1
|
Leusmann S, Ménová P, Shanin E, Titz A, Rademacher C. Glycomimetics for the inhibition and modulation of lectins. Chem Soc Rev 2023; 52:3663-3740. [PMID: 37232696 PMCID: PMC10243309 DOI: 10.1039/d2cs00954d] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Indexed: 05/27/2023]
Abstract
Carbohydrates are essential mediators of many processes in health and disease. They regulate self-/non-self- discrimination, are key elements of cellular communication, cancer, infection and inflammation, and determine protein folding, function and life-times. Moreover, they are integral to the cellular envelope for microorganisms and participate in biofilm formation. These diverse functions of carbohydrates are mediated by carbohydrate-binding proteins, lectins, and the more the knowledge about the biology of these proteins is advancing, the more interfering with carbohydrate recognition becomes a viable option for the development of novel therapeutics. In this respect, small molecules mimicking this recognition process become more and more available either as tools for fostering our basic understanding of glycobiology or as therapeutics. In this review, we outline the general design principles of glycomimetic inhibitors (Section 2). This section is then followed by highlighting three approaches to interfere with lectin function, i.e. with carbohydrate-derived glycomimetics (Section 3.1), novel glycomimetic scaffolds (Section 3.2) and allosteric modulators (Section 3.3). We summarize recent advances in design and application of glycomimetics for various classes of lectins of mammalian, viral and bacterial origin. Besides highlighting design principles in general, we showcase defined cases in which glycomimetics have been advanced to clinical trials or marketed. Additionally, emerging applications of glycomimetics for targeted protein degradation and targeted delivery purposes are reviewed in Section 4.
Collapse
Affiliation(s)
- Steffen Leusmann
- Chemical Biology of Carbohydrates (CBCH), Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research, 66123 Saarbrücken, Germany.
- Department of Chemistry, Saarland University, 66123 Saarbrücken, Germany
- Deutsches Zentrum für Infektionsforschung (DZIF), Standort Hannover-Braunschweig, Germany
| | - Petra Ménová
- University of Chemistry and Technology, Prague, Technická 5, 16628 Prague 6, Czech Republic
| | - Elena Shanin
- Department of Pharmaceutical Sciences, University of Vienna, Josef-Holaubek-Platz 2, 1090 Vienna, Austria.
- Department of Microbiology, Immunobiology and Genetics, Max F. Perutz Laboratories, University of Vienna, Biocenter 5, 1030 Vienna, Austria
| | - Alexander Titz
- Chemical Biology of Carbohydrates (CBCH), Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research, 66123 Saarbrücken, Germany.
- Department of Chemistry, Saarland University, 66123 Saarbrücken, Germany
- Deutsches Zentrum für Infektionsforschung (DZIF), Standort Hannover-Braunschweig, Germany
| | - Christoph Rademacher
- Department of Pharmaceutical Sciences, University of Vienna, Josef-Holaubek-Platz 2, 1090 Vienna, Austria.
- Department of Microbiology, Immunobiology and Genetics, Max F. Perutz Laboratories, University of Vienna, Biocenter 5, 1030 Vienna, Austria
| |
Collapse
|
2
|
Laderach DJ, Compagno D. Inhibition of galectins in cancer: Biological challenges for their clinical application. Front Immunol 2023; 13:1104625. [PMID: 36703969 PMCID: PMC9872792 DOI: 10.3389/fimmu.2022.1104625] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 12/16/2022] [Indexed: 01/11/2023] Open
Abstract
Galectins play relevant roles in tumor development, progression and metastasis. Accordingly, galectins are certainly enticing targets for medical intervention in cancer. To date, however, clinical trials based on galectin inhibitors reported inconclusive results. This review summarizes the galectin inhibitors currently being evaluated and discusses some of the biological challenges that need to be addressed to improve these strategies for the benefit of cancer patients.
