1
|
Schuermans S, Kestens C, Marques PE. Systemic mechanisms of necrotic cell debris clearance. Cell Death Dis 2024; 15:557. [PMID: 39090111 PMCID: PMC11294570 DOI: 10.1038/s41419-024-06947-5] [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: 03/27/2024] [Revised: 07/23/2024] [Accepted: 07/24/2024] [Indexed: 08/04/2024]
Abstract
Necrosis is an overarching term that describes cell death modalities caused by (extreme) adverse conditions in which cells lose structural integrity. A guaranteed consequence of necrosis is the production of necrotic cell remnants, or debris. Necrotic cell debris is a strong trigger of inflammation, and although inflammatory responses are required for tissue healing, necrotic debris may lead to uncontrolled immune responses and collateral damage. Besides local phagocytosis by recruited leukocytes, there is accumulating evidence that extracellular mechanisms are also involved in necrotic debris clearance. In this review, we focused on systemic clearance mechanisms present in the bloodstream and vasculature that often cooperate to drive the clearance of cell debris. We reviewed the contribution and cooperation of extracellular DNases, the actin-scavenger system, the fibrinolytic system and reticuloendothelial cells in performing clearance of necrotic debris. Moreover, associations of the (mis)functioning of these clearance systems with a variety of diseases were provided, illustrating the importance of the mechanisms of clearance of dead cells in the organism.
Collapse
Affiliation(s)
- Sara Schuermans
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Caine Kestens
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Pedro Elias Marques
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium.
| |
Collapse
|
2
|
Zhang N, Ru B, Hu J, Xu L, Wan Q, Liu W, Cai W, Zhu T, Ji Z, Guo R, Zhang L, Li S, Tong X. Recent advances of CREKA peptide-based nanoplatforms in biomedical applications. J Nanobiotechnology 2023; 21:77. [PMID: 36869341 PMCID: PMC9985238 DOI: 10.1186/s12951-023-01827-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 02/22/2023] [Indexed: 03/05/2023] Open
Abstract
Nanomedicine technology is a rapidly developing field of research and application that uses nanoparticles as a platform to facilitate the diagnosis and treatment of diseases. Nanoparticles loaded with drugs and imaging contrast agents have already been used in clinically, but they are essentially passive delivery carriers. To make nanoparticles smarter, an important function is the ability to actively locate target tissues. It enables nanoparticles to accumulate in target tissues at higher concentrations, thereby improving therapeutic efficacy and reducing side effects. Among the different ligands, the CREKA peptide (Cys-Arg-Glu-Lys-Ala) is a desirable targeting ligand and has a good targeting ability for overexpressed fibrin in different models, such as cancers, myocardial ischemia-reperfusion, and atherosclerosis. In this review, the characteristic of the CREKA peptide and the latest reports regarding the application of CREKA-based nanoplatforms in different biological tissues are described. In addition, the existing problems and future application perspectives of CREKA-based nanoplatforms are also addressed.
Collapse
Affiliation(s)
- Nannan Zhang
- Laboratory Medicine Center, Clinical Research Institute, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
- Center for Rehabilitation Medicine, Department of Pain Management, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
| | - Bin Ru
- Laboratory Medicine Center, Clinical Research Institute, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
- Center for Rehabilitation Medicine, Department of Pain Management, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
| | - Jiaqi Hu
- Laboratory Medicine Center, Clinical Research Institute, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
- Center for Rehabilitation Medicine, Department of Pain Management, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
| | - Langhai Xu
- Laboratory Medicine Center, Clinical Research Institute, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
- Center for Rehabilitation Medicine, Department of Pain Management, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
| | - Quan Wan
- Laboratory Medicine Center, Clinical Research Institute, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
- Center for Rehabilitation Medicine, Department of Pain Management, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
| | - Wenlong Liu
- Laboratory Medicine Center, Clinical Research Institute, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
- Center for Rehabilitation Medicine, Department of Pain Management, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
| | - WenJun Cai
- Laboratory Medicine Center, Clinical Research Institute, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
- Center for Rehabilitation Medicine, Department of Pain Management, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
| | - Tingli Zhu
- Laboratory Medicine Center, Clinical Research Institute, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
- Center for Rehabilitation Medicine, Department of Pain Management, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
| | - Zhongwei Ji
- Laboratory Medicine Center, Clinical Research Institute, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
- Center for Rehabilitation Medicine, Department of Pain Management, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
| | - Ran Guo
- Laboratory Medicine Center, Clinical Research Institute, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
- Center for Rehabilitation Medicine, Department of Pain Management, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
| | - Lin Zhang
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China.
| | - Shun Li
- Laboratory Medicine Center, Clinical Research Institute, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China.
- Center for Rehabilitation Medicine, Department of Pain Management, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China.
| | - Xiangmin Tong
- Laboratory Medicine Center, Clinical Research Institute, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China.
| |
Collapse
|
3
|
Browning JL, Bhawan J, Tseng A, Crossland N, Bujor AM, Akassoglou K, Assassi S, Skaug B, Ho J. Extensive and Persistent Extravascular Dermal Fibrin Deposition Characterizes Systemic Sclerosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.16.523256. [PMID: 36711912 PMCID: PMC9882194 DOI: 10.1101/2023.01.16.523256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
Systemic sclerosis (SSc) is an autoimmune disease characterized by progressive multiorgan fibrosis. While the cause of SSc remains unknown, a perturbed vasculature is considered a critical early step in the pathogenesis. Using fibrinogen as a marker of vascular leakage, we found extensive extravascular fibrinogen deposition in the dermis of both limited and diffuse systemic sclerosis disease, and it was present in both early and late-stage patients. Based on a timed series of excision wounds, retention on the fibrin deposit of the splice variant domain, fibrinogen αEC, indicated a recent event, while fibrin networks lacking the αEC domain were older. Application of this timing tool to SSc revealed considerable heterogeneity in αEC domain distribution providing unique insight into disease activity. Intriguingly, the fibrinogen-αEC domain also accumulated in macrophages. These observations indicate that systemic sclerosis is characterized by ongoing vascular leakage resulting in extensive interstitial fibrin deposition that is either continually replenished and/or there is impaired fibrin clearance. Unresolved fibrin deposition might then incite chronic tissue remodeling.
Collapse
Affiliation(s)
- Jeffrey L Browning
- Department of Microbiology, Boston University Chobanian & Avedesian School of Medicine, Boston, MA
- Department of Rheumatology, Boston University Chobanian & Avedesian School of Medicine, Boston, MA
| | - Jag Bhawan
- Department of Dermatopathology, Boston University Chobanian & Avedesian School of Medicine, Boston, MA
- Department of Pathology and Laboratory Medicine, Boston University Chobanian & Avedesian School of Medicine, Boston, MA
| | - Anna Tseng
- Department of Pathology and Laboratory Medicine, Boston University Chobanian & Avedesian School of Medicine, Boston, MA
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA
| | - Nicholas Crossland
- Department of Pathology and Laboratory Medicine, Boston University Chobanian & Avedesian School of Medicine, Boston, MA
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA
| | - Andreea M Bujor
- Department of Rheumatology, Boston University Chobanian & Avedesian School of Medicine, Boston, MA
| | - Katerina Akassoglou
- Gladstone Institute of Neurological Disease San Francisco California USA
- Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA
| | - Shervin Assassi
- Division of Rheumatology, University of Texas Health Science Center, Houston, TX
| | - Brian Skaug
- Division of Rheumatology, University of Texas Health Science Center, Houston, TX
| | - Jonathan Ho
- Department of Dermatopathology, Boston University Chobanian & Avedesian School of Medicine, Boston, MA
- Section Dermatology University of the West Indies, Mona Jamaica
| |
Collapse
|
4
|
Tang X, Xu Q, Yang S, Huang X, Wang L, Huang F, Luo J, Zhou X, Wu A, Mei Q, Zhao C, Wu J. Toll-like Receptors and Thrombopoiesis. Int J Mol Sci 2023; 24:1010. [PMID: 36674552 PMCID: PMC9864288 DOI: 10.3390/ijms24021010] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 12/27/2022] [Accepted: 01/03/2023] [Indexed: 01/06/2023] Open
Abstract
Platelets are the second most abundant blood component after red blood cells and can participate in a variety of physiological and pathological functions. Beyond its traditional role in hemostasis and thrombosis, it also plays an indispensable role in inflammatory diseases. However, thrombocytopenia is a common hematologic problem in the clinic, and it presents a proportional relationship with the fatality of many diseases. Therefore, the prevention and treatment of thrombocytopenia is of great importance. The expression of Toll-like receptors (TLRs) is one of the most relevant characteristics of thrombopoiesis and the platelet inflammatory function. We know that the TLR family is found on the surface or inside almost all cells, where they perform many immune functions. Of those, TLR2 and TLR4 are the main stress-inducing members and play an integral role in inflammatory diseases and platelet production and function. Therefore, the aim of this review is to present and discuss the relationship between platelets, inflammation and the TLR family and extend recent research on the influence of the TLR2 and TLR4 pathways and the regulation of platelet production and function. Reviewing the interaction between TLRs and platelets in inflammation may be a research direction or program for the treatment of thrombocytopenia-related and inflammatory-related diseases.
Collapse
Affiliation(s)
- Xiaoqin Tang
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Qian Xu
- Department of Physiology, School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, China
| | - Shuo Yang
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Xinwu Huang
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Long Wang
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- Institute of Cardiovascular Research, the Key Laboratory of Medical Electrophysiology, Ministry of Education of China, Luzhou 646000, China
| | - Feihong Huang
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- Institute of Cardiovascular Research, the Key Laboratory of Medical Electrophysiology, Ministry of Education of China, Luzhou 646000, China
| | - Jiesi Luo
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- Institute of Cardiovascular Research, the Key Laboratory of Medical Electrophysiology, Ministry of Education of China, Luzhou 646000, China
| | - Xiaogang Zhou
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- Institute of Cardiovascular Research, the Key Laboratory of Medical Electrophysiology, Ministry of Education of China, Luzhou 646000, China
| | - Anguo Wu
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- Institute of Cardiovascular Research, the Key Laboratory of Medical Electrophysiology, Ministry of Education of China, Luzhou 646000, China
| | - Qibing Mei
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- Institute of Cardiovascular Research, the Key Laboratory of Medical Electrophysiology, Ministry of Education of China, Luzhou 646000, China
| | - Chunling Zhao
- Department of Physiology, School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, China
| | - Jianming Wu
- Department of Physiology, School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, China
- Institute of Cardiovascular Research, the Key Laboratory of Medical Electrophysiology, Ministry of Education of China, Luzhou 646000, China
| |
Collapse
|
5
|
Wang CC, Wei SC, Luo SC. Recent Advances and Biomedical Applications of Peptide-Integrated Conducting Polymers. ACS APPLIED BIO MATERIALS 2022; 5:1916-1933. [PMID: 35119258 DOI: 10.1021/acsabm.1c01194] [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] [Indexed: 12/14/2022]
Abstract
Conducting polymers (CPs) are of great interests to researchers around the world in biomedical applications owing to their unique electrical and mechanical properties. Besides, they are easy to fabricate and have long-term stability. These features make CPs a powerful building block of modern biomaterials. Peptide functionalization has been a versatile tool for the development of CP-based biomaterials. With the aid of peptide modifications, the biocompatibility, target selectivity, and cellular interactions of CPs can be greatly improved. Reflecting these aspects, an increasing number of studies on peptide-integrated conducting polymers have been reported recently. In this review, various kinds of peptide immobilization strategies on CPs are introduced. Moreover, the aims of peptide modification are discussed in three aspects: enhancing the specific selectivity, avoiding nonspecific adhesion, and mimicking the environment of extracellular matrix. We highlighted recent studies in the applications of peptide-integrated CPs in electrochemical sensors, antifouling surfaces, and conductive biointerfaces. These studies have shown great potentials from the integration of peptide and CPs as a versatile platform for advanced biological and clinical applications in the near future.
