1
|
Xu H, Ma H, Li Y, Bi S, Cai K, Wu L, Zhang L, Guan H, Li C, Yang J, Qiu P. Propylene glycol alginate sodium sulfate suppressed lung metastasis by blocking P-selectin to recruit CD4 regulatory T cells. Int J Biol Macromol 2024; 279:134976. [PMID: 39179086 DOI: 10.1016/j.ijbiomac.2024.134976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 08/12/2024] [Accepted: 08/21/2024] [Indexed: 08/26/2024]
Abstract
P-selectin has been shown to enhance growth and metastasis of mouse tumors by promoting regulatory T cell (Treg) infiltration into the tumors. Theoretically, a P-selectin antagonist could suppress the process. Popylene glycol alginate sodium sulfate (PSS) is a heparin-like marine drug, which was originally approved to treat cardiovascular disease in China. Previously, we reported that PSS was an effective P-selectin antagonist in vitro. However, it is unknown whether PSS can regulate Treg infiltration and its effect on lung metastasis in vivo. Our results showed that PSS at 30 mg/kg significantly suppressed lung metastasis and improved overall survival, with potency comparable to the positive control LMWH. Mechanistic study indicated that PSS blocked tumor cells adhesion and activated platelets by directly binding with activated platelet's P-selectin. Compared to the model group, PSS decreased the percent of Tregs by 63 % in lungs after treating for 21 days while increasing CD8+ T cells (1.59-fold) and Granzyme B+ CD8 T cells (2.08-fold)' percentage for generating an adaptive response for systemic tumor suppression. The study indicated that the P-selectin antagonist, PSS, suppressed lung metastasis by inhibiting the infiltration of regulatory T cells (Treg) into the tumors.
Collapse
Affiliation(s)
- Huixin Xu
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Rd, Qingdao, Shandong 266003, China; Marine Biomedical Research Institute of Qiangdao, 23 Hongkong East Rd, Qingdao, Shandong 266003, China
| | - He Ma
- College of Veterinary Medicine, Qingdao Agricultural University, 700 Changcheng Rd, Qingdao, Shandong,266109, China
| | - Yannan Li
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Rd, Qingdao, Shandong 266003, China
| | - Shijie Bi
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Rd, Qingdao, Shandong 266003, China
| | - Kaiyu Cai
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Rd, Qingdao, Shandong 266003, China
| | - Lijuan Wu
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Rd, Qingdao, Shandong 266003, China
| | - Lei Zhang
- Research Center of Traditional Chinese Medicine and Clinical Pharmacy, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan 250014, China
| | - Huashi Guan
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Rd, Qingdao, Shandong 266003, China; Marine Biomedical Research Institute of Qiangdao, 23 Hongkong East Rd, Qingdao, Shandong 266003, China
| | - Chunxia Li
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Rd, Qingdao, Shandong 266003, China
| | - Jinbo Yang
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Rd, Qingdao, Shandong 266003, China; Marine Biomedical Research Institute of Qiangdao, 23 Hongkong East Rd, Qingdao, Shandong 266003, China
| | - Peiju Qiu
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Rd, Qingdao, Shandong 266003, China; Marine Biomedical Research Institute of Qiangdao, 23 Hongkong East Rd, Qingdao, Shandong 266003, China
| |
Collapse
|
2
|
Ying Y, Hao W. Corrigendum: Immunomodulatory function and anti-tumor mechanism of natural polysaccharides: a review. Front Immunol 2024; 14:1361355. [PMID: 38264646 PMCID: PMC10804138 DOI: 10.3389/fimmu.2023.1361355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Accepted: 12/29/2023] [Indexed: 01/25/2024] Open
Abstract
[This corrects the article DOI: 10.3389/fimmu.2023.1147641.].
Collapse
Affiliation(s)
- Yang Ying
- Cancer Institute, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, Hangzhou, Zhejiang, China
- Zhejiang Provincial Clinical Research Center for Cancer, Cancer Center of Zhejiang University, Hangzhou, Zhejiang, China
| | - Wu Hao
- Cancer Institute, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, Hangzhou, Zhejiang, China
- Zhejiang Provincial Clinical Research Center for Cancer, Cancer Center of Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
3
|
Ying Y, Hao W. Immunomodulatory function and anti-tumor mechanism of natural polysaccharides: A review. Front Immunol 2023; 14:1147641. [PMID: 36969152 PMCID: PMC10035574 DOI: 10.3389/fimmu.2023.1147641] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 02/22/2023] [Indexed: 03/11/2023] Open
Abstract
Polysaccharides extracted from natural resources have attracted extensive attention in biomedical research and pharmaceutical fields, due to their medical values in anti-tumor, immunomodulation, drug delivery, and many other aspects. At present, a variety of natural polysaccharides have been developed as adjuvant drugs in clinical application. Benefit from their structural variability, polysaccharides have great potential in regulating cellular signals. Some polysaccharides exert direct anti-tumor effects by inducing cell cycle arrest and apoptosis, while the majority of polysaccharides can regulate the host immune system and indirectly inhibit tumors by activating either non-specific or specific immune responses. As the essential of microenvironment in the process of tumor development has been gradually revealed, some polysaccharides were found to inhibit the proliferation and metastasis of tumor cells via tumoral niche modulation. Here, we focused on natural polysaccharides with biomedical application potential, reviewed the recent advancement in their immunomodulation function and highlighted the importance of their signaling transduction feature for the antitumor drug development.