Collapse
Affiliation(s)
- Diego José Laderach
- Molecular and Functional Glyco-Oncology Laboratory, Instituto de Química Biológica de la Facutad de Ciencias Exactas y Naturales (IQUIBICEN-CONICET), Buenos Aires, Argentina,Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina,Departamento de Ciencias Básicas, Universidad Nacional de Luján, Luján, Argentina,*Correspondence: Diego José Laderach,
| | - Daniel Compagno
- Molecular and Functional Glyco-Oncology Laboratory, Instituto de Química Biológica de la Facutad de Ciencias Exactas y Naturales (IQUIBICEN-CONICET), Buenos Aires, Argentina,Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
3
|
Unraveling How Tumor-Derived Galectins Contribute to Anti-Cancer Immunity Failure. Cancers (Basel) 2021; 13:cancers13184529. [PMID: 34572756 PMCID: PMC8469970 DOI: 10.3390/cancers13184529] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Revised: 08/16/2021] [Accepted: 08/24/2021] [Indexed: 12/15/2022] Open
Abstract
Simple Summary This review compiles our current knowledge of one of the main pathways activated by tumors to escape immune attack. Indeed, it integrates the current understanding of how tumor-derived circulating galectins affect the elicitation of effective anti-tumor immunity. It focuses on several relevant topics: which are the main galectins produced by tumors, how soluble galectins circulate throughout biological liquids (taking a body-settled gradient concentration into account), the conditions required for the galectins’ functions to be accomplished at the tumor and tumor-distant sites, and how the physicochemical properties of the microenvironment in each tissue determine their functions. These are no mere semantic definitions as they define which functions can be performed in said tissues instead. Finally, we discuss the promising future of galectins as targets in cancer immunotherapy and some outstanding questions in the field. Abstract Current data indicates that anti-tumor T cell-mediated immunity correlates with a better prognosis in cancer patients. However, it has widely been demonstrated that tumor cells negatively manage immune attack by activating several immune-suppressive mechanisms. It is, therefore, essential to fully understand how lymphocytes are activated in a tumor microenvironment and, above all, how to prevent these cells from becoming dysfunctional. Tumors produce galectins-1, -3, -7, -8, and -9 as one of the major molecular mechanisms to evade immune control of tumor development. These galectins impact different steps in the establishment of the anti-tumor immune responses. Here, we carry out a critical dissection on the mechanisms through which tumor-derived galectins can influence the production and the functionality of anti-tumor T lymphocytes. This knowledge may help us design more effective immunotherapies to treat human cancers.
Collapse
|
4
|
Dings RPM, Kumar N, Mikkelson S, Gabius HJ, Mayo KH. Simulating cellular galectin networks by mixing galectins in vitro reveals synergistic activity. Biochem Biophys Rep 2021; 28:101116. [PMID: 34485713 PMCID: PMC8408429 DOI: 10.1016/j.bbrep.2021.101116] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 08/09/2021] [Accepted: 08/23/2021] [Indexed: 12/31/2022] Open
Abstract
Background Even though members of the family of adhesion/growth-regulatory galectins are increasingly detected to be co-expressed, they are still being routinely tested separately. The recent discovery of heterodimer formation among galectins-1, -3, and -7 in mixtures prompts further study of their functional activities in mixtures. Methods Cell agglutination, galectin binding to cells, as well as effects on cell proliferation, onset of apoptosis and migration were determined in assays using various cell types and mixtures of galectins-1, -3, and -7. Results Evidence for a more than additive increases of experimental parameters was consistently obtained. Conclusion Testing galectins in mixtures simulates the situation of co-expression in situ and reveals unsuspected over-additive activities. This new insight is relevant for analyzing galectin functionality in (patho)physiological conditions. Galectins-1, -3, and -7 form heterodimers in solution. Mixtures of galectins simulates galectin co-expression in situ. Mixtures display synergistic activities in vitro. Cell agglutination, apoptosis, proliferation, migration affected. Findings are relevant for galectin functionality in vivo.
