lim
- Stem Cell Laboratory, Haematology Unit, Cancer Research Centre, Institute for Medical Research, Kuala Lumpur 50588, Malaysia.
| | - Zubaidah Zakaria
- Stem Cell Laboratory, Haematology Unit, Cancer Research Centre, Institute for Medical Research, Kuala Lumpur 50588, Malaysia.
| | - Syahril Abdullah
- UPM-MAKNA Cancer Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia.
- Medical Genetics Laboratory, Department of Biomedical Sciences, Faculty of Medicine & Health Sciences, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia.
| |
Collapse
|
38
|
Kih M, Lee EJ, Lee NK, Kim YK, Lee KE, Jeong C, Yang Y, Kim DH, Kim IS. Designed trimer-mimetic TNF superfamily ligands on self-assembling nanocages. Biomaterials 2018; 180:67-77. [PMID: 30025246 DOI: 10.1016/j.biomaterials.2018.07.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 07/05/2018] [Accepted: 07/06/2018] [Indexed: 12/19/2022]
Abstract
Presentation of an endogenous bioactive ligand in its native form is a key factor in controlling and determining its bioactivity, stability, and therapeutic efficacy. In this study, we developed a novel strategy for presenting trimeric ligands on nanocages by designing, optimizing and testing based on the rational design, high-resolution structural analysis and agonistic activity assays in vitro and in vivo. We successfully designed a nanocage that presents the TNF superfamily member, TRAIL (TNF-related apoptosis-inducing ligand) in its native-like trimeric structure. The native structure of TRAIL complexes was mimicked on the resulting trimeric TRAIL-presenting nanocages (TTPNs) by inserting sufficient spacing, determined from three-dimensional structural models, to provide optimal access to the corresponding receptors. The efficacy of TTPNs as an anti-tumor agent was confirmed in preclinical studies, which revealed up to 330-fold increased affinity, 62.5-fold enhanced apoptotic activity, and improved pharmacokinetic characteristics and stability compared with the monomeric form of TRAIL (mTRAIL). In this latter context, TTPNs exhibited greater than 90% stability over 1 mo, whereas ∼50% of mTRAIL aggregated within 2 d. Consistent with their enhanced stability and ultra-high affinity for the TRAIL receptor, TTPNs effectively induced apoptosis of tumor cells in vivo, leading to effective inhibition of tumor growth. Although TRAIL was used here as a proof-of-concept, all members of the TNF superfamily share the TNF homology domain (THD) and have similar distances between ecto-domain C-termini. Thus, other TNF superfamily ligands could be genetically substituted for the TRAIL ligand on the surface of this biomimetic delivery platform.
Collapse
Affiliation(s)
- Minwoo Kih
- KU-KIST Graduate School of Converging Science and Technology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea; Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, 5 Hwarang-ro 14-gil, Seongbuk-gu, Seoul, 02792, Republic of Korea
| | - Eun Jung Lee
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, 5 Hwarang-ro 14-gil, Seongbuk-gu, Seoul, 02792, Republic of Korea; Department of Chemical Engineering, School of Applied Chemical Engineering, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Na Kyeong Lee
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, 5 Hwarang-ro 14-gil, Seongbuk-gu, Seoul, 02792, Republic of Korea; Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea
| | - Yoon Kyoung Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea; Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, 5 Hwarang-ro 14-gil, Seongbuk-gu, Seoul, 02792, Republic of Korea
| | - Kyung Eun Lee
- Advanced Analysis Center, Korea Institute of Science and Technology, 5 Hwarang-ro 14-gil, Seongbuk-gu, Seoul, 02792, Republic of Korea
| | - Cherlhyun Jeong
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, 5 Hwarang-ro 14-gil, Seongbuk-gu, Seoul, 02792, Republic of Korea
| | - Yoosoo Yang
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, 5 Hwarang-ro 14-gil, Seongbuk-gu, Seoul, 02792, Republic of Korea
| | - Dong-Hwee Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea
| | - In-San Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea; Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, 5 Hwarang-ro 14-gil, Seongbuk-gu, Seoul, 02792, Republic of Korea.