Collapse
Affiliation(s)
- Chi-Cha Wang
- Department of Materials Science and Engineering, National Taiwan University, No. 1, Sec. 4, Roosevelt Road, Taipei 10617, Taiwan
| | - Shu-Chen Wei
- Department of Internal Medicine, National Taiwan University Hospital and College of Medicine, No.1 Jen Ai Road, Section 1, Taipei 10051, Taiwan
| | - Shyh-Chyang Luo
- Department of Materials Science and Engineering, National Taiwan University, No. 1, Sec. 4, Roosevelt Road, Taipei 10617, Taiwan.,Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes (NHRI), Miaoli County, 35053 Taiwan
| |
Collapse
|
6
|
Cao Y. The impact of the hypoxia-VEGF-vascular permeability on COVID-19-infected patients. EXPLORATION (BEIJING, CHINA) 2021; 1:20210051. [PMID: 35434726 PMCID: PMC8653011 DOI: 10.1002/exp.20210051] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 09/07/2021] [Indexed: 01/08/2023]
Abstract
Effective treatment of patients with severe COVID-19 to reduce mortality remains one of the most challenging medical issues in controlling unpredictable emergencies caused by the global pandemics. Unfortunately, such effective therapies are not available at this time of writing. In this article, I discuss the possibility of repurposing clinically available anti-VEGF (vascular endothelial growth factor) drugs that are routinely used in oncology and ophthalmology areas for effective treatment of patients with severe and critical COVID-19. Our preliminary findings from a clinical trial support the therapeutic concept of using anti-VEGF for treating patients with severe COVID-19 to reduce mortality. The aim of this article is to further provide mechanistic insights into the role of VEGF in causing pathological changes during COVID-19 infection.
Collapse
Affiliation(s)
- Yihai Cao
- Department of Microbiology, Tumor and Cell BiologyKarolinska InstituteStockholmSweden
| |
Collapse
|
7
|
Thachil J, Khorana A, Carrier M. Similarities and perspectives on the two C's-Cancer and COVID-19. J Thromb Haemost 2021; 19:1161-1167. [PMID: 33725410 PMCID: PMC8250039 DOI: 10.1111/jth.15294] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 03/02/2021] [Accepted: 03/02/2021] [Indexed: 12/14/2022]
Abstract
COVID-19 continues to dominate the health-care burden in the twenty-first century. While health-care professionals around the world try their best to minimize the mortality from this pandemic, we also continue to battle the high mortality from different types of cancer. For the hemostasis and thrombosis specialist, these two conditions present some unusual similarities including the high rate of thrombosis and marked elevation of D-dimers. In this forum article, we discuss these similarities and provide some considerations for future research and therapeutic trials.
Collapse
Affiliation(s)
- Jecko Thachil
- Department of Haematology, Manchester University Hospitals, Manchester, UK
| | - Alok Khorana
- Department of Hematology and Medical Oncology, Taussig Cancer Institute and Case Comprehensive Cancer Center, Cleveland, Ohio, USA
| | - Marc Carrier
- Cleveland Clinic, Cleveland, Ohio, USA
- Department of Medicine, University of Ottawa, The Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| |
Collapse
|
8
|
An Integrated Approach of the Potential Underlying Molecular Mechanistic Paradigms of SARS-CoV-2-Mediated Coagulopathy. Indian J Clin Biochem 2021; 36:387-403. [PMID: 33875909 PMCID: PMC8047580 DOI: 10.1007/s12291-021-00972-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Accepted: 04/03/2021] [Indexed: 02/06/2023]
Abstract
Coronavirus disease 2019 (Covid-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a pandemic disease which has affected more than 6.2 million people globally, with numbers mounting considerably daily. However, till date, no specific treatment modalities are available for Covid-19 and also not much information is known about this disease. Recent studies have revealed that SARS-CoV-2 infection is associated with the generation of thrombosis and coagulopathy. Fundamentally, it has been believed that a diverse array of signalling pathways might be responsible for the activation of coagulation cascade during SARS-CoV-2 infection. Henceforth, a detailed understanding of these probable underlying molecular mechanistic pathways causing thrombosis in Covid-19 disease deserves an urgent exploration. Therefore, in this review, the hypothetical crosstalk between distinct signalling pathways including apoptosis, inflammation, hypoxia and angiogenesis attributable for the commencement of thrombotic events during SARS-CoV-2 infection has been addressed which might further unravel promising therapeutic targets in Covid-19 disease.
Collapse
|
9
|
Liu YT, Goel S, Kai M, Moran Guerrero JA, Nguyen T, Mai J, Shen H, Ziemys A, Yokoi K. Seed- and Soil-Dependent Differences in Murine Breast Tumor Microenvironments Dictate Anti-PD-L1 IgG Delivery and Therapeutic Efficacy. Pharmaceutics 2021; 13:pharmaceutics13040530. [PMID: 33920216 PMCID: PMC8069710 DOI: 10.3390/pharmaceutics13040530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 04/06/2021] [Accepted: 04/08/2021] [Indexed: 11/16/2022] Open
Abstract
We sought to determine if Stephen Paget’s “seed and soil” hypothesis of organ-preference patterns of cancer metastasis can explain the development of heterogeneity in a tumor microenvironment (TME) as well as immunotherapeutic delivery and efficacy. We established single-cell-derived clones (clones 1 and 16) from parental 4T1 murine breast cancer cells to create orthotopic primary and liver metastasis models to deconvolute polyclonal complexity cancer cells and the difference in TME-derived heterogeneities. Tumor-bearing mice were treated with anti-PD-L1 IgG or a control antibody, and immunofluorescent imaging and quantification were then performed to evaluate the therapeutic efficacy on tumor growth, the delivery of therapy to tumors, the development of blood vessels, the expression of PD-L1, the accumulation of immune cells, and the amount of coagulation inside tumors. The quantification showed an inverse correlation between the amount of delivered therapy and therapeutic efficacy in parental-cell-derived tumors. In contrast, tumors originating from clone 16 cells accumulated a significantly greater amount of therapy and responded better than clone-1-derived tumors. This difference was greater when tumors grew in the liver than the primary site. A similar trend was found in PD-L1 expression and immune cell accumulation. However, the change in the number of blood vessels was not significant. In addition, the amount of coagulation was more abundant in clone-1-derived tumors when compared to others. Thus, our findings reconfirmed the seed- and soil-dependent differences in PD-L1 expression, therapeutic delivery, immune cell accumulation, and tumor coagulation, which can constitute a heterogeneous delivery and response of immunotherapy in polyclonal tumors growing in different organs.
Collapse
|
10
|
Dvorak HF. Reconciling VEGF With VPF: The Importance of Increased Vascular Permeability for Stroma Formation in Tumors, Healing Wounds, and Chronic Inflammation. Front Cell Dev Biol 2021; 9:660609. [PMID: 33834026 PMCID: PMC8021773 DOI: 10.3389/fcell.2021.660609] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 03/04/2021] [Indexed: 11/30/2022] Open
Abstract
It is widely believed that vascular endothelial growth factor (VEGF) induces angiogenesis by its direct mitogenic and motogenic actions on vascular endothelial cells. However, these activities are only detected when endothelial cells are cultured at very low (0.1%) serum concentrations and would not be expected to take place at the much higher serum levels found in angiogenic sites in vivo. This conundrum can be resolved by recalling VEGF’s original function, that of an extremely potent vascular permeability factor (VPF). In vivo VPF/VEGF increases microvascular permeability such that whole plasma leaks into the tissues where it undergoes clotting by tissue factor that is expressed on tumor and host connective tissue cells to deposit fibrin and generate serum. By providing tissue support and by reprogramming the gene expression patterns of cells locally, fibrin and serum can together account for the formation of vascular connective tissue stroma. In sum, by increasing vascular permeability, VPF/VEGF triggers the “wound healing response,” setting in motion a fundamental pathophysiological process that induces the mature stroma that is found not only in healing wounds but also in solid tumors and chronic inflammatory diseases. Once initiated by increased vascular permeability, this response may be difficult to impede, perhaps contributing to the limited success of anti-VEGF therapies in treating cancer.
Collapse
Affiliation(s)
- Harold F Dvorak
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
11
|
Tucker TA, Idell S. The Contribution of the Urokinase Plasminogen Activator and the Urokinase Receptor to Pleural and Parenchymal Lung Injury and Repair: A Narrative Review. Int J Mol Sci 2021; 22:ijms22031437. [PMID: 33535429 PMCID: PMC7867090 DOI: 10.3390/ijms22031437] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 01/26/2021] [Accepted: 01/26/2021] [Indexed: 12/28/2022] Open
Abstract
Pleural and parenchymal lung injury have long been characterized by acute inflammation and pathologic tissue reorganization, when severe. Although transitional matrix deposition is a normal part of the injury response, unresolved fibrin deposition can lead to pleural loculation and scarification of affected areas. Within this review, we present a brief discussion of the fibrinolytic pathway, its components, and their contribution to injury progression. We review how local derangements of fibrinolysis, resulting from increased coagulation and reduced plasminogen activator activity, promote extravascular fibrin deposition. Further, we describe how pleural mesothelial cells contribute to lung scarring via the acquisition of a profibrotic phenotype. We also discuss soluble uPAR, a recently identified biomarker of pleural injury, and its diagnostic value in the grading of pleural effusions. Finally, we provide an in-depth discussion on the clinical importance of single-chain urokinase plasminogen activator (uPA) for the treatment of loculated pleural collections.
Collapse
Affiliation(s)
| | - Steven Idell
- Correspondence: ; Tel.: +1-903-877-7556; Fax: +1-903-877-7316
| |
Collapse
|
12
|
Han X, Wang L, Li T, Zhang J, Zhang D, Li J, Xia Y, Liu Y, Tan W. Beyond Blocking: Engineering RNAi-Mediated Targeted Immune Checkpoint Nanoblocker Enables T-Cell-Independent Cancer Treatment. ACS NANO 2020; 14:17524-17534. [PMID: 33290659 DOI: 10.1021/acsnano.0c08022] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
The emergence of immune checkpoint blockade to activate host T cells to attack tumor cells has revolutionized the cancer treatment landscape over the past decade. However, sustained response has only been achieved in a small proportion of patients. This can be attributed to physiological barriers, such as T-cell heterogeneity and immunosuppressive tumor microenvironments. To this can be added obstacles intrinsic to traditional antibody-driven blockade methods, including the inability to inhibit checkpoint translocation from cytoplasm, systemic immune toxicity, and "bite back" effect on T cells. Using non-small cell lung cancer (NSCLC) as the cancer model, here we report an unconventional, yet powerful, tumor-targeted checkpoint blocking strategy by RNAi nanoengineering for T-cell-independent cancer therapy. Unlike antibodies, such nanoblocker silences both membranous and cytoplasmic PD-L1 in cancer cells, thus eliminating the binding step. Moreover, it is demonstrated that silencing of PD-L1 by the nanoblocker can cause the direct programmed cell death of NSCLC H460 cells, without the need of T-cell intervention. In vivo results from xenograft tumor models further demonstrate that tumor-homing peptide modification enables the nanoblocker to accumulate in the tumor tissue, downregulate the PD-L1 expression, and inhibit the tumor growth more efficiently than the nontargeted group. These findings may offer an effective means toward overcoming barriers against traditional checkpoint blockade and provide different insights into the molecular mechanism(s) underlying immunotherapy.