Collapse
Affiliation(s)
- Ruud P M Dings
- Department of Biochemistry, Molecular Biology & Biophysics, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Nigam Kumar
- Department of Biochemistry, Molecular Biology & Biophysics, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Sterling Mikkelson
- Department of Biochemistry, Molecular Biology & Biophysics, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Hans-Joachim Gabius
- Institute of Physiological Chemistry, Faculty of Veterinary Medicine, Ludwig-Maximilians-University, Veterinarstr. 13, Munich, 80539, Germany
| | - Kevin H Mayo
- Department of Biochemistry, Molecular Biology & Biophysics, University of Minnesota, Minneapolis, MN, 55455, USA
| |
Collapse
|
5
|
Jenkins SV, Alimohammadi M, Terry AS, Griffin RJ, Tackett AJ, Leung JW, Vang KB, Byrum SD, Dings RP. Dysbiotic stress increases the sensitivity of the tumor vasculature to radiotherapy and c-Met inhibitors. Angiogenesis 2021; 24:597-611. [PMID: 33629198 PMCID: PMC8295215 DOI: 10.1007/s10456-021-09771-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 01/22/2021] [Accepted: 02/03/2021] [Indexed: 10/22/2022]
Abstract
Antibiotic-induced microbial imbalance, or dysbiosis, has systemic and long-lasting effects on the host and response to cancer therapies. However, the effects on tumor endothelial cells are largely unknown. Therefore, the goal of the current study was to generate matched B16-F10 melanoma associated endothelial cell lines isolated from mice with and without antibiotic-induced dysbiosis. After validating endothelial cell markers on a genomic and proteomic level, functional angiogenesis assays (i.e., migration and tube formation) also confirmed their vasculature origin. Subsequently, we found that tumor endothelial cells derived from dysbiotic mice (TEC-Dys) were more sensitive to ionizing radiotherapy in the range of clinically-relevant hypofractionated doses, as compared to tumor endothelial cells derived from orthobiotic mice (TEC-Ortho). In order to identify tumor vasculature-associated drug targets during dysbiosis, we used tandem mass tag mass spectroscopy and focused on the statistically significant cellular membrane proteins overexpressed in TEC-Dys. By these criteria c-Met was the most differentially expressed protein, which was validated histologically by comparing tumors with or without dysbiosis. Moreover, in vitro, c-Met inhibitors Foretinib, Crizotinib and Cabozantinib were significantly more effective against TEC-Dys than TEC-Ortho. In vivo, Foretinib inhibited tumor growth to a greater extent during dysbiosis as compared to orthobiotic conditions. Thus, we surmise that tumor response in dysbiotic patients may be greatly improved by targeting dysbiosis-induced pathways, such as c-Met, distinct from the many targets suppressed due to dysbiosis.