Collapse
Affiliation(s)
- Yang Ying
- Cancer Institute, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, Hangzhou, Zhejiang, China
- Zhejiang Provincial Clinical Research Center for Cancer, Cancer Center of Zhejiang University, Hangzhou, Zhejiang, China
| | - Wu Hao
- Cancer Institute, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, Hangzhou, Zhejiang, China
- Zhejiang Provincial Clinical Research Center for Cancer, Cancer Center of Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
4
|
Advances in oral absorption of polysaccharides: Mechanism, affecting factors, and improvement strategies. Carbohydr Polym 2022; 282:119110. [DOI: 10.1016/j.carbpol.2022.119110] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 01/04/2022] [Accepted: 01/05/2022] [Indexed: 02/08/2023]
|
5
|
Apical sodium-dependent bile acid transporter, drug target for bile acid related diseases and delivery target for prodrugs: Current and future challenges. Pharmacol Ther 2020; 212:107539. [PMID: 32201314 DOI: 10.1016/j.pharmthera.2020.107539] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Accepted: 03/11/2020] [Indexed: 02/06/2023]
|
6
|
Lanzi C, Cassinelli G. Heparan Sulfate Mimetics in Cancer Therapy: The Challenge to Define Structural Determinants and the Relevance of Targets for Optimal Activity. Molecules 2018; 23:E2915. [PMID: 30413079 PMCID: PMC6278363 DOI: 10.3390/molecules23112915] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 11/06/2018] [Accepted: 11/06/2018] [Indexed: 12/21/2022] Open
Abstract
Beyond anticoagulation, the therapeutic potential of heparin derivatives and heparan sulfate (HS) mimetics (functionally defined HS mimetics) in oncology is related to their ability to bind and modulate the function of a vast array of HS-binding proteins with pivotal roles in cancer growth and progression. The definition of structural/functional determinants and the introduction of chemical modifications enabled heparin derivatives to be identified with greatly reduced or absent anticoagulant activity, but conserved/enhanced anticancer activity. These studies paved the way for the disclosure of structural requirements for the inhibitory effects of HS mimetics on heparanase, selectins, and growth factor receptor signaling, as well as for the limitation of side effects. Actually, HS mimetics affect the tumor biological behavior via a multi-target mechanism of action based on their effects on tumor cells and various components of the tumor microenvironment. Emerging evidence indicates that immunomodulation can participate in the antitumor activity of these agents. Significant ability to enhance the antitumor effects of combination treatments with standard therapies was shown in several tumor models. While the first HS mimetics are undergoing early clinical evaluation, an improved understanding of the molecular contexts favoring the antitumor action in certain malignancies or subgroups is needed to fully exploit their potential.
Collapse
Affiliation(s)
- Cinzia Lanzi
- Molecular Pharmacology Unit, Department of Applied Research and Technological Development, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy.
| | - Giuliana Cassinelli
- Molecular Pharmacology Unit, Department of Applied Research and Technological Development, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy.
| |
Collapse
|
7
|
Manandhar S, Park J, Kothandan VK, Lee J, Alam F, Jee JP, Hwang J, Byun Y, Hwang SR. Properties of Heparinoids Premixed with Tumor-Derived Extracellular Vesicles. Bioconjug Chem 2018; 29:3757-3767. [DOI: 10.1021/acs.bioconjchem.8b00637] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
| | - Jooho Park
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, 5, Hwarang-ro 14-gil, Seongbuk-gu, Seoul 02792, Republic of Korea
| | | | | | - Farzana Alam
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas 79106, United States
| | | | | | - Youngro Byun
- College of Pharmacy, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Republic of Korea
| | | |
Collapse
|
8
|
Hwang YH, Jeong MJ, Kim MJ, Kim JK, Lee DY. Enhancement of T 2 -weighted MR contrast using heparin for cell tracking in vivo. J IND ENG CHEM 2017. [DOI: 10.1016/j.jiec.2017.06.043] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
9
|
Strategies to Overcome Heparins' Low Oral Bioavailability. Pharmaceuticals (Basel) 2016; 9:ph9030037. [PMID: 27367704 PMCID: PMC5039490 DOI: 10.3390/ph9030037] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Revised: 06/15/2016] [Accepted: 06/23/2016] [Indexed: 01/10/2023] Open
Abstract
Even after a century, heparin is still the most effective anticoagulant available with few side effects. The poor oral absorption of heparins triggered the search for strategies to achieve oral bioavailability since this route has evident advantages over parenteral administration. Several approaches emerged, such as conjugation of heparins with bile acids and lipids, formulation with penetration enhancers, and encapsulation of heparins in micro and nanoparticles. Some of these strategies appear to have potential as good delivery systems to overcome heparin’s low oral bioavailability. Nevertheless, none have reached the market yet. Overall, this review aims to provide insights regarding the oral bioavailability of heparin.
