1
|
Rasel MSI, Mohona FA, Akter W, Kabir S, Chowdhury AA, Chowdhury JA, Hassan MA, Al Mamun A, Ghose DK, Ahmad Z, Khan FS, Bari MF, Rahman MS, Amran MS. Exploration of Site-Specific Drug Targeting-A Review on EPR-, Stimuli-, Chemical-, and Receptor-Based Approaches as Potential Drug Targeting Methods in Cancer Treatment. JOURNAL OF ONCOLOGY 2022; 2022:9396760. [PMID: 36284633 PMCID: PMC9588330 DOI: 10.1155/2022/9396760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 07/21/2022] [Indexed: 11/17/2022]
Abstract
Cancer has been one of the most dominant causes of mortality globally over the last few decades. In cancer treatment, the selective targeting of tumor cells is indispensable, making it a better replacement for conventional chemotherapies by diminishing their adverse side effects. While designing a drug to be delivered selectively in the target organ, the drug development scientists should focus on various factors such as the type of cancer they are dealing with according to which drug, targeting moieties, and pharmaceutical carriers should be targeted. All published articles have been collected regarding cancer and drug-targeting approaches from well reputed databases including MEDLINE, Embase, Cochrane Library, CENTRAL and ClinicalTrials.gov, Science Direct, PubMed, Scopus, Wiley, and Springer. The articles published between January 2010 and December 2020 were considered. Due to the existence of various mechanisms, it is challenging to choose which one is appropriate for a specific case. Moreover, a combination of more than one approach is often utilized to achieve optimal drug effects. In this review, we have summarized and highlighted central mechanisms of how the targeted drug delivery system works in the specific diseased microenvironment, along with the strategies to make an approach more effective. We have also included some pictorial illustrations to have a precise idea about different types of drug targeting. The core contribution of this work includes providing a cancer drug development scientist with a broad preliminary idea to choose the appropriate approach among the various targeted drug delivery mechanisms. Also, the study will contribute to improving anticancer treatment approaches by providing a pathway for lesser side effects observed in conventional chemotherapeutic techniques.
Collapse
Affiliation(s)
- Md. Shamiul Islam Rasel
- Department of Pharmacy, Faculty of Pharmacy, University of Dhaka, Shahbag, Dhaka 1000, Bangladesh
| | - Farhana Afrin Mohona
- Department of Pharmacy, Faculty of Pharmacy, University of Dhaka, Shahbag, Dhaka 1000, Bangladesh
| | - Wahida Akter
- College of Pharmacy, University of Houston, Houston, USA
| | - Shaila Kabir
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Dhaka, Shahbag, Dhaka 1000, Bangladesh
| | - Abu Asad Chowdhury
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Dhaka, Shahbag, Dhaka 1000, Bangladesh
| | - Jakir Ahmed Chowdhury
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Dhaka, Shahbag, Dhaka 1000, Bangladesh
| | - Md. Abul Hassan
- Department of Science & Technology, Tokushima University Graduate School, Tokushima, Japan
| | - Abdullah Al Mamun
- Molecular Pharmacology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035 Zhejiang, China
| | - Dipayon Krisna Ghose
- Department of Biochemistry and Molecular Biology, Jagannath University, Dhaka 1100, Bangladesh
| | - Zubair Ahmad
- Unit of Bee Research and Honey Production, King Khalid University, Abha 61413, Saudi Arabia
- Department of Biology, College of Arts and Sciences, King Khalid University, Abha 61413, Saudi Arabia
| | - Farhat S. Khan
- Department of Biology, College of Arts and Sciences, King Khalid University, Abha 61413, Saudi Arabia
| | - Md. Fazlul Bari
- Department of Biochemistry and Molecular Biology, Trust University, Barishal, Ruiya, Nobogram Road, Barishal 8200, Bangladesh
| | - Md. Sohanur Rahman
- Department of Biochemistry and Molecular Biology, Trust University, Barishal, Ruiya, Nobogram Road, Barishal 8200, Bangladesh
| | - Md. Shah Amran
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Dhaka, Shahbag, Dhaka 1000, Bangladesh
| |
Collapse
|
2
|
Zi Y, Yang K, He J, Wu Z, Liu J, Zhang W. Strategies to enhance drug delivery to solid tumors by harnessing the EPR effects and alternative targeting mechanisms. Adv Drug Deliv Rev 2022; 188:114449. [PMID: 35835353 DOI: 10.1016/j.addr.2022.114449] [Citation(s) in RCA: 87] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 05/03/2022] [Accepted: 07/06/2022] [Indexed: 12/20/2022]
Abstract
The Enhanced Permeability and Retention (EPR) effect has been recognized as the central paradigm in tumor-targeted delivery in the last decades. In the wake of this concept, nanotechnologies have reached phenomenal levels in research. However, clinical tumors display a poor manifestation of EPR effect. Factors including tumor heterogeneity, complicating tumor microenvironment, and discrepancies between laboratory models and human tumors largely contribute to poor efficiency in tumor-targeted delivery and therapeutic failure in clinical translation. In this article, approaches for evaluation of EPR effect in human tumor were overviewed as guidance to employ EPR effect for cancer treatment. Strategies to augment EPR-mediated tumoral delivery are discussed in different dimensions including enhancement of vascular permeability, depletion of tumor extracellular matrix and optimization of nanoparticle design. Besides, the recent development in alternative tumor-targeted delivery mechanisms are highlighted including transendothelial pathway, endogenous cell carriers and non-immunogenic bacteria-mediated delivery. In addition, the emerging preclinical models better reflect human tumors are introduced. Finally, more rational applications of EPR effect in other disease and field are proposed. This article elaborates on fundamental reasons for the gaps between theoretical expectation and clinical outcomes, attempting to provide some perspective directions for future development of cancer nanomedicines in this still evolving landscape.
Collapse
Affiliation(s)
- Yixuan Zi
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, PR China
| | - Kaiyun Yang
- School of Pharmacy, University of Auckland, Private Bag 92019, Auckland, New Zealand
| | - Jianhua He
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, PR China
| | - Zimei Wu
- School of Pharmacy, University of Auckland, Private Bag 92019, Auckland, New Zealand.
| | - Jianping Liu
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, PR China.
| | - Wenli Zhang
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, PR China.
| |
Collapse
|
3
|
Exploiting the dynamics of the EPR effect and strategies to improve the therapeutic effects of nanomedicines by using EPR effect enhancers. Adv Drug Deliv Rev 2020; 157:142-160. [PMID: 32553783 DOI: 10.1016/j.addr.2020.06.005] [Citation(s) in RCA: 404] [Impact Index Per Article: 80.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 06/03/2020] [Accepted: 06/04/2020] [Indexed: 12/18/2022]
Abstract
The enhanced permeability and retention (EPR) effect is a unique phenomenon of solid tumors that is related to their particular anatomical and pathophysiological characteristics, e.g. defective vascular architecture; large gaps between endothelial cells in blood vessels; abundant vascular mediators such as bradykinin, nitric oxide, carbon monoxide, and vascular endothelial growth factor; and impaired lymphatic recovery. These features lead to tumor tissues showing considerable extravasation of plasma components and nanomedicines. These data comprise the basic theory underlying the development of macromolecular agents or nanomedicines. The EPR effect is not necessarily valid for all solid tumors, because tumor blood flow and vascular permeability vary greatly. Tumor blood flow is frequently obstructed as tumor size increases, as often seen clinically; early stage, small tumors show a more uniform EPR effect, whereas advanced large tumor show heterogeneity in EPR effect. Accordingly, it would be very important to apply enhancers of EPR effect in clinical setting to make EPR effect more uniform. In this review, we discuss the EPR effect: its history, factors involved, and dynamics and heterogeneity. Strategies to overcome the EPR effect's heterogeneity may guarantee better therapeutic outcomes of drug delivery to advanced cancers.
Collapse
|
4
|
Biomimetic nano-NOS mediated local NO release for inhibiting cancer-associated platelet activation and disrupting tumor vascular barriers. Biomaterials 2020; 255:120141. [PMID: 32505753 DOI: 10.1016/j.biomaterials.2020.120141] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 05/20/2020] [Accepted: 05/21/2020] [Indexed: 02/07/2023]
Abstract
Platelets attribute to the hypercoagulation of blood and maintenance of the tumor vascular integrity, resulting in limited intratumoral perfusion of nanoparticle into solid tumors. To overcome these adversities, we herein present an antiplatelet strategy based on erythrocyte membrane-enveloped proteinic nanoparticles that biomimic nitric oxide synthase (NOS)with co-loading of l-Arginine (LA) and photosensitizer IR783 for local NO release and inhibition of the activation of tumor-associated platelets specifically, thereby enhancing vascular permeability and accumulation of the nanoparticles in tumors. A cRGD-immobolized membrane structure is constructed to actively target platelets and cancer cells respectively, through overexpressed integrin receptors such as integrin αIIbβ3 and αvβ3, accelerating the inhibition of platelet activation and endocytosis of nanoparticles by tumor cells. Bio-mimicking the arginine/NO pathway in vivo, synergistical delivery of LA and IR783 enables LA molecules readily oxidize to NO with O2 that is mediated by activated IR783, the resulted NO not only retards the activity of platelets to disrupt the vascular integrity of tumor but also enhances toxicity to cancer cells. In addition, NIR-controlled release localizes the NO spatiotemporally to tumor-associated platelets and prevents undesirable systemic bleeding substantially. The reduction of the hypercoagulable state is further demonstrated by the down-regulation of tissues factor (TF) expression in tumor cells. Our study describes a promising approach to combat cancer, which advances the biomimetic NOS system as the potent therapeutic forces toward clinic applications.
