1
|
Wang Q, Sun J, Jiang H, Yu M. Emerging roles of extracellular vesicles in oral and maxillofacial areas. Int J Oral Sci 2025; 17:11. [PMID: 39900916 PMCID: PMC11791077 DOI: 10.1038/s41368-024-00341-9] [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: 05/03/2024] [Revised: 12/02/2024] [Accepted: 12/03/2024] [Indexed: 02/05/2025] Open
Abstract
The oral and maxillofacial region is a highly complex area composed of multiple tissue types and bears various critical functions of the human body. Diseases in this region pose significant diagnostic and management challenges; therefore, exploring new strategies for early diagnosis, targeted treatment, and tissue reconstruction is key to improving patient prognosis and quality of life. Extracellular vesicles are a group of heterogeneous lipid-bilayer membrane structures secreted by most cell types, including exosomes, microvesicles, and apoptotic bodies. Present in various body fluids and tissues, they act as messengers via the transfer of nucleic acids, proteins, and metabolites to recipient cells. To date, studies have revealed the different roles of extracellular vesicles in physiological or pathological processes, as well as applications in disease diagnosis, prognosis, and treatment. The importance and tissue specificity of the dental and maxillofacial tissues indicate that extracellular vesicles derived from this region are promising for further research. This paper reviews the published data on extracellular vesicles derived from cells, body fluids, and tissues in oral and maxillofacial regions, summarizes the latest advances in extracellular vesicles from extensive sources, and concludes with a focus on the current research progress and application prospects of engineered exosomes in oral science.
Collapse
Affiliation(s)
- Qianting Wang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of the Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, China
| | - Jiayu Sun
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of the Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, China
| | - Haci Jiang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of the Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, China
| | - Mengfei Yu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of the Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, China.
| |
Collapse
|
2
|
Farooqi AA, Turgambayeva A, Kamalbekova G, Suleimenova R, Latypova N, Ospanova S, Ospanova D, Abdikadyr Z, Zhussupov S. TRAIL as a Warrior in Nano-Sized Trojan Horse: Anticancer and Anti-Metastatic Effects of Nano-Formulations of TRAIL in Cell Culture and Animal Model Studies. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1977. [PMID: 39768856 PMCID: PMC11677168 DOI: 10.3390/medicina60121977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 11/17/2024] [Accepted: 11/28/2024] [Indexed: 01/11/2025]
Abstract
Cancer is a therapeutically challenging and genomically complicated disease. Pioneering studies have uncovered multifaceted aspects of cancer, ranging from intra- and inter-tumor heterogeneity, drug resistance, and genetic/epigenetic mutations. Loss of apoptosis is another critical aspect that makes cancer cells resistant to death. A substantial fraction of mechanistic information gleaned from cutting-edge studies has enabled researchers to develop near-to-complete resolution of the apoptotic pathway. Within the exciting frontiers of apoptosis, TRAIL (tumor necrosis factor-related apoptosis-inducing ligand) has garnered phenomenal appreciation by interdisciplinary researchers principally because of its unique capability to target cancer cells. TRAIL-based monotherapies and combinatorial therapies have reached phase II and phase III clinical trials. Rapidly upgrading the list of clinical trials substantiates the clinically valuable role of TRAIL-based therapeutics in cancer therapy. However, there is a growing concern about the poor bioavailability and rapid clearance of TRAIL-based therapeutics. Excitingly, the charismatic field of nanotechnology offers solutions for different problems, and we have witnessed remarkable breakthroughs in the efficacy of TRAIL-based therapeutics using nanotechnological approaches. In this review, we have attempted to provide a summary about different nanotechnologically assisted delivery methods for TRAIL-based therapeutics in cell culture studies and animal model studies for the inhibition/prevention of cancer.
Collapse
Affiliation(s)
- Ammad Ahmad Farooqi
- Department of Molecular Oncology, Institute of Biomedical and Genetic Engineering (IBGE), Islamabad 44090, Pakistan
| | - Assiya Turgambayeva
- Department of Public Health and Management, Astana Medical University, Astana 010000, Kazakhstan; (A.T.); (S.O.)
| | - Gulnara Kamalbekova
- Department of Family Medicine, Astana Medical University, Astana 010000, Kazakhstan; (G.K.); (N.L.)
| | - Roza Suleimenova
- Department of Public Health and Hygiene, Astana Medical University, Astana 010000, Kazakhstan;
| | - Natalya Latypova
- Department of Family Medicine, Astana Medical University, Astana 010000, Kazakhstan; (G.K.); (N.L.)
| | - Sholpan Ospanova
- Department of Public Health and Management, Astana Medical University, Astana 010000, Kazakhstan; (A.T.); (S.O.)
| | - Dinara Ospanova
- Faculty of Medicine and Healthcare, Al-Farabi Kazakh National University, 71 Al-Farabi Ave, Almaty 050040, Kazakhstan;
| | - Zhanat Abdikadyr
- Department of Biostatistics, Bioinformatics and Information Technologies, Astana Medical University, Astana 010000, Kazakhstan;
| | - Sabit Zhussupov
- Department of Surgery, Semey Medical University, Semey 071400, Kazakhstan;
| |
Collapse
|
3
|
Xu X, Xu L, Wen C, Xia J, Zhang Y, Liang Y. Programming assembly of biomimetic exosomes: An emerging theranostic nanomedicine platform. Mater Today Bio 2023; 22:100760. [PMID: 37636982 PMCID: PMC10450992 DOI: 10.1016/j.mtbio.2023.100760] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 08/03/2023] [Accepted: 08/04/2023] [Indexed: 08/29/2023] Open
Abstract
Exosomes have emerged as a promising cell-free therapeutic approach. However, challenges in large-scale production, quality control, and heterogeneity must be overcome before they can be used clinically. Biomimetic exosomes containing key components of natural exosomes have been assembled through extrusion, artificial synthesis, and liposome fusion to address these limitations. These exosome-mimetics (EMs) possess similar morphology and function but provide higher yields, faster large-scale production, and similar size compared to conventional exosomes. This article provides an overview of the chemical and biological properties of various synthetic exosome systems, including nanovesicles (NVs), EMs, and hybrid exosomes. We highlight recent advances in the production and applications of nanobiotechnology and discuss the advantages, limitations, and potential clinical applications of programming assembly of exosome mimetics.