Collapse
Affiliation(s)
- Xiaoyan Han
- Molecular Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha 410082, China
| | - Linlin Wang
- Molecular Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha 410082, China
| | - Ting Li
- Molecular Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha 410082, China
| | - Jiahui Zhang
- Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, Shanghai 200025, China
| | - Dailiang Zhang
- Molecular Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha 410082, China
| | - Jili Li
- Molecular Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha 410082, China
| | - Yinghao Xia
- Molecular Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha 410082, China
| | - Yanlan Liu
- Molecular Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha 410082, China
| | - Weihong Tan
- Molecular Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha 410082, China
- Institute of Molecular Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, College of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200040, China
- Institute of Cancer and Basic Medicine (IBMC), Chinese Academy of Sciences; The Cancer Hospital of the University of Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| |
Collapse
|
13
|
Ferreira TH, de Oliveira Freitas LB, Fernandes RS, dos Santos VM, Resende JM, Cardoso VN, de Barros ALB, de Sousa EMB. Boron nitride nanotube-CREKA peptide as an effective target system to metastatic breast cancer. JOURNAL OF PHARMACEUTICAL INVESTIGATION 2019. [DOI: 10.1007/s40005-019-00467-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
14
|
Abstract
Creating an optimal microenvironment that supports angiogenesis, cell-cell cross talk, cell migration, and differentiation is crucial for pulp/dentin regeneration. It was shown that dental stem cells being seeded onto a scaffold and transplanted in vivo could give rise to a new tissue similar to that of the native pulp. However, the unique structure of the tooth with a pulp space encased within hard dentin allows only a single blood supply from a small apical opening located at the apex of the root canals. Therefore, a further strategy that can address this limitation such as the incorporation of endothelial/endothelial progenitor cells or cells with high angiogenic potential into the transplant is required so that the added cells can contribute to the vascularization within the implant. However, the placement of 2 or more different cell types inside 3-dimensional porous scaffolds is technologically challenging. In contrast to the conventional scaffolding approach, self-assembly of monodispersed cells into 3-dimensional tissue mimics permits true physiological interactions between and among different types of cells without any influence from a secondary material. In this review, we discuss potential strategies that can be used in vasculature engineering in dental pulp regeneration with a specific emphasis on combining prevascularization and scaffold-based or scaffold-free approaches.
Collapse
Affiliation(s)
| | - Chengfei Zhang
- Endodontology, Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
15
|
Spicer CD, Jumeaux C, Gupta B, Stevens MM. Peptide and protein nanoparticle conjugates: versatile platforms for biomedical applications. Chem Soc Rev 2018; 47:3574-3620. [PMID: 29479622 PMCID: PMC6386136 DOI: 10.1039/c7cs00877e] [Citation(s) in RCA: 303] [Impact Index Per Article: 43.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Peptide- and protein-nanoparticle conjugates have emerged as powerful tools for biomedical applications, enabling the treatment, diagnosis, and prevention of disease. In this review, we focus on the key roles played by peptides and proteins in improving, controlling, and defining the performance of nanotechnologies. Within this framework, we provide a comprehensive overview of the key sequences and structures utilised to provide biological and physical stability to nano-constructs, direct particles to their target and influence their cellular and tissue distribution, induce and control biological responses, and form polypeptide self-assembled nanoparticles. In doing so, we highlight the great advances made by the field, as well as the challenges still faced in achieving the clinical translation of peptide- and protein-functionalised nano-drug delivery vehicles, imaging species, and active therapeutics.
Collapse
Affiliation(s)
- Christopher D Spicer
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Scheeles Väg 2, Stockholm, Sweden.
| | | | | | | |
Collapse
|
16
|
Xiong H, Du S, Zhang P, Jiang Z, Zhou J, Yao J. Primary tumor and pre-metastatic niches co-targeting “peptides-lego” hybrid hydroxyapatite nanoparticles for metastatic breast cancer treatment. Biomater Sci 2018; 6:2591-2604. [DOI: 10.1039/c8bm00706c] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Hybrid hydroxyapatite nanoparticles orchestrating tumor metastasis resisting therapy (TMRT) and tumor metastasis targeting therapy (TMTT).
Collapse
Affiliation(s)
- Hui Xiong
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Druggability of Biopharmaceuticals
- Department of Pharmaceutics
- China Pharmaceutical University
- Nanjing 210009
- China
| | - Shi Du
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Druggability of Biopharmaceuticals
- Department of Pharmaceutics
- China Pharmaceutical University
- Nanjing 210009
- China
| | - Ping Zhang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Druggability of Biopharmaceuticals
- Department of Pharmaceutics
- China Pharmaceutical University
- Nanjing 210009
- China
| | - Zhijie Jiang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Druggability of Biopharmaceuticals
- Department of Pharmaceutics
- China Pharmaceutical University
- Nanjing 210009
- China
| | - Jianping Zhou
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Druggability of Biopharmaceuticals
- Department of Pharmaceutics
- China Pharmaceutical University
- Nanjing 210009
- China
| | - Jing Yao
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Druggability of Biopharmaceuticals
- Department of Pharmaceutics
- China Pharmaceutical University
- Nanjing 210009
- China
| |
Collapse
|
17
|
Zhang B, Hu Y, Pang Z. Modulating the Tumor Microenvironment to Enhance Tumor Nanomedicine Delivery. Front Pharmacol 2017; 8:952. [PMID: 29311946 PMCID: PMC5744178 DOI: 10.3389/fphar.2017.00952] [Citation(s) in RCA: 95] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Accepted: 12/15/2017] [Indexed: 12/18/2022] Open
Abstract
Nanomedicines including liposomes, micelles, and nanoparticles based on the enhanced permeability and retention (EPR) effect have become the mainstream for tumor treatment owing to their superiority over conventional anticancer agents. Advanced design of nanomedicine including active targeting nanomedicine, tumor-responsive nanomedicine, and optimization of physicochemical properties to enable highly effective delivery of nanomedicine to tumors has further improved their therapeutic benefits. However, these strategies still could not conquer the delivery barriers of a tumor microenvironment such as heterogeneous blood flow, dense extracellular matrix, abundant stroma cells, and high interstitial fluid pressure, which severely impaired vascular transport of nanomedicines, hindered their effective extravasation, and impeded their interstitial transport to realize uniform distribution inside tumors. Therefore, modulation of tumor microenvironment has now emerged as an important strategy to improve nanomedicine delivery to tumors. Here, we review the existing strategies and approaches for tumor microenvironment modulation to improve tumor perfusion for helping more nanomedicines to reach the tumor site, to facilitate nanomedicine extravasation for enhancing transvascular transport, and to improve interstitial transport for optimizing the distribution of nanomedicines. These strategies may provide an avenue for the development of new combination chemotherapeutic regimens and reassessment of previously suboptimal agents.
Collapse
Affiliation(s)
- Bo Zhang
- School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai, China
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu Hu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhiqing Pang
- School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai, China
| |
Collapse
|
18
|
Deletion of endothelial cell-specific liver kinase B1 increases angiogenesis and tumor growth via vascular endothelial growth factor. Oncogene 2017; 36:4277-4287. [PMID: 28346429 PMCID: PMC5532072 DOI: 10.1038/onc.2017.61] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Revised: 01/18/2017] [Accepted: 02/09/2017] [Indexed: 12/25/2022]
Abstract
Liver kinase B1 (LKB1) is a serine/threonine protein kinase ubiquitously expressed in mammalian cells. It was first identified in Peutz-Jeghers syndrome as a tumor suppressor gene. Whether endothelial LKB1 regulates angiogenesis and tumor growth is unknown. In this study, we generated endothelial cell-specific LKB1-knockout (LKB1endo−/−) mice by crossbreeding vascular endothelial-cadherin-Cre mice with LKB1flox/flox mice. Vascular endothelial growth factor (VEGF) level was highly co-stained in endothelial cells but not macrophages in LKB1endo−/− mice. Consistently, LKB1endo−/− mouse tissues including the lung, skin, kidney, and liver showed increased vascular permeability. Tumors implanted in LKB1endo−/− mice but not macrophage-specific LKB1-knockout mice grew faster and showed enhanced vascular permeability and increased angiogenesis as compared with those implanted in wild-type mice. Injection of VEGF-neutralizing antibody but not the isotype-matched control antibody decreased endothelial-cell angiogenesis and tumor growth in vivo. Furthermore, LKB1 deletion enhanced mouse retinal and cell angiogenesis, and knockdown of VEGF by small-interfering RNA decreased endothelial cell proliferation and migration. Re-expression of LKB1 or knockdown of VEGF receptor 2 decreased the over-proliferation and -migration observed in LKB1endo−/− cells. Mechanistically, LKB1 could bind to the VEGF transcription factor, specificity protein 1 (Sp1), which then inhibited the binding of Sp1 to the VEGF promoter to reduce VEGF expression. Endothelial LKB1 may regulate endothelial angiogenesis and tumor growth by modulating Sp1-mediated VEGF expression.
Collapse
|
19
|
Heinolainen K, Karaman S, D'Amico G, Tammela T, Sormunen R, Eklund L, Alitalo K, Zarkada G. VEGFR3 Modulates Vascular Permeability by Controlling VEGF/VEGFR2 Signaling. Circ Res 2017; 120:1414-1425. [PMID: 28298294 DOI: 10.1161/circresaha.116.310477] [Citation(s) in RCA: 111] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 03/08/2017] [Accepted: 03/14/2017] [Indexed: 01/02/2023]
Abstract
RATIONALE Vascular endothelial growth factor (VEGF) is the main driver of angiogenesis and vascular permeability via VEGF receptor 2 (VEGFR2), whereas lymphangiogenesis signals are transduced by VEGFC/D via VEGFR3. VEGFR3 also regulates sprouting angiogenesis and blood vessel growth, but to what extent VEGFR3 signaling controls blood vessel permeability remains unknown. OBJECTIVE To investigate the role of VEGFR3 in the regulation of VEGF-induced vascular permeability. METHODS AND RESULTS Long-term global Vegfr3 gene deletion in adult mice resulted in increased fibrinogen deposition in lungs and kidneys, indicating enhanced vascular leakage at the steady state. Short-term deletion of Vegfr3 in blood vascular endothelial cells increased baseline leakage in various tissues, as well as in tumors, and exacerbated vascular permeability in response to VEGF, administered via intradermal adenoviral delivery or through systemic injection of recombinant protein. VEGFR3 gene silencing upregulated VEGFR2 protein levels and phosphorylation in cultured endothelial cells. Consistent with elevated VEGFR2 activity, vascular endothelial cadherin showed reduced localization at endothelial cell-cell junctions in postnatal retinas after Vegfr3 deletion, or after VEGFR3 silencing in cultured endothelial cells. Furthermore, concurrent deletion of Vegfr2 prevented VEGF-induced excessive vascular leakage in mice lacking Vegfr3. CONCLUSIONS VEGFR3 limits VEGFR2 expression and VEGF/VEGFR2 pathway activity in quiescent and angiogenic blood vascular endothelial cells, thereby preventing excessive vascular permeability.