Collapse
|
5
|
Valero N, Mosquera J, Torres M, Duran A, Velastegui M, Reyes J, Fernandez M, Fernandez G, Veliz T. Increased serum ferritin and interleukin-18 levels in children with dengue. Braz J Microbiol 2019; 50:649-656. [PMID: 31243722 DOI: 10.1007/s42770-019-00105-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 06/12/2019] [Indexed: 12/12/2022] Open
Abstract
Activated monocytes/macrophages that produce a cytokine storm play an important role in the pathogenesis of dengue. Interleukin-18 (IL-18) is a proinflammatory cytokine produced by monocyte/macrophages that is increased during dengue. Ferritin is an acute-phase reactant and expressed by cells of the reticulo-endothelial system in response to infection by dengue virus. The aims of this study were to analyze the simultaneous expression of both IL-18 and ferritins in children infected by diverse serotypes of dengue virus (DENV) and determine their association with dengue severity. In this regard, children with dengue (n = 25) and healthy controls with similar age and sex (n = 20) were analyzed for circulating ferritin and cytokines. Monocytes were isolated by Hystopaque gradient and co-cultured with DENV-2. IL-18 and ferritin contents in blood, and IL-18 in culture supernatants were determined by ELISA. Increased levels of ferritin and IL-18 (p < 0.0001) were observed in dengue patients, not associated to NS1expression or type of infection (primary or secondary). Highest values of both molecules (p < 0.001) were observed in dengue with warning signs and severe dengue. Differential effect on IL-18/ferritin production was observed associated to viral serotype infection. There were no correlations between ferritin vs. IL-18 production, ferritin vs. NS1 status, and IL-18 vs. NS1 status. Viral-infected monocyte cultures showed increased production of IL-18 (p < 0.001). In conclusion, increased circulating ferritin and IL-18 are expressed in children infected by different serotypes of DENV associated with dengue severity.
Collapse
Affiliation(s)
- Nereida Valero
- Instituto de Investigaciones Clínicas "Dr. Américo Negrette", Facultad de Medicina, Universidad del Zulia, Apartado Postal 23, Maracaibo, Zulia, 4001-A, Venezuela. .,Universidad Estatal del Sur de Manabi, Jipijapa, Ecuador.
| | - Jesus Mosquera
- Instituto de Investigaciones Clínicas "Dr. Américo Negrette", Facultad de Medicina, Universidad del Zulia, Apartado Postal 23, Maracaibo, Zulia, 4001-A, Venezuela
| | - Mariana Torres
- Instituto de Investigaciones Clínicas "Dr. Américo Negrette", Facultad de Medicina, Universidad del Zulia, Apartado Postal 23, Maracaibo, Zulia, 4001-A, Venezuela
| | - Anyelo Duran
- Instituto de Investigaciones Clínicas "Dr. Américo Negrette", Facultad de Medicina, Universidad del Zulia, Apartado Postal 23, Maracaibo, Zulia, 4001-A, Venezuela
| | - Maria Velastegui
- Instituto de Investigaciones Clínicas "Dr. Américo Negrette", Facultad de Medicina, Universidad del Zulia, Apartado Postal 23, Maracaibo, Zulia, 4001-A, Venezuela
| | - Javier Reyes
- Universidad Estatal del Sur de Manabi, Jipijapa, Ecuador
| | - Miriam Fernandez
- Facultad Ciencias de la Salud, Universidad Técnica de Ambato, Ambato, Ecuador
| | - Gerardo Fernandez
- Facultad Ciencias de la Salud, Universidad Técnica de Ambato, Ambato, Ecuador
| | - Teresa Veliz
- Universidad Estatal del Sur de Manabi, Jipijapa, Ecuador
| |
Collapse
|
6
|
da Costa PLN, Wynne D, Fifis T, Nguyen L, Perini M, Christophi C. The kallikrein-Kinin system modulates the progression of colorectal liver metastases in a mouse model. BMC Cancer 2018; 18:382. [PMID: 29618333 PMCID: PMC5885419 DOI: 10.1186/s12885-018-4260-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Accepted: 03/20/2018] [Indexed: 01/06/2023] Open
Abstract
Background The Kallikrein-Kinin System (KKS) has been found to play a role in tumor progression in several cancers. The KKS metabolic cascade depends on signalling through two cross talking receptors; bradykinin receptor 1 (B1R) and bradykinin receptor 2 (B2R). Activation of the Kinin receptor is responsible for multiple pathophysiologic functions including increase of vascular permeability and induction of host inflammatory responses that exert diverse effects on tumor growth. Methods B1R and B2R expression on mouse and human CRC cell lines was investigated. Changes in tumor growth and progression was assessed in male CBA mice bearing colorectal liver metastases (CRLM) following treatment with B1R or B2R blockers. In vitro cultures of human SW-480 and mouse colorectal cancer (MoCR) cell lines were examined for changes in their proliferation and migration properties following treatment with B1R or B2R blockers. Results Both colorectal cancer cell lines tested strongly positive for B1R and B2R expression. Inhibition of both receptors retarded tumor growth but only B1R blockade significantly reduced tumor load and increased tumor apoptosis. Blockade of either receptor reduced tumor vascularization in vivo and significantly inhibited proliferation and migration of colorectal cancer cells in vitro. Conclusion Taken together, the present study demonstrated that kinin receptor blockade inhibited tumor growth and reduced its invading properties suggesting that KKS manipulation could be a novel target in colorectal cancer therapy. Electronic supplementary material The online version of this article (10.1186/s12885-018-4260-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Patricia Luiza Nunes da Costa
- Department of Surgery, University of Melbourne, Austin Health, Lance Townsend Building Level 8, Studley Rd, Heidelberg, VIC, 3084, Australia.,Laboratório de Oncologia Experimental, Faculdade de Medicina da Universidade de São Paulo and Instituto do Câncer do Estado de São Paulo, São Paulo, Brazil
| | - David Wynne
- Department of Surgery, University of Melbourne, Austin Health, Lance Townsend Building Level 8, Studley Rd, Heidelberg, VIC, 3084, Australia
| | - Theodora Fifis
- Department of Surgery, University of Melbourne, Austin Health, Lance Townsend Building Level 8, Studley Rd, Heidelberg, VIC, 3084, Australia.
| | - Linh Nguyen
- Department of Surgery, University of Melbourne, Austin Health, Lance Townsend Building Level 8, Studley Rd, Heidelberg, VIC, 3084, Australia
| | - Marcos Perini
- Department of Surgery, University of Melbourne, Austin Health, Lance Townsend Building Level 8, Studley Rd, Heidelberg, VIC, 3084, Australia
| | - Christopher Christophi
- Department of Surgery, University of Melbourne, Austin Health, Lance Townsend Building Level 8, Studley Rd, Heidelberg, VIC, 3084, Australia
| |
Collapse
|
7
|
Abstract
History of the EPR (enhanced permeability and retention) effect is discussed, which goes back to the analyses of molecular pathology in bacterial infection and edema (extravasation) formation. The first mediator we found for extravasation was bradykinin. Later on, were found nitric oxide and superoxide, then formation of peroxynitrite, that activates procollagenase. In this inflammatory setting many other vascular mediators are involved that are also common to cancer vasculature. Obviously cancer vasculature is defective architechtally, and this makes macromolecular drugs more permeable through the vascular wall. The importance of this pathophysiological event of EPR effect can be applied to macromolecular drug-delivery, or tumor selective delivery, which takes hours to achieve in the primary as well as metastatic tumors, not to mention of the inflamed tissues. The retention of the EPR means that such drugs will be retained in tumor tissues more than days to weeks. This was demonstrated initially, and most dramatically, using SMANCS, a protein-polymer conjugated-drug dissolved in lipid contrast medium (Lipiodol) by administering intraarterially. For disseminating the EPR concept globally, or in the scientific community, Professor Ruth Duncan played a key role at the early stage, as she worked extensively on polymer- therapeutics, and knew its importance.
Collapse
Affiliation(s)
- Hiroshi Maeda
- a Kumamoto University and BioDynamic Research Foundation , Kumamoto , Japan.,b Department of Molecular Pathology, Division of Health Sciences, Graduate School of Medicine , Osaka University , Osaka , Japan
| |
Collapse
|
8
|
Atukorale PU, Covarrubias G, Bauer L, Karathanasis E. Vascular targeting of nanoparticles for molecular imaging of diseased endothelium. Adv Drug Deliv Rev 2017; 113:141-156. [PMID: 27639317 DOI: 10.1016/j.addr.2016.09.006] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2016] [Revised: 09/02/2016] [Accepted: 09/08/2016] [Indexed: 01/08/2023]
Abstract
This review seeks to highlight the enormous potential of targeted nanoparticles for molecular imaging applications. Being the closest point-of-contact, circulating nanoparticles can gain direct access to targetable molecular markers of disease that appear on the endothelium. Further, nanoparticles are ideally suitable to vascular targeting due to geometrically enhanced multivalent attachment on the vascular target. This natural synergy between nanoparticles, vascular targeting and molecular imaging can provide new avenues for diagnosis and prognosis of disease with quantitative precision. In addition to the obvious applications of targeting molecular signatures of vascular diseases (e.g., atherosclerosis), deep-tissue diseases often manifest themselves by continuously altering and remodeling their neighboring blood vessels (e.g., cancer). Thus, the remodeled endothelium provides a wide range of targets for nanoparticles and molecular imaging. To demonstrate the potential of molecular imaging, we present a variety of nanoparticles designed for molecular imaging of cancer or atherosclerosis using different imaging modalities.