| |
Collapse
|
39
|
Danish L, Imig D, Allgöwer F, Scheurich P, Pollak N. Bcl-2-mediated control of TRAIL-induced apoptotic response in the non-small lung cancer cell line NCI-H460 is effective at late caspase processing steps. PLoS One 2018; 13:e0198203. [PMID: 29927992 PMCID: PMC6013189 DOI: 10.1371/journal.pone.0198203] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Accepted: 05/15/2018] [Indexed: 11/29/2022] Open
Abstract
Dysregulation of the mitochondrial signaling pathway of apoptosis induction represents a major hurdle in tumor therapy. The objective of the presented work was to investigate the role of the intrinsic (mitochondrial) apoptotic pathway in the non-small lung cancer cell line NCI-H460 upon induction of apoptosis using the highly bioactive TRAIL derivative Db-scTRAIL. NCI-H460 cells were TRAIL sensitive but an only about 3 fold overexpression of Bcl-2 was sufficient to induce a highly TRAIL resistant phenotype, confirming that the mitochondrial pathway is crucial for TRAIL-induced apoptosis induction. TRAIL resistance was paralleled by a strong inhibition of caspase-8, -9 and -3 activities and blocked their full processing. Notably, especially the final cleavage steps of the initiator caspase-8 and the executioner caspase-3 were effectively blocked by Bcl-2 overexpression. Caspase-9 knockdown failed to protect NCI-H460 cells from TRAIL-induced cell death, suggesting a minor role of this initiator caspase in this apoptotic pathway. Rather, knockdown of the XIAP antagonist Smac resulted in enhanced caspase-3 degradation after stimulation of cells with TRAIL. Of note, downregulation of XIAP had only limited effects on TRAIL sensitivity of wild-type NCI-H460 cells, but resensitized Bcl-2 overexpressing cells for TRAIL-induced apoptosis. In particular, XIAP knockdown in combination with TRAIL allowed the final cleavage step of caspase-3 to generate the catalytically active p17 fragment, whose production was otherwise blocked in Bcl-2 overexpressing cells. Together, our data strongly suggest that XIAP-mediated inhibition of final caspase-3 processing is the last and major hurdle in TRAIL-induced apoptosis in NCI-H460 cells, which can be overcome by Smac in a Bcl-2 level dependent manner. Quantitative investigation of the XIAP/Smac interplay using a mathematical model approach corroborates our experimental data strengthening the suggested roles of XIAP and Smac as critical determinants for TRAIL sensitivity.
Collapse
Affiliation(s)
- Lubna Danish
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - Dirke Imig
- Institute of Systems Theory and Automatic Control, University of Stuttgart, Stuttgart, Germany
| | - Frank Allgöwer
- Institute of Systems Theory and Automatic Control, University of Stuttgart, Stuttgart, Germany
| | - Peter Scheurich
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - Nadine Pollak
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
- Institute of Systems Theory and Automatic Control, University of Stuttgart, Stuttgart, Germany
- * E-mail:
| |
Collapse
|
40
|
Ralff MD, El-Deiry WS. TRAIL pathway targeting therapeutics. EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2018; 3:197-204. [PMID: 30740527 DOI: 10.1080/23808993.2018.1476062] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Introduction Despite decades of focused research efforts, cancer remains a significant cause of morbidity and mortality. Tumor necrosis factor(TNF)-related apoptosis-inducing ligand (TRAIL) is capable of inducing cell death selectively in cancer cells while sparing normal cells. Areas covered In this review, the authors cover TRA therapy and strategies that have been undertaken to improve their efficacy, as well as unconventional approaches to TRAIL pathway activation including TRAIL-inducing small molecules. They also discuss mechanisms of resistance to TRAIL and the use of combination strategies to overcome it. Expert commentary Targeting the TRAIL pathway has been of interest in oncology, and although initial clinical trials of TRAIL receptor agonists (TRAs) showed limitations, novel approaches represent the future of TRAIL-based therapy.
Collapse
Affiliation(s)
- Marie D Ralff
- MD/PhD Program, Lewis Katz School of Medicine, Temple University, Philadelphia, PA.,Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Department of Hematology/Oncology and Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, PA
| | - Wafik S El-Deiry
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Department of Hematology/Oncology and Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, PA
| |
Collapse
|
41
|
Amirijavid S, Entezari M. Comparison of the effects of three kinds of IgYs, (normal, nanoliposomal and nanoparticle conjugated), which are produced against the small domains of DR5 protein on cancer cells. IET Nanobiotechnol 2018; 12:436-440. [PMID: 29768226 DOI: 10.1049/iet-nbt.2017.0123] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Cancer treatment with several kinds of drugs, especially targets the apoptotic pathways nowadays. TNF-related apoptosis-inducing ligand (TRAIL) as one of the important members of death receptors, significantly trigger induction of apoptosis in cancer cells. Three conserved domains of Death receptor (DR5) protein extracellular domain, which are fortified cysteine, were chosen and chemically synthesised. Hens were immunised with nano-liposomal peptides, and as a result the purified Immunoglobulin (IgYs) remarkably killed the cancerous MCF7 cells. The flow cytometric assay, confirmed the apoptotic death. Among several kinds of carriers that were used in this research, the nano-liposomal and nanoparticle conjugated, both were acceptable choices for drug delivery. Furthermore, the IgY against DR5's small peptides with such carriers successfully reached the target and significantly killed the cancer cells via apoptosis.