Collapse
Affiliation(s)
- Xiao Xu
- Department of Joint Surgery and Sports Medicine, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, 272029, China
| | - Limei Xu
- Department of Hematology, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, 272029, China
| | - Caining Wen
- Department of Joint Surgery and Sports Medicine, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, 272029, China
| | - Jiang Xia
- Department of Chemistry, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Yuanmin Zhang
- Department of Joint Surgery and Sports Medicine, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, 272029, China
- Jining Medical University, Jining, Shandong, 272067, China
| | - Yujie Liang
- Department of Joint Surgery and Sports Medicine, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, 272029, China
- Jining Medical University, Jining, Shandong, 272067, China
| |
Collapse
|
4
|
Marco-Brualla J, de Miguel D, Martínez-Lostao L, Anel A. DR5 Up-Regulation Induced by Dichloroacetate Sensitizes Tumor Cells to Lipid Nanoparticles Decorated with TRAIL. J Clin Med 2023; 12:jcm12020608. [PMID: 36675536 PMCID: PMC9864242 DOI: 10.3390/jcm12020608] [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: 12/13/2022] [Revised: 01/04/2023] [Accepted: 01/10/2023] [Indexed: 01/14/2023] Open
Abstract
Cancer resistance to treatments is a challenge that researchers constantly seek to overcome. For instance, TNF-related apoptosis-inducing ligand (TRAIL) is a potential good prospect as an anti-cancer therapy, as it attacks tumor cells but not normal cells. However, treatments based in soluble TRAIL provided incomplete clinical results and diverse formulations have been developed to improve its bioactivity. In previous works, we generated a new TRAIL formulation based in its attachment to the surface of unilamellar nanoliposomes (LUV-TRAIL). This formulation greatly increased apoptosis in a wide selection of tumor cell types, albeit a few of them remained resistant. On the other hand, it has been described that a metabolic shift in cancer cells can also alter its sensitivity to other treatments. In this work, we sought to increase the sensitivity of several tumor cell types resistant to LUV-TRAIL by previous exposure to the metabolic drug dichloroacetate (DCA), which forces oxidative phosphorylation. Results showed that DCA + LUV-TRAIL had a synergistic effect on both lung adenocarcinoma A549, colorectal HT29, and breast cancer MCF7 cells. Despite DCA inducing intracellular changes in a cell-type specific way, the increase in cell death by apoptosis was clearly correlated with an increase in death receptor 5 (DR5) surface expression in all cell lines. Therefore, DCA-induced metabolic shift emerges as a suitable option to overcome TRAIL resistance in cancer cells.
Collapse
Affiliation(s)
- Joaquín Marco-Brualla
- Apoptosis, Immunity and Cancer Group, Department of Biochemistry and Molecular and Cell Biology, Aragon Health Research Institute (IIS-Aragón) & University of Zaragoza, 50009 Zargoza, Spain
| | - Diego de Miguel
- Apoptosis, Immunity and Cancer Group, Department of Biochemistry and Molecular and Cell Biology, Aragon Health Research Institute (IIS-Aragón) & University of Zaragoza, 50009 Zargoza, Spain
| | | | - Alberto Anel
- Apoptosis, Immunity and Cancer Group, Department of Biochemistry and Molecular and Cell Biology, Aragon Health Research Institute (IIS-Aragón) & University of Zaragoza, 50009 Zargoza, Spain
- Correspondence:
| |
Collapse
|
5
|
Razeghian E, Suksatan W, Sulaiman Rahman H, Bokov DO, Abdelbasset WK, Hassanzadeh A, Marofi F, Yazdanifar M, Jarahian M. Harnessing TRAIL-Induced Apoptosis Pathway for Cancer Immunotherapy and Associated Challenges. Front Immunol 2021; 12:699746. [PMID: 34489946 PMCID: PMC8417882 DOI: 10.3389/fimmu.2021.699746] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Accepted: 08/05/2021] [Indexed: 01/04/2023] Open
Abstract
The immune cytokine tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) has attracted rapidly evolving attention as a cancer treatment modality because of its competence to selectively eliminate tumor cells without instigating toxicity in vivo. TRAIL has revealed encouraging promise in preclinical reports in animal models as a cancer treatment option; however, the foremost constraint of the TRAIL therapy is the advancement of TRAIL resistance through a myriad of mechanisms in tumor cells. Investigations have documented that improvement of the expression of anti-apoptotic proteins and survival or proliferation involved signaling pathways concurrently suppressing the expression of pro-apoptotic proteins along with down-regulation of expression of TRAILR1 and TRAILR2, also known as death receptor 4 and 5 (DR4/5) are reliable for tumor cells resistance to TRAIL. Therefore, it seems that the development of a therapeutic approach for overcoming TRAIL resistance is of paramount importance. Studies currently have shown that combined treatment with anti-tumor agents, ranging from synthetic agents to natural products, and TRAIL could result in induction of apoptosis in TRAIL-resistant cells. Also, human mesenchymal stem/stromal cells (MSCs) engineered to generate and deliver TRAIL can provide both targeted and continued delivery of this apoptosis-inducing cytokine. Similarly, nanoparticle (NPs)-based TRAIL delivery offers novel platforms to defeat barricades to TRAIL therapeutic delivery. In the current review, we will focus on underlying mechanisms contributed to inducing resistance to TRAIL in tumor cells, and also discuss recent findings concerning the therapeutic efficacy of combined treatment of TRAIL with other antitumor compounds, and also TRAIL-delivery using human MSCs and NPs to overcome tumor cells resistance to TRAIL.
Collapse
Affiliation(s)
- Ehsan Razeghian
- Human Genetics Division, Medical Biotechnology Department, National Institute of Genetics Engineering and Biotechnology (NIGEB), Tehran, Iran
| | - Wanich Suksatan
- Faculty of Nursing, HRH Princess Chulabhorn College of Medical Science, Chulabhorn Royal Academy, Bangkok, Thailand
| | - Heshu Sulaiman Rahman
- Department of Physiology, College of Medicine, University of Suleimanyah, Suleimanyah, Iraq
- Department of Medical Laboratory Sciences, Komar University of Science and Technology, Sulaimaniyah, Iraq
| | - Dmitry O. Bokov
- Institute of Pharmacy, Sechenov First Moscow State Medical University, Moscow, Russia
- Laboratory of Food Chemistry, Federal Research Center of Nutrition, Biotechnology and Food Safety, Moscow, Russia
| | - Walid Kamal Abdelbasset
- Department of Health and Rehabilitation Sciences, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al Kharj, Saudi Arabia
- Department of Physical Therapy, Kasr Al-Aini Hospital, Cairo University, Giza, Egypt
| | - Ali Hassanzadeh
- Department of Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Faroogh Marofi
- Immunology Research Center (IRC), Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahboubeh Yazdanifar
- Stem Cell Transplantation and Regenerative Medicine, Department of Pediatrics, Stanford University School of Medicine, Palo Alto, CA, United States
| | - Mostafa Jarahian
- Toxicology and Chemotherapy Unit (G401), German Cancer Research Center, Heidelberg, Germany
| |
Collapse
|
6
|
Villarreal-Leal RA, Cooke JP, Corradetti B. Biomimetic and immunomodulatory therapeutics as an alternative to natural exosomes for vascular and cardiac applications. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2021; 35:102385. [PMID: 33774130 PMCID: PMC8238887 DOI: 10.1016/j.nano.2021.102385] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 02/21/2021] [Accepted: 03/03/2021] [Indexed: 02/07/2023]
Abstract
Inflammation is a central mechanism in cardiovascular diseases (CVD), where sustained oxidative stress and immune responses contribute to cardiac remodeling and impairment. Exosomes are extracellular vesicles released by cells to communicate with their surroundings and to modulate the tissue microenvironment. Recent evidence indicates their potential as cell-free immunomodulatory therapeutics for CVD, preventing cell death and fibrosis while inducing wound healing and angiogenesis. Biomimetic exosomes are semi-synthetic particles engineered using essential moieties present in natural exosomes (lipids, RNA, proteins) to reproduce their therapeutic effects while improving on scalability and standardization due to the ample range of moieties available to produce them. In this review, we provide an up-to-date description of the use of exosomes for CVD and offer our vision on the areas of opportunity for the development of biomimetic strategies. We also discuss the current limitations to overcome in the process towards their translation into clinic.