Collapse
|
9
|
Effect of Nitric Oxide on the Antifungal Activity of Oxidative Stress and Azoles Against Candida albicans. Indian J Microbiol 2016; 56:214-218. [PMID: 27570314 DOI: 10.1007/s12088-016-0580-x] [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] [Received: 03/10/2016] [Accepted: 03/31/2016] [Indexed: 12/26/2022] Open
Abstract
Nitric oxide (NO) is a small molecule with a wide range of biological activities in mammalian and bacteria. However, the role of NO in fungi, especially Candida albicans, is not clear. In this study, we confirmed the generation of endogenous NO in C. albicans, and found that the production of endogenous NO in C. albicans was associated with nitric oxide synthase pathway. Our results further indicated that the production of endogenous NO in C. albicans was reduced under oxidative stress such as menadione or H2O2 treatment. Meanwhile, exogenous NO donor, sodium nitroprusside (SNP), synergized with H2O2 against C. albicans. Interestingly, SNP could inhibit the antifungal effect of azoles against C. albicans in vitro, suggesting that NO might be involved in the resistance of C. albicans to antifungals. Collectively, this study demonstrated the production of endogenous NO in C. albicans, and indicated that NO may play an important role in the response of C. albicans to oxidative stress and azoles.
Collapse
|
10
|
van de Weg CAM, Huits RMHG, Pannuti CS, Brouns RM, van den Berg RWA, van den Ham HJ, Martina BEE, Osterhaus ADME, Netea MG, Meijers JCM, van Gorp ECM, Kallas EG. Hyperferritinaemia in dengue virus infected patients is associated with immune activation and coagulation disturbances. PLoS Negl Trop Dis 2014; 8:e3214. [PMID: 25299654 PMCID: PMC4191960 DOI: 10.1371/journal.pntd.0003214] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2014] [Accepted: 08/25/2014] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND During a dengue outbreak on the Caribbean island Aruba, highly elevated levels of ferritin were detected in dengue virus infected patients. Ferritin is an acute-phase reactant and hyperferritinaemia is a hallmark of diseases caused by extensive immune activation, such as haemophagocytic lymphohistiocytosis. The aim of this study was to investigate whether hyperferritinaemia in dengue patients was associated with clinical markers of extensive immune activation and coagulation disturbances. METHODOLOGY/PRINCIPAL FINDINGS Levels of ferritin, standard laboratory markers, sIL-2R, IL-18 and coagulation and fibrinolytic markers were determined in samples from patients with uncomplicated dengue in Aruba. Levels of ferritin were significantly increased in dengue patients compared to patients with other febrile illnesses. Moreover, levels of ferritin associated significantly with the occurrence of viraemia. Hyperferritinaemia was also significantly associated with thrombocytopenia, elevated liver enzymes and coagulation disturbances. The results were validated in a cohort of dengue virus infected patients in Brazil. In this cohort levels of ferritin and cytokine profiles were determined. Increased levels of ferritin in dengue virus infected patients in Brazil were associated with disease severity and a pro-inflammatory cytokine profile. CONCLUSIONS/SIGNIFICANCE Altogether, we provide evidence that ferritin can be used as a clinical marker to discriminate between dengue and other febrile illnesses. The occurrence of hyperferritinaemia in dengue virus infected patients is indicative for highly active disease resulting in immune activation and coagulation disturbances. Therefore, we recommend that patients with hyperferritinaemia are monitored carefully.
Collapse
Affiliation(s)
| | - Ralph M. H. G. Huits
- Emergency Department and Department of Internal Medicine, Dr. Horacio E. Oduber Hospitaal, Oranjestad, Aruba
| | - Cláudio S. Pannuti
- Instituto de Medicina Tropical de São Paulo e Departamento de Moléstias Infecciosas e Parasitárias (LIM-52), Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | | | | | | | - Byron E. E. Martina
- Department of Viroscience, Erasmus Medical Center, Rotterdam, The Netherlands
| | | | - Mihai G. Netea
- Department of Experimental Internal Medicine, Radboud University, Nijmegen, The Netherlands
| | - Joost C. M. Meijers
- Department of Experimental Vascular Medicine, Academic Medical Center, Amsterdam, The Netherlands
| | - Eric C. M. van Gorp
- Department of Viroscience, Erasmus Medical Center, Rotterdam, The Netherlands
- * E-mail:
| | - Esper G. Kallas
- Disciplina de Imunologia Clínica e Alergia (LIM-60), Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
11
|
Ishida T, Kiwada H. Alteration of tumor microenvironment for improved delivery and intratumor distribution of nanocarriers. Biol Pharm Bull 2014; 36:692-7. [PMID: 23649327 DOI: 10.1248/bpb.b13-00121] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Nanocarrier-based cancer chemotherapeutics are thought to increase therapeutic efficiency and reduce the side effects of associated chemotherapeutic agents by altering the agents' pharmacokinetics and tissue distribution following intravenous administration. In spite of these favorable properties, nanocarrier-based cancer chemotherapeutics are not always effective because of their heterogeneous intratumoral localization. Homogeneous distribution of nanocarriers in a tumor would improve the efficacy of nanocarrier-based cancer chemotherapeutics. In this article, we describe and discuss some trials that attempt to manipulate the barriers in the tumor microenvironment that hinder extravasation through the tumor vasculature and penetration of nanocarriers in solid tumors. Alterations of the tumor microenvironment that relate directly to the intratumoral distribution of nanocarriers may be potential strategies to improve the delivery of nanocarrier-based cancer chemotherapeutics.
Collapse
Affiliation(s)
- Tatsuhiro Ishida
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Health Biosciences, The University of Tokushima, Tokushima 770–8503, Japan.
| | | |
Collapse
|
12
|
Proteomics-based identification of plasma proteins and their association with the host-pathogen interaction in chronic typhoid carriers. Int J Infect Dis 2013; 19:59-66. [PMID: 24291468 PMCID: PMC7129176 DOI: 10.1016/j.ijid.2013.10.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2013] [Revised: 10/08/2013] [Accepted: 10/09/2013] [Indexed: 01/03/2023] Open
Abstract
Background Current diagnostic tests are inadequate to detect typhoid cases, as well as the chronic carrier state, the sole reservoir of Salmonella enterica serovar Typhi. The current study was conducted to find new molecular signatures of pathogen/disease to understand the mechanism behind the host–pathogen interaction in enteric fever. Methods Proteomics-based studies were done to determine the expression of differentially expressed proteins in the plasma of controls, acute typhoid cases, and chronic typhoid carriers. Further, transcriptome-based analysis using reverse-transcriptase PCR (RT-PCR) was done in controls, acute typhoid cases, and chronic typhoid carriers. Results Results showed the upregulation of proprotein convertase subtilisin, furin, haptoglobin, and albumin in the plasma of chronic typhoid carriers. The elevation in mRNA expression of four differentially expressed proteins confirms the changes at the transcriptional level. Further, the increase in albumin and haptoglobin in chronic typhoid carriers shows their role in free radical generation, inflammation, and monocyte cell signaling. Conclusion Through proteomics techniques, this study identified four proteins in the chronic typhoid carrier host that may have a role in the disease pathogenesis of enteric fever.
Collapse
|
13
|
Maeda H. The link between infection and cancer: tumor vasculature, free radicals, and drug delivery to tumors via the EPR effect. Cancer Sci 2013; 104:779-89. [PMID: 23495730 PMCID: PMC7657157 DOI: 10.1111/cas.12152] [Citation(s) in RCA: 117] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2013] [Accepted: 03/10/2013] [Indexed: 12/16/2022] Open
Abstract
This review focuses primarily on my own research, including pathogenic mechanisms of microbial infection, vascular permeability in infection and tumors, and effects of nitric oxide (NO), superoxide anion radical (O₂⁻), and 8-nitroguanosine in the enhanced permeability and retention (EPR) effect for the tumor-selective delivery of macromolecular agents (nanomedicines). Infection-induced vascular permeability is mediated by activation of the kinin-generating protease cascade (kallikrein-kinin) triggered by exogenous microbial proteases. A similar mechanism operates in cancer tissues and in carcinomatosis of the pleural and peritoneal cavities. Infection also stimulates O₂⁻ generation via activation of xanthine oxidase while generating NO by inducing NO synthase. These chemicals function in mutation and carcinogenesis and promote inflammation, in which peroxynitrite (a product of O₂⁻ and NO) activates MMP, damages DNA and RNA, and regenerates 8-nitroguanosine and 8-oxoguanosine. We showed vascular permeability by using macromolecular drugs, which are not simply extravasated through the vascular wall into the tumor interstitium but remain there for prolonged periods. We thus discovered the EPR effect, which led to the rational development of tumor-selective delivery of polymer conjugates, micellar and liposomal drugs, and genes. Our styrene-maleic acid copolymer conjugated with neocarzinostatin was the first agent of its kind used to treat hepatoma. The EPR effect occurs not only because of defective vascular architecture but also through the generation of various vascular mediators such as kinin, NO, and vascular endothelial growth factor. Although most solid tumors, including human tumors, show the EPR effect, heterogeneity of tumor tissue may impede drug delivery. This review describes the barriers and countermeasures for improved drug delivery to tumors by using nanomedicines.