Collapse
Affiliation(s)
| | - Maliheh Entezari
- Department of Genetics, Tehran Medical Sciences Branch, Islamic Azad University, Tehran, Iran.
| |
Collapse
|
42
|
Legler K, Hauser C, Egberts JH, Willms A, Heneweer C, Boretius S, Röcken C, Glüer CC, Becker T, Kluge M, Hill O, Gieffers C, Fricke H, Kalthoff H, Lemke J, Trauzold A. The novel TRAIL-receptor agonist APG350 exerts superior therapeutic activity in pancreatic cancer cells. Cell Death Dis 2018; 9:445. [PMID: 29670075 PMCID: PMC5906476 DOI: 10.1038/s41419-018-0478-0] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Revised: 03/02/2018] [Accepted: 03/07/2018] [Indexed: 12/12/2022]
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) has raised attention as a novel anticancer therapeutic as it induces apoptosis preferentially in tumor cells. However, first-generation TRAIL-receptor agonists (TRAs), comprising recombinant TRAIL and agonistic receptor-specific antibodies, have not demonstrated anticancer activity in clinical studies. In fact, cancer cells are often resistant to conventional TRAs. Therefore, in addition to TRAIL-sensitizing strategies, next-generation TRAs with superior apoptotic activity are warranted. APG350 is a novel, highly potent TRAIL-receptor agonist with a hexavalent binding mode allowing the clustering of six TRAIL-receptors per drug molecule. Here we report on preclinical in vitro and in vivo studies testing the activity of APG350 on pancreatic ductal adenocarcinoma (PDAC) cells. We found that APG350 potently induced apoptosis of Colo357, PancTuI and Panc89 cells in vitro. In addition, APG350 treatment activated non-canonical TRAIL signaling pathways (MAPK, p38, JNK, ERK1/ERK2 and NF-κB) and induced the secretion of IL-8. Stable overexpression of Bcl-xL inhibited APG350-induced cell death and augmented activation of non-canonical pathways. Intriguingly, pre-treatment of Bcl-xL-overexpressing cells with the BH3-mimic Navitoclax restored their sensitivity to APG350. To study the effects of APG350 on PDAC cells in vivo, we applied two different orthotopic xenotransplantation mouse models, with and without primary tumor resection, representing adjuvant and palliative treatment regimes, respectively. APG350 treatment of established tumors (palliative treatment) significantly reduced tumor burden. These effects, however, were not seen in tumors with enforced overexpression of Bcl-xL. Upon primary tumor resection and subsequent APG350 treatment (adjuvant therapy), APG350 limited recurrent tumor growth and metastases. Importantly, therapeutic efficacy of APG350 treatment was more effective compared with treatment with soluble TRAIL in both models. In conclusion, APG350 represents a promising next-generation TRA for the treatment of PDAC. Moreover, our results suggest that combining APG350 with Navitoclax might be a succesfull strategy for cancers harboring mitochondrial apoptosis resistance.
Collapse
Affiliation(s)
- Karen Legler
- Institute for Experimental Cancer Research, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Charlotte Hauser
- Clinic for General Surgery, Visceral, Thoracic, Transplantation and Pediatric Surgery, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Jan-Hendrik Egberts
- Clinic for General Surgery, Visceral, Thoracic, Transplantation and Pediatric Surgery, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Anna Willms
- Institute for Experimental Cancer Research, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Carola Heneweer
- Clinic for Diagnostic Radiology and Neuroradiology, University Hospital Schleswig-Holstein, Kiel, Germany.,Department of Radiology, University Hospital Cologne, Cologne, Germany
| | - Susann Boretius
- Clinic for Diagnostic Radiology and Neuroradiology, University Hospital Schleswig-Holstein, Kiel, Germany.,Functional Imaging Laboratory, German Primate Center, Leibniz Institute for Primate Research and Georg-August-University Göttingen, Göttingen, Germany
| | - Christoph Röcken
- Institute of Pathology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Claus-Christian Glüer
- Section Biomedical Imaging, Department of Diagnostic Radiology und Neuroradiology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Thomas Becker
- Clinic for General Surgery, Visceral, Thoracic, Transplantation and Pediatric Surgery, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Michael Kluge
- APOGENIX AG, Im Neuenheimer Feld 584, Heidelberg, Germany.,Affimed GmbH, Im Neuenheimer Feld 582, Heidelberg, Germany
| | - Oliver Hill
- APOGENIX AG, Im Neuenheimer Feld 584, Heidelberg, Germany
| | | | - Harald Fricke
- APOGENIX AG, Im Neuenheimer Feld 584, Heidelberg, Germany
| | - Holger Kalthoff
- Institute for Experimental Cancer Research, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Johannes Lemke
- Clinic of General and Visceral Surgery, University Hospital Ulm, Ulm, Germany
| | - Anna Trauzold
- Institute for Experimental Cancer Research, Christian-Albrechts-University of Kiel, Kiel, Germany. .,Clinic for General Surgery, Visceral, Thoracic, Transplantation and Pediatric Surgery, University Hospital Schleswig-Holstein, Kiel, Germany.