Collapse
Affiliation(s)
- Ramiro A Villarreal-Leal
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey, Nuevo León, Mexico
| | - John P Cooke
- RNA Therapeutics Program, Department of Cardiovascular Sciences (R.S., J.P.C.), Houston Methodist Research Institute, TX, USA; Houston Methodist DeBakey Heart and Vascular Center (J.P.C.), Houston Methodist Hospital, TX, USA
| | - Bruna Corradetti
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, USA; Center of NanoHealth, Swansea University Medical School, Swansea, UK.
| |
Collapse
|
7
|
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
|
8
|
Importance of TRAIL Molecular Anatomy in Receptor Oligomerization and Signaling. Implications for Cancer Therapy. Cancers (Basel) 2019; 11:cancers11040444. [PMID: 30934872 PMCID: PMC6521207 DOI: 10.3390/cancers11040444] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [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: 12/12/2022] Open
Abstract
(TNF)-related apoptosis-inducing ligand (TRAIL) is able to activate the extrinsic apoptotic pathway upon binding to DR4/TRAIL-R1 and/or DR5/TRAIL-R2 receptors. Structural data indicate that TRAIL functions as a trimer that can engage three receptor molecules simultaneously, resulting in receptor trimerization and leading to conformational changes in TRAIL receptors. However, receptor conformational changes induced by the binding of TRAIL depend on the molecular form of this death ligand, and not always properly trigger the apoptotic cascade. In fact, TRAIL exhibits a much stronger pro-apoptotic activity when is found as a transmembrane protein than when it occurs as a soluble form and this enhanced biological activity is directly linked to its ability to cluster TRAIL receptors in supra-molecular structures. In this regard, cells involved in tumor immunosurveillance, such as activated human T cells, secrete endogenous TRAIL as a transmembrane protein associated with lipid microvesicles called exosomes upon T-cell reactivation. Consequently, it seems clear that a proper oligomerization of TRAIL receptors, which leads to a strong apoptotic signaling, is crucial for inducing apoptosis in cancer cells upon TRAIL treatment. In this review, the current knowledge of oligomerization status of TRAIL receptors is discussed as well as the implications for cancer treatment when using TRAIL-based therapies.
Collapse
|
9
|
Wen Z, Liu F, Chen Q, Xu Y, Li H, Sun S. Recent development in biodegradable nanovehicle delivery system-assisted immunotherapy. Biomater Sci 2019; 7:4414-4443. [DOI: 10.1039/c9bm00961b] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
A schematic illustration of BNDS biodegradation and release antigen delivery for assisting immunotherapy.
Collapse
Affiliation(s)
- Zhenfu Wen
- Shaanxi Key Laboratory of Natural Products & Chemical Biology
- College of Chemistry & Pharmacy
- Northwest A&F University
- Yangling
- P. R. China
| | - Fengyu Liu
- State Key Laboratory of Fine Chemicals
- School of Chemistry
- Dalian University of Technology
- Ganjingzi District
- P. R. China
| | | | - Yongqian Xu
- Shaanxi Key Laboratory of Natural Products & Chemical Biology
- College of Chemistry & Pharmacy
- Northwest A&F University
- Yangling
- P. R. China
| | - Hongjuan Li
- Shaanxi Key Laboratory of Natural Products & Chemical Biology
- College of Chemistry & Pharmacy
- Northwest A&F University
- Yangling
- P. R. China
| | - Shiguo Sun
- Shaanxi Key Laboratory of Natural Products & Chemical Biology
- College of Chemistry & Pharmacy
- Northwest A&F University
- Yangling
- P. R. China
| |
Collapse
|
10
|
Shamili FH, Bayegi HR, Salmasi Z, Sadri K, Mahmoudi M, Kalantari M, Ramezani M, Abnous K. Exosomes derived from TRAIL-engineered mesenchymal stem cells with effective anti-tumor activity in a mouse melanoma model. Int J Pharm 2018; 549:218-229. [PMID: 30075248 DOI: 10.1016/j.ijpharm.2018.07.067] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 07/24/2018] [Accepted: 07/30/2018] [Indexed: 02/07/2023]
Abstract
Exosomes are biological nano-sized vesicles (~30-200 nm in diameter) that are produced by a wide range of cells and play several roles in cell-cell communications. These vesicles contain membrane and cytoplasmic components of producing cells. Mesenchymal stem cells (MSCs) are the ideal producer of exosomes. The secreted vesicles from MSCs are promising biological vehicles for cell-free therapy in regenerative medicine, cancer therapy and targeted delivery of therapeutic agents to the tumor cells. Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is a promising member of the TNF family with selective effect on cancerous cells. Recent evidences showed that the membrane TRAIL-armed exosomes possess anti-tumor activity. However, the effect of in vivo administration of TRAIL-armed exosomes has not been reported so far. In the current study, mesenchymal stem cells expressing TRAIL/GFP proteins were prepared with the help of a non-viral vector based on polyethylenimine 25 kDa. Then, exosomes containing TRAIL protein (Exo-TRAIL) were isolated from the supernatant of genetically engineered MSCs and characterized. Antitumor activity of both MSC-derived exosomes and Exo-TRAIL was investigated in vitro and in vivo in three models. The results indicated that the co-injection of both Exo-TRAIL and tumor cells delayed the tumor appearance. Besides, the tumor volume/weight was efficiently decreased in tumor bearing mice. Moreover, it was shown that multi-dose injections of Exo-TRAIL reduced the tumor size while single dose treatment with Exo-TRAIL did not show significant anti-tumor activity. To conclude, these results suggested that MSC-derived Exo-TRAIL has a potential capacity for cancer treatment. [corrected].