Collapse
Affiliation(s)
- Hiroshi Maeda
- Institute of Drug Delivery System Research, Sojo University, Kumamoto, Japan.
| |
Collapse
|
14
|
Kashuba E, Bailey J, Allsup D, Cawkwell L. The kinin-kallikrein system: physiological roles, pathophysiology and its relationship to cancer biomarkers. Biomarkers 2013; 18:279-96. [PMID: 23672534 DOI: 10.3109/1354750x.2013.787544] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The kinin-kallikrein system (KKS) is an endogenous multiprotein cascade, the activation of which leads to triggering of the intrinsic coagulation pathway and enzymatic hydrolysis of kininogens with the consequent release of bradykinin-related peptides. This system plays a crucial role in inflammation, vasodilation, smooth muscle contraction, cardioprotection, vascular permeability, blood pressure control, coagulation and pain. In this review, we will outline the physiology and pathophysiology of the KKS and also highlight the association of this system with carcinogenesis and cancer progression.
Collapse
Affiliation(s)
- Elena Kashuba
- Postgraduate Medical Institute, University of Hull, Hull, UK
| | | | | | | |
Collapse
|
15
|
Maeda H, Nakamura H, Fang J. The EPR effect for macromolecular drug delivery to solid tumors: Improvement of tumor uptake, lowering of systemic toxicity, and distinct tumor imaging in vivo. Adv Drug Deliv Rev 2013. [DOI: '10.1016/j.addr.2012.10.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2023]
|
16
|
Maeda H, Nakamura H, Fang J. The EPR effect for macromolecular drug delivery to solid tumors: Improvement of tumor uptake, lowering of systemic toxicity, and distinct tumor imaging in vivo. Adv Drug Deliv Rev 2013; 65:71-9. [PMID: 23088862 DOI: 10.1016/j.addr.2012.10.002] [Citation(s) in RCA: 1710] [Impact Index Per Article: 142.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2012] [Revised: 10/11/2012] [Accepted: 10/16/2012] [Indexed: 02/06/2023]
Abstract
The EPR effect results from the extravasation of macromolecules or nanoparticles through tumor blood vessels. We here provide a historical review of the EPR effect, including its features, vascular mediators found in both cancer and inflamed tissue. In addition, architectural and physiological differences of tumor blood vessels vs that of normal tissue are commented. Furthermore, methods of augmentation of the EPR effect are described, that result in better tumor delivery and improved therapeutic effect, where nitroglycerin, angiotensin I-converting enzyme (ACE) inhibitor, or angiotensin II-induced hypertension are employed. Consequently, better therapeutic effect and reduced systemic toxicity are generally observed. Obviously, the EPR effect based delivery of nanoprobes are also useful for tumor-selective imaging agents with using fluorescent or radio nuclei in nanoprobes. We also commented a key difference between passive tumor targeting and the EPR effect in tumors, particularly as related to drug retention in tumors: passive targeting of low-molecular-weight X-ray contrast agents involves a retention period of less than a few minutes, whereas the EPR effect of nanoparticles involves a prolonged retention time-days to weeks.
Collapse
Affiliation(s)
- Hiroshi Maeda
- DDS Research Institute, Sojo University, Ikeda, Kumamoto, Japan.
| | | | | |
Collapse
|
17
|
Azzopardi EA, Ferguson EL, Thomas DW. The enhanced permeability retention effect: a new paradigm for drug targeting in infection. J Antimicrob Chemother 2012; 68:257-74. [PMID: 23054997 DOI: 10.1093/jac/dks379] [Citation(s) in RCA: 198] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Multidrug-resistant, Gram-negative infection is a major global determinant of morbidity, mortality and cost of care. The advent of nanomedicine has enabled tailored engineering of macromolecular constructs, permitting increasingly selective targeting, alteration of volume of distribution and activity/toxicity. Macromolecules tend to passively and preferentially accumulate at sites of enhanced vascular permeability and are then retained. This enhanced permeability and retention (EPR) effect, whilst recognized as a major breakthrough in anti-tumoral targeting, has not yet been fully exploited in infection. Shared pathophysiological pathways in both cancer and infection are evident and a number of novel nanomedicines have shown promise in selective, passive, size-mediated targeting to infection. This review describes the similarities and parallels in pathophysiological pathways at molecular, cellular and circulatory levels between inflammation/infection and cancer therapy, where use of this principle has been established.
Collapse
Affiliation(s)
- Ernest A Azzopardi
- School of Dentistry, Cardiff University, Heath Park, Cardiff CF14 4XY, Wales, UK.
| | | | | |
Collapse
|
18
|
Côté J, Bovenzi V, Savard M, Dubuc C, Fortier A, Neugebauer W, Tremblay L, Müller-Esterl W, Tsanaclis AM, Lepage M, Fortin D, Gobeil F. Induction of selective blood-tumor barrier permeability and macromolecular transport by a biostable kinin B1 receptor agonist in a glioma rat model. PLoS One 2012; 7:e37485. [PMID: 22629405 PMCID: PMC3357387 DOI: 10.1371/journal.pone.0037485] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2012] [Accepted: 04/24/2012] [Indexed: 12/24/2022] Open
Abstract
Treatment of malignant glioma with chemotherapy is limited mostly because of delivery impediment related to the blood-brain tumor barrier (BTB). B1 receptors (B1R), inducible prototypical G-protein coupled receptors (GPCR) can regulate permeability of vessels including possibly that of brain tumors. Here, we determine the extent of BTB permeability induced by the natural and synthetic peptide B1R agonists, LysdesArg9BK (LDBK) and SarLys[dPhe8]desArg9BK (NG29), in syngeneic F98 glioma-implanted Fischer rats. Ten days after tumor inoculation, we detected the presence of B1R on tumor cells and associated vasculature. NG29 infusion increased brain distribution volume and uptake profiles of paramagnetic probes (Magnevist and Gadomer) at tumoral sites (T1-weighted imaging). These effects were blocked by B1R antagonist and non-selective cyclooxygenase inhibitors, but not by B2R antagonist and non-selective nitric oxide synthase inhibitors. Consistent with MRI data, systemic co-administration of NG29 improved brain tumor delivery of Carboplatin chemotherapy (ICP-Mass spectrometry). We also detected elevated B1R expression in clinical samples of high-grade glioma. Our results documented a novel GPCR-signaling mechanism for promoting transient BTB disruption, involving activation of B1R and ensuing production of COX metabolites. They also underlined the potential value of synthetic biostable B1R agonists as selective BTB modulators for local delivery of different sized-therapeutics at (peri)tumoral sites.
Collapse
Affiliation(s)
- Jérôme Côté
- Department of Pharmacology, University Hospital, Frankfurt, Germany
- Department of Nuclear Medicine and Radiobiology, University Hospital, Frankfurt, Germany
- Institute of Pharmacology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Veronica Bovenzi
- Department of Pharmacology, University Hospital, Frankfurt, Germany
- Institute of Pharmacology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Martin Savard
- Department of Pharmacology, University Hospital, Frankfurt, Germany
- Institute of Pharmacology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Céléna Dubuc
- Department of Pharmacology, University Hospital, Frankfurt, Germany
- Institute of Pharmacology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Audrey Fortier
- Department of Pharmacology, University Hospital, Frankfurt, Germany
| | | | - Luc Tremblay
- Department of Nuclear Medicine and Radiobiology, University Hospital, Frankfurt, Germany
| | | | - Ana-Maria Tsanaclis
- Department of Pathology, Centre Hospitalier Universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Martin Lepage
- Department of Nuclear Medicine and Radiobiology, University Hospital, Frankfurt, Germany
- Institute of Pharmacology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - David Fortin
- Department of Surgery, Université de Sherbrooke, Sherbrooke, Quebec, Canada
- Institute of Pharmacology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Fernand Gobeil
- Department of Pharmacology, University Hospital, Frankfurt, Germany
- Institute of Pharmacology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Quebec, Canada
- * E-mail:
| |
Collapse
|
19
|
Guevara-Lora I. Kinin-mediated inflammation in neurodegenerative disorders. Neurochem Int 2012; 61:72-8. [PMID: 22554400 DOI: 10.1016/j.neuint.2012.04.013] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2012] [Revised: 04/01/2012] [Accepted: 04/10/2012] [Indexed: 01/06/2023]
Abstract
The mediatory role of kinins in both acute and chronic inflammation within nervous tissues has been widely described. Bradykinin, the major representative of these bioactive peptides, is one of a few mediators of inflammation that directly stimulates afferent nerves due to the broad expression of specific kinin receptors in cell types in these tissues. Moreover, kinins may be delivered to a site of injury not only after their production at the endothelium surface but also following their local production through the enzymatic degradation of kininogens at the surface of nerve cells. A strong correlation between inflammatory processes and neurodegeneration has been established. The activation of nerve cells, particularly microglia, in response to injury, trauma or infection initiates a number of reactions in the neuronal neighborhood that can lead to cell death after the prolonged action of inflammatory substances. In recent years, there has been a growing interest in the effects of kinins on neuronal destruction. In these studies, the overexpression of proteins involved in kinin generation or of kinin receptors has been observed in several neurologic disorders including neurodegenerative diseases such Alzheimer's disease and multiple sclerosis as well as disorders associated with a deficiency in cell communication such as epilepsy. This review is focused on recent findings that provide reliable evidence of the mediatory role of kinins in the inflammatory responses associated with different neurological disorders. A deeper understanding of the role of kinins in neurodegenerative diseases is likely to promote the future development of new therapeutic strategies for the control of these disorders. An example of this could be the prospective use of kinin receptor antagonists.