Collapse
|
10
|
Nurunnabi M, Khatun Z, Revuri V, Nafiujjaman M, Cha S, Cho S, Moo Huh K, Lee YK. Design and strategies for bile acid mediated therapy and imaging. RSC Adv 2016. [DOI: 10.1039/c6ra10978k] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Bioinspired materials have received substantial attention across biomedical, biological, and drug delivery research because of their high biocompatibility and lower toxicity compared with synthetic materials.
Collapse
Affiliation(s)
- Md Nurunnabi
- Department of Polymer Science & Engineering
- Chungnam National University
- Daejeon 305-764
- Republic of Korea
- Department of Chemical & Biological Engineering
| | - Zehedina Khatun
- Department of Polymer Science & Engineering
- Chungnam National University
- Daejeon 305-764
- Republic of Korea
| | - Vishnu Revuri
- Department of Green Bioengineering
- Korea National University of Transportation
- Chungju 380-702
- Republic of Korea
| | - Md Nafiujjaman
- Department of Green Bioengineering
- Korea National University of Transportation
- Chungju 380-702
- Republic of Korea
| | - Seungbin Cha
- Department of Biomedical Chemistry
- Konkuk University
- Chungju-si
- Republic of Korea
| | - Sungpil Cho
- KB Biomed Inc
- Chungju 380-702
- Republic of Korea
| | - Kang Moo Huh
- Department of Polymer Science & Engineering
- Chungnam National University
- Daejeon 305-764
- Republic of Korea
| | - Yong-kyu Lee
- Department of Chemical & Biological Engineering
- Korea National University of Transportation
- Chungju 380-702
- Republic of Korea
- Department of Green Bioengineering
| |
Collapse
|
11
|
Teng L, Fu H, Deng C, Chen J, Chen J. Modulating the SDF-1/CXCL12-induced cancer cell growth and adhesion by sulfated K5 polysaccharides in vitro. Biomed Pharmacother 2015. [PMID: 26211579 DOI: 10.1016/j.biopha.2015.05.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Stromal cell-derived factor-1 (SDF-1)/chemokine (CXC motif) ligand 12 (CXCL12) is involved in the process of tumor progression. Sulfated K5 polysaccharides have shown anti-cancer activity by acting on multiple targets, though it remains unclear whether sulfated K5 polysaccharides would disrupt SDF-1/CXCL12-stimulated cancer biology. This study aimed to investigate the effects of sulfated K5 polysaccharides on cell growth, adhesion in murine B16 melanoma cells and the underlying mechanism by targeting SDF-1/CXCL12. Results indicated that K5-NS,OS inhibited the proliferation of B16 melanoma cells, induced the cell cycle arrest mainly at the G0/G1 phase, and suppressed cancer cell proliferation or adhesion induced by SDF-1/CXCL12. It was possible that K5-NS,OS appeared to interact with CXCL12 and block the subsequent biological functions. This work suggests that the existence of O- and N-sulfate groups is more effective in targeting CXCL12 and exhibiting anti-cancer activity.
Collapse
Affiliation(s)
- Liping Teng
- Wuxi Medical School, Jiangnan University, Wuxi 214122, PR China; The Key Laboratory of Industrial Biotechnology, Ministry of Education, Jiangnan University, Wuxi 214122, PR China
| | - Haitian Fu
- School of Pharmaceutical Science, Jiangnan University, Wuxi 214122, PR China
| | - Chao Deng
- Wuxi Medical School, Jiangnan University, Wuxi 214122, PR China
| | - Jingxiao Chen
- School of Pharmaceutical Science, Jiangnan University, Wuxi 214122, PR China
| | - Jinghua Chen
- The Key Laboratory of Industrial Biotechnology, Ministry of Education, Jiangnan University, Wuxi 214122, PR China; School of Pharmaceutical Science, Jiangnan University, Wuxi 214122, PR China.