| |
Collapse
|
43
|
Wu ZF, Wang Y, Yang P, Hou JZ, Zhang JY, Hu Y, Zeng ZC. Toll-like receptor 4 and its associated proteins as prognostic factors for HCC treated by post-radiotherapy surgery. Oncol Lett 2018; 15:9599-9608. [PMID: 29928336 PMCID: PMC6004720 DOI: 10.3892/ol.2018.8583] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2015] [Accepted: 10/18/2017] [Indexed: 01/08/2023] Open
Abstract
Locally advanced hepatocellular carcinoma (HCC) treated by radiotherapy (RT) may be suited for further treatment with surgery. As a critical mediator of the post-RT immune response, Toll-like receptor 4 (TLR4) and its associated proteins may serve as prognostic factors for patients with HCC treated by post-RT surgery. In the present study, a total of 20 patients with HCC treated by post-RT surgery were enrolled. Resected tumor and peritumoral liver tissues were used to construct tissue microarrays that were assessed with immunohistochemical staining for the expression levels of TLR4, tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) and vascular endothelial growth factor receptor 2 (VEGFR2). The overall (OS) and disease-free (DFS) survival outcomes for each patient were assessed, and the severity of radiation-induced liver diseases (RILDs) was detected. The patients with low TLR4 or TRAIL expression exhibited significantly better OS times than those with high TLR4 (P=0.003) or TRAIL (P=0.007) expression, whereas the median DFS times for patients with low VEGFR2 or TRAIL were significantly longer than those with high VEGFR2 (P=0.003) or TRAIL (P=0.008) expression. No significant differences in OS or DFS times were identified according to the expression of TLR4, VEGFR2 or TRAIL in peritumoral liver tissue, although more severe RILDs were identified in patients with the high expression of these factors in the peritumoral liver tissue post-RT (P<0.05). Therefore, the expression levels of TLR4 and its associated proteins in HCC tumors may be suitable as prognostic factors for patients with HCC treated by post-RT surgery. The inhibition of TLR4, VEGFR2 and TRAIL expression in HCC and non-tumor liver tissue may lessen the severity of RILDs and improve survival outcomes in the future.
Collapse
Affiliation(s)
- Zhi-Feng Wu
- Department of Radiation Oncology, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| | - Ying Wang
- Department of Ultrasonography, Huashan Hospital, Fudan University, Shanghai 200032, P.R. China
| | - Ping Yang
- Department of Radiation Oncology, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| | - Jia-Zhou Hou
- Department of Radiation Oncology, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| | - Jian-Ying Zhang
- Department of Radiation Oncology, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| | - Yong Hu
- Department of Radiation Oncology, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| | - Zhao-Chong Zeng
- Department of Radiation Oncology, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| |
Collapse
|
44
|
Li SS, Tang QL, Wang SH, Chen YH, Liu JJ, Yang XM. Simultaneously Targeting Bcl-2 and Akt Pathways Reverses Resistance of Nasopharyngeal Carcinoma to TRAIL Synergistically. TUMORI JOURNAL 2018; 97:762-70. [DOI: 10.1177/030089161109700614] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Aims and Background Despite progress in treatment techniques, the five-year survival rate of nasopharyngeal carcinoma (NPC) is disappointing. Tumor necrosis factor-related apoptosis inducing ligand (TRAIL) can selectively induce apoptosis in most tumor cells while sparing normal cells. Given the antiapoptotic functions of Bcl-2 and Akt, we examined the effects of targeting these pathways alone or simultaneously on TRAIL apoptosis in NPC cell lines. Methods and Study Design We first tested the cytotoxic effect of TRAIL and the expression of death receptors, Bcl-2, Akt, and p-Akt on four NPC cell lines by MTT and Western blotting, respectively. Small interfering RNAs (siRNAs) targeting Bcl-2 and PI3–K inhibitor (LY294002) were used alone or combined with TRAIL in the cell lines and cytotoxicity was examined by MTT. Apoptosis rates, mitochondrial transmembrane potential, and apoptotic pathway signals were detected by flow cytometric analysis, DiOC6(3) assays, and Western blotting after the various combination treatments on CNE-2, the cell line that was most resistant to TRAIL. Results Although no direct correlation between the sensitivity to TRAIL and the relative expression levels of Bcl-2 and activated Akt was found in the NPC cell lines examined, siRNA mediated the downregulation of Bcl-2 and LY294002-induced inactivation of Akt, increasing the sensitivity of all examined NPC cell lines to TRAIL. Synergistic enhancement of TRAIL-mediated cytotoxicity was observed in combination treatment of Bcl-2 siRNA and LY294002 compared to cells treated with each treatment alone. The synergistic effects were mediated through increased apoptotic signaling of the mitochondrial pathway, as was evident from the more increased mitochondrial depolarization, activation of caspase-9 and caspase-3, and suppression of XIAP. Conclusions This study provides proof of principle that TRAIL combined with simultaneously targeting the Bcl-2 and Akt signaling pathways may have potential as a novel future treatment strategy for NPC.