Collapse
Affiliation(s)
- Krista Heinolainen
- From the Wihuri Research Institute and Translational Cancer Biology Research Program, Biomedicum Helsinki, University of Helsinki, Finland (K.H., S.K., G.D'A., T.T., K.A., G.Z.); Biocenter Oulu and Department of Pathology, University of Oulu and Oulu University Hospital, Finland (R.S.); and Oulu Center for Cell-Matrix Research, Biocenter Oulu and Faculty of Biochemistry and Molecular Medicine, University of Oulu, Finland (L.E.)
| | - Sinem Karaman
- From the Wihuri Research Institute and Translational Cancer Biology Research Program, Biomedicum Helsinki, University of Helsinki, Finland (K.H., S.K., G.D'A., T.T., K.A., G.Z.); Biocenter Oulu and Department of Pathology, University of Oulu and Oulu University Hospital, Finland (R.S.); and Oulu Center for Cell-Matrix Research, Biocenter Oulu and Faculty of Biochemistry and Molecular Medicine, University of Oulu, Finland (L.E.)
| | - Gabriela D'Amico
- From the Wihuri Research Institute and Translational Cancer Biology Research Program, Biomedicum Helsinki, University of Helsinki, Finland (K.H., S.K., G.D'A., T.T., K.A., G.Z.); Biocenter Oulu and Department of Pathology, University of Oulu and Oulu University Hospital, Finland (R.S.); and Oulu Center for Cell-Matrix Research, Biocenter Oulu and Faculty of Biochemistry and Molecular Medicine, University of Oulu, Finland (L.E.)
| | - Tuomas Tammela
- From the Wihuri Research Institute and Translational Cancer Biology Research Program, Biomedicum Helsinki, University of Helsinki, Finland (K.H., S.K., G.D'A., T.T., K.A., G.Z.); Biocenter Oulu and Department of Pathology, University of Oulu and Oulu University Hospital, Finland (R.S.); and Oulu Center for Cell-Matrix Research, Biocenter Oulu and Faculty of Biochemistry and Molecular Medicine, University of Oulu, Finland (L.E.)
| | - Raija Sormunen
- From the Wihuri Research Institute and Translational Cancer Biology Research Program, Biomedicum Helsinki, University of Helsinki, Finland (K.H., S.K., G.D'A., T.T., K.A., G.Z.); Biocenter Oulu and Department of Pathology, University of Oulu and Oulu University Hospital, Finland (R.S.); and Oulu Center for Cell-Matrix Research, Biocenter Oulu and Faculty of Biochemistry and Molecular Medicine, University of Oulu, Finland (L.E.)
| | - Lauri Eklund
- From the Wihuri Research Institute and Translational Cancer Biology Research Program, Biomedicum Helsinki, University of Helsinki, Finland (K.H., S.K., G.D'A., T.T., K.A., G.Z.); Biocenter Oulu and Department of Pathology, University of Oulu and Oulu University Hospital, Finland (R.S.); and Oulu Center for Cell-Matrix Research, Biocenter Oulu and Faculty of Biochemistry and Molecular Medicine, University of Oulu, Finland (L.E.)
| | - Kari Alitalo
- From the Wihuri Research Institute and Translational Cancer Biology Research Program, Biomedicum Helsinki, University of Helsinki, Finland (K.H., S.K., G.D'A., T.T., K.A., G.Z.); Biocenter Oulu and Department of Pathology, University of Oulu and Oulu University Hospital, Finland (R.S.); and Oulu Center for Cell-Matrix Research, Biocenter Oulu and Faculty of Biochemistry and Molecular Medicine, University of Oulu, Finland (L.E.)
| | - Georgia Zarkada
- From the Wihuri Research Institute and Translational Cancer Biology Research Program, Biomedicum Helsinki, University of Helsinki, Finland (K.H., S.K., G.D'A., T.T., K.A., G.Z.); Biocenter Oulu and Department of Pathology, University of Oulu and Oulu University Hospital, Finland (R.S.); and Oulu Center for Cell-Matrix Research, Biocenter Oulu and Faculty of Biochemistry and Molecular Medicine, University of Oulu, Finland (L.E.).
| |
Collapse
|
20
|
Ando M, Fujimoto M, Takahashi Y, Nishikawa M, Hamana A, Takakura Y. Targeted Delivery of Interferon Gamma Using a Recombinant Fusion Protein of a Fibrin Clot-Binding Peptide With Interferon Gamma for Cancer Gene Therapy. J Pharm Sci 2016; 106:892-897. [PMID: 27939375 DOI: 10.1016/j.xphs.2016.11.018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Revised: 11/25/2016] [Accepted: 11/29/2016] [Indexed: 11/28/2022]
Abstract
Accelerated formation of fibrin clots in a tumor microenvironment can be used for targeted delivery of interferon gamma (IFNγ) to tumor cells. Here, we selected cysteine-arginine-glutamic acid-lysine-alanine (CREKA) as the fibrin clot-binding peptide and designed 2 types of fusion proteins for tumor targeting. The CREKA peptide was fused to IFNγ's C-terminus, with or without a matrix metalloproteinase-2 (MMP2)-cleavable linker (IFNγ-mmp-CREKA or IFNγ-CREKA, respectively). The former was designed to release IFNγ from IFNγ-mmp-CREKA bound to fibrin clots, to ensure IFNγ's function in the tumor milieu. IFNγ-activated sequence-dependent reporter gene expression in B16-BL6 cells revealed that the biological activities of IFNγ-CREKA and IFNγ were comparable, whereas that of IFNγ-mmp-CREKA was approximately 60% that of IFNγ. Plasma concentrations of IFNγ-CREKA and IFNγ-mmp-CREKA remained at effective levels for at least 4 weeks after gene transfer into mice. After gene transfer to tumor-bearing mice, intratumoral concentration of IFNγ in pCpG-IFNγ-mmp-CREKA group was tended to be higher than those of the other groups. Inhibition of colon-26 tumor growth was significantly more with gene transfer of IFNγ-mmp-CREKA than with IFNγ or IFNγ-CREKA. These results indicate that targeted delivery of IFNγ to fibrin clots through IFNγ-mmp-CREKA fusion can enhance the therapeutic efficacy of IFNγ in cancer gene therapy.
Collapse
Affiliation(s)
- Mitsuru Ando
- Department of Biopharmaceutics and Drug Metabolism, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto 606-8501, Japan
| | - Mai Fujimoto
- Department of Biopharmaceutics and Drug Metabolism, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto 606-8501, Japan
| | - Yuki Takahashi
- Department of Biopharmaceutics and Drug Metabolism, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto 606-8501, Japan
| | - Makiya Nishikawa
- Department of Biopharmaceutics and Drug Metabolism, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto 606-8501, Japan
| | - Atsushi Hamana
- Department of Biopharmaceutics and Drug Metabolism, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto 606-8501, Japan
| | - Yoshinobu Takakura
- Department of Biopharmaceutics and Drug Metabolism, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto 606-8501, Japan.
| |
Collapse
|
21
|
Kobayashi A, Oda T, Maeda H. Protein Binding of Macromolecular Anticancer Agent SMANCS: Characterization of Poly(styrene-co-maleic acid) Derivatives as an Albumin Binding Ligand. J BIOACT COMPAT POL 2016. [DOI: 10.1177/088391158800300401] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
We have studied the interaction of macromolecular anticancer agent SMANCS, a conjugate of partially half-butyl-esterified styrene-co-maleic acid polymer[butyl-SMA]- and neocarzinostatin (NCS), with various serum proteins by the fluorescence polarization method. Comparatively strong binding of FITC-labeled SMANCS (F-SMANCS) to human serum albumin (HSA) and weak binding to fibrinogen were observed, while other serum proteins did not exhibit any appreciable binding profile. From Scatchard polt analysis, the asso ciation constant of binding for F-SMANCS to HSA at 37 °C, pH 7.4 was calculated to be 2.19 × 106 M-1 and the number of moles of F-SMANCS bound to 1 mol of HSA was 3.2 Binding of F-NCS to HSA was not observed. The F-SMANCS bound to HSA was effectively displaced by butyl-SMA, but not by NCS. This evidence supports that SMANCS binds to HSA through butyl-SMA, not through NCS portion. A role of the alkyl ester groups of SMA derivatives to HSA binding was investigated by competitive inhibition using butyl-SMA, ethyl-SMA, H-SMA and short chain butyl-SMA. The degree of competitive in hibition was stronger in the following order: butyl-(n = 6) > ethyl (n = 6) = short butyl-(n = 4) > H-SMA. The number of carbons introduced by esterification and the hydrophobicity of SMA derivatives both positively in fluence the binding affinity to HSA. The binding site for SMANCS on HSA was investigated. From the competitive inhibition of known standard drugs (war farin, diazepam and digitoxin) and endogenous substance (bilirubin) in the albumin binding. The binding site on HSA appears to be in the close vicinity to the warfarin or diazepam binding site, and might partially overlap with the bilirubin binding site. These data support that the prolonged plasma half-life of SMANCS in vivo reported previously can be attributed to this albumin binding.
Collapse
Affiliation(s)
- Akira Kobayashi
- Department of Microbiology Kumamoto University Medical School Kumamoto 860, Japan
| | - Tatsuya Oda
- Department of Microbiology Kumamoto University Medical School Kumamoto 860, Japan
| | - Hiroshi Maeda
- Department of Microbiology Kumamoto University Medical School Kumamoto 860, Japan
| |
Collapse
|
22
|
Zhang B, Jiang T, She X, Shen S, Wang S, Deng J, Shi W, Mei H, Hu Y, Pang Z, Jiang X. Fibrin degradation by rtPA enhances the delivery of nanotherapeutics to A549 tumors in nude mice. Biomaterials 2016; 96:63-71. [PMID: 27149664 DOI: 10.1016/j.biomaterials.2016.04.015] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 04/13/2016] [Accepted: 04/14/2016] [Indexed: 11/26/2022]
Abstract
Effective drug delivery to a tumor depends on favorable blood perfusion within the tumor. As an important component of tumor extracellular matrix, fibrin is abundant near tumor vessels. Inspired by the distinct distribution pattern and vessel-dependent production of fibrin, we hypothesized that fibrin depletion in tumors decompresses tumor vessels to improve tumor blood perfusion and accordingly enhance drug delivery to tumors rich in vessels. In the present study, we attempted to employ a clinically used thrombolytic drug, recombinant tissue plasminogen activator (rtPA), to modulate fibrin deposition in tumors. We then combined this drug with a nanoparticle drug delivery system for tumor therapy. RtPA treatment (25 mg/kg/d i. p. administration for two weeks) successfully depleted fibrin deposition and enhanced blood perfusion within A549 tumor xenografts. Furthermore, rtPA treatment also improved the in vivo delivery of 115-nm nanoparticles to tumor tissues. Finally, rtPA combined with therapeutic agent-loaded nanoparticles resulted in the most effective shrinkage of A549 tumor xenografts compared with the control groups. Overall, the present study provides a new strategy to enhance the delivery of nanotherapeutics to tumors rich in vessels.