Collapse
Affiliation(s)
- Ibeth Guevara-Lora
- Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland.
| |
Collapse
|
20
|
Figueroa CD, Ehrenfeld P, Bhoola KD. Kinin receptors as targets for cancer therapy. Expert Opin Ther Targets 2012; 16:299-312. [DOI: 10.1517/14728222.2012.662957] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
21
|
Maeda H. Vascular permeability in cancer and infection as related to macromolecular drug delivery, with emphasis on the EPR effect for tumor-selective drug targeting. PROCEEDINGS OF THE JAPAN ACADEMY. SERIES B, PHYSICAL AND BIOLOGICAL SCIENCES 2012; 88:53-71. [PMID: 22450535 PMCID: PMC3365245 DOI: 10.2183/pjab.88.53] [Citation(s) in RCA: 172] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2011] [Accepted: 01/17/2012] [Indexed: 05/18/2023]
Abstract
Tumor and inflammation have many common features. One hallmark of both is enhanced vascular permeability, which is mediated by various factors including bradykinin, nitric oxide (NO), peroxynitrite, prostaglandins etc. A unique characteristic of tumors, however, is defective vascular anatomy. The enhanced vascular permeability in tumors is also distinctive in that extravasated macromolecules are not readily cleared. We utilized the enhanced permeability and retention (EPR) effect of tumors for tumor selective delivery of macromolecular drugs. Consequently, such drugs, nanoparticles or lipid particles, when injected intravenously, selectively accumulate in tumor tissues and remain there for long periods. The EPR effect of tumor tissue is frequently inhomogeneous and the heterogeneity of the EPR effect may reduce the tumor delivery of macromolecular drugs. Therefore, we developed methods to augment the EPR effect without inducing adverse effects for instance raising the systemic blood pressure by infusing angiotensin II during arterial injection of SMANCS/Lipiodol. This method was validated in clinical setting. Further, benefits of utilization of NO-releasing agent such as nitroglycerin or angiotensin-converting enzyme (ACE) inhibitors were demonstrated. The EPR effect is thus now widely accepted as the most basic mechanism for tumor-selective targeting of macromolecular drugs, or so-called nanomedicine.
Collapse
Affiliation(s)
- Hiroshi Maeda
- Institute of Drug Delivery System Research, School of Pharmaceutical Sciences, Sojo University, Kumamoto, Japan.
| |
Collapse
|
22
|
Greish K. Enhanced permeability and retention effect for selective targeting of anticancer nanomedicine: are we there yet? DRUG DISCOVERY TODAY. TECHNOLOGIES 2012; 9:e71-e174. [PMID: 24064277 DOI: 10.1016/j.ddtec.2011.11.010] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
|
23
|
Grainger SJ, Serna JV, Sunny S, Zhou Y, Deng CX, El-Sayed MEH. Pulsed ultrasound enhances nanoparticle penetration into breast cancer spheroids. Mol Pharm 2010; 7:2006-19. [PMID: 20957996 DOI: 10.1021/mp100280b] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Effective treatment of solid tumors requires homogeneous distribution of anticancer drugs within the entire tumor volume to deliver lethal concentrations to resistant cancer cells and tumor-initiating cancer stem cells. However, penetration of small molecular weight chemotherapeutic agents and drug-loaded polymeric and lipid particles into the hypoxic and necrotic regions of solid tumors remains a significant challenge. This article reports the results of pulsed ultrasound enhanced penetration of nanosized fluorescent particles into MCF-7 breast cancer spheroids (300-350 μm diameter) as a function of particle size and charge. With pulsed ultrasound application in the presence of microbubbles, small (20 nm) particles achieve 6-20-fold higher penetration and concentration in the spheroid's core compared to those not exposed to ultrasound. Increase in particle size to 40 and 100 nm results in their effective penetration into the spheroid's core to 9- and 3-fold, respectively. In addition, anionic carboxylate particles achieved higher penetration (2.3-, 3.7-, and 4.7-fold) into the core of MCF-7 breast cancer spheroids compared to neutral (2.2-, 1.9-, and 2.4-fold) and cationic particles (1.5-, 1.4-, and 1.9-fold) upon US exposure for 30, 60, and 90 s under the same experimental conditions. These results demonstrate the feasibility of utilizing pulsed ultrasound to increase the penetration of nanosized particles into MCF-7 spheroids mimicking tumor tissue. The effects of particle properties on the penetration enhancement were also illustrated.
Collapse
Affiliation(s)
- Stephanie J Grainger
- Department of Biomedical Engineering, University of Michigan, 1101 Beal Avenue, Ann Arbor, Michigan 48109, United States
| | | | | | | | | | | |
Collapse
|
24
|
Enhanced permeability and retention (EPR) effect for anticancer nanomedicine drug targeting. Methods Mol Biol 2010; 624:25-37. [PMID: 20217587 DOI: 10.1007/978-1-60761-609-2_3] [Citation(s) in RCA: 532] [Impact Index Per Article: 35.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Effective cancer therapy remains one of the most challenging tasks to the scientific community, with little advancement on overall cancer survival landscape during the last two decades. A major limitation inherent to most conventional anticancer chemotherapeutic agents is their lack of tumor selectivity. One way to achieve selective drug targeting to solid tumors is to exploit abnormalities of tumor vasculature, namely hypervascularization, aberrant vascular architecture, extensive production of vascular permeability factors stimulating extravasation within tumor tissues, and lack of lymphatic drainage. Due to their large size, nano-sized macromolecular anticancer drugs administered intravenously (i.v.) escape renal clearance. Being unable to penetrate through tight endothelial junctions of normal blood vessels, their concentration builds up in the plasma rendering them long plasma half-life. More importantly, they can selectively extravasate in tumor tissues due to its abnormal vascular nature. Overtime the tumor concentration will build up reaching several folds higher than that of the plasma due to lack of efficient lymphatic drainage in solid tumor, an ideal application for EPR-based selective anticancer nanotherapy. Indeed, this selective high local concentration of nano-sized anticancer drugs in tumor tissues has proven superior in therapeutic effect with minimal side effects in both preclinical and clinical settings.
Collapse
|
25
|
Segal E, Satchi-Fainaro R. Design and development of polymer conjugates as anti-angiogenic agents. Adv Drug Deliv Rev 2009; 61:1159-76. [PMID: 19699248 DOI: 10.1016/j.addr.2009.06.005] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2009] [Accepted: 06/12/2009] [Indexed: 12/17/2022]
Abstract
Angiogenesis, the formation of new blood vessels from pre-existing vasculature, is one of the central key steps in tumor progression and metastasis. Consequently, it became an important target in cancer therapy, making novel angiogenesis inhibitors a new modality of anticancer agents. Although relative to conventional chemotherapy, anti-angiogenic agents display a safer toxicity profile, the vast majority of these agents are low-molecular-weight compounds exhibiting poor pharmacokinetic profile with short half-life in the bloodstream and high overall clearance rate. The "Polymer Therapeutics" field has significantly improved the therapeutic potential of low-molecular-weight drugs and proteins for cancer treatment. Drugs can be conjugated to polymeric carriers that can be either directly conjugated to targeting proteins or peptides or derivatized with adapters conjugated to a targeting moiety. This approach holds a significant promise for the development of new targeted anti-angiogenic therapies as well as for the optimization of existing anti-angiogenic drugs or polypeptides. Here we overview the innovative approach of targeting tumor angiogenesis using polymer therapeutics.
Collapse
|
26
|
Molina L, Matus CE, Astroza A, Pavicic F, Tapia E, Toledo C, Perez JA, Nualart F, Gonzalez CB, Burgos RA, Figueroa CD, Ehrenfeld P, Poblete MT. Stimulation of the bradykinin B(1) receptor induces the proliferation of estrogen-sensitive breast cancer cells and activates the ERK1/2 signaling pathway. Breast Cancer Res Treat 2009; 118:499-510. [PMID: 19184415 DOI: 10.1007/s10549-009-0314-4] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2008] [Accepted: 01/08/2009] [Indexed: 12/11/2022]
Abstract
Kinin peptides exert multiple biological effects by binding to two types of G protein-coupled receptors known as B(1) (B(1)R) and B(2) receptors. Expression of the B(1)R in human breast cancer was recently reported, but up to now the consequences of its stimulation are unknown. Our aims were (1) to investigate the capacity of B(1)R to trigger cell proliferation in breast cancer cells, (2) to explore some of the downstream events occurring after B(1)R stimulation that may be linked to cell proliferation, and (3) to determine whether human breast tumors express potentially active B(1)R assessed by the binding of a radiolabeled agonist. Breast cancer cells expressed both the mRNA and the immunoreactive protein of B(1)R that once stimulated triggered cell proliferation at nanomolar concentrations of the ligand. Inhibitor studies suggested that the proliferative effects depend on the activity of epidermal growth factor receptor and subsequent ERK1/2 mitogen-activated protein kinases phosphorylation. B(1)R binding sites, were detected in 3/4 fibroadenomas, in 4/4 ductal carcinomas in situ and in 11/13 invasive ductal carcinomas. The B(1)R-epidermal growth factor receptor crosstalk may be a key interaction that maintains tumor growth, and antagonism of B(1)R may be a valuable alternative for the treatment of breast cancer.