Collapse
Affiliation(s)
- Shi-Sheng Li
- Department of Otolaryngology, Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Qing-Lai Tang
- Department of Otolaryngology, Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Shu-hui Wang
- Department of Otolaryngology, Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yue-Hong Chen
- Department of Otolaryngology, Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jia-Jia Liu
- Department of Otolaryngology, Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xin-Ming Yang
- Department of Otolaryngology, Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
45
|
Sun LF, An DQ, Niyazi GL, Ma WH, Xu ZW, Xie Y. Effects of Tianxiangdan Granule treatment on atherosclerosis via NF‑κB and p38 MAPK signaling pathways. Mol Med Rep 2017; 17:1642-1650. [PMID: 29257205 PMCID: PMC5780105 DOI: 10.3892/mmr.2017.8067] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Accepted: 04/07/2017] [Indexed: 01/26/2023] Open
Abstract
The present study aimed to determine the effects of Tianxiangdan Granule on nuclear factor (NF)-κB p65 and p38 mitogen-activated protein kinase (MAPK) inflammatory signaling pathways, and explored the possible mechanism underlying the effects of Tianxiangdan Granule on prevention and treatment of atherosclerosis. A total of 48 apolipoprotein E−/− mice (age, 8 weeks) were selected and divided into two groups: The normal control group (n=12) and the modeling group (n=36). In the modeling group, mice were fed a high-fat diet and were maintained in an artificial climate box, in order to stimulate the climate and eating habit characteristics of Xinjiang. Every morning, ApoE−/− mice in the modeling group were placed in the artificial climate box at 10:00 am and were taken out at 09:00 pm and placed back in the room temperature environment. The temperature of the artificial climate box was set at 6±2°C, relative humidity was controlled at 25–32.8% and the light-dark cycle was 12 h/day. The purpose of this method was to establish the Huizhuo Tanzu type atherosclerosis model. Following successful generation of the model, mice in the modeling group were randomly divided into three groups: Model group (n=10), Tianxiangdan group (n=10) and atorvastatin group (n=10). After 12 weeks, mice were sacrificed and the serum levels of interleukin (IL)-1β and tumor necrosis factor (TNF)-α in each group were detected. Furthermore, the expression levels of NF-κB p65 and p38 MAPK in aortic tissue were detected. The results indicated that the concentrations of IL-1β and TNF-α were significantly higher in mice in the model group compared with in the normal control group (P<0.01), whereas the concentrations of IL-1β and TNF-α were lower in the Tianxiangdan and atorvastatin groups compared with in the model group (P<0.01). Furthermore, the protein expression levels of phosphorylated (p)-NF-κB p65 and p-p38 MAPK protein were higher in aortic tissues from the model group compared with in the normal control group (P<0.01), p-NF-κB p65 and p-p38 MAPK protein expression was reduced in the atorvastatin and Tianxiangdan groups compared with in the model group. The present study indicated that the mechanism underlying the effects of Tianxiangdan Granule on the prevention and treatment of atherosclerosis may be as follows: Tianxiangdan Granule may decrease the expression of the inflammatory cytokines IL-1β and TNF-α, and suppress activation of the NF-κB p65 and p38 MAPK signaling pathways.