Collapse
Affiliation(s)
- Bo Zhang
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, Hubei, 430022, PR China
| | - Ting Jiang
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, Hubei, 430022, PR China
| | - Xiaojian She
- School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, 826 Zhangheng Road, Shanghai, 201203, PR China
| | - Shun Shen
- School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, 826 Zhangheng Road, Shanghai, 201203, PR China
| | - Sheng Wang
- School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, 826 Zhangheng Road, Shanghai, 201203, PR China
| | - Jun Deng
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, Hubei, 430022, PR China
| | - Wei Shi
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, Hubei, 430022, PR China
| | - Heng Mei
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, Hubei, 430022, PR China; Collaborative Innovation Center of Hematology, Huazhong University of Science and Technology, PR China
| | - Yu Hu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, Hubei, 430022, PR China; Collaborative Innovation Center of Hematology, Huazhong University of Science and Technology, PR China.
| | - Zhiqing Pang
- School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, 826 Zhangheng Road, Shanghai, 201203, PR China.
| | - Xinguo Jiang
- School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, 826 Zhangheng Road, Shanghai, 201203, PR China
| |
Collapse
|
23
|
Abstract
Similarities between tumors and the inflammatory response associated with wound healing have been recognized for more than 150 years and continue to intrigue. Some years ago, based on our then recent discovery of vascular permeability factor (VPF)/VEGF, I suggested that tumors behaved as wounds that do not heal. More particularly, I proposed that tumors co-opted the wound-healing response to induce the stroma they required for maintenance and growth. Work over the past few decades has supported this hypothesis and has put it on a firmer molecular basis. In outline, VPF/VEGF initiates a sequence of events in both tumors and wounds that includes the following: increased vascular permeability; extravasation of plasma, fibrinogen and other plasma proteins; activation of the clotting system outside the vascular system; deposition of an extravascular fibrin gel that serves as a provisional stroma and a favorable matrix for cell migration; induction of angiogenesis and arterio-venogenesis; subsequent degradation of fibrin and its replacement by "granulation tissue" (highly vascular connective tissue); and, finally, vascular resorption and collagen synthesis, resulting in the formation of dense fibrous connective tissue (called "scar tissue" in wounds and "desmoplasia" in cancer). A similar sequence of events also takes place in a variety of important inflammatory diseases that involve cellular immunity.
Collapse
Affiliation(s)
- Harold F Dvorak
- The Center for Vascular Biology Research and the Departments of Pathology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
24
|
The inflammatory actions of coagulant and fibrinolytic proteases in disease. Mediators Inflamm 2015; 2015:437695. [PMID: 25878399 PMCID: PMC4387953 DOI: 10.1155/2015/437695] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2014] [Revised: 03/02/2015] [Accepted: 03/16/2015] [Indexed: 12/30/2022] Open
Abstract
Aside from their role in hemostasis, coagulant and fibrinolytic proteases are important mediators of inflammation in diseases such as asthma, atherosclerosis, rheumatoid arthritis, and cancer. The blood circulating zymogens of these proteases enter damaged tissue as a consequence of vascular leak or rupture to become activated and contribute to extravascular coagulation or fibrinolysis. The coagulants, factor Xa (FXa), factor VIIa (FVIIa), tissue factor, and thrombin, also evoke cell-mediated actions on structural cells (e.g., fibroblasts and smooth muscle cells) or inflammatory cells (e.g., macrophages) via the proteolytic activation of protease-activated receptors (PARs). Plasmin, the principle enzymatic mediator of fibrinolysis, also forms toll-like receptor-4 (TLR-4) activating fibrin degradation products (FDPs) and can release latent-matrix bound growth factors such as transforming growth factor-β (TGF-β). Furthermore, the proteases that convert plasminogen into plasmin (e.g., urokinase plasminogen activator) evoke plasmin-independent proinflammatory actions involving coreceptor activation. Selectively targeting the receptor-mediated actions of hemostatic proteases is a strategy that may be used to treat inflammatory disease without the bleeding complications of conventional anticoagulant therapies. The mechanisms by which proteases of the coagulant and fibrinolytic systems contribute to extravascular inflammation in disease will be considered in this review.
Collapse
|
25
|
Wang C, Wang X, Zhong T, Zhao Y, Zhang WQ, Ren W, Huang D, Zhang S, Guo Y, Yao X, Tang YQ, Zhang X, Zhang Q. The antitumor activity of tumor-homing peptide-modified thermosensitive liposomes containing doxorubicin on MCF-7/ADR: in vitro and in vivo. Int J Nanomedicine 2015; 10:2229-48. [PMID: 25834435 PMCID: PMC4372005 DOI: 10.2147/ijn.s79840] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Clotted plasma proteins are present on the walls of tumor vessels and in tumor stroma. Tumor-homing peptide Cys-Arg-Glu-Lys-Ala (CREKA) could recognize the clotted plasma proteins in tumor vessels. Thermosensitive liposomes could immediately release the encapsulated drug in the vasculature of the heated tumor. In this study, we designed a novel form of targeted thermosensitive liposomes, CREKA-modified lysolipid-containing thermosensitive liposomes (LTSLs), containing doxorubicin (DOX) (DOX-LTSL-CREKA), to investigate the hypothesis that DOX-LTSL-CREKA might target the clotted plasma proteins in tumor vessels as well as tumor stroma and then exhibit burst release of the encapsulated DOX at the heated tumor site. We also hypothesized that the high local drug concentration produced by these thermosensitive liposomes after local hyperthermia treatment will be useful for treatment of multidrug resistance. The multidrug-resistant human breast adenocarcinoma (MCF-7/ADR) cell line was chosen as a tumor cell model, and the targeting and immediate release characteristics of DOX-LTSL-CREKA were investigated in vitro and in vivo. Furthermore, the antitumor activity of DOX-LTSL-CREKA was evaluated in MCF-7/ADR tumor-bearing nude mice in vivo. The targeting effect of the CREKA-modified thermosensitive liposomes on the clotted plasma proteins was confirmed in our in vivo imaging and immunohistochemistry experiments. The burst release of this delivery system was observed in our in vitro temperature-triggered DOX release and flow cytometry analysis and also by confocal microscopy experiments. The antitumor activity of the DOX-LTSL-CREKA was confirmed in tumor-bearing nude mice in vivo. Our findings suggest that the combination of targeting the clotted plasma proteins in the tumor vessel wall as well as tumor stroma by using CREKA peptide and temperature-triggered drug release from liposomes by using thermosensitive liposomes offers an attractive strategy for chemotherapeutic drug delivery to tumors.
Collapse
Affiliation(s)
- Chao Wang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing, People's Republic of China
| | - Xin Wang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing, People's Republic of China
| | - Ting Zhong
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing, People's Republic of China
| | - Yang Zhao
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing, People's Republic of China
| | - Wei-Qiang Zhang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing, People's Republic of China
| | - Wei Ren
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing, People's Republic of China
| | - Dan Huang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing, People's Republic of China
| | - Shuang Zhang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing, People's Republic of China
| | - Yang Guo
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing, People's Republic of China
| | - Xin Yao
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing, People's Republic of China
| | - Yi-Qun Tang
- Department of Clinical Pharmacy, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Xuan Zhang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing, People's Republic of China
| | - Qiang Zhang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing, People's Republic of China ; State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, People's Republic of China
| |
Collapse
|
26
|
Al-Hilal TA, Alam F, Park JW, Kim K, Kwon IC, Ryu GH, Byun Y. Prevention effect of orally active heparin conjugate on cancer-associated thrombosis. J Control Release 2014; 195:155-61. [DOI: 10.1016/j.jconrel.2014.05.027] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2014] [Revised: 05/08/2014] [Accepted: 05/16/2014] [Indexed: 12/11/2022]
|
27
|
Wu J, Zhao J, Zhang B, Qian Y, Gao H, Yu Y, Wei Y, Yang Z, Jiang X, Pang Z. Polyethylene glycol-polylactic acid nanoparticles modified with cysteine-arginine-glutamic acid-lysine-alanine fibrin-homing peptide for glioblastoma therapy by enhanced retention effect. Int J Nanomedicine 2014; 9:5261-71. [PMID: 25419130 PMCID: PMC4235507 DOI: 10.2147/ijn.s72649] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
For a nanoparticulate drug-delivery system, crucial challenges in brain-glioblastoma therapy are its poor penetration and retention in the glioblastoma parenchyma. As a prevailing component in the extracellular matrix of many solid tumors, fibrin plays a critical role in the maintenance of glioblastoma morphology and glioblastoma cell differentiation and proliferation. We developed a new drug-delivery system by conjugating polyethylene glycol–polylactic acid nanoparticles (NPs) with cysteine–arginine–glutamic acid–lysine–alanine (CREKA; TNPs), a peptide with special affinity for fibrin, to mediate glioblastoma-homing and prolong NP retention at the tumor site. In vitro binding tests indicated that CREKA significantly enhanced specific binding of NPs with fibrin. In vivo fluorescence imaging of glioblastoma-bearing nude mice, ex vivo brain imaging, and glioblastoma distribution demonstrated that TNPs had higher accumulation and longer retention in the glioblastoma site over unmodified NPs. Furthermore, pharmacodynamic results showed that paclitaxel-loaded TNPs significantly prolonged the median survival time of intracranial U87 glioblastoma-bearing nude mice compared with controls, Taxol, and NPs. These findings suggested that TNPs were able to target the glioblastoma and enhance retention, which is a valuable strategy for tumor therapy.
Collapse
Affiliation(s)
- Junzhu Wu
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai, People's Republic of China ; School of Pharmacy, Dali University, Xiaguan, People's Republic of China
| | - Jingjing Zhao
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai, People's Republic of China ; School of Pharmacy, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Bo Zhang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai, People's Republic of China
| | - Yong Qian
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai, People's Republic of China
| | - Huile Gao
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai, People's Republic of China
| | - Yuan Yu
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai, People's Republic of China
| | - Yan Wei
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai, People's Republic of China
| | - Zhi Yang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai, People's Republic of China
| | - Xinguo Jiang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai, People's Republic of China
| | - Zhiqing Pang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai, People's Republic of China
| |
Collapse
|
28
|
Abstract
Systemic administration of antiangiogenic drugs that target components of the vascular endothelial growth factor A (VEGF-A; VEGF) signal transduction pathway has become a viable therapeutic option for patients with various types of cancer. Nevertheless, these drugs can drive alterations in healthy vasculatures, which in turn are associated with adverse effects in healthy tissues. VEGF is crucial for vascular homeostasis and the maintenance of vascular integrity and architecture in endocrine organs. Given these critical physiological functions, systemic delivery of drugs that target VEGF signalling can block VEGF-mediated vascular functions in endocrine organs, such as the thyroid gland, and lead to endocrine dysfunction, including hypothyroidism, adrenal insufficiency and altered insulin sensitivity. This Review discusses emerging evidence from preclinical and clinical studies that contributes to understanding the mechanisms that underlie the vascular changes and subsequent modulations of endocrine function that are induced by targeted inhibition of VEGF signalling. Understanding these mechanisms is crucial for the design of antiangiogenic drugs with minimal associated adverse effects that will enable effective treatment of patients with cancer.
Collapse
Affiliation(s)
- Yihai Cao
- Department of Microbiology, Tumour and Cell Biology, Karolinska Institutet, Nobels vag 16, 17177 Stockholm, Sweden
| |
Collapse
|
29
|
Patel AR, Chougule MB, Lim E, Francis KP, Safe S, Singh M. Theranostic tumor homing nanocarriers for the treatment of lung cancer. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2014; 10:1053-1063. [PMID: 24355163 PMCID: PMC4061286 DOI: 10.1016/j.nano.2013.12.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2013] [Revised: 11/25/2013] [Accepted: 12/07/2013] [Indexed: 12/31/2022]
Abstract
UNLABELLED The drugs/strategies to selectively inhibit tumor blood supply have generated interest in recent years for enhancement of cancer therapeutics. The objective of this study was to formulate tumor homing PEGylated CREKA peptide conjugated theranostic nanoparticles of DIM-C-pPhC6H5 (DIM-P) and investigate in vivo antitumor activity as well as evaluate the targeted efficiency to lung tumors using imaging techniques. DIM-P loaded Nanoparticles (NCs-D) were prepared using lipids, and DOGS-NTA-Ni and the surface of NCs-D were modified with PEGylated CREKA peptide (PCNCs-D). PCNCs-D showed 3 fold higher binding to clotted plasma proteins in tumor vasculature compared to NCs-D. PCNCs-D showed 26%±4% and 22%±5% increase in tumor reduction compared to NCs-D in metastatic and orthotopic models respectively. In-vivo imaging studies showed ~40 folds higher migration of PCNCs-Di in tumor vasculature than NCs-Di. Our studies demonstrate the role of PCNCs-D as theranostic tumor homing drug delivery and imaging systems for lung cancer diagnosis and treatment. FROM THE CLINICAL EDITOR This study demonstrates a very efficient delivery system to address lung cancer growth through blood supply inhibition.