Collapse
Affiliation(s)
- Fazileh Hosseini Shamili
- Department of Immunology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Houshang Rafatpanah Bayegi
- Department of Immunology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Immunology Research Center, Inflammation and Inflammatory Diseases Division, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Zahra Salmasi
- Nanotechnology research center, Pharmaceutical technology institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Kayvan Sadri
- Nuclear Medicine Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahmoud Mahmoudi
- Department of Immunology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahmoudreza Kalantari
- Department of Pathology, Faculty of Medicine, Ghaem Hospital, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Ramezani
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Khalil Abnous
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
11
|
Shi Y, Pang X, Wang J, Liu G. NanoTRAIL-Oncology: A Strategic Approach in Cancer Research and Therapy. Adv Healthc Mater 2018. [PMID: 29527836 DOI: 10.1002/adhm.201800053] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
TRAIL is a member of the tumor necrosis factor superfamily that can largely trigger apoptosis in a wide variety of cancer cells, but not in normal cells. However, insufficient exposure to cancer tissues or cells and drug resistance has severely impeded the clinical application of TRAIL. Recently, nanobiotechnology has brought about a revolution in advanced drug delivery for enhanced anticancer therapy using TRAIL. With the help of materials science, immunology, genetic engineering, and protein engineering, substantial progress is made by expressing fusion proteins with TRAIL, engineering TRAIL on biological membranes, and loading TRAIL into functional nanocarriers or conjugating it onto their surfaces. Thus, the nanoparticle-based TRAIL (nanoTRAIL) opens up intriguing opportunities for efficient and safe bioapplications. In this review, the mechanisms of action and biological function of TRAIL, as well as the current status of TRAIL treatment, are comprehensively discussed. The application of functional nanotechnology combined with TRAIL in cancer therapy is also discussed.
Collapse
Affiliation(s)
- Yesi Shi
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics and Center for Molecular Imaging and Translational Medicine; School of Public Health; Xiamen University; Xiamen 361102 China
| | - Xin Pang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics and Center for Molecular Imaging and Translational Medicine; School of Public Health; Xiamen University; Xiamen 361102 China
| | - Junqing Wang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics and Center for Molecular Imaging and Translational Medicine; School of Public Health; Xiamen University; Xiamen 361102 China
- Collaborative Innovation Center of Guangxi Biological Medicine and the; Medical and Scientific Research Center; Guangxi Medical University; Nanning 530021 China
| | - Gang Liu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics and Center for Molecular Imaging and Translational Medicine; School of Public Health; Xiamen University; Xiamen 361102 China
| |
Collapse
|
12
|
Gallego-Lleyda A, De Miguel D, Anel A, Martinez-Lostao L. Lipid Nanoparticles Decorated with TNF-Related Aptosis-Inducing Ligand (TRAIL) Are More Cytotoxic than Soluble Recombinant TRAIL in Sarcoma. Int J Mol Sci 2018; 19:ijms19051449. [PMID: 29757258 PMCID: PMC5983602 DOI: 10.3390/ijms19051449] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2018] [Revised: 04/29/2018] [Accepted: 05/11/2018] [Indexed: 01/25/2023] Open
Abstract
Sarcomas are rare and heterogeneous cancers classically associated with a poor outcome. Sarcomas are 1% of the cancer but recent estimations indicate that sarcomas account for 2% of the estimated cancer-related deaths. Traditional treatment with surgery, radiotherapy, and chemotherapy has improved the outcome for some types of sarcomas. However, novel therapeutic strategies to treat sarcomas are necessary. TNF-related apoptosis-inducing ligand (TRAIL) is a death ligand initially described as capable of inducing apoptosis on tumor cell while sparing normal cells. Only few clinical trials have used TRAIL-based treatments in sarcoma, but they show only low or moderate efficacy of TRAIL. Consequently, novel TRAIL formulations with an improved TRAIL bioactivity are necessary. Our group has developed a novel TRAIL formulation based on tethering this death ligand on a lipid nanoparticle surface (LUV-TRAIL) resembling the physiological secretion of TRAIL as a trasmembrane protein inserted into the membrane of exosomes. We have already demonstrated that LUV-TRAIL shows an improved cytotoxic activity when compared to soluble recombinant TRAIL both in hematological malignancies and epithelial-derived cancers. In the present study, we have tested LUV-TRAIL in several human sarcoma tumor cell lines with different sensitivity to soluble recombinant TRAIL, finding that LUV-TRAIL was more efficient than soluble recombinant TRAIL. Moreover, combined treatment of LUV-TRAIL with distinct drugs proved to be especially effective, sensitizing even more resistant cell lines to TRAIL.
Collapse
Affiliation(s)
- Ana Gallego-Lleyda
- Departamento de Bioquímica, Biología Molecular y Celular, Universidad de Zaragoza, 50009 Zaragoza, Spain.
- Instituto de Investigación Sanitaria de Aragón (ISS), 50009 Zaragoza, Spain.
| | - Diego De Miguel
- Departamento de Bioquímica, Biología Molecular y Celular, Universidad de Zaragoza, 50009 Zaragoza, Spain.
- Cell Death, Cancer and Inflammation, University College of London, London WC1E 6BT, UK.
| | - Alberto Anel
- Departamento de Bioquímica, Biología Molecular y Celular, Universidad de Zaragoza, 50009 Zaragoza, Spain.
- Instituto de Investigación Sanitaria de Aragón (ISS), 50009 Zaragoza, Spain.
| | - Luis Martinez-Lostao
- Instituto de Investigación Sanitaria de Aragón (ISS), 50009 Zaragoza, Spain.
- Servicio de Inmunología, Hospital Clínico Universitario Lozano Blesa, 50009 Zaragoza, Spain.
- Departamento de Microbiología, Medicina Preventiva y Salud Pública, Universidad de Zaragoza, 50009 Zaragoza, Spain.
- Instituto de Nanociencia de Aragón, 50009 Zaragoza, Spain.
| |
Collapse
|
13
|
García-Manrique P, Matos M, Gutiérrez G, Pazos C, Blanco-López MC. Therapeutic biomaterials based on extracellular vesicles: classification of bio-engineering and mimetic preparation routes. J Extracell Vesicles 2018; 7:1422676. [PMID: 29372017 PMCID: PMC5774402 DOI: 10.1080/20013078.2017.1422676] [Citation(s) in RCA: 134] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Accepted: 12/22/2017] [Indexed: 12/19/2022] Open
Abstract
Extracellular vesicles (EVs) are emerging as novel theranostic tools. Limitations related to clinical uses are leading to a new research area on design and manufacture of artificial EVs. Several strategies have been reported in order to produce artificial EVs, but there has not yet been a clear criterion by which to differentiate these novel biomaterials. In this paper, we suggest for the first time a systematic classification of the terms used to build up the artificial EV landscape, based on the preparation method. This could be useful to guide the derivation to clinical trial routes and to clarify the literature. According to our classification, we have reviewed the main strategies reported to date for their preparation, including key points such as: cargo loading, surface targeting strategies, purification steps, generation of membrane fragments for the construction of biomimetic materials, preparation of synthetic membranes inspired in EV composition and subsequent surface decoration.