Collapse
Affiliation(s)
- Long-Fei Sun
- Coronary Care Unit, Affiliated Hospital of Traditional Chinese Medicine, Xinjiang Medical University, Urumqi, Xinjiang 830000, P.R. China
| | - Dong-Qing An
- Department of Traditional Chinese Medicine, College of Traditional Chinese Medicine, Xinjiang Medical University, Urumqi, Xinnjiang 830011, P.R. China
| | - Gu-Lijiamali Niyazi
- Rehabilitation Unit, Affiliated Hospital of Traditional Chinese Medicine, Xinjiang Medical University, Urumqi, Xinjiang 830000, P.R. China
| | - Wen-Hui Ma
- Department of Traditional Chinese Medicine, College of Traditional Chinese Medicine, Xinjiang Medical University, Urumqi, Xinnjiang 830011, P.R. China
| | - Zheng-Wei Xu
- Department of Traditional Chinese Medicine, College of Traditional Chinese Medicine, Xinjiang Medical University, Urumqi, Xinnjiang 830011, P.R. China
| | - Yang Xie
- Department of Traditional Chinese Medicine, College of Traditional Chinese Medicine, Xinjiang Medical University, Urumqi, Xinnjiang 830011, P.R. China
| |
Collapse
|
46
|
Keresztes A, Streicher JM. Synergistic interaction of the cannabinoid and death receptor systems - a potential target for future cancer therapies? FEBS Lett 2017; 591:3235-3251. [PMID: 28948607 DOI: 10.1002/1873-3468.12863] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 09/12/2017] [Accepted: 09/19/2017] [Indexed: 01/16/2023]
Abstract
Cannabinoid receptors have been shown to interact with other receptors, including tumor necrosis factor receptor superfamily (TNFRS) members, to induce cancer cell death. When cannabinoids and death-inducing ligands (including TNF-related apoptosis-inducing ligand) are administered together, they have been shown to synergize and demonstrate enhanced antitumor activity in vitro. Certain cannabinoid ligands have been shown to sensitize cancer cells and synergistically interact with members of the TNFRS, thus suggesting that the combination of cannabinoids with death receptor (DR) ligands induces additive or synergistic tumor cell death. This review summarizes recent findings on the interaction of the cannabinoid and DR systems and suggests possible clinical co-application of cannabinoids and DR ligands in the treatment of various malignancies.
Collapse
Affiliation(s)
- Attila Keresztes
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, USA
| | - John M Streicher
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, USA
| |
Collapse
|
47
|
Li X, Zhang X, Gong P, Xia F, Li L, Yang Z, Li J. TLR2 -/- Mice Display Decreased Severity of Giardiasis via Enhanced Proinflammatory Cytokines Production Dependent on AKT Signal Pathway. Front Immunol 2017; 8:1186. [PMID: 28979269 PMCID: PMC5611375 DOI: 10.3389/fimmu.2017.01186] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 09/07/2017] [Indexed: 02/05/2023] Open
Abstract
Giardia infection is one of the most common causes of waterborne diarrheal disease in a wide array of mammalian hosts, including humans globally. Although numerous studies have indicated that adaptive immune responses are important for Giardia defense, however, whether the host innate immune system such as TLRs recognizes Giardia remains poorly understood. TLR2 plays a crucial role in pathogen recognition, innate immunity activation, and the eventual pathogen elimination. In this study, we investigated the role of TLR2 as a non-protective inflammatory response on controlling the severity of giardiasis. RT-PCR analysis suggested that TLR2 expression was increased in vitro. We demonstrated that Giardia lamblia-induced cytokines expression by the activation of p38 and ERK pathways via TLR2. Interestingly, the expression of IL-12 p40, TNF-α, and IL-6, but not IFN-γ, was enhanced in TLR2-blocked and TLR2−/− mouse macrophages exposed to G. lamblia trophozoites compared with wild-type (WT) mouse macrophages. Further analysis demonstrated that G. lamblia trophozoites reduced cytokines secretion by activating AKT pathway in WT mouse macrophages. Immunohistochemical staining in G. lamblia cysts infected TLR2−/− and WT mice showed that TLR2 was highly expressed in duodenum in infected WT mice. Also, infected TLR2−/− and AKT-blocked mice showed an increased production of IL-12 p40 and IFN-γ compared with infected WT mice at the early stage during infection. Interestingly, infected TLR2−/− and AKT-blocked mice displayed a decreased parasite burden, an increased weight gain rate, and short parasite persistence. Histological morphometry showed shortened villus length, hyperplastic crypt and decreased ratio of villus height/crypt depth in infected WT mice compared with in infected TLR2−/− and AKT-blocked mice. Together, our results suggested that TLR2 deficiency leads to alleviation of giardiasis and reduction of parasite burden through the promotion of proinflammatory cytokines production. For the first time, our results demonstrated that TLR2 played a negative role in host defense against Giardia.