| |
Collapse
|
12
|
Antiangiogenic and anticancer effect of an orally active low molecular weight heparin conjugates and its application to lung cancer chemoprevention. J Control Release 2015; 199:122-31. [DOI: 10.1016/j.jconrel.2014.12.015] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2014] [Revised: 11/24/2014] [Accepted: 12/14/2014] [Indexed: 01/15/2023]
|
13
|
Kim JY, Alam F, Chung SW, Park J, Jeon OC, Kim SY, Son WC, Byun Y. Combinational chemoprevention effect of celecoxib and an oral antiangiogenic LHD4 on colorectal carcinogenesis in mice. Anticancer Drugs 2014; 25:1061-71. [DOI: 10.1097/cad.0000000000000141] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
14
|
Pisano C, Vlodavsky I, Ilan N, Zunino F. The potential of heparanase as a therapeutic target in cancer. Biochem Pharmacol 2014; 89:12-9. [PMID: 24565907 DOI: 10.1016/j.bcp.2014.02.010] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2013] [Revised: 02/13/2014] [Accepted: 02/14/2014] [Indexed: 12/19/2022]
Abstract
Heparanase has generated substantial interest as therapeutic target for antitumor therapy, because its activity is implicated in malignant behavior of cancer cells and in tumor progression. Increased heparanase expression was found in numerous tumor types and correlates with poor prognosis. Heparanase, an endoglucuronidase responsible for heparan sulfate cleavage, regulates the structure and function of heparan sulfate proteoglycans, leading to disassembly of the extracellular matrix. The action of heparanase is involved in multiple regulatory events related, among other effects, to augmented bioavailability of growth factors and cytokines. Inhibitors of heparanase suppress tumor growth, angiogenesis and metastasis by modulating growth factor-mediated signaling, ECM barrier function and cell interactions in the tumor microenvironment. Therefore, targeting heparanase has potential implications for anti-tumor, anti-angiogenic and anti-inflammatory therapies. Current approaches for heparanase inhibition include development of chemically modified heparins, small molecule inhibitors and neutralizing antibodies. The available evidence supports the emerging utility of heparanase inhibition as a promising antitumor strategy, specifically in rational combination with other agents. The recent studies with compounds designed to block heparanase (e.g., modified heparins) provide a rational basis for their therapeutic application and optimization.
Collapse
Affiliation(s)
| | - Israel Vlodavsky
- Cancer and Vascular Biology Research Center Rappaport, Faculty of Medicine, Technion, Haifa, Israel
| | - Neta Ilan
- Cancer and Vascular Biology Research Center Rappaport, Faculty of Medicine, Technion, Haifa, Israel
| | - Franco Zunino
- Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy.
| |
Collapse
|
15
|
Nikitovic D, Mytilinaiou M, Berdiaki A, Karamanos NK, Tzanakakis GN. Heparan sulfate proteoglycans and heparin regulate melanoma cell functions. Biochim Biophys Acta Gen Subj 2014; 1840:2471-81. [PMID: 24486410 DOI: 10.1016/j.bbagen.2014.01.031] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2013] [Revised: 01/17/2014] [Accepted: 01/20/2014] [Indexed: 12/12/2022]
Abstract
BACKGROUND The solid melanoma tumor consists of transformed melanoma cells, and the associated stromal cells including fibroblasts, endothelial cells, immune cells, as well as, soluble macro- and micro-molecules of the extracellular matrix (ECM) forming the complex network of the tumor microenvironment. Heparan sulfate proteoglycans (HSPGs) are an important component of the melanoma tumor ECM. Importantly, there appears to be both a quantitative and a qualitative shift in the content of HSPGs, in parallel to the nevi-radial growth phase-vertical growth phase melanoma progression. Moreover, these changes in HSPG expression are correlated to modulations of key melanoma cell functions. SCOPE OF REVIEW This review will critically discuss the roles of HSPGs/heparin in melanoma development and progression. MAJOR CONCLUSIONS We have correlated HSPGs' expression and distribution with melanoma cell signaling and functions as well as angiogenesis. GENERAL SIGNIFICANCE The current knowledge of HSPGs/heparin biology in melanoma provides a foundation we can utilize in the ongoing search for new approaches in designing anti-tumor therapy. This article is part of a Special Issue entitled Matrix-mediated cell behaviour and properties.
Collapse
Affiliation(s)
- D Nikitovic
- Department of Anatomy, Histology, Embryology, Medical School, University of Crete, Heraklion 71003, Greece
| | - M Mytilinaiou
- Department of Anatomy, Histology, Embryology, Medical School, University of Crete, Heraklion 71003, Greece
| | - Ai Berdiaki
- Department of Anatomy, Histology, Embryology, Medical School, University of Crete, Heraklion 71003, Greece
| | - N K Karamanos
- Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras 26110, Greece
| | - G N Tzanakakis
- Department of Anatomy, Histology, Embryology, Medical School, University of Crete, Heraklion 71003, Greece.
| |
Collapse
|
16
|
Al-Hilal TA, Alam F, Byun Y. Oral drug delivery systems using chemical conjugates or physical complexes. Adv Drug Deliv Rev 2013; 65:845-64. [PMID: 23220326 DOI: 10.1016/j.addr.2012.11.002] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2012] [Revised: 11/25/2012] [Accepted: 11/27/2012] [Indexed: 02/07/2023]
Abstract
Oral delivery of therapeutics is extremely challenging. The digestive system is designed in a way that naturally allows the degradation of proteins or peptides into small molecules prior to absorption. For systemic absorption, the intact drug molecules must traverse the impending harsh gastrointestinal environment. Technologies, such as enteric coating, with oral dosage formulation strategies have successfully provided the protection of non-peptide based therapeutics against the harsh, acidic condition of the stomach. However, these technologies showed limited success on the protection of therapeutic proteins and peptides. Importantly, inherent permeability coefficient of the therapeutics is still a major problem that has remained unresolved for decades. Addressing this issue in the context, we summarize the strategies that are developed in enhancing the intestinal permeability of a drug molecule either by modifying the intestinal epithelium or by modifying the drug itself. These modifications have been pursued by using a group of molecules that can be conjugated to the drug molecule to alter the cell permeability of the drug or mixed with the drug molecule to alter the epithelial barrier function, in order to achieve the effective drug permeation. This article will address the current trends and future perspectives of the oral delivery strategies.