Collapse
Affiliation(s)
- Pablo García-Manrique
- Department of Physical and Analytical Chemistry, University of Oviedo, Oviedo, Spain
- Department of Chemical and Environmental Engineering, University of Oviedo, Oviedo, Spain
| | - María Matos
- Department of Chemical and Environmental Engineering, University of Oviedo, Oviedo, Spain
| | - Gemma Gutiérrez
- Department of Chemical and Environmental Engineering, University of Oviedo, Oviedo, Spain
| | - Carmen Pazos
- Department of Chemical and Environmental Engineering, University of Oviedo, Oviedo, Spain
| | | |
Collapse
|
14
|
Virumbrales-Muñoz M, Ayuso JM, Olave M, Monge R, de Miguel D, Martínez-Lostao L, Le Gac S, Doblare M, Ochoa I, Fernandez LJ. Multiwell capillarity-based microfluidic device for the study of 3D tumour tissue-2D endothelium interactions and drug screening in co-culture models. Sci Rep 2017; 7:11998. [PMID: 28931839 PMCID: PMC5607255 DOI: 10.1038/s41598-017-12049-4] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Accepted: 08/31/2017] [Indexed: 12/12/2022] Open
Abstract
The tumour microenvironment is very complex, and essential in tumour development and drug resistance. The endothelium is critical in the tumour microenvironment: it provides nutrients and oxygen to the tumour and is essential for systemic drug delivery. Therefore, we report a simple, user-friendly microfluidic device for co-culture of a 3D breast tumour model and a 2D endothelium model for cross-talk and drug delivery studies. First, we demonstrated the endothelium was functional, whereas the tumour model exhibited in vivo features, e.g., oxygen gradients and preferential proliferation of cells with better access to nutrients and oxygen. Next, we observed the endothelium structure lost its integrity in the co-culture. Following this, we evaluated two drug formulations of TRAIL (TNF-related apoptosis inducing ligand): soluble and anchored to a LUV (large unilamellar vesicle). Both diffused through the endothelium, LUV-TRAIL being more efficient in killing tumour cells, showing no effect on the integrity of endothelium. Overall, we have developed a simple capillary force-based microfluidic device for 2D and 3D cell co-cultures. Our device allows high-throughput approaches, patterning different cell types and generating gradients without specialised equipment. We anticipate this microfluidic device will facilitate drug screening in a relevant microenvironment thanks to its simple, effective and user-friendly operation.
Collapse
Affiliation(s)
- María Virumbrales-Muñoz
- Department of Biomedical Engineering, Wisconsin Institutes for Medical Research, University of Wisconsin-Madison, 1111 Highland Avenue, Madison, 53705, Wisconsin, United States
| | - José María Ayuso
- Department of Biomedical Engineering, Wisconsin Institutes for Medical Research, University of Wisconsin-Madison, 1111 Highland Avenue, Madison, 53705, Wisconsin, United States.,Medical Engineering, Morgridge Institute for Research, 330 N Orchard Street, Madison, 53715, Wisconsin, United States
| | - Marta Olave
- Group of Applied Mechanics and Bioengineering (AMB), Centro de Investigación Biomédica en Red. Bioingenieria, biomateriales y nanomedicina (CIBER-BBN), Mariano Esquillor Gómez, Zaragoza, 50018, Spain.,Aragon Institute of Engineering Research (I3A), University of Zaragoza, Mariano Esquillor Gómez, Zaragoza, 50009, Spain.,Aragon Institute of Biomedical Research, Instituto de Salud Carlos III, Mariano Esquillor Gómez, Zaragoza, 50009, Spain
| | - Rosa Monge
- Group of Applied Mechanics and Bioengineering (AMB), Centro de Investigación Biomédica en Red. Bioingenieria, biomateriales y nanomedicina (CIBER-BBN), Mariano Esquillor Gómez, Zaragoza, 50018, Spain.,Aragon Institute of Engineering Research (I3A), University of Zaragoza, Mariano Esquillor Gómez, Zaragoza, 50009, Spain.,Aragon Institute of Biomedical Research, Instituto de Salud Carlos III, Mariano Esquillor Gómez, Zaragoza, 50009, Spain.,BEONCHIP S.L., Mariano Esquillor Gómez, Zaragoza, 50018, Spain
| | - Diego de Miguel
- Centre for Cell Death, Cancer and Inflammation (CCCI), UCL Cancer Institute, University College of London, Gower Street, London, WC1E 6BT, UK.,Department of Biochemistry, Molecular and Cell Biology, University of Zaragoza, Calle de Pedro Cerbuna, 12, Zaragoza, 50009, Spain.,Aragon Institute of Biomedical Research (IIS Aragón), Instituto de Salud Carlos III, Avda. San Juan Bosco 13, Zaragoza, 50018, Spain
| | - Luis Martínez-Lostao
- Aragon Institute of Biomedical Research (IIS Aragón), Instituto de Salud Carlos III, Avda. San Juan Bosco 13, Zaragoza, 50018, Spain.,Department of Microbiology, Preventive Medicine and Public Health, University of Zaragoza, Domingo Miral, Zaragoza, 50009, Spain.,Department of Immunology, University Clinical Hospital Lozano Blesa, Padre Arrupe, Zaragoza, 50009, Spain.,Institute of Nanoscience of Aragón (INA), Mariano Esquillor Gómez, Zaragoza, 50009, Spain
| | - Séverine Le Gac
- Applied Microfluidics for BioEngineering Research, MESA+ Institute for Nanotechnology, MIRA Institute for Biomedical Research and Technical Medicine, University of Twente, Enschede, The Netherlands
| | - Manuel Doblare
- Group of Applied Mechanics and Bioengineering (AMB), Centro de Investigación Biomédica en Red. Bioingenieria, biomateriales y nanomedicina (CIBER-BBN), Mariano Esquillor Gómez, Zaragoza, 50018, Spain.,Aragon Institute of Engineering Research (I3A), University of Zaragoza, Mariano Esquillor Gómez, Zaragoza, 50009, Spain.,Aragon Institute of Biomedical Research, Instituto de Salud Carlos III, Mariano Esquillor Gómez, Zaragoza, 50009, Spain
| | - Ignacio Ochoa
- Group of Applied Mechanics and Bioengineering (AMB), Centro de Investigación Biomédica en Red. Bioingenieria, biomateriales y nanomedicina (CIBER-BBN), Mariano Esquillor Gómez, Zaragoza, 50018, Spain. .,Aragon Institute of Engineering Research (I3A), University of Zaragoza, Mariano Esquillor Gómez, Zaragoza, 50009, Spain. .,Aragon Institute of Biomedical Research, Instituto de Salud Carlos III, Mariano Esquillor Gómez, Zaragoza, 50009, Spain.
| | - Luis J Fernandez
- Group of Applied Mechanics and Bioengineering (AMB), Centro de Investigación Biomédica en Red. Bioingenieria, biomateriales y nanomedicina (CIBER-BBN), Mariano Esquillor Gómez, Zaragoza, 50018, Spain. .,Aragon Institute of Engineering Research (I3A), University of Zaragoza, Mariano Esquillor Gómez, Zaragoza, 50009, Spain. .,Aragon Institute of Biomedical Research, Instituto de Salud Carlos III, Mariano Esquillor Gómez, Zaragoza, 50009, Spain.