Collapse
Affiliation(s)
- Xin Li
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Xichen Zhang
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Pengtao Gong
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Feifei Xia
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Ling Li
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Zhengtao Yang
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Jianhua Li
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| |
Collapse
|
48
|
Gamie Z, Kapriniotis K, Papanikolaou D, Haagensen E, Da Conceicao Ribeiro R, Dalgarno K, Krippner-Heidenreich A, Gerrand C, Tsiridis E, Rankin KS. TNF-related apoptosis-inducing ligand (TRAIL) for bone sarcoma treatment: Pre-clinical and clinical data. Cancer Lett 2017; 409:66-80. [PMID: 28888998 DOI: 10.1016/j.canlet.2017.08.036] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 08/24/2017] [Accepted: 08/28/2017] [Indexed: 01/25/2023]
Abstract
Bone sarcomas are rare, highly malignant mesenchymal tumours that affect teenagers and young adults, as well as older patients. Despite intensive, multimodal therapy, patients with bone sarcomas have poor 5-year survival, close to 50%, with lack of improvement over recent decades. TNF-related apoptosis-inducing ligand (TRAIL), a member of the tumour necrosis factor (TNF) ligand superfamily (TNFLSF), has been found to induce apoptosis in cancer cells while sparing nontransformed cells, and may therefore offer a promising new approach to treatment. We cover the existing preclinical and clinical evidence about the use of TRAIL and other death receptor agonists in bone sarcoma treatment. In vitro studies indicate that TRAIL and other death receptor agonists are generally potent against bone sarcoma cell lines. Ewing's sarcoma cell lines present the highest sensitivity, whereas osteosarcoma and chondrosarcoma cell lines are considered less sensitive. In vivo studies also demonstrate satisfactory results, especially in Ewing's sarcoma xenograft models. However, the few clinical trials in the literature show only low or moderate efficacy of TRAIL in treating bone sarcoma. Potential strategies to overcome the in vivo resistance reported include co-administration with other drugs and the potential to deliver TRAIL on the surface of primed mesenchymal or immune cells and the use of targeted single chain antibodies such as scFv-scTRAIL.
Collapse
Affiliation(s)
- Zakareya Gamie
- Northern Institute for Cancer Research, Paul O'Gorman Building, Medical School, Newcastle University, Framlington Place, Newcastle Upon Tyne, NE2 4HH, UK.
| | - Konstantinos Kapriniotis
- Academic Orthopedic Department, "PapaGeorgiou" General Hospital, Thessaloniki, Greece; CORE-Center for Orthopedic Research at CIRI-AUTh, Aristotle University Medical School, Thessaloniki, Hellas, Greece.
| | - Dimitra Papanikolaou
- Academic Orthopedic Department, "PapaGeorgiou" General Hospital, Thessaloniki, Greece; CORE-Center for Orthopedic Research at CIRI-AUTh, Aristotle University Medical School, Thessaloniki, Hellas, Greece.
| | - Emma Haagensen
- Northern Institute for Cancer Research, Paul O'Gorman Building, Medical School, Newcastle University, Framlington Place, Newcastle Upon Tyne, NE2 4HH, UK.
| | - Ricardo Da Conceicao Ribeiro
- School of Mechanical and Systems Engineering, Stephenson Building, Claremont Road, Newcastle Upon Tyne, NE1 7RU, UK.
| | - Kenneth Dalgarno
- School of Mechanical and Systems Engineering, Stephenson Building, Claremont Road, Newcastle Upon Tyne, NE1 7RU, UK.
| | - Anja Krippner-Heidenreich
- Northern Institute for Cancer Research, Paul O'Gorman Building, Medical School, Newcastle University, Framlington Place, Newcastle Upon Tyne, NE2 4HH, UK.
| | - Craig Gerrand
- North of England Bone and Soft Tissue Tumour Service, Freeman Hospital, Freeman Road, Newcastle Upon Tyne, NE7 7DN, UK.
| | - Eleftherios Tsiridis
- Academic Orthopedic Department, "PapaGeorgiou" General Hospital, Thessaloniki, Greece; CORE-Center for Orthopedic Research at CIRI-AUTh, Aristotle University Medical School, Thessaloniki, Hellas, Greece; Secretary General European Hip Society, Austria.
| | - Kenneth Samora Rankin
- Northern Institute for Cancer Research, Paul O'Gorman Building, Medical School, Newcastle University, Framlington Place, Newcastle Upon Tyne, NE2 4HH, UK.
| |
Collapse
|
49
|
Liu H, Su D, Zhang J, Ge S, Li Y, Wang F, Gravel M, Roulston A, Song Q, Xu W, Liang JG, Shore G, Wang X, Liang P. Improvement of Pharmacokinetic Profile of TRAIL via Trimer-Tag Enhances its Antitumor Activity in vivo. Sci Rep 2017; 7:8953. [PMID: 28827692 PMCID: PMC5566391 DOI: 10.1038/s41598-017-09518-1] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Accepted: 07/24/2017] [Indexed: 12/11/2022] Open
Abstract
TNF-related apoptosis-inducing ligand (TRAIL/Apo2L) has long been considered a tantalizing target for cancer therapy because it mediates activation of the extrinsic apoptosis pathway in a tumor-specific manner by binding to and trimerizing its functional receptors DR4 or DR5. Despite initial promise, both recombinant human TRAIL (native TRAIL) and dimeric DR4/DR5 agonist monoclonal antibodies (mAbs) failed in multiple human clinical trials. Here we show that in-frame fusion of human C-propeptide of α1(I) collagen (Trimer-Tag) to the C-terminus of mature human TRAIL leads to a disulfide bond-linked homotrimer which can be expressed at high levels as a secreted protein from CHO cells. The resulting TRAIL-Trimer not only retains similar bioactivity and receptor binding kinetics as native TRAIL in vitro which are 4-5 orders of magnitude superior to that of dimeric TRAIL-Fc, but also manifests more favorable pharmacokinetic and antitumor pharmacodynamic profiles in vivo than that of native TRAIL. Taken together, this work provides direct evidence for the in vivo antitumor efficacy of TRAIL being proportional to systemic drug exposure and suggests that the previous clinical failures may have been due to rapid systemic clearance of native TRAIL and poor apoptosis-inducing potency of dimeric agonist mAbs despite their long serum half-lives.