Collapse
Affiliation(s)
- Samir V. Jenkins
- Department of Radiation Oncology, University of Arkansas for Medical Sciences, Little Rock, AR USA
| | - Mohammad Alimohammadi
- Department of Radiation Oncology, University of Arkansas for Medical Sciences, Little Rock, AR USA
| | - Alexia S. Terry
- Department of Radiation Oncology, University of Arkansas for Medical Sciences, Little Rock, AR USA
| | - Robert J. Griffin
- Department of Radiation Oncology, University of Arkansas for Medical Sciences, Little Rock, AR USA
| | - Alan J. Tackett
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR USA,Arkansas Children's Research Institute, 13 Children's Way, Little Rock, AR 72202, USA
| | - Justin W. Leung
- Department of Radiation Oncology, University of Arkansas for Medical Sciences, Little Rock, AR USA
| | - Kieng B. Vang
- Center for Integrative Nanotechnology Sciences, University of Arkansas at Little Rock, Arkansas, Little Rock, AR USA
| | - Stephanie D. Byrum
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR USA,Arkansas Children's Research Institute, 13 Children's Way, Little Rock, AR 72202, USA
| | - Ruud P.M. Dings
- Department of Radiation Oncology, University of Arkansas for Medical Sciences, Little Rock, AR USA,Corresponding Author: Ruud P.M. Dings, 4301 W. Markham Street, Mail Slot #771, Little Rock, AR 72205. ; Phone: +1 501 526 7876
| |
Collapse
|
6
|
Miller MC, Zheng Y, Suylen D, Ippel H, Cañada FJ, Berbís MA, Jiménez-Barbero J, Tai G, Gabius HJ, Mayo KH. Targeting the CRD F-face of Human Galectin-3 and Allosterically Modulating Glycan Binding by Angiostatic PTX008 and a Structurally Optimized Derivative. ChemMedChem 2020; 16:713-723. [PMID: 33156953 DOI: 10.1002/cmdc.202000742] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Revised: 10/31/2020] [Indexed: 12/25/2022]
Abstract
Calix[4]arene PTX008 is an angiostatic agent that inhibits tumor growth in mice by binding to galectin-1, a β-galactoside-binding lectin. To assess the affinity profile of PTX008 for galectins, we used 15 N,1 H HSQC NMR spectroscopy to show that PTX008 also binds to galectin-3 (Gal-3), albeit more weakly. We identified the contact site for PTX008 on the F-face of the Gal-3 carbohydrate recognition domain. STD NMR revealed that the hydrophobic phenyl ring crown of the calixarene is the binding epitope. With this information, we performed molecular modeling of the complex to assist in improving the rather low affinity of PTX008 for Gal-3. By removing the N-dimethyl alkyl chain amide groups, we produced PTX013 whose reduced alkyl chain length and polar character led to an approximately eightfold stronger binding than PTX008. PTX013 also binds Gal-1 more strongly than PTX008, whereas neither interacts strongly, if at all, with Gal-7. In addition, PTX013, like PTX008, is an allosteric inhibitor of galectin binding to the canonical ligand lactose. This study broadens the scope for galectin targeting by calixarene-based compounds and opens the perspective for selective galectin blocking.
Collapse
Affiliation(s)
- Michelle C Miller
- Department of Biochemistry, Molecular Biology & Biophysics, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Yi Zheng
- School of Life Science, Northeast Normal University, 130024, Changchun, People's Republic of China
| | - Dennis Suylen
- Department of Biochemistry and CARIM, Maastricht University, 6229HX, Maastricht, The Netherlands
| | - Hans Ippel
- Department of Biochemistry and CARIM, Maastricht University, 6229HX, Maastricht, The Netherlands
| | - F Javier Cañada
- NMR and Molecular Recognition Group, Centro de Investigaciones Biológicas Margarita Salas (CSIC), C/Ramiro de Maeztu 9, 28040, Madrid, Spain
| | - M Alvaro Berbís
- NMR and Molecular Recognition Group, Centro de Investigaciones Biológicas Margarita Salas (CSIC), C/Ramiro de Maeztu 9, 28040, Madrid, Spain