| |
Collapse
|
15
|
Mu LM, Ju RJ, Liu R, Bu YZ, Zhang JY, Li XQ, Zeng F, Lu WL. Dual-functional drug liposomes in treatment of resistant cancers. Adv Drug Deliv Rev 2017; 115:46-56. [PMID: 28433739 DOI: 10.1016/j.addr.2017.04.006] [Citation(s) in RCA: 97] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2016] [Revised: 04/15/2017] [Accepted: 04/18/2017] [Indexed: 12/26/2022]
Abstract
Efficacy of regular chemotherapy is significantly hampered by multidrug resistance (MDR) and severe systemic toxicity. The reduced toxicity has been evidenced after administration of drug liposomes, consisting of the first generation of regular drug liposomes, the second generation of long-circulation drug liposomes, and the third generation of targeting drug liposomes. However, MDR of cancers remains as an unsolved issue. The objective of this article is to review the dual-functional drug liposomes, which demonstrate the potential in overcoming MDR. Herein, dual-functional drug liposomes are referring to the drug-containing phospholipid bilayer vesicles that possess a dual-function of providing the basic efficacy of drug and the extended effect of the drug carrier. They exhibit unique roles in treatment of resistant cancer via circumventing drug efflux caused by adenosine triphosphate binding cassette (ABC) transporters, eliminating cancer stem cells, destroying mitochondria, initiating apoptosis, regulating autophagy, destroying supply channels, utilizing microenvironment, and silencing genes of the resistant cancer. As the prospect of an estimation, dual-functional drug liposomes would exhibit more strength in their extended function, hence deserving further investigation for clinical validation.
Collapse
|
16
|
Belkahla H, Herlem G, Picaud F, Gharbi T, Hémadi M, Ammar S, Micheau O. TRAIL-NP hybrids for cancer therapy: a review. NANOSCALE 2017; 9:5755-5768. [PMID: 28443893 DOI: 10.1039/c7nr01469d] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Cancer is a worldwide health problem. It is now considered as a leading cause of morbidity and mortality in developed countries. In the last few decades, considerable progress has been made in anti-cancer therapies, allowing the cure of patients suffering from this disease, or at least helping to prolong their lives. Several cancers, such as those of the lung and pancreas, are still devastating in the absence of therapeutic options. In the early 90s, TRAIL (Tumor Necrosis Factor-related apoptosis-inducing ligand), a cytokine belonging to the TNF superfamily, attracted major interest in oncology owing to its selective anti-tumor properties. Clinical trials using soluble TRAIL or antibodies targeting the two main agonist receptors (TRAIL-R1 and TRAIL-R2) have, however, failed to demonstrate their efficacy in the clinic. TRAIL is expressed on the surface of natural killer or CD8+ T activated cells and contributes to tumor surveillance. Nanoparticles functionalized with TRAIL mimic membrane-TRAIL and exhibit stronger antitumoral properties than soluble TRAIL or TRAIL receptor agonist antibodies. This review provides an update on the association and the use of nanoparticles associated with TRAIL for cancer therapy.
Collapse
Affiliation(s)
- H Belkahla
- Nanomedicine Lab, EA 4662, Université de Bourgogne Franche-Comté, Besançon, France
| | | | | | | | | | | | | |
Collapse
|
17
|
Naoum GE, Buchsbaum DJ, Tawadros F, Farooqi A, Arafat WO. Journey of TRAIL from Bench to Bedside and its Potential Role in Immuno-Oncology. Oncol Rev 2017; 11:332. [PMID: 28584572 PMCID: PMC5432952 DOI: 10.4081/oncol.2017.332] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2016] [Revised: 02/17/2017] [Accepted: 02/27/2017] [Indexed: 12/21/2022] Open
Abstract
Induction of apoptosis in cancer cells has increasingly been the focus of many therapeutic approaches in oncology field. Since its identification as a TNF family member, TRAIL (TNF-related apoptosis-inducing ligand) paved a new path in apoptosis inducing cancer therapies. Its selective ability to activate extrinsic and intrinsic cell death pathways in cancer cells only, independently from p53 mutations responsible for conventional therapeutics resistance, spotted TRAIL as a potent cancer apoptotic agent. Many recombinant preparations of TRAIL and death receptor targeting monoclonal antibodies have been developed and being tested pre-clinically and clinically both as a single agent and in combinations. Of note, the monoclonal antibodies were not the only type of antibodies developed to target TRAIL receptors. Recent technology has brought forth several single chain variable domains (scFv) designs fused recombinantly to TRAIL as well. Also, it is becoming progressively more understandable that field of nanotechnology has revolutionized cancer diagnosis and therapy. The recent breakthroughs in materials science and protein engineering have helped considerably in strategically loading drugs into nanoparticles or conjugating drugs to their surface. In this review we aim to comprehensively highlight the molecular knowledge of TRAIL in the context of its pathway, receptors and resistance factors. We also aim to review the clinical trials that have been done using TRAIL based therapies and to review various scFv designs, the arsenal of nano-carriers and molecules available to selectively target tumor cells with TRAIL.
Collapse
Affiliation(s)
| | | | | | | | - Waleed O. Arafat
- Alexandria Comprehensive Cancer Center, Alexandria, Egypt
- Univeristy of Alabama, Birmingham, AL, USA
- University of Alexandria, Faculty of Medicine, Egypt
| |
Collapse
|
18
|
Hendriks D, Choi G, de Bruyn M, Wiersma VR, Bremer E. Antibody-Based Cancer Therapy: Successful Agents and Novel Approaches. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2017; 331:289-383. [PMID: 28325214 DOI: 10.1016/bs.ircmb.2016.10.002] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Since their discovery, antibodies have been viewed as ideal candidates or "magic bullets" for use in targeted therapy in the fields of cancer, autoimmunity, and chronic inflammatory disorders. A wave of antibody-dedicated research followed, which resulted in the clinical approval of a first generation of monoclonal antibodies for cancer therapy such as rituximab (1997) and cetuximab (2004), and infliximab (2002) for the treatment of autoimmune diseases. More recently, the development of antibodies that prevent checkpoint-mediated inhibition of T cell responses invigorated the field of cancer immunotherapy. Such antibodies induced unprecedented long-term remissions in patients with advanced stage malignancies, most notably melanoma and lung cancer, that do not respond to conventional therapies. In this review, we will recapitulate the development of antibody-based therapy, and detail recent advances and new functions, particularly in the field of cancer immunotherapy. With the advent of recombinant DNA engineering, a number of rationally designed molecular formats of antibodies and antibody-derived agents have become available, and we will discuss various molecular formats including antibodies with improved effector functions, bispecific antibodies, antibody-drug conjugates, antibody-cytokine fusion proteins, and T cells genetically modified with chimeric antigen receptors. With these exciting advances, new antibody-based treatment options will likely enter clinical practice and pave the way toward more successful control of malignant diseases.