Collapse
Affiliation(s)
- Luis Molina
- Laboratorio de Patologia Celular, Instituto de Anatomia, Histologia y Patologia, Universidad Austral de Chile, Valdivia, Chile
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Greish K. Enhanced permeability and retention of macromolecular drugs in solid tumors: a royal gate for targeted anticancer nanomedicines. J Drug Target 2007; 15:457-64. [PMID: 17671892 DOI: 10.1080/10611860701539584] [Citation(s) in RCA: 433] [Impact Index Per Article: 24.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Over the past two decades cancer has ascended the causes of human death to be number one or two in many nations world wide. A major limitation inherent to most conventional anticancer chemotherapeutic agents is their lack of tumor selectivity. One way to achieve selective drug targeting to solid tumors is to exploit abnormalities of tumor vasculature, namely, hypervascularisation; aberrant vascular architecture; extensive production of vascular permeability factors stimulating extravasation within tumor tissues; and lack of lymphatic drainage. Maeda and his colleagues have extensively studied tumor vascular abnormalities in terms of active and selective delivery of anticancer drugs to tumor tissues, notably defining the enhanced permeability and retention effect (EPR effect) of macromolecular drugs in solid tumors. Due to their large molecular size, nanosized macromolecular anticancer drugs administered intravenously (i.v.) escape renal clearance. Often they can not penetrate the tight endothelial junctions of normal blood vessels, but they can extravasate in tumour vasculature and become trapped in the tumor vicinity. With time the tumor concentration will build up reaching several folds higher than that of the plasma due to lack of efficient lymphatic drainage in solid tumor; an ideal application for EPR-based selective anticancer drug delivery. Establishing this principle hastened development of various polymer conjugates and polymeric micelles as well as multifunctional nanoparticles for targeted cancer chemotherapy. Indeed this selective high local concentration of nanosized anticancer drugs in tumor tissues has proven superior in therapeutic effect with minimal side effects in both preclinical and clinical settings. In this review the mechanisms and factors involved in the EPR effect, as well as the uniqueness of nanoscale drugs for tumor targeting through EPR effect, will be discussed in detail.
Collapse
Affiliation(s)
- Khaled Greish
- Laboratory of Microbiology and Oncology, Faculty of Pharmaceutical Sciences, Sojo University, Kumamoto, Japan.
| |
Collapse
|
28
|
Ballet S, De Wachter R, Van Rompaey K, Tömböly C, Feytens D, Töth G, Quartara L, Cucchi P, Meini S, Tourwé D. Bradykinin analogs containing the 4-amino-2-benzazepin-3-one scaffold at the C-terminus. J Pept Sci 2007; 13:164-70. [PMID: 17266049 DOI: 10.1002/psc.827] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
High affinity peptide ligands for the bradykinin (BK) B(2) subtype receptor have been shown to adopt a beta-turn conformation of the C-terminal tetrapeptide (H-Arg(1)-Pro(2)-Pro(3)-Gly(4)-Phe(5)-Ser(6)-Pro(7)-Phe(8)-Arg(9)-OH). We investigated the replacement of the Pro(7)-Phe(8) dipeptide moiety in BK or the D-Tic(7)-Oic(8) subunit in HOE140 (H-D-Arg(0)-Arg(1)-Pro(2)-Hyp(3)-Gly(4)-Thi(5)-Ser(6)-D-Tic(7)-Oic(8)-Arg(9)-OH) by 4-amino-1,2,4,5-tetrahydro-2-benzazepin-3-one templates (Aba). Binding studies to the human B(2) receptor showed a correlation between the affinities of the BK analogs and the propensity of the templates to adopt a beta-turn conformation. The L-spiro-Aba-Gly containing HOE140 analog BK10 has the best affinity, which correlates with the known turn-inducing property of this template. All the compounds did not modify basal inositolphosphate (IP) output in B(2)-expressing CHO cells up to 10 microM concentration. The antagonist properties were confirmed by the guinea pig ileum smooth muscle contractility assay. The new amino-benzazepinone (Aba) substituted BK analogs were found to be surmountable antagonists.
Collapse
Affiliation(s)
- S Ballet
- Department of Organic Chemistry, Vrije Universiteit Brussel, Pleinlaan 2, B-1050 Brussels, Belgium
| | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Zhang H, Gu YT, Xue YX. Bradykinin-induced blood-brain tumor barrier permeability increase is mediated by adenosine 5'-triphosphate-sensitive potassium channel. Brain Res 2007; 1144:33-41. [PMID: 17331483 DOI: 10.1016/j.brainres.2007.01.133] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2006] [Revised: 01/15/2007] [Accepted: 01/21/2007] [Indexed: 10/23/2022]
Abstract
Bradykinin has been shown to selectively transiently increase the permeability of the blood-brain barrier (BBB). This study was performed to determine whether ATP-sensitive potassium (K(ATP)) channels mediate the increase in permeability of brain tumor microvessels induced by BK. Using a rat brain glioma (C6) model, we found increased expression of K(ATP) channels at tumor sites via Western blot analysis, after intracarotid infusion of bradykinin at a dose of 10 microg/kg/min for 15 min. A significant increase (73.58%) of the integrated density value (IDV) of the K(ATP) channel Kir6.2 subunit was observed in rats with glioma after 10 min of bradykinin perfusion. The over-expression of K(ATP) channels with bradykinin was significantly attenuated by the K(ATP) channel antagonist glibenclamide. Immunohistochemistry and immunolocalization experiments showed that the over-expression of K(ATP) channels was more obvious near tumor capillaries of 10 microm in diameter. I(KATP) modulation by bradykinin in cultured C6 cells was also studied using the patch-clamp technique in a whole-cell configuration. Administration of bradykinin led to a significant opening of K(ATP) channels in a time-dependent manner. This led to the conclusion that the bradykinin-mediated BBB permeability increase is due to accelerated formation of K(ATP) channels, which are thus as an important target in the biochemical regulation of this process.
Collapse
Affiliation(s)
- Hua Zhang
- Department of Neurobiology, China Medical University, Shenyang, 110001, Liaoning Province, P.R. China
| | | | | |
Collapse
|
30
|
Iyer AK, Khaled G, Fang J, Maeda H. Exploiting the enhanced permeability and retention effect for tumor targeting. Drug Discov Today 2007; 11:812-8. [PMID: 16935749 DOI: 10.1016/j.drudis.2006.07.005] [Citation(s) in RCA: 1288] [Impact Index Per Article: 71.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2006] [Revised: 05/31/2006] [Accepted: 07/10/2006] [Indexed: 12/14/2022]
Abstract
Of the tumor targeting strategies, the enhanced permeability and retention (EPR) effect of macromolecules is a key mechanism for solid tumor targeting, and considered a gold standard for novel drug design. In this review, we discuss various endogenous factors that can positively impact the EPR effect in tumor tissues. Further, we discuss ways to augment the EPR effect by use of exogenous agents, as well as practical methods available in the clinical setting. Some innovative examples developed by researchers to combat cancer by the EPR mechanism are also discussed.
Collapse
Affiliation(s)
- Arun K Iyer
- Laboratory of Microbiology and Oncology, Faculty of Pharmaceutical Sciences, Sojo University, Ikeda 862-0082, Japan
| | | | | | | |
Collapse
|
31
|
Zorin NA, Zorina VN, Zorina RM. Role of proteins of the macroglobulin family in regulation of tumor growth. Russ J Dev Biol 2006. [DOI: 10.1134/s1062360406010024] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
32
|
Richards GP, Parveen S. A survey for phosphoglucose isomerase with lysyl aminopeptidase activity in Vibrionaceae and non-Vibrio pathogens. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2005; 1748:128-33. [PMID: 15752701 DOI: 10.1016/j.bbapap.2004.12.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2004] [Revised: 12/22/2004] [Accepted: 12/23/2004] [Indexed: 11/23/2022]
Abstract
Phosphoglucose isomerase (PGI) with a novel lysyl aminopeptidase (LysAP) activity was recently purified and characterized from Vibrio vulnificus. We showed that it cleaves the amino-terminal lysyl residue from des-Arg(10)-kallidin to produce des-Arg(9)-bradykinin, suggesting that it plays a role in virulence. A survey was conducted to determine the presence of this potential virulence-enhancing enzyme among twenty-three halotolerant human and fish pathogens from eleven species within the Vibrionaceae family, including V. vulnificus, V. parahaemolyticus, V. cholerae, Aeromonas hydrophila, and Plesiomonas shigelloides. In addition, fourteen species of non-Vibrionaceae pathogens were screened for LysAP activity. Cell lysates were partially purified by anion exchange chromatography and fractions were screened for LysAP and isomerase activities. PGI-LysAP activity was detected in chromatographic fractions from all the Vibrio species tested, but was not detected in any of the non-Vibrionaceae pathogens. Levels of isomerase and LysAP activity correlated (R(2)=0.92) for nine strains of V. vulnificus. Since the Vibrionaceae represent an important family of human and fish pathogens, our identification of PGI-LysAP activity in a broad array of vibrios may lead to the development of improved analytical methods for their identification as well as interventions to reduce the high morbidity and mortality associated with some Vibrionaceae infections in clinical, veterinary, and aquaculture settings.