| | - Jesús Jiménez-Barbero
- NMR and Molecular Recognition Group, Centro de Investigaciones Biológicas Margarita Salas (CSIC), C/Ramiro de Maeztu 9, 28040, Madrid, Spain.,CIC bioGUNE, Bizkaia Technological Park, Building 801 A, 48160, Derio, Spain.,Ikerbasque, Basque Foundation for Science, 28009, Bilbao, Spain
| | - Guihua Tai
- School of Life Science, Northeast Normal University, 130024, Changchun, People's Republic of China
| | - Hans-Joachim Gabius
- Institute of Physiological Chemistry, Faculty of Veterinary Medicine, Ludwig-Maximillians-University, 80539, Munich, Germany
| | - Kevin H Mayo
- Department of Biochemistry, Molecular Biology & Biophysics, University of Minnesota, Minneapolis, MN, 55455, USA
| |
Collapse
|
7
|
Design, synthesis and evaluation of calix[4]arene-based carbonyl amide derivatives with antitumor activities. Eur J Med Chem 2020; 210:112984. [PMID: 33183867 DOI: 10.1016/j.ejmech.2020.112984] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Revised: 10/18/2020] [Accepted: 10/30/2020] [Indexed: 11/23/2022]
Abstract
Calixarenes, with potential functionalization on the upper and lower rim, have been explored in recent years for the design and construction of anticancer agents in the field of drugs and pharmaceuticals. Herein, optimization of bis [N-(2-hydroxyethyl) aminocarbonylmethoxyl substituted calix [4] arene (CLX-4) using structure-based drug design and traditional medicinal chemistry led to the discovery of series of calix [4]arene carbonyl amide derivatives 5a-5t. Evaluation of the cytotoxicity of 5a-5t employing MTT assay in MCF-7, MDA-MB-231 (human breast cancer cells), HT29 (human colon carcinoma cells), HepG2 (human hepatocellular carcinoma cells), A549 (human lung adenocarcinoma cells) and HUVEC (Human Umbilical Vein Endothelial) cells demonstrated that the most promising compound 5h displayed the most superior inhibitory effect against A549 and MDA-MB-231 cells, which were 3.2 times and 6.8 times of CLX-4, respectively. In addition, the cell inhibition rate (at 10 μM) against normal HUVEC cells in vitro was only 9.6%, indicating the safty of compound 5h. Moreover, compound 5h could inhibit the migration of MDA-MB-231 cell in wound healing assay. Further mechanism studies significantly indicated that compound 5h could block MDA-MB-231 cell cycle arrest in G0/G1 phase by down regulating cyclin D1 and CDK4, and induce apoptosis by up-regulation of Bax, down-regulation of Caspase-3, PARP and Bcl-2 proteins, resulting in the reduction of DNA synthesis and cell division arrest. This work provides worthy of further exploration for the promising calixarene-based anticancer drugs.
Collapse
|
8
|
Bertuzzi S, Quintana JI, Ardá A, Gimeno A, Jiménez-Barbero J. Targeting Galectins With Glycomimetics. Front Chem 2020; 8:593. [PMID: 32850631 PMCID: PMC7426508 DOI: 10.3389/fchem.2020.00593] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 06/08/2020] [Indexed: 01/06/2023] Open
Abstract
Among glycan-binding proteins, galectins, β-galactoside-binding lectins, exhibit relevant biological roles and are implicated in many diseases, such as cancer and inflammation. Their involvement in crucial pathologies makes them interesting targets for drug discovery. In this review, we gather the last approaches toward the specific design of glycomimetics as potential drugs against galectins. Different approaches, either using specific glycomimetic molecules decorated with key functional groups or employing multivalent presentations of lactose and N-acetyl lactosamine analogs, have provided promising results for binding and modulating different galectins. The review highlights the results obtained with these approximations, from the employment of S-glycosyl compounds to peptidomimetics and multivalent glycopolymers, mostly employed to recognize and/or detect hGal-1 and hGal-3.