Collapse
Affiliation(s)
- D Hendriks
- Department of Surgery, Translational Surgical Oncology, University of Groningen, University Medical Center Groningen (UMCG), Groningen, The Netherlands
| | - G Choi
- Department of Hematology, Section Immunohematology, University of Groningen, University Medical Center Groningen (UMCG), Groningen, The Netherlands
| | - M de Bruyn
- Department of Obstetrics & Gynecology, University of Groningen, University Medical Center Groningen (UMCG), Groningen, The Netherlands
| | - V R Wiersma
- Department of Hematology, Section Immunohematology, University of Groningen, University Medical Center Groningen (UMCG), Groningen, The Netherlands.
| | - E Bremer
- Department of Hematology, Section Immunohematology, University of Groningen, University Medical Center Groningen (UMCG), Groningen, The Netherlands; University of Exeter Medical School, Exeter, UK.
| |
Collapse
|
19
|
Abstract
This review focuses on summarizing the existing work about nanomaterial-based cancer immunotherapy in detail.
Collapse
Affiliation(s)
- Lijia Luo
- Key Laboratory of Magnetic Materials and Devices
- CAS & Key Laboratory of Additive Manufacturing Materials of Zhejiang Province, & Division of Functional Materials and Nanodevices
- Ningbo Institute of Materials Technology and Engineering
- Chinese Academy of Sciences
- Ningbo
| | - Rui Shu
- University of Chinese Academy of Sciences
- Beijing 100049
- China
- Key Laboratory of Marine Materials and Related Technology
- CAS & Ningbo Institute of Materials Technology and Engineering
| | - Aiguo Wu
- Key Laboratory of Magnetic Materials and Devices
- CAS & Key Laboratory of Additive Manufacturing Materials of Zhejiang Province, & Division of Functional Materials and Nanodevices
- Ningbo Institute of Materials Technology and Engineering
- Chinese Academy of Sciences
- Ningbo
| |
Collapse
|
20
|
High-order TRAIL oligomer formation in TRAIL-coated lipid nanoparticles enhances DR5 cross-linking and increases antitumour effect against colon cancer. Cancer Lett 2016; 383:250-260. [DOI: 10.1016/j.canlet.2016.10.005] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Revised: 09/29/2016] [Accepted: 10/02/2016] [Indexed: 02/02/2023]
|
21
|
Erkoc P, Cingöz A, Bagci-Onder T, Kizilel S. Quinacrine Mediated Sensitization of Glioblastoma (GBM) Cells to TRAIL through MMP-Sensitive PEG Hydrogel Carriers. Macromol Biosci 2016; 17. [DOI: 10.1002/mabi.201600267] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Revised: 09/05/2016] [Indexed: 12/13/2022]
Affiliation(s)
- Pelin Erkoc
- Biomedical Sciences and Engineering; Koc University; 34450 Sariyer Istanbul Turkey
| | - Ahmet Cingöz
- School of Medicine; Koc University; 34450 Sariyer Istanbul Turkey
| | - Tugba Bagci-Onder
- Biomedical Sciences and Engineering; Koc University; 34450 Sariyer Istanbul Turkey
- School of Medicine; Koc University; 34450 Sariyer Istanbul Turkey
| | - Seda Kizilel
- Biomedical Sciences and Engineering; Koc University; 34450 Sariyer Istanbul Turkey
- Chemical and Biological Engineering; Koc University; 34450 Sariyer Istanbul Turkey
| |
Collapse
|
22
|
Hendriks D, He Y, Koopmans I, Wiersma VR, van Ginkel RJ, Samplonius DF, Helfrich W, Bremer E. Programmed Death Ligand 1 (PD-L1)-targeted TRAIL combines PD-L1-mediated checkpoint inhibition with TRAIL-mediated apoptosis induction. Oncoimmunology 2016; 5:e1202390. [PMID: 27622071 PMCID: PMC5007955 DOI: 10.1080/2162402x.2016.1202390] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Revised: 06/07/2016] [Accepted: 06/12/2016] [Indexed: 01/18/2023] Open
Abstract
Antibodies that block PD-L1/PD-1 immune checkpoints restore the activity of functionally-impaired antitumor T cells. These antibodies show unprecedented clinical benefit in various advanced cancers, particularly in melanoma. However, only a subset of cancer patients responds to current PD-L1/PD-1-blocking strategies, highlighting the need for further advancements in PD-L1/PD-1-based immunotherapy. Here, we report on a novel approach designed to combine PD-L1 checkpoint inhibition with the tumor-selective induction of apoptosis by TNF-related Apoptosis Inducing Ligand (TRAIL). In brief, a new bi-functional fusion protein, designated anti-PD-L1:TRAIL, was constructed comprising a PD-L1-blocking antibody fragment genetically fused to the extracellular domain of the pro-apoptotic tumoricidal protein TRAIL. Treatment of PD-L1-expressing cancer cells with anti-PD-L1:TRAIL induced PD-L1-directed TRAIL-mediated cancer cell death. Treatment of T cells with anti-PD-L1:TRAIL augmented T cell activation, as evidenced by increased proliferation, secretion of IFNγ and enhanced killing of cancer cell lines and primary patient-derived cancer cells in mixed T cell/cancer cell culture experiments. Of note, elevated levels of IFNγ further upregulated PD-L1 on cancer cells and simultaneously sensitized cancer cells to TRAIL-mediated apoptosis by anti-PD-L1:TRAIL. Additionally, anti-PD-L1:TRAIL converted immunosuppressive PD-L1-expressing myeloid cells into pro-apoptotic effector cells that triggered TRAIL-mediated cancer cell death. In conclusion, combining PD-L1 checkpoint inhibition with TRAIL-mediated induction of apoptosis using anti-PD-L1:TRAIL yields promising multi-fold and mutually reinforcing anticancer activity that may be exploited to enhance the efficacy of therapeutic PD-L1/PD-1 checkpoint inhibition.