Collapse
Affiliation(s)
- Apurva R Patel
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, USA
| | - Mahavir B Chougule
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Hawai'i at Hilo, Hilo, HI, USA
| | - Ed Lim
- Calipers-Life Sciences & Technology, A Perkin Elmer Company, Alameda, CA, USA
| | - Kevin P Francis
- Calipers-Life Sciences & Technology, A Perkin Elmer Company, Alameda, CA, USA
| | - Stephen Safe
- Institute of Biosciences and Technology, Texas A&M University, Houston, TX, USA
| | - Mandip Singh
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, USA.
| |
Collapse
|
30
|
Galectin-1 induces vascular permeability through the neuropilin-1/vascular endothelial growth factor receptor-1 complex. Angiogenesis 2014; 17:839-49. [PMID: 24719187 DOI: 10.1007/s10456-014-9431-8] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2014] [Accepted: 04/04/2014] [Indexed: 10/25/2022]
Abstract
Galectin-1 (Gal-1) is a β-galactoside-binding lectin that regulates endothelial cell migration, proliferation, and adhesion. However, the effect of Gal-1 on vascular permeability and the underlying mechanisms are unclear. We found that high Gal-1 expression was associated with elevated tumor vascular permeability in specimens of oral squamous cell carcinoma. Using transendothelial passage of FITC-dextran and a Miles assay, we demonstrated that Gal-1 increased vascular permeability extracellularly through its carbohydrate recognition domain. Mechanism dissection revealed that the neuropilin (NRP)-1/vascular endothelial growth factor receptor- (VEGFR)-1 complex was required for Gal-1-regulated vascular permeability. Activation of VEGFR-1 triggered activation of Akt which led to a reduction in vascular endothelial-cadherin at cell-cell junctions and resulted in cytoskeletal rearrangement. Both inhibition of Gal-1 secreted from cancer cells and administration of an anti-Gal-1 antibody in the tumor microenvironment suppressed tumor growth and vascular permeability in xenograft models. In conclusion, our results demonstrate a novel function of Gal-1 of increasing vascular permeability through the NRP-1/VEGFR1 and Akt signaling pathway and indicate that targeting Gal-1 by an anti-Gal-1 antibody is a feasible therapy for vascular hyperpermeability and cancer.
Collapse
|
31
|
Boulaftali Y, Hess PR, Kahn ML, Bergmeier W. Platelet immunoreceptor tyrosine-based activation motif (ITAM) signaling and vascular integrity. Circ Res 2014; 114:1174-84. [PMID: 24677237 PMCID: PMC4000726 DOI: 10.1161/circresaha.114.301611] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2013] [Accepted: 02/18/2014] [Indexed: 01/27/2023]
Abstract
Platelets are well-known for their critical role in hemostasis, that is, the prevention of blood loss at sites of mechanical vessel injury. Inappropriate platelet activation and adhesion, however, can lead to thrombotic complications, such as myocardial infarction and stroke. To fulfill its role in hemostasis, the platelet is equipped with various G protein-coupled receptors that mediate the response to soluble agonists such as thrombin, ADP, and thromboxane A2. In addition to G protein-coupled receptors, platelets express 3 glycoproteins that belong to the family of immunoreceptor tyrosine-based activation motif receptors: Fc receptor γ chain, which is noncovalently associated with the glycoprotein VI collagen receptor, C-type lectin 2, the receptor for podoplanin, and Fc receptor γII A, a low-affinity receptor for immune complexes. Although both genetic and chemical approaches have documented a critical role for platelet G protein-coupled receptors in hemostasis, the contribution of immunoreceptor tyrosine-based activation motif receptors to this process is less defined. Studies performed during the past decade, however, have identified new roles for platelet immunoreceptor tyrosine-based activation motif signaling in vascular integrity in utero and at sites of inflammation. The purpose of this review is to summarize recent findings on how platelet immunoreceptor tyrosine-based activation motif signaling controls vascular integrity, both in the presence and absence of mechanical injury.
Collapse
Affiliation(s)
- Yacine Boulaftali
- From the McAllister Heart Institute (Y.B., W.B.) and Department of Biochemistry and Biophysics (W.B.), University of North Carolina, Chapel Hill; and Department of Medicine and Division of Cardiology, University of Pennsylvania, Philadelphia (P.R.H., M.L.K.)
| | | | | | | |
Collapse
|
32
|
Zhang B, Shen S, Liao Z, Shi W, Wang Y, Zhao J, Hu Y, Yang J, Chen J, Mei H, Hu Y, Pang Z, Jiang X. Targeting fibronectins of glioma extracellular matrix by CLT1 peptide-conjugated nanoparticles. Biomaterials 2014; 35:4088-98. [PMID: 24513320 DOI: 10.1016/j.biomaterials.2014.01.046] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Accepted: 01/19/2014] [Indexed: 01/14/2023]
Abstract
The abundant extracellular matrix (ECM) in the glioma microenvironment play a critical role in the maintenance of glioma morphology, glioma cells differentiation and proliferation, but little has been done to understand the feasibility of ECM as the therapeutic target for glioma therapy. In this study, a drug delivery system targeting fibronectins (FNs), a prevailing component in the ECM of many solid tumors, was constructed for glioma therapy based on the interaction between the abundant FNs in glioma tissues and the FNs-targeting moiety CLT1 peptide. CLT1 peptide was successfully conjugated to PEG-PLA nanoparticles (CNP). FNs were demonstrated to be highly expressed in the ECM of glioma spheroids in vitro and glioma tissues in vivo. CLT1 modification favored targeting nanoparticles penetration into the core of glioma spheroids and consequently induced more severe inhibitive effects on glioma spheroids growth than traditional NP. In vivo imaging, ex vivo imaging and glioma tissue slides showed that CNP enhanced nanoparticles retention in glioma site, distributed more extensively and more deeply into glioma tissues than that of conventional NP, and mainly located in glioma cells rather than in extracellular matrix as conventional NP. Pharmacodynamics outcomes revealed that the median survival time of glioma-bearing mice models treated with paclitaxel-loaded CNP (CNP-PTX) was significantly prolonged when compared with that of any other group. TUNEL assay demonstrated that more extensive cell apoptosis was induced by CNP-PTX treatment compared with other treatments. Altogether, these promising results indicated that this ECM-targeting drug delivery system enhanced retention and glioma cell uptake of nanoparticles and might have a great potential for glioma therapy in clinical applications.
Collapse
Affiliation(s)
- Bo Zhang
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, Hubei 430022, PR China
| | - Shun Shen
- School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, 826 Zhangheng Road, Shanghai 201203, PR China
| | - Ziwei Liao
- School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, 826 Zhangheng Road, Shanghai 201203, PR China
| | - Wei Shi
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, Hubei 430022, PR China
| | - Yu Wang
- School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, 826 Zhangheng Road, Shanghai 201203, PR China
| | - Jingjing Zhao
- School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, 826 Zhangheng Road, Shanghai 201203, PR China
| | - Yue Hu
- College of Pharmacy, Jiamusi University, Jiamusi 154007, PR China
| | - Jiarong Yang
- College of Pharmacy, Jiamusi University, Jiamusi 154007, PR China
| | - Jun Chen
- School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, 826 Zhangheng Road, Shanghai 201203, PR China
| | - Heng Mei
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, Hubei 430022, PR China
| | - Yu Hu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, Hubei 430022, PR China.
| | - Zhiqing Pang
- School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, 826 Zhangheng Road, Shanghai 201203, PR China.
| | - Xinguo Jiang
- School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, 826 Zhangheng Road, Shanghai 201203, PR China
| |
Collapse
|
33
|
Abstract
Glioblastoma-targeted drug delivery systems facilitate efficient delivery of chemotherapeutic agents to malignant gliomas, while minimizing systemic toxicity and side effects. Taking advantage of the fibrin deposition that is characteristic of tumors, we constructed spherical, Cy7-labeled, targeting micelles to glioblastoma through the addition of the fibrin-binding pentapeptide, cysteine–arginine–glutamic acid–lysine–alanine, or CREKA. Conjugation of the CREKA peptide to Cy7-micelles increased the average particle size and zeta potential. Upon intravenous administration to GL261 glioma bearing mice, Cy7-micelles passively accumulated at the brain tumor site via the enhanced permeability and retention (EPR) effect, and Cy7-CREKA-micelles displayed enhanced tumor homing via active targeting as early as 1 h after administration, as confirmed via in vivo and ex vivo imaging and immunohistochemistry. Biodistribution of micelles showed an accumulation within the liver and kidneys, leading to micelle elimination via renal clearance and the reticuloendothelial system (RES). Histological evaluation showed no signs of cytotoxicity or tissue damage, confirming the safety and utility of this nanoparticle system for delivery to glioblastoma. Our findings offer strong evidence for the glioblastoma-targeting potential of CREKA-micelles and provide the foundation for CREKA-mediated, targeted therapy of glioma.
Collapse
|
34
|
Ruoslahti E. Peptides as targeting elements and tissue penetration devices for nanoparticles. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2012; 24:3747-56. [PMID: 22550056 PMCID: PMC3947925 DOI: 10.1002/adma.201200454] [Citation(s) in RCA: 321] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2012] [Revised: 02/29/2012] [Indexed: 04/14/2023]
Abstract
The use of nanoparticles in medicine (nanomedicine) has recently become an intensely studied field. Nanoparticles carrying drugs and imaging agents have already reached the clinic, but they are essentially passive delivery vehicles, not what are referred to as "smart" nanoparticles. An important function to add to make nanoparticles smarter is active homing to the target tissue. It makes nanoparticles accumulate in the target tissue at higher concentrations than would be the case without this feature, increasing therapeutic efficacy and reducing side effects. This review discusses the recent developments in the nanoparticle targeting field with emphasis on peptides that home to vascular "zip codes" in target tissues and provide a tissue- and cell-penetrating function.
Collapse
Affiliation(s)
- Erkki Ruoslahti
- Center for Nanomedicine, UCSB, Biology II Bldg., University of California, Santa Barbara, CA 93106-9610, USA.
| |
Collapse
|
35
|
Schäfer B, Piliponsky AM, Oka T, Song CH, Gerard NP, Gerard C, Tsai M, Kalesnikoff J, Galli SJ. Mast cell anaphylatoxin receptor expression can enhance IgE-dependent skin inflammation in mice. J Allergy Clin Immunol 2012; 131:541-8.e1-9. [PMID: 22728083 DOI: 10.1016/j.jaci.2012.05.009] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2011] [Revised: 05/07/2012] [Accepted: 05/11/2012] [Indexed: 12/29/2022]
Abstract
BACKGROUND Mast cells express receptors for complement anaphylatoxins C3a and C5a (ie, C3a receptor [C3aR] and C5a receptor [C5aR]), and C3a and C5a are generated during various IgE-dependent immediate hypersensitivity reactions in vivo. However, it is not clear to what extent mast cell expression of C3aR or C5aR influences C3a- or C5a-induced cutaneous responses or IgE-dependent mast cell activation and passive cutaneous anaphylaxis (PCA) in vivo. OBJECTIVE We sought to assess whether mouse skin mast cell expression of C3aR or C5aR influences (1) the cells' responsiveness to intradermal injections of C3a or C5a or (2) the extent of IgE-dependent mast cell degranulation and PCA in vivo. METHODS We measured the magnitude of cutaneous responses to intradermal injections of C3a or C5a and the extent of IgE-dependent mast cell degranulation and PCA responses in mice containing mast cells that did or did not express C3aR or C5aR. RESULTS The majority of the skin swelling induced by means of intradermal injection of C3a or C5a required that mast cells at the site expressed C3aR or C5aR, respectively, and the extent of IgE-dependent degranulation of skin mast cells and IgE-dependent PCA was significantly reduced when mast cells lacked either C3aR or C5aR. IgE-dependent PCA responses associated with local increases in C3a levels occurred in antibody-deficient mice but not in mice deficient in FcɛRIγ. CONCLUSION Expression of C3aR and C5aR by skin mast cells contributes importantly to the ability of C3a and C5a to induce skin swelling and can enhance mast cell degranulation and inflammation during IgE-dependent PCA in vivo.