Collapse
Affiliation(s)
- Taslim A Al-Hilal
- College of Pharmacy, Seoul National University, Seoul 151-742, South Korea
| | | | | |
Collapse
|
17
|
Salvador JAR, Carvalho JFS, Neves MAC, Silvestre SM, Leitão AJ, Silva MMC, Sá e Melo ML. Anticancer steroids: linking natural and semi-synthetic compounds. Nat Prod Rep 2013; 30:324-74. [PMID: 23151898 DOI: 10.1039/c2np20082a] [Citation(s) in RCA: 197] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Steroids, a widespread class of natural organic compounds occurring in animals, plants and fungi, have shown great therapeutic value for a broad array of pathologies. The present overview is focused on the anticancer activity of steroids, which is very representative of a rich structural molecular diversity and ability to interact with various biological targets and pathways. This review encompasses the most relevant discoveries on steroid anticancer drugs and leads through the last decade and comprises 668 references.
Collapse
Affiliation(s)
- Jorge A R Salvador
- Laboratory of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Coimbra, Polo das Ciências da Saúde, 3000-508, Coimbra, Portugal.
| | | | | | | | | | | | | |
Collapse
|
18
|
Tumour vasculature targeting agents in hybrid/conjugate drugs. Angiogenesis 2013; 16:503-24. [DOI: 10.1007/s10456-013-9347-8] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2013] [Accepted: 03/19/2013] [Indexed: 12/28/2022]
|
19
|
The metastasis-promoting roles of tumor-associated immune cells. J Mol Med (Berl) 2013; 91:411-29. [PMID: 23515621 DOI: 10.1007/s00109-013-1021-5] [Citation(s) in RCA: 269] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2013] [Revised: 03/06/2013] [Accepted: 03/07/2013] [Indexed: 12/12/2022]
Abstract
Tumor metastasis is driven not only by the accumulation of intrinsic alterations in malignant cells, but also by the interactions of cancer cells with various stromal cell components of the tumor microenvironment. In particular, inflammation and infiltration of the tumor tissue by host immune cells, such as tumor-associated macrophages, myeloid-derived suppressor cells, and regulatory T cells, have been shown to support tumor growth in addition to invasion and metastasis. Each step of tumor development, from initiation through metastatic spread, is promoted by communication between tumor and immune cells via the secretion of cytokines, growth factors, and proteases that remodel the tumor microenvironment. Invasion and metastasis require neovascularization, breakdown of the basement membrane, and remodeling of the extracellular matrix for tumor cell invasion and extravasation into the blood and lymphatic vessels. The subsequent dissemination of tumor cells to distant organ sites necessitates a treacherous journey through the vasculature, which is fostered by close association with platelets and macrophages. Additionally, the establishment of the pre-metastatic niche and specific metastasis organ tropism is fostered by neutrophils and bone marrow-derived hematopoietic immune progenitor cells and other inflammatory cytokines derived from tumor and immune cells, which alter the local environment of the tissue to promote adhesion of circulating tumor cells. This review focuses on the interactions between tumor cells and immune cells recruited to the tumor microenvironment and examines the factors allowing these cells to promote each stage of metastasis.
Collapse
|
20
|
The antineoplastic effect of low-molecular-weight heparins - a literature review. Contemp Oncol (Pozn) 2013; 17:6-13. [PMID: 23788954 PMCID: PMC3685354 DOI: 10.5114/wo.2013.33766] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2012] [Revised: 04/15/2012] [Accepted: 05/07/2012] [Indexed: 11/17/2022] Open
Abstract
There is some evidence for the antitumor effect of heparins, especially the low-molecular-weight ones. The authors discuss the potential mechanism of this antineoplastic effect and present results from several in vitro and in vivo experiments. The clinical trials concerning the impact of low-molecular-weight heparins on the tumor and on the patients' survival are described. The objective was to find out if heparins could be administered as an antitumor drug, independently of their anticoagulatory properties. The antitumor role of tissue factor, heparinase, chemokines, stromal proteins, cellular interactions as well as angiogenesis and immunology seems certain. The results of the available studies seem promising but large clinical trials are necessary in order to confirm the antineoplastic effect of the low-molecular-weight heparins and to approve them for standard anticancer treatment. It could be a breakthrough in modern oncology.