Collapse
Affiliation(s)
- Djoke Hendriks
- University of Groningen, University Medical Center Groningen (UMCG), Department of Surgery, Laboratory for Translational Surgical Oncology , Groningen, the Netherlands
| | - Yuan He
- University of Groningen, University Medical Center Groningen (UMCG), Department of Surgery, Laboratory for Translational Surgical Oncology , Groningen, the Netherlands
| | - Iris Koopmans
- University of Groningen, University Medical Center Groningen (UMCG), Department of Surgery, Laboratory for Translational Surgical Oncology , Groningen, the Netherlands
| | - Valerie R Wiersma
- University of Groningen, University Medical Center Groningen (UMCG), Department of Surgery, Laboratory for Translational Surgical Oncology , Groningen, the Netherlands
| | - Robert J van Ginkel
- University of Groningen, University Medical Center Groningen (UMCG), Department of Surgery, Laboratory for Translational Surgical Oncology , Groningen, the Netherlands
| | - Douwe F Samplonius
- University of Groningen, University Medical Center Groningen (UMCG), Department of Surgery, Laboratory for Translational Surgical Oncology , Groningen, the Netherlands
| | - Wijnand Helfrich
- University of Groningen, University Medical Center Groningen (UMCG), Department of Surgery, Laboratory for Translational Surgical Oncology , Groningen, the Netherlands
| | - Edwin Bremer
- University of Exeter Medical School, St Luke's Campus, Exeter, Devon, UK; University of Groningen, University Medical Center Groningen, Department of Experimental Hematology, Section Immunohematology, Cancer Research Center Groningen (CRCG), Groningen, the Netherlands
| |
Collapse
|
23
|
De Miguel D, Gallego-Lleyda A, Ayuso JM, Erviti-Ardanaz S, Pazo-Cid R, del Agua C, Fernández LJ, Ochoa I, Anel A, Martinez-Lostao L. TRAIL-coated lipid-nanoparticles overcome resistance to soluble recombinant TRAIL in non-small cell lung cancer cells. NANOTECHNOLOGY 2016; 27:185101. [PMID: 27001952 DOI: 10.1088/0957-4484/27/18/185101] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
PURPOSE Non-small cell lung cancer (NSCLC) is one the types of cancer with higher prevalence and mortality. Apo2-Ligand/TRAIL is a TNF family member able to induce apoptosis in tumor cells but not in normal cells. It has been tested in clinical trials against different types of human cancer including NSCLC. However, results of clinical trials have shown a limited efficacy of TRAIL-based therapies. Recently we have demonstrated that artificial lipid nanoparticles coated with bioactive Apo2L/TRAIL (LUV-TRAIL) greatly improved TRAIL cytotoxic ability being capable of killing chemoresistant hematological cancer cells. In the present work we have extended the study to NSCLC. METHODS/PATIENTS LUV-TRAIL-induced cytotoxicity was assessed on different NSCLC cell lines with different sensitivity to soluble TRAIL and on primary human tumor cells from three patients suffering from NSCLC cancer. We also tested LUV-TRAIL-cytotoxic ability in combination with several anti-tumor agents. RESULTS LUV-TRAIL exhibited a greater cytotoxic effect compared to soluble TRAIL both in A549 cells and primary human NSCLC cells. LUV-TRAIL-induced cell death was dependent on caspase-8 and caspase-3 activation. Moreover, combination of LUV-TRAIL with other anti-tumor agents such as flavopiridol, and SNS-032 clearly enhanced LUV-TRAIL-induced cytotoxicity against NSCLC cancer cells. CONCLUSION The novel formulation of TRAIL based on displaying it on the surface of lipid nanoparticles greatly increases its anti-tumor activity and has clinical potential in cancer treatment.
Collapse
Affiliation(s)
- Diego De Miguel
- Departamento de Bioquímica, Biología Molecular y Celular, Universidad de Zaragoza, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
de Miguel D, Lemke J, Anel A, Walczak H, Martinez-Lostao L. Onto better TRAILs for cancer treatment. Cell Death Differ 2016; 23:733-47. [PMID: 26943322 PMCID: PMC4832109 DOI: 10.1038/cdd.2015.174] [Citation(s) in RCA: 241] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Revised: 12/11/2015] [Accepted: 12/17/2015] [Indexed: 01/01/2023] Open
Abstract
Tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL), also known as Apo-2 ligand (Apo2L), is a member of the TNF cytokine superfamily. By cross-linking TRAIL-Receptor (TRAIL-R) 1 or TRAIL-R2, also known as death receptors 4 and 5 (DR4 and DR5), TRAIL has the capability to induce apoptosis in a wide variety of tumor cells while sparing vital normal cells. The discovery of this unique property among TNF superfamily members laid the foundation for testing the clinical potential of TRAIL-R-targeting therapies in the cancer clinic. To date, two of these therapeutic strategies have been tested clinically: (i) recombinant human TRAIL and (ii) antibodies directed against TRAIL-R1 or TRAIL-R2. Unfortunately, however, these TRAIL-R agonists have basically failed as most human tumors are resistant to apoptosis induction by them. It recently emerged that this is largely due to the poor agonistic activity of these agents. Consequently, novel TRAIL-R-targeting agents with increased bioactivity are currently being developed with the aim of rendering TRAIL-based therapies more active. This review summarizes these second-generation novel formulations of TRAIL and other TRAIL-R agonists, which exhibit enhanced cytotoxic capacity toward cancer cells, thereby providing the potential of being more effective when applied clinically than first-generation TRAIL-R agonists.
Collapse
Affiliation(s)
- D de Miguel
- Departamento de Bioquímica, Biología Molecular y Celular, Facultad de Ciencias, Universidad de Zaragoza, Zaragoza, Spain
- Instituto de Investigación Sanitaria de Aragón, Zaragoza, Spain
| | - J Lemke
- UCL Cancer Institute, Faculty of Medical Sciences, University College London, London, UK
| | - A Anel
- Departamento de Bioquímica, Biología Molecular y Celular, Facultad de Ciencias, Universidad de Zaragoza, Zaragoza, Spain
- Instituto de Investigación Sanitaria de Aragón, Zaragoza, Spain
| | - H Walczak
- UCL Cancer Institute, Faculty of Medical Sciences, University College London, London, UK
| | - L Martinez-Lostao
- Departamento de Bioquímica, Biología Molecular y Celular, Facultad de Ciencias, Universidad de Zaragoza, Zaragoza, Spain
- Instituto de Investigación Sanitaria de Aragón, Zaragoza, Spain
- Instituto de Nanociencia de Aragón, Zaragoza, Spain
| |
Collapse
|
25
|
Chester C, Fritsch K, Kohrt HE. Natural Killer Cell Immunomodulation: Targeting Activating, Inhibitory, and Co-stimulatory Receptor Signaling for Cancer Immunotherapy. Front Immunol 2015; 6:601. [PMID: 26697006 PMCID: PMC4667030 DOI: 10.3389/fimmu.2015.00601] [Citation(s) in RCA: 129] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2015] [Accepted: 11/09/2015] [Indexed: 01/11/2023] Open
Abstract
There is compelling clinical and experimental evidence to suggest that natural killer (NK) cells play a critical role in the recognition and eradication of tumors. Efforts at using NK cells as antitumor agents began over two decades ago, but recent advances in elucidating NK cell biology have accelerated the development of NK cell-targeting therapeutics. NK cell activation and the triggering of effector functions is governed by a complex set of activating and inhibitory receptors. In the early phases of cancer immune surveillance, NK cells directly identify and lyse cancer cells. Nascent transformed cells elicit NK cell activation and are eliminated. However, as tumors progress, cancerous cells develop immunosuppressive mechanisms that circumvent NK cell-mediated killing, allowing for tumor escape and proliferation. Therapeutic intervention aims to reverse tumor-induced NK cell suppression and sustain NK cells’ tumorlytic capacities. Here, we review tumor–NK cell interactions, discuss the mechanisms by which NK cells generate an antitumor immune response, and discuss NK cell-based therapeutic strategies targeting activating, inhibitory, and co-stimulatory receptors.
Collapse
Affiliation(s)
- Cariad Chester
- Division of Oncology, Department of Medicine, Stanford University , Stanford, CA , USA ; Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine , Stanford, CA , USA
| | - Katherine Fritsch
- Division of Oncology, Department of Medicine, Stanford University , Stanford, CA , USA
| | - Holbrook E Kohrt
- Division of Oncology, Department of Medicine, Stanford University , Stanford, CA , USA
| |
Collapse
|