Collapse
Affiliation(s)
- Beatrix Schäfer
- Department of Pathology, Stanford University School of Medicine, Stanford, Calif 94305-5324, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Yu MK, Park J, Jon S. Targeting strategies for multifunctional nanoparticles in cancer imaging and therapy. Theranostics 2012; 2:3-44. [PMID: 22272217 PMCID: PMC3263514 DOI: 10.7150/thno.3463] [Citation(s) in RCA: 547] [Impact Index Per Article: 42.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2011] [Accepted: 09/28/2011] [Indexed: 12/11/2022] Open
Abstract
Nanomaterials offer new opportunities for cancer diagnosis and treatment. Multifunctional nanoparticles harboring various functions including targeting, imaging, therapy, and etc have been intensively studied aiming to overcome limitations associated with conventional cancer diagnosis and therapy. Of various nanoparticles, magnetic iron oxide nanoparticles with superparamagnetic property have shown potential as multifunctional nanoparticles for clinical translation because they have been used asmagnetic resonance imaging (MRI) constrast agents in clinic and their features could be easily tailored by including targeting moieties, fluorescence dyes, or therapeutic agents. This review summarizes targeting strategies for construction of multifunctional nanoparticles including magnetic nanoparticles-based theranostic systems, and the various surface engineering strategies of nanoparticles for in vivo applications.
Collapse
Affiliation(s)
| | | | - Sangyong Jon
- Cell Dynamics Research Center, School of Life Sciences, Gwangju Institute of Science and Technology, 261 Chemdangwagi-ro, Gwangju 500-712, Republic of Korea
| |
Collapse
|
37
|
Takagi M. Toll-like receptor--a potent driving force behind rheumatoid arthritis. J Clin Exp Hematop 2011; 51:77-92. [PMID: 22104306 DOI: 10.3960/jslrt.51.77] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Toll like receptor (TLR), one of the key functions of innate immune system, can recognize not only exogenous pathogen-associated molecular patterns, namely PAMPs, but also endogenous molecules created upon tissue injury, sterile inflammation and degeneration. Endogenous TLR ligands are called as damage-associated molecular patters (DAMPs), including endogenous molecules released by activated and necrotic cells, and extracellular matrix molecules. DAMPs are also known as alarmins. TLR research has brought about new insights in the rheumatic diseases. Previous reports suggest that TLRs and the signal pathways intensively contribute to the pathogenesis of rheumatoid arthritis (RA) and other arthritic conditions with interaction of various TLR ligands. Accumulated knowledge of TLR system is summarized to overlook TLRs and the signaling pathway in arthritis conditions, with special reference to RA.
Collapse
Affiliation(s)
- Michiaki Takagi
- Department of Orthopaedic Surgery, Yamagata University School of Medicine, Japan.
| |
Collapse
|
38
|
Monaco C, Terrando N, Midwood KS. Toll-like receptor signaling: common pathways that drive cardiovascular disease and rheumatoid arthritis. Arthritis Care Res (Hoboken) 2011; 63:500-11. [PMID: 21452263 DOI: 10.1002/acr.20382] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Claudia Monaco
- Kennedy Institute of Rheumatology, Imperial College, London, UK.
| | | | | |
Collapse
|
39
|
Evaluation of a fibrin-binding gadolinium chelate peptide tetramer in a brain glioma model. Invest Radiol 2011; 46:169-77. [PMID: 21150792 DOI: 10.1097/rli.0b013e3181f7a0b0] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
PURPOSE To compare a fibrin-targeted, high relaxivity gadolinium tetramer, EP-2104R, in terms of magnitude of contrast enhancement (CE) and temporal time course, to a conventional extracellular gadolinium chelate, in a brain glioma model at 1.5-T magnetic resonance imaging. METHODS Six rats were evaluated, with each animal receiving (for separate studies) 0.05 mmol/kg gadopentetate dimeglumine (Gd DTPA or Magnevist) and 0.0125 mmol/kg of EP-2104R, with the 2 magnetic resonance examinations separated in each animal by 24 hours. The compound (EP-2104R) was synthesized using published methodology, being comprised of an 11 amino acid peptide derivatized at both the C- and N-termini with Gd-DOTA-like (Dotarem-like) moieties. T1-weighted scans were acquired precontrast and for 5 consecutive 2-minute intervals postcontrast, and subsequently at 15 and 20 minutes postcontrast. RESULTS Maximum tumor contrast-to-noise and CE both occurred at 1 minute versus at 5 minutes following administration of Gd DTPA versus EP-2104R, respectively. Utilizing an equivalent dose on a Gd ion per body weight basis, signal-to-noise, contrast-to-noise, and CE were greater for EP-2104R at all time points postcontrast, yielding overall statistically significantly greater levels of all 3 parameters with the latter. With EP-2104R, improvements in CE ranged between 87% and 391%, increasing at each measured time postcontrast with the exception of a slight decrease from 15 to 20 minutes postadministration. Histopathology confirmed, using immunofluorescence technique, abnormally increased fibrin within the tumor. CONCLUSIONS Statistically significantly greater brain tumor enhancement was noted with greater lesion enhancement at all observed time points postcontrast for EP-2104R utilizing an equivalent concentration to Gd DTPA on a per gadolinium ion basis. These findings together with the prolonged time course of enhancement suggest possible fibrin-binding and altered distribution kinetics.
Collapse
|
40
|
Agemy L, Sugahara KN, Kotamraju VR, Gujraty K, Girard OM, Kono Y, Mattrey RF, Park JH, Sailor MJ, Jimenez AI, Cativiela C, Zanuy D, Sayago FJ, Aleman C, Nussinov R, Ruoslahti E. Nanoparticle-induced vascular blockade in human prostate cancer. Blood 2010; 116:2847-56. [PMID: 20587786 PMCID: PMC2974592 DOI: 10.1182/blood-2010-03-274258] [Citation(s) in RCA: 130] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2010] [Accepted: 06/03/2010] [Indexed: 11/20/2022] Open
Abstract
The tumor-homing pentapeptide CREKA (Cys-Arg-Glu-Lys-Ala) specifically homes to tumors by binding to fibrin and fibrin-associated clotted plasma proteins in tumor vessels. Previous results show that CREKA-coated superparamagnetic iron oxide particles can cause additional clotting in tumor vessels, which creates more binding sites for the peptide. We have used this self-amplifying homing system to develop theranostic nanoparticles that simultaneously serve as an imaging agent and inhibit tumor growth by obstructing tumor circulation through blood clotting. The CREKA nanoparticles were combined with nanoparticles coated with another tumor-homing peptide, CRKDKC, and nanoparticles with an elongated shape (nanoworms) were used for improved binding efficacy. The efficacy of the CREKA peptide was then increased by replacing some residues with nonproteinogenic counterparts, which increased the stability of the peptide in the circulation. Treatment of mice bearing orthotopic human prostate cancer tumors with the targeted nanoworms caused extensive clotting in tumor vessels, whereas no clotting was observed in the vessels of normal tissues. Optical and magnetic resonance imaging confirmed tumor-specific targeting of the nanoworms, and ultrasound imaging showed reduced blood flow in tumor vessels. Treatment of mice with prostate cancer with multiple doses of the nanoworms induced tumor necrosis and a highly significant reduction in tumor growth.
Collapse
Affiliation(s)
- Lilach Agemy
- Vascular Mapping Laboratory, Center for Nanomedicine, Sanford-Burnham Medical Research Institute at the University of California at Santa Barbara (UCSB), Santa Barbara, CA, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Tan M, Wu X, Jeong EK, Chen Q, Lu ZR. Peptide-targeted Nanoglobular Gd-DOTA monoamide conjugates for magnetic resonance cancer molecular imaging. Biomacromolecules 2010; 11:754-61. [PMID: 20131758 DOI: 10.1021/bm901352v] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Effective imaging of a cancer molecular biomarker is critical for accurate cancer diagnosis and prognosis. CLT1 peptide was observed to specifically bind to the fibrin-fibronectin complexes presented in tumor extracellular matrix. In this study, we synthesized and evaluated CLT1 peptide-targeted nanoglobular Gd-DOTA monoamide conjugates for magnetic resonance (MR) imaging of the fibrin-fibronectin complexes in tumor. The targeted nanoglobular contrast agents were prepared by conjugating peptide CLT1 to G2 and G3 nanoglobule (lysine dendrimers with a cubic silsesquioxane core) Gd-DOTA monoamide conjugates via click chemistry. The T(1) relaxivities of peptide-targeted G2 and G3 nanoglobules were 7.92 and 8.20 mM(-1) s(-1) at 3T, respectively. Approximately 2 peptides and 25 Gd-DOTA chelates were conjugated onto the surface of 32 amine groups of G2 nanoglobule, and 3 peptides and 43 Gd-DOTA chelates onto the surface of 64 amine groups of G3 nanoglobule. The peptide-targeted nanoglobular contrast agents showed greater contrast enhancement than the corresponding nontargeted agents in tumor at a dose of 0.03 mmol-Gd/kg in female athymic mice bearing MDA-MB-231 human breast carcinoma xenografts. The targeted MRI contrast agents have a potential for specific cancer molecular imaging with MRI.
Collapse
Affiliation(s)
- Mingqian Tan
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio 44106, USA
| | | | | | | | | |
Collapse
|
42
|
DAMPening inflammation by modulating TLR signalling. Mediators Inflamm 2010; 2010. [PMID: 20706656 PMCID: PMC2913853 DOI: 10.1155/2010/672395] [Citation(s) in RCA: 682] [Impact Index Per Article: 45.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2009] [Accepted: 04/20/2010] [Indexed: 12/12/2022] Open
Abstract
Damage-associated molecular patterns (DAMPs) include endogenous intracellular molecules released by activated or necrotic cells and extracellular matrix (ECM) molecules that are upregulated upon injury or degraded following tissue damage. DAMPs are vital danger signals that alert our immune system to tissue damage upon both infectious and sterile insult. DAMP activation of Toll-like receptors (TLRs) induces inflammatory gene expression to mediate tissue repair. However, DAMPs have also been implicated in diseases where excessive inflammation plays a key role in pathogenesis, including rheumatoid arthritis (RA), cancer, and atherosclerosis. TLR activation by DAMPs may initiate positive feedback loops where increasing tissue damage perpetuates pro-inflammatory responses leading to chronic inflammation. Here we explore the current knowledge about distinct signalling cascades resulting from self TLR activation. We also discuss the involvement of endogenous TLR activators in disease and highlight how specifically targeting DAMPs may yield therapies that do not globally suppress the immune system.