Collapse
Affiliation(s)
- Gary P Richards
- United States Department of Agriculture, Agricultural Research Service, Microbial Food Safety Research Unit, Delaware State University, Dover, Delaware 19901, USA.
| | | |
Collapse
|
33
|
Richards GP, Hammer CH, Garfield MK, Parveen S. Characterization of a lysyl aminopeptidase activity associated with phosphoglucose isomerase of Vibrio vulnificus. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2004; 1700:219-29. [PMID: 15262231 DOI: 10.1016/j.bbapap.2004.05.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2004] [Revised: 05/07/2004] [Accepted: 05/14/2004] [Indexed: 11/29/2022]
Abstract
Phosphoglucose isomerase (PGI) is a multifunctional enzyme involved in glycolysis and gluconeogenesis and, in mammalian cells, functions as neuroleukin, autocrine motility factor (AMF), and differentiation and maturation factor (MF). We isolated and characterized PGI with a novel lysyl aminopeptidase (LysAP) activity (PGI-LysAP) from Vibrio vulnificus. Mass spectrometry revealed that PGI-LysAP is a heterodimer consisting of 23.4- and 60.8-kDa subunits. Only the heterodimer displayed LysAP activity. PGI-LysAP has a pI around 6.0 and high specificity toward the synthetic, fluorogenic substrate l-lysyl-7-amino-4-methylcoumarin. LysAP activity is optimal at pH 8.0, is 64% higher at 37 degrees C than at 21 degrees C, does not directly correlate with virulence, and is strongly inhibited by serine protease and metalloprotease inhibitors. PGI-LysAP was also identified in Vibrio parahaemolyticus and V. cholerae, but was absent from non-Vibrio human pathogens. Sequencing of the pgi gene revealed 1653 bp coding for a 550-amino-acid protein. Cloned and expressed PGI formed a homodimer with isomerase activity, but not LysAP activity. The finding of LysAP activity associated with heterodimeric PGI should foster a broad search for putative substrates in an effort to elucidate the role of PGI-LysAP in bacteria and its roles in the pathophysiology of diseases.
Collapse
Affiliation(s)
- Gary P Richards
- Agricultural Research Service, US Department of Agriculture, Delaware State University, James W.W. Baker Center, Dover, DE 19901, USA.
| | | | | | | |
Collapse
|
34
|
Hu HZ, Gao N, Liu S, Ren J, Xia Y, Wood JD. Metabotropic signal transduction for bradykinin in submucosal neurons of guinea pig small intestine. J Pharmacol Exp Ther 2004; 309:310-9. [PMID: 14718601 DOI: 10.1124/jpet.103.059204] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Intracellular recording methods with "sharp" microelectrodes were used to study signal transduction mechanisms underlying the excitatory action of bradykinin (BK) in morphologically identified neurons in the small intestinal submucosal plexus. Exposure to BK evoked slowly activating membrane depolarization and enhanced excitability associated with increased input resistance in AH-type and decreased input resistance in S-type neurons. Preincubation with pertussis toxin did not affect the BK-evoked responses. Pretreatment with the cyclooxygenase inhibitors indomethacin or piroxicam suppressed or abolished the BK-evoked responses. Application of prostaglandin (PG) E(2) or PG analogs evoked BK-like depolarizing responses in the submucosal plexus with a potency order of PGE(2) > PGE(1) > 17-phenyl trinor-PGE(2) > PGI(2) > sulprostone > PGF(2alpha). Depolarizing responses to bradykinin or PGE(2) in S-type neurons were suppressed in the presence of the phospholipase C inhibitor U73122 [(1-6-[([17beta]-3-methoxyestra-1,3,5[10]-tren-17-71)amino]hexyl)-1H-pyrrole-2,5-dione)], but not the inactive analog U73343 [(1-6-[([17beta]-3-methoxyestra-1,3,5[10]trien-17yl)amino]hexyl)-2,5-pyrrolidinedione)]. The inositol-1,4,5-trisphosphate receptor antagonist 2-aminoethoxy-diphenylborane and the calmodulin inhibitor W-7, but not ryanodine, suppressed both bradykinin- and PGE(2)-evoked responses. KN-62, an inhibitor of calmodulin kinases, or GF109203X, a specific protein kinase C inhibitor, suppressed both BK- and PGE(2)-evoked depolarizing responses. Selective protein kinase A inhibitors did not alter BK- or PGE(2)-evoked depolarizing responses in S neurons. The results suggest that BK stimulates synthesis and release of PGE(2), which acts at EP(1) receptors to evoke depolarizing responses in submucosal neurons. The postreceptor transduction cascade includes activation of phospholipase C, inositol-1,4,5-trisphosphate production, intraneuronal Ca2+ mobilization, activation of protein kinase C and/or calmodulin kinases, and phosphorylation of cationic channels.
Collapse
Affiliation(s)
- Hong-Zhen Hu
- Department of Physiology and Cell Biology, College of Medicine and Public Health, The Ohio State University, 304 Hamilton Hall, 1645 Neil Avenue, Columbus, OH 43210-1218, USA
| | | | | | | | | | | |
Collapse
|
35
|
Legat FJ, Maier A, Dittrich P, Zenahlik P, Kern T, Nuhsbaumer S, Frossard M, Salmhofer W, Kerl H, Müller M. Penetration of fosfomycin into inflammatory lesions in patients with cellulitis or diabetic foot syndrome. Antimicrob Agents Chemother 2003; 47:371-4. [PMID: 12499216 PMCID: PMC148983 DOI: 10.1128/aac.47.1.371-374.2003] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2002] [Revised: 07/26/2002] [Accepted: 10/23/2002] [Indexed: 11/20/2022] Open
Abstract
We investigated the distribution of the broad-spectrum antibiotic fosfomycin in infected soft tissue of patients with uncomplicated cellulitis of the lower extremities or diabetic foot infection using in vivo microdialysis. Our findings suggest that fosfomycin exhibits good and similar penetration into the fluid in the interstitial space in inflamed and noninflamed soft tissue in patients.
Collapse
Affiliation(s)
- F J Legat
- Department of Dermatology, Division of Thoracic and Hyperbaric Surgery, Karl-Franzens-University Graz Medical School, A-8036 Graz, Austria.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Abstract
Macromolecular drugs (also referred to as polymeric drugs) are a diverse group of drugs including polymer-conjugated drugs, polymeric micelles, liposomal drugs and solid phase depot formulations of various agents. In this review we will consider only water-soluble macromolecular drugs. In common, such drugs have high molecular weights, more than 40 kDa, which enables them to overcome renal excretion. Consequently, this group of drugs can attain prolonged plasma or local half-lives. The prolonged circulating time of these macromolecules enables them to utilise the vascular abnormalities of solid tumour tissues, a phenomenon called the enhanced permeability and retention (EPR) effect. The EPR effect facilitates extravasation of polymeric drugs more selectively at tumour tissues, and this selective targeting to solid tumour tissues may lead to superior therapeutic benefits with fewer systemic adverse effects. This contrasts with conventional low-molecular-weight drugs, where intratumour concentration diminishes rapidly in parallel with plasma concentration. The EPR effect is also operative in inflammatory tissues, which justifies the development and use of this class of drugs in infectious and inflammatory conditions. At the present time, several polymeric drugs have been approved by regulatory agencies. These include zinostatin stimalamer (copolymer styrene maleic acid-conjugated neocarzinostatin, or SMANCS) and polyethyleneglycol-conjugated interferon-alpha-2a. This article discusses these and other polymeric drugs in the setting of targeting to solid tumours.
Collapse
Affiliation(s)
- Khaled Greish
- Department of Microbiology, Kumamoto University School of Medicine, Kumamoto, Japan
| | | | | | | | | |
Collapse
|
37
|
Dziadulewicz EK, Ritchie TJ, Hallett A, Snell CR, Davies JW, Wrigglesworth R, Dunstan AR, Bloomfield GC, Drake GS, McIntyre P, Brown MC, Burgess GM, Lee W, Davis C, Yaqoob M, Phagoo SB, Phillips E, Perkins MN, Campbell EA, Davis AJ, Rang HP. Nonpeptide bradykinin B2 receptor antagonists: conversion of rodent-selective bradyzide analogues into potent, orally-active human bradykinin B2 receptor antagonists. J Med Chem 2002; 45:2160-72. [PMID: 12014954 DOI: 10.1021/jm0111088] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The 1-(2-nitrophenyl)thiosemicarbazide (TSC) derivative, (S)-1-[4-(4-benzhydrylthiosemicarbazido)-3-nitrobenzenesulfonyl]pyrrolidine-2-carboxylic acid [2-[(2-dimethylaminoethyl)methylamino]ethyl]amide (bradyzide; (S)-4), was recently disclosed as a novel, potent, orally active nonpeptide bradykinin (BK) B2 receptor antagonist. The compound inhibited the specific binding of [3H]BK to NG108-15 cell membrane preparations (rodent neuroblastoma-glioma) expressing B2 receptors with a K(i) of 0.5 +/- 0.2 nM. Compound (S)-4 also demonstrated oral efficacy against Freund's complete adjuvant (FCA)-induced mechanical hyperalgesia in rats with an ED50 value of 0.84 micromol/kg. After we optimized the terminal binding determinants projecting from the TSC framework, we found that it was possible to replace the potentially toxicophoric nitro and divalent sulfur moieties with only a 15-fold loss in binding affinity ((S)-14a). However, bradyzide and its congeners were found to have much lower affinities for cloned human B2 receptors, expressed in Cos-7 cells. The hitherto synthesized TSC series was screened against the human B2 receptor, and the dibenzosuberane (DBS) pharmacophore emerged as the key structural requirement for potency. Incorporation of this group resulted in a series of derivatives ((S)-14d,e and 19b-d) with K(i) ranges of 10.7-176 nM in NG108-15 cells (expressing the rodent B2 receptor) and 0.79-253 nM in Cos-7 cells (expressing the human B2 receptor). There was no evidence of agonist activity with any of the nonpeptides in any of the cell lines tested. In vivo, oral administration of compound 19c reversed FCA-induced and turpentine-induced mechanical hyperalgesia in rodents with ED50 values of 0.027 and 0.32 micromol/kg, respectively. The selectivity profiles of compounds (S)-14f and (S)-14g were also assessed to determine the conformational and/or steric preferences of the double-ring arrangement. The affinity of (S)-14 g for the human B2 receptor suggested that it may be a hydrophobic interaction with the ethane bridge of the DBS moiety that accounts for the increased potency of compounds (S)-14d,e and 19b,c at this receptor, by favoring a binding mode inaccessible to the unsubstituted diphenylmethyl derivative, (S)-4.