Collapse
|
21
|
Baldwin AD, Kiick KL. Reversible maleimide-thiol adducts yield glutathione-sensitive poly(ethylene glycol)-heparin hydrogels. Polym Chem 2013; 4:133-143. [PMID: 23766781 DOI: 10.1039/c2py20576a] [Citation(s) in RCA: 123] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
We have recently reported that retro Michael-type addition reactions can be employed for producing labile chemical linkages with tunable sensitivity to physiologically relevant reducing potentials. We reasoned that such strategies would also be useful in the design of glutathione-sensitive hydrogels for a variety of targeted delivery and tissue engineering applications. In this report, we describe hydrogels in which maleimide-functionalized low molecular weight heparin (LMWH) is crosslinked with various thiol-functionalized poly(ethylene glycol) (PEG) multi-arm star polymers. Judicious selection of the chemical identity of the thiol permits tuning of degradation via previously unstudied, but versatile chemical methods. Thiol pKa and hydrophobicity affected both the gelation and degradation of these hydrogels. Maleimide-thiol crosslinking reactions and retro Michael-type addition reactions were verified with 1H NMR during the crosslinking and degradation of hydrogels. PEGs esterified with phenylthiol derivatives, specifically 4-mercaptophenylpropionic acid or 2,2-dimethyl-3-(4-mercaptophenyl)propionic acid, induced sensitivity to glutathione as shown by a decrease in hydrogel degradation time of 4-fold and 5-fold respectively, measured via spectrophotometric quantification of LMWH. The degradation proceeded through the retro Michael-type addition of the succinimide thioether linkage, with apparent pseudo-first order reaction constants derived from oscillatory rheology experiments of 0.039 ± 0.006 h-1 and 0.031 ± 0.003 h-1. The pseudo-first order retro reaction constants were approximately an order of magnitude slower than the degradation rate constants for hydrogels crosslinked via disulfide linkages, indicating the potential use of these Michael-type addition products for reduction-mediated release and/or degradation, with increased blood stability and prolonged drug delivery timescales compared to disulfide moieties.
Collapse
Affiliation(s)
- Aaron D Baldwin
- Department of Materials Science and Engineering, University of Delaware, 201 DuPont Hall, Newark, DE 19716, USA
| | | |
Collapse
|
22
|
Li J, King MR. Adhesion receptors as therapeutic targets for circulating tumor cells. Front Oncol 2012; 2:79. [PMID: 22837985 PMCID: PMC3402858 DOI: 10.3389/fonc.2012.00079] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2012] [Accepted: 07/07/2012] [Indexed: 12/12/2022] Open
Abstract
Metastasis contributes to >90% of cancer-associated mortality. Though primary tumors can be removed by surgical resection or chemo/radiotherapy, metastatic disease is a great challenge to treatment due to its systemic nature. As metastatic “seeds,” circulating tumor cells (CTCs) are believed to be responsible for dissemination from a primary tumor to anatomically distant organs. Despite the possibility of physical trapping of CTCs in microvessels, recent advances have provided insights into the involvement of a variety of adhesion molecules on CTCs. Such adhesion molecules facilitate direct interaction with the endothelium in specific tissues or indirectly through leukocytes. Importantly, significant progress has been made in understanding how these receptors confer enhanced invasion and survival advantage during hematogenous circulation of CTCs through recruitment of macrophages, neutrophils, platelets, and other cells. This review highlights the identification of novel adhesion molecules and how blocking their function can compromise successful seeding and colonization of CTCs in new microenvironment. Encouraged by existing diagnostic tools to identify and isolate CTCs, strategic targeting of these adhesion molecules to deliver conventional chemotherapeutics or novel apoptotic signals is discussed for the neutralization of CTCs in the circulation.
Collapse
Affiliation(s)
- Jiahe Li
- Department of Biomedical Engineering, Cornell University Ithaca, NY, USA
| | | |
Collapse
|
23
|
Maraveyas A, Johnson MJ, Xiao YP, Noble S. Malignant melanoma as a target malignancy for the study of the anti-metastatic properties of the heparins. Cancer Metastasis Rev 2011; 29:777-84. [PMID: 20936327 PMCID: PMC2962791 DOI: 10.1007/s10555-010-9263-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
The outlook for metastatic melanoma to the brain is dismal. New therapeutic avenues are therefore needed. The anti-metastatic mechanisms that may underpin the effects of low molecular weight heparins (LMWHs) in in vitro and preclinical melanoma models warrant translating to a clinical setting. This review outlines a rationale that supports our proposal that metastatic melanoma to the brain is a clinical setting in which to study the anti-metastatic potential of LMWHs. Prevention or delay of brain metastases in melanoma is a clinically relevant and measurable target. Studies to explore the effect of anticoagulants on cancer survival are underway in other malignancies such as lung, pancreas, ovary, breast, and stomach cancer. However, no study to our knowledge has a methodology that could produce clinical evidence in support of a mechanism for whatever benefit may be seen. The setting we propose would allow translation of the molecular knowledge of the metastatic pathways mediated by platelets and the selectins—all potential targets of heparin—in a “time to appearance” of brain metastases endpoint. Since brain metastases are so common and they have a singularly adverse impact on survival, the “biological neuroprotection” model we propose in metastatic melanoma could provide the translational evidence to support the benefit of LMWHs in melanoma. More significantly, this would open the door to a wider “anti-metastatic” approach that could have much greater impact in patients with minimal disease being treated in adjuvant settings for the more common malignancies such as breast and colon cancer.