Collapse
|
43
|
Drexler SK, Sacre SM, Foxwell BM. Toll-like receptors: a new target in rheumatoid arthritis? Expert Rev Clin Immunol 2010; 2:585-99. [PMID: 20477615 DOI: 10.1586/1744666x.2.4.585] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Rheumatoid arthritis (RA) is one of the most prevalent autoimmune diseases. It is characterized by chronic inflammation of the joint leading to its destruction. Although the initiating cause remains elusive, environmental factors and genetic background are known to contribute to the etiology of RA. The role of Toll-like receptors (TLRs) in innate immunity and their ability to recognize microbial products has been well characterized. TLRs are able to recognize endogenous molecules released upon cell damage and necrosis, and are present in RA synovial fluid. Although it appears unlikely that a pathogen underlies the pathogenesis or progression of RA, the release of endogenous TLR ligands during inflammation may activate TLRs and perpetuate the disease. An increasing body of circumstantial evidence implicates TLR signaling in RA, although, at present, their involvement is not defined comprehensively. Targeting individual TLRs or their signaling transducers may provide a more specific therapy without global suppression of the immune system.
Collapse
Affiliation(s)
- Stefan K Drexler
- Imperial College of Science, Technology and Medicine, Kennedy Institute of Rheumatology Division, Faculty of Medicine, 1 Aspenlea Road, Hammersmith, London,W6 8LH, UK.
| | | | | |
Collapse
|
44
|
Abstract
The various types of cells that comprise the tumor mass all carry molecular markers that are not expressed or are expressed at much lower levels in normal cells. These differentially expressed molecules can be used as docking sites to concentrate drug conjugates and nanoparticles at tumors. Specific markers in tumor vessels are particularly well suited for targeting because molecules at the surface of blood vessels are readily accessible to circulating compounds. The increased concentration of a drug in the site of disease made possible by targeted delivery can be used to increase efficacy, reduce side effects, or achieve some of both. We review the recent advances in this delivery approach with a focus on the use of molecular markers of tumor vasculature as the primary target and nanoparticles as the delivery vehicle.
Collapse
Affiliation(s)
- Erkki Ruoslahti
- Vascular Mapping Center, Sanford-Burnham Medical Research Institute, University of California, Santa Barbara, Santa Barbara, CA 93106, USA.
| | | | | |
Collapse
|
45
|
Tabata M, Kadomatsu T, Fukuhara S, Miyata K, Ito Y, Endo M, Urano T, Zhu HJ, Tsukano H, Tazume H, Kaikita K, Miyashita K, Iwawaki T, Shimabukuro M, Sakaguchi K, Ito T, Nakagata N, Yamada T, Katagiri H, Kasuga M, Ando Y, Ogawa H, Mochizuki N, Itoh H, Suda T, Oike Y. Angiopoietin-like protein 2 promotes chronic adipose tissue inflammation and obesity-related systemic insulin resistance. Cell Metab 2009; 10:178-88. [PMID: 19723494 DOI: 10.1016/j.cmet.2009.08.003] [Citation(s) in RCA: 284] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2009] [Revised: 06/05/2009] [Accepted: 08/10/2009] [Indexed: 01/22/2023]
Abstract
Recent studies of obesity have provided new insights into the mechanisms underlying insulin resistance and metabolic dysregulation. Numerous efforts have been made to identify key regulators of obesity-linked adipose tissue inflammation and insulin resistance. We found that angiopoietin-like protein 2 (Angptl2) was secreted by adipose tissue and that its circulating level was closely related to adiposity, systemic insulin resistance, and inflammation in both mice and humans. Angptl2 activated an inflammatory cascade in endothelial cells via integrin signaling and induced chemotaxis of monocytes/macrophages. Constitutive Angptl2 activation in vivo induced inflammation of the vasculature characterized by abundant attachment of leukocytes to the vessel walls and increased permeability. Angptl2 deletion ameliorated adipose tissue inflammation and systemic insulin resistance in diet-induced obese mice. Conversely, Angptl2 overexpression in adipose tissue caused local inflammation and systemic insulin resistance in nonobese mice. Thus, Angptl2 is a key adipocyte-derived inflammatory mediator that links obesity to systemic insulin resistance.
Collapse
Affiliation(s)
- Mitsuhisa Tabata
- Department of Molecular Genetics, Graduate School of Medical Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto 860-8556, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
von Känel R, Thayer JF, Fischer JE. Nighttime vagal cardiac control and plasma fibrinogen levels in a population of working men and women. Ann Noninvasive Electrocardiol 2009; 14:176-84. [PMID: 19419403 DOI: 10.1111/j.1542-474x.2009.00293.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
BACKGROUND Elevated plasma fibrinogen levels have prospectively been associated with an increased risk of coronary artery disease in different populations. Plasma fibrinogen is a measure of systemic inflammation crucially involved in atherosclerosis. The vagus nerve curtails inflammation via a cholinergic antiinflammatory pathway. We hypothesized that lower vagal control of the heart relates to higher plasma fibrinogen levels. METHODS Study participants were 559 employees (age 17-63 years; 89% men) of an airplane manufacturing plant in southern Germany. All subjects underwent medical examination, blood sampling, and 24-hour ambulatory heart rate recording while kept on their work routine. The root mean square of successive differences in RR intervals during the night period (nighttime RMSSD) was computed as the heart rate variability index of vagal function. RESULTS After controlling for demographic, lifestyle, and medical factors, nighttime RMSSD explained 1.7% (P = 0.001), 0.8% (P = 0.033), and 7.8% (P = 0.007), respectively, of the variance in fibrinogen levels in all subjects, men, and women. Nighttime RMSSD and fibrinogen levels were stronger correlated in women than in men. In all workers, men, and women, respectively, there was a mean +/- SEM increase of 0.41 +/- 0.13 mg/dL, 0.28 +/- 0.13 mg/dL, and 1.16 +/- 0.41 mg/dL fibrinogen for each millisecond decrease in nighttime RMSSD. CONCLUSIONS Reduced vagal outflow to the heart correlated with elevated plasma fibrinogen levels independent of the established cardiovascular risk factors. This relationship seemed comparably stronger in women than men. Such an autonomic mechanism might contribute to the atherosclerotic process and its thrombotic complications.
Collapse
Affiliation(s)
- Roland von Känel
- Department of General Internal Medicine, Bern University Hospital, Inselspital, Bern, Switzerland.
| | | | | |
Collapse
|
47
|
MyD88-dependent nuclear factor-κB activation is involved in fibrinogen-induced hypertrophic response of cardiomyocytes. J Hypertens 2009; 27:1084-93. [DOI: 10.1097/hjh.0b013e3283293c93] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
48
|
Lindahl U, Pejler G. Heparin-like polysaccharides in intra- and extravascular coagulation reactions. ACTA MEDICA SCANDINAVICA. SUPPLEMENTUM 2009; 715:139-44. [PMID: 2954435 DOI: 10.1111/j.0954-6820.1987.tb09914.x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
49
|
Von Känel R, Bellingrath S, Kudielka BM. Association between longitudinal changes in depressive symptoms and plasma fibrinogen levels in school teachers. Psychophysiology 2009; 46:473-80. [PMID: 19298625 DOI: 10.1111/j.1469-8986.2009.00788.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Depression and anxiety previously predicted coronary artery disease (CAD) risk. Inflammation contributes to CAD and shows an association with depression. We followed 57 teachers (mean 49+/-8 years) over 21 months and investigated whether changes in depressive and anxiety symptoms relate to those in the CAD risk and inflammation marker fibrinogen and vice versa. Increase in depressive symptoms and in fibrinogen levels were significantly correlated. While controlling for baseline depression rendered the association between changes in depression and fibrinogen nonsignificant, taking into account baseline fibrinogen levels maintained the predictive value of fibrinogen change for depression change. Anxiety and fibrinogen changes were not significantly correlated. This dynamic relationship between depression and the inflammatory biomarker fibrinogen might advance our knowledge about psychobiological mechanisms underlying both CAD and sickness behavior.
Collapse
Affiliation(s)
- Roland Von Känel
- Division of Psychosomatic Medicine, Inselspital, Bern University Hospital, and University of Bern, Bern, Switzerland
| | | | | |
Collapse
|
50
|
Park JH, von Maltzahn G, Zhang L, Derfus AM, Simberg D, Harris TJ, Ruoslahti E, Bhatia SN, Sailor MJ. Systematic surface engineering of magnetic nanoworms for in vivo tumor targeting. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2009; 5:694-700. [PMID: 19263431 PMCID: PMC3058937 DOI: 10.1002/smll.200801789] [Citation(s) in RCA: 204] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
In the design of nanoparticles that can target disease tissue in vivo, parameters such as targeting ligand density, type of target receptor, and nanoparticle shape can play an important role in determining the extent of accumulation. Herein, a systematic study of these parameters for the targeting of mouse xenograft tumors is performed using superparamagnetic iron oxide as a model nanoparticle system. The type of targeting peptide (recognizing cell surface versus extracellular matrix), the surface coverage of the peptide, its attachment chemistry, and the shape of the nanomaterial [elongated (nanoworm, NW) versus spherical (nanosphere, NS)] are varied. Nanoparticle circulation times and in vivo tumor-targeting efficiencies are quantified in two xenograft models of human tumors (MDA-MB-435 human carcinoma and HT1080 human fibrosarcoma). It is found that the in vivo tumor-targeting ability of the NW is superior to that of the NS, that the smaller, neutral CREKA targeting group is more effective than the larger, positively charged F3 molecule, that a maximum in tumor-targeting efficiency and blood half-life is observed with approximately 60 CREKA peptides per NW for either the HT1080 or the MDA-MB-435 tumor types, and that incorporation of a 5-kDa polyethylene glycol linker improves targeting to both tumor types relative to a short linker. It is concluded that the blood half-life of a targeting molecule-nanomaterial ensemble is a key consideration when selecting the appropriate ligand and nanoparticle chemistry for tumor targeting.
Collapse
Affiliation(s)
- Ji-Ho Park
- Materials Science and Engineering Program, Department of Chemistry and Biochemistry, University of California, San Diego, 9500 Gilman, La Jolla, CA 92093 (USA)
| | - Geoffrey von Maltzahn
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139 (USA)
| | - Lianglin Zhang
- Cancer Research Center, Burnham Institute for Medical Research, 10901 N. Torrey Pines Rd., La Jolla, CA 92037 (USA)
- Vascular Mapping Center, Burnham Institute for Medical Research (at UCSB) Bio II, Rm. #3119, University of California, Santa Barbara, Santa Barbara, CA 93106 (USA)
| | - Austin M. Derfus
- Department of Bioengineering, University of California, San Diego, 9500 Gilman, La Jolla, CA 92093 (USA)
| | - Dmitri Simberg
- Cancer Research Center, Burnham Institute for Medical Research, 10901 N. Torrey Pines Rd., La Jolla, CA 92037 (USA)
- Vascular Mapping Center, Burnham Institute for Medical Research (at UCSB) Bio II, Rm. #3119, University of California, Santa Barbara, Santa Barbara, CA 93106 (USA)
| | - Todd J. Harris
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139 (USA)
| | - Erkki Ruoslahti
- Cancer Research Center, Burnham Institute for Medical Research, 10901 N. Torrey Pines Rd., La Jolla, CA 92037 (USA)
- Vascular Mapping Center, Burnham Institute for Medical Research (at UCSB) Bio II, Rm. #3119, University of California, Santa Barbara, Santa Barbara, CA 93106 (USA)
| | - Sangeeta N. Bhatia
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139 (USA)
| | - Michael J. Sailor
- Materials Science and Engineering Program, Department of Chemistry and Biochemistry, University of California, San Diego, 9500 Gilman, La Jolla, CA 92093 (USA)
| |
Collapse
|