Collapse
Affiliation(s)
- Haipeng Liu
- Department of Biochemistry & Molecular Biology, College of Life Sciences, Sichuan University, Chengdu, China
| | - Danmei Su
- Department of Biochemistry & Molecular Biology, College of Life Sciences, Sichuan University, Chengdu, China
| | - Jinlong Zhang
- Department of Biochemistry & Molecular Biology, College of Life Sciences, Sichuan University, Chengdu, China
| | - Shuaishuai Ge
- Department of Biochemistry & Molecular Biology, College of Life Sciences, Sichuan University, Chengdu, China
| | - Youwei Li
- Department of Biochemistry & Molecular Biology, College of Life Sciences, Sichuan University, Chengdu, China
| | - Fei Wang
- Department of Biochemistry & Molecular Biology, College of Life Sciences, Sichuan University, Chengdu, China
| | - Michel Gravel
- Laboratory for Therapeutic Development, Rosalind and Morris Goodman Cancer Research Centre, McGill University, Montreal (QC), Canada
| | - Anne Roulston
- Laboratory for Therapeutic Development, Rosalind and Morris Goodman Cancer Research Centre, McGill University, Montreal (QC), Canada
| | - Qin Song
- Clover Biopharmaceuticals, Chengdu, China
| | - Wei Xu
- Clover Biopharmaceuticals, Chengdu, China
| | | | - Gordon Shore
- Laboratory for Therapeutic Development, Rosalind and Morris Goodman Cancer Research Centre, McGill University, Montreal (QC), Canada
| | - Xiaodong Wang
- National Institute of Biological Sciences, Beijing, China
| | - Peng Liang
- Department of Biochemistry & Molecular Biology, College of Life Sciences, Sichuan University, Chengdu, China.
- Clover Biopharmaceuticals, Chengdu, China.
- GenHunter Corporation, 624 Grassmere Park, Nashville, TN, 37211, USA.
| |
Collapse
|
50
|
von Karstedt S, Montinaro A, Walczak H. Exploring the TRAILs less travelled: TRAIL in cancer biology and therapy. Nat Rev Cancer 2017; 17:352-366. [PMID: 28536452 DOI: 10.1038/nrc.2017.28] [Citation(s) in RCA: 406] [Impact Index Per Article: 50.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The discovery that the tumour necrosis factor-related apoptosis-inducing ligand (TRAIL) can induce apoptosis of cancer cells without causing toxicity in mice has led to the in-depth study of pro-apoptotic TRAIL receptor (TRAIL-R) signalling and the development of biotherapeutic drug candidates that activate TRAIL-Rs. The outcome of clinical trials with these TRAIL-R agonists has, however, been disappointing so far. Recent evidence indicates that many cancers, in addition to being TRAIL resistant, use the endogenous TRAIL-TRAIL-R system to their own advantage. However, novel insight on two fronts - how resistance of cancer cells to TRAIL-based pro-apoptotic therapies might be overcome, and how the pro-tumorigenic effects of endogenous TRAIL might be countered - gives reasonable hope that the TRAIL system can be harnessed to treat cancer. In this Review we assess the status quo of our understanding of the biology of the TRAIL-TRAIL-R system - as well as the gaps therein - and discuss the opportunities and challenges in effectively targeting this pathway.
Collapse
Affiliation(s)
- Silvia von Karstedt
- Centre for Cell Death, Cancer and Inflammation, UCL Cancer Institute, University College London, 72 Huntley Street, London WC1E 6DD, UK
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Antonella Montinaro
- Centre for Cell Death, Cancer and Inflammation, UCL Cancer Institute, University College London, 72 Huntley Street, London WC1E 6DD, UK
| | - Henning Walczak
- Centre for Cell Death, Cancer and Inflammation, UCL Cancer Institute, University College London, 72 Huntley Street, London WC1E 6DD, UK
| |
Collapse
|