Collapse
|
24
|
High antiangiogenic and low anticoagulant efficacy of orally active low molecular weight heparin derivatives. J Control Release 2010; 148:317-26. [PMID: 20869408 DOI: 10.1016/j.jconrel.2010.09.014] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2010] [Revised: 07/02/2010] [Accepted: 09/14/2010] [Indexed: 11/23/2022]
Abstract
Heparin, an anticoagulant that is widely used clinically, is also known to bind to several kinds of proteins through electrostatic interactions because of its polyanionic character. These interactions are mediated by the physicochemical properties of heparin such as sequence composition, sulfation patterns, charge distribution, overall charge density, and molecular size. Although this electrostatic character mediates its binding to many proteins related with tumor progression, thereby providing its antiangiogenic property, the administration of heparin for treating cancer is limited in clinical applications due to several drawbacks, such as its low oral absorption, unsatisfactory therapeutic effects, and strong anticoagulant activity which induces hemorrhaging. Here, we evaluated novel, orally active, low molecular weight heparin (LMWH) derivatives (LHD) conjugated with deoxycholic acid (DOCA) that show reduced anticoagulant activity and enhanced antiangiogenic activity. The chemical conjugate of LMWH and DOCA was synthesized by conjugating the amine group of N-deoxycholylethylamine (EtDOCA) with the carboxylic groups of heparin at various DOCA conjugation ratios. The LMWH-DOCA conjugate series (LHD1, LHD1.5, LHD2, and LHD4) were further formulated with poloxamer 407 as a solubilizer for oral administration. An in vitro endothelial tubular formation and in vivo Matrigel plug assay were performed to verify the antiangiogenic potential of LHD. Finally, we evaluated tumor growth inhibition of oral LHD administration in a SCC7 model as well as in A549 human cancer cell lines in a mouse xenograft model. Increasing DOCA conjugation ratios showed decreased anticoagulant activity, eventually to zero. LHD could block angiogenesis in the tubular formation assay and the Matrigel plug assay. In particular, oral administration of LHD4, which has 4 molecules of DOCA per mole of LMWH, inhibited tumor growth in SCC7 mice model as well as A549 mice xenograft model. LHD4 was orally absorbable, showed minimal anticoagulant activity and inhibits tumor growth via antiangiogenesis. These findings demonstrate the therapeutic potential of LHD4 as a new oral anti-cancer drug.
Collapse
|
25
|
Kim SK, Lee S, Jin S, Moon HT, Jeon OC, Lee DY, Byun Y. Diabetes Correction in Pancreatectomized Canines by Orally Absorbable Insulin−Deoxycholate Complex. Mol Pharm 2010; 7:708-17. [DOI: 10.1021/mp9002688] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Sang Kyoon Kim
- College of Pharmacy, Seoul National University, Seoul 151-742, South Korea, Mediplex Corporation, Seoul 135-729, South Korea, Department of Bioengineering, College of Engineering, Hanyang University, Seoul 133-791, South Korea, and Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul 151-742, South Korea
| | - Seulki Lee
- College of Pharmacy, Seoul National University, Seoul 151-742, South Korea, Mediplex Corporation, Seoul 135-729, South Korea, Department of Bioengineering, College of Engineering, Hanyang University, Seoul 133-791, South Korea, and Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul 151-742, South Korea
| | - Sunji Jin
- College of Pharmacy, Seoul National University, Seoul 151-742, South Korea, Mediplex Corporation, Seoul 135-729, South Korea, Department of Bioengineering, College of Engineering, Hanyang University, Seoul 133-791, South Korea, and Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul 151-742, South Korea
| | - Hyun Tae Moon
- College of Pharmacy, Seoul National University, Seoul 151-742, South Korea, Mediplex Corporation, Seoul 135-729, South Korea, Department of Bioengineering, College of Engineering, Hanyang University, Seoul 133-791, South Korea, and Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul 151-742, South Korea
| | - Ok Cheol Jeon
- College of Pharmacy, Seoul National University, Seoul 151-742, South Korea, Mediplex Corporation, Seoul 135-729, South Korea, Department of Bioengineering, College of Engineering, Hanyang University, Seoul 133-791, South Korea, and Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul 151-742, South Korea
| | - Dong Yun Lee
- College of Pharmacy, Seoul National University, Seoul 151-742, South Korea, Mediplex Corporation, Seoul 135-729, South Korea, Department of Bioengineering, College of Engineering, Hanyang University, Seoul 133-791, South Korea, and Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul 151-742, South Korea
| | - Youngro Byun
- College of Pharmacy, Seoul National University, Seoul 151-742, South Korea, Mediplex Corporation, Seoul 135-729, South Korea, Department of Bioengineering, College of Engineering, Hanyang University, Seoul 133-791, South Korea, and Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul 151-742, South Korea
| |
Collapse
|
26
|
Borsig L. Antimetastatic activities of heparins and modified heparins. Experimental evidence. Thromb Res 2010; 125 Suppl 2:S66-71. [DOI: 10.1016/s0049-3848(10)70017-7] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
27
|
Lee DY, Lee SW, Kim SK, Lee M, Chang HW, Moon HT, Byun Y, Kim SY. Antiangiogenic Activity of Orally Absorbable Heparin Derivative in Different Types of Cancer Cells. Pharm Res 2009; 26:2667-76. [DOI: 10.1007/s11095-009-9989-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2009] [Accepted: 09/28/2009] [Indexed: 10/20/2022]
|
28
|
Abstract
It was nearly 100 years since heparin was discovered, but the role of this widely used anticoagulant is still remarkably thought provoking now. During pathological processes such as atherosclerosis, inflammation, cancer and infection, phenomena of cell adhesion are ubiquitous and complicated. Heparin exerts anti-adhesion activity appearing as a common mechanism of its potential polypharmacology in those diseases. Furthermore, heparin can bind a variety of signalling molecules such as growth factors, cell surface proteins of pathogens and most notably, cell adhesion molecules. These signalling molecules are involved in cell communication, acting as ligands, receptors and second messengers. Considering that heparan sulphate glycosaminoglycan is increasingly recognized as a key mediator in many cellular processes, the structural similarity with heparan sulphate suggests that heparin is a multifunctional intervenor in cell communication.