Collapse
|
38
|
Wu J, Akaike T, Hayashida K, Miyamoto Y, Nakagawa T, Miyakawa K, Müller-Esterl W, Maeda H. Identification of bradykinin receptors in clinical cancer specimens and murine tumor tissues. Int J Cancer 2002; 98:29-35. [PMID: 11857381 DOI: 10.1002/ijc.10142] [Citation(s) in RCA: 77] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Bradykinin (BK) has multiple pathophysiologic functions such as induction of vascular permeability and mitogenesis, and it triggers the release of other mediators such as nitric oxide in inflammatory and cancer tissues. To explore the pathophysiologic roles of BK in tumor, we examined the distribution of BK B2 receptors in human adenocarcinoma (lung, stomach), lymphoma (lymph node), hepatoma, squamous cell carcinoma (lung) and carcinoid (duodenum), and in mouse colon adenocarcinoma 38 (C-38) and sarcoma 180 (S-180) tumor tissues. Immunohistochemical staining of tumor tissues with an anti-BK B2 receptor antibody, or autoradiography with the B2 receptor antagonist [125I]HOE 140 (D-Arg-[Hyp Thi D-Tic Oic8]-BK) and the B2 receptor agonist [3H]BK indicated the presence of B2 receptors in all human tumor cells and murine S-180 and C-38 cells. Specific binding of [3H]HOE 140 was observed in S-180 cells with a Kd of 2.1 nM. Binding of [125I]HOE 140 to S-180 cells was competed by an excess amount (20-100 times) of nonradiolabeled HOE 140 or BK, but not by BK B1 receptor agonist des-Arg9-BK. These results provide direct evidence that the BK B2 receptor is expressed in human cancer and experimental murine tumors, which suggests a potential role for BK in inducing pathologic signal transduction in cancer growth and progression, nitric oxide production and vascular permeability enhancement in tumors. BK antagonists may thus have applications in the modulation of cancer growth and in paraneoplastic syndromes.
Collapse
Affiliation(s)
- Jun Wu
- Department of Microbiology, Kumamoto University School of Medicine, Kumamoto, Japan
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Stewart JM, Gera L, York EJ, Chan DC, Whalley EJ, Bunn PA, Vavrek RJ. Metabolism-resistant bradykinin antagonists: development and applications. Biol Chem 2001; 382:37-41. [PMID: 11258669 DOI: 10.1515/bc.2001.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Bradykinin plays many roles in normal and pathological physiology, but rapid enzymatic degradation made elucidation of its functions extremely difficult. Development of effective degradation-resistant antagonists made it possible to delineate these roles and to open the way for development of new drugs to control pathology due to excess production of bradykinin. Presently available peptide bradykinin antagonists are extremely potent, are completely resistant to enzymatic degradation, and are orally available. Non-peptide bradykinin antagonists have also been discovered. Development of bradykinin antagonists as drugs for cancer, inflammation and trauma is anticipated.
Collapse
Affiliation(s)
- J M Stewart
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Denver 80262, USA
| | | | | | | | | | | | | |
Collapse
|
40
|
Stewart JM, Gera L, York EJ, Chan DC, Bunn P. Bradykinin antagonists: present progress and future prospects. IMMUNOPHARMACOLOGY 1999; 43:155-61. [PMID: 10596848 DOI: 10.1016/s0162-3109(99)00102-2] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Bradykinin (BK) antagonist peptides have been powerful tools for delineating roles of kinins in both normal and pathological physiology and offer promise of drug development for a variety of inflammatory conditions and cancers. At the present time, potent peptide antagonists are available that are either specific for BK B1 or B2 receptors, or are effective on both receptor classes. Non-peptide BK B2 antagonists are now being announced and are under investigation in several companies. The best peptide B1-B2 peptide antagonist is stable against all kininases, is orally available, and has a very long lifetime in vivo. Certain dimers of this antagonist, as well as several smaller molecules, are active against several cancers, both in vitro and in vivo.
Collapse
Affiliation(s)
- J M Stewart
- Department of Biochemistry and Molecular Genetics, University of Colorado Medical School, Denver 80262, USA.
| | | | | | | | | |
Collapse
|
41
|
Anbar M. Clinical thermal imaging today. IEEE ENGINEERING IN MEDICINE AND BIOLOGY MAGAZINE : THE QUARTERLY MAGAZINE OF THE ENGINEERING IN MEDICINE & BIOLOGY SOCIETY 1998; 17:25-33. [PMID: 9672807 DOI: 10.1109/51.687960] [Citation(s) in RCA: 75] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- M Anbar
- School of Medicine and Biomedical Sciences, SUNY, Buffalo, USA.
| |
Collapse
|
42
|
Nitric oxide and oxygen radicals in infection, inflammation, and cancer. BIOCHEMISTRY. BIOKHIMIIA 1998. [PMID: 9721338 DOI: 10.1007/978-1-4615-5081-5_18] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
In recent years, accumulated evidence indicates that free radical species and nitric oxide (NO) or its derivatives are the key denominators in carcinogenesis. Our present topics discussed in this article will focus on the biological significance of free radical generation induced by viral and bacterial infections. In influenza virus infection in mice, the level of xanthine oxidase (XO) at the infected sites was elevated to a great extent. The timing of paralleled induction of XO with that of inducible NO synthase (iNOS) indicates efficient simultaneous reaction: NO + O2*- --> ONOO- (peroxynitrite). Peroxynitrite formation was identified by immunostaining of nitrotyrosine at the local site of infected organs. Peroxynitrite exhibits unique chemical reactivities such as protein nitration, DNA-strand breakage, guanine nitration, etc., which may then bring about not only cytotoxic effect but also mutagenesis. Numbers of evidence in vitro and in vivo show that treatment with chemical carcinogens such as carbon tetrachloride and heterocyclic amines also generated superoxide. The chronic inflammatory reactions, e.g., zymosan- and silica-induced granuloma, revealed very similar free radical generation in vivo. In addition, most experimental solid tumors have elevated levels of iNOS in the tumor tissue, and NO thus generated facilitates vascular permeability, which accelerates nutritional supply to the tumor tissue and hence sustains the rapid tumor growth. These circumstantial evidences suggest that inflammatory responses induced by various pathogens would accelerate mutagenesis as well as tissue damage, whereas NO also sustains more effectively solid tumor growth when normal cells are transformed to tumor or carcinoma cells by the host-derived free radical species.
Collapse
|
43
|
Stewart JM, Gera L, Chan DC, Whalley ET, Hanson WL, Zuzack JS. Potent, long-acting, orally-active bradykinin antagonists for a wide range of applications. IMMUNOPHARMACOLOGY 1997; 36:167-72. [PMID: 9228542 DOI: 10.1016/s0162-3109(97)00017-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Actions of bradykinin (Arg-Pro-Pro-Gly-Phe-Ser-Pro-Phe-Arg; BK) are mediated by constitutively expressed B2 receptors, that require the full BK peptide chain, and by B1 receptors, induced in inflammation, that use BK(1-8) as ligand. In addition to many physiological and pathophysiological functions, the growth factor activity of BK evidently allows it to act as an autocrine stimulant for small cell lung cancer. A new group of BK antagonists containing the novel amino acid a-(2-indanyl)glycine provides extremely potent broad-spectrum as well as selective antagonists for all these functions.
Collapse
Affiliation(s)
- J M Stewart
- Department of Biochemistry, University of Colorado Medical School, Denver 80262, USA.
| | | | | | | | | | | |
Collapse
|
44
|
Stewart JM, Gera L, Chan DC, Whalley ET, Hanson WL, Zuzack JS. Potent, long-acting bradykinin antagonists for a wide range of applications. Can J Physiol Pharmacol 1997. [DOI: 10.1139/y97-084] [Citation(s) in RCA: 45] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
45
|
Affiliation(s)
- H Maeda
- Department of Microbiology, Kumamoto University School of Medicine, Japan
| |
Collapse
|