Collapse
Affiliation(s)
- Xianxiang Xu
- Department of Pharmacology of Chinese Materia Medica, China Pharmaceutical University, Nanjing, China
| | | |
Collapse
|
29
|
Cui ZY, Park MJ, Lee J, Ahn JS, Ahn MJ, Seo SW, Park JW, Byun Y, Park K. Modest anti-cancer activity of a bile acid acylated heparin derivative in a PC14PE6 induced orthotopic lung cancer model. Cancer Res Treat 2009; 41:80-6. [PMID: 19707505 DOI: 10.4143/crt.2009.41.2.80] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2009] [Accepted: 05/19/2009] [Indexed: 11/21/2022] Open
Abstract
PURPOSE A novel chemically modified heparin derivative, heparin-deoxycholic acid nano-particles, has lower anticoagulant activity, and was recently reported to have significant anti-tumor effects on squamous head and neck cancer cells. Therefore, the aim of this study was to evaluate the anti-tumor effects of heparin-deoxycholic acid nano-particles in a human lung adenocarcinoma cell line. MATERIALS AND METHODS An orthotopic lung cancer model in 16 mice was developed using intra-thoracic injections of 0.5x10(6) PC14PE6 cells. Ten days after inoculation, the mice were divided into two groups. PBS and Heparin-DOCA particles were injected once a day every 3 days in the tail vein, for a total of 5 injections. The body weight and survival of each mouse were monitored and the tumor size in the lung was measured by SPECT-CT before and after heparin-DOCA nano-particle treatment. RESULTS IThe HD particles had no significant cytotoxicity when the PC9 cells were treated in vitro. There was no statistical difference in tumor size, body weight and survival between the HD treated and control groups in vivo. Furthermore, there was no difference in the amount of CD31 between tumor tissues in the two study groups. CONCLUSION HD synthesized with unfractionated heparin had no apparent inhibitory effects on tumor growth in a PC14PE6 cell induced orthotopic lung cancer mouse model. The HD particles did not significantly inhibit tumor-induced angiogenesis at the tumor sites.
Collapse
Affiliation(s)
- Zheng Yun Cui
- Medical Nanoelement Development Center, Seoul, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Borensztajn K, Bijlsma MF, Reitsma PH, Peppelenbosch MP, Spek CA. Coagulation factor Xa inhibits cancer cell migration via protease-activated receptor-1 activation. Thromb Res 2009; 124:219-25. [PMID: 19250659 DOI: 10.1016/j.thromres.2009.01.015] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2008] [Revised: 12/16/2008] [Accepted: 01/21/2009] [Indexed: 01/05/2023]
Abstract
Cell migration is critically important in (patho)physiological processes. The metastatic potential of cancer cells partly depends on activation of the coagulation cascade. The aim of the present study was to determine whether coagulation factor X (FXa) can regulate the migration and invasion of cancer cells. Quite unexpectedly, we found that FXa markedly diminished the migration of different cancer cell lines of various origins (breast, lung and colon cancer cells). We showed that FXa mediated inhibition of cancer cell migration was specific, as it was inhibited by TAP (a specific FXa inhibitor) but not by Hirudin (a specific thrombin inhibitor). Moreover, the FXa effect was dose dependent, with a maximal inhibitory effect reached at 0.75 U/ml FXa (corresponding to 130.5 nM). Next, we determined that FXa acted via protease-activated receptor (PAR)-1-dependent signaling, and PAR-1 desensitization, as well as knocking-down PAR-1 expression, abolished the FXa effects. Finally, we showed that Gialpha was not involved in FXa mediated inhibition of cell migration as its effects were not reverted by pertussis toxin. These results suggest that, beyond its role in blood coagulation, FXa plays a key role in cancer cell migration. They also shed light on an unexpected role of PAR-1, which appears to be a Janus-like receptor in cancer cell biology.
Collapse
Affiliation(s)
- Keren Borensztajn
- Department of Cell Biology, University of Groningen, Groningen, The Netherlands.
| | | | | | | | | |
Collapse
|