Collapse
Affiliation(s)
- Sara Bertuzzi
- CIC bioGUNE, Basque Research Technology Alliance, Derio, Spain
| | - Jon I Quintana
- CIC bioGUNE, Basque Research Technology Alliance, Derio, Spain
| | - Ana Ardá
- CIC bioGUNE, Basque Research Technology Alliance, Derio, Spain
| | - Ana Gimeno
- CIC bioGUNE, Basque Research Technology Alliance, Derio, Spain
| | - Jesús Jiménez-Barbero
- CIC bioGUNE, Basque Research Technology Alliance, Derio, Spain.,Ikerbasque, Basque Foundation for Science, Bilbao, Spain.,Department of Organic Chemistry II, Faculty of Science and Technology, University of the Basque Country - UPV-EHU, Leioa, Spain
| |
Collapse
|
9
|
Goud NS, Soukya PSL, Ghouse M, Komal D, Alvala R, Alvala M. Human Galectin-1 and Its Inhibitors: Privileged Target for Cancer and HIV. Mini Rev Med Chem 2019; 19:1369-1378. [PMID: 30834831 DOI: 10.2174/1389557519666190304120821] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 11/07/2018] [Accepted: 02/22/2019] [Indexed: 01/18/2023]
Abstract
Galectin 1(Gal-1), a β-galactoside binding mammalian lectin of 14KDa, is implicated in many signalling pathways, immune responses associated with cancer progression and immune disorders. Inhibition of human Gal-1 has been regarded as one of the potential therapeutic approaches for the treatment of cancer, as it plays a major role in tumour development and metastasis by modulating various biological functions viz. apoptosis, angiogenesis, migration, cell immune escape. Gal-1 is considered as a biomarker in diagnosis, prognosis and treatment condition. The overexpression of Gal-1 is well established and seen in many types of cancer progression like osteosarcoma, breast, lung, prostate, melanoma, etc. Gal-1 greatly accelerates the binding kinetics of HIV-1 to susceptible cells, leading to faster viral entry and a more robust viral replication by specific binding of CD4 cells. Hence, the Gal-1 is considered a promising molecular target for the development of new therapeutic drugs for cancer and HIV. The present review laid emphasis on structural insights and functional role of Gal-1 in the disease, current Gal-1 inhibitors and future prospects in the design of specific Gal-1 inhibitors.
Collapse
Affiliation(s)
- Narella Sridhar Goud
- Department of Medicinal Chemistry, National Institute of pharmaceutical Education and Research (NIPER) - Hyderabad, Balanagar, 500037, India
| | - P S Lakshmi Soukya
- Department of Medicinal Chemistry, National Institute of pharmaceutical Education and Research (NIPER) - Hyderabad, Balanagar, 500037, India
| | - Mahammad Ghouse
- Department of Medicinal Chemistry, National Institute of pharmaceutical Education and Research (NIPER) - Hyderabad, Balanagar, 500037, India
| | - Daipule Komal
- Department of Medicinal Chemistry, National Institute of pharmaceutical Education and Research (NIPER) - Hyderabad, Balanagar, 500037, India
| | - Ravi Alvala
- G. Pulla Reddy College of pharmacy, Hyderabad, 500028, India
| | - Mallika Alvala
- Department of Medicinal Chemistry, National Institute of pharmaceutical Education and Research (NIPER) - Hyderabad, Balanagar, 500037, India
| |
Collapse
|
10
|
Dasgupta A, Acharya K. Mushrooms: an emerging resource for therapeutic terpenoids. 3 Biotech 2019; 9:369. [PMID: 31588393 DOI: 10.1007/s13205-019-1906-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 09/19/2019] [Indexed: 12/22/2022] Open
Abstract
Mankind has always been fascinated with nature and have heavily explored natural products since the ancient times. Evolution of diseases led to research on synthetic structure, specificity and activity-guided treatment. To combat threats of new developing diseases and the deleterious side effects posed by modern therapy, researchers have once again looked back towards natural resources. Although plants have been the main source of natural drugs, lower fungi are being recently paid attention to. Among them, mushrooms have emerged as an under-explored yet immensely rich resource, especially for bioactive terpenoids. A lot of research is going on around the world with mushroom-derived terpenoids especially their medicinal properties, some of which have even been used in pre- and post-clinical studies. From the literatures that are available, it was found that mushroom terpenoids have activity against a wide range of diseases. In this review, we have summarized different mushroom-derived terpenoids and their therapeutic properties.
Collapse
Affiliation(s)
- Adhiraj Dasgupta
- Molecular and Applied Mycology and Plant Pathology Laboratory, Department of Botany, University of Calcutta, 35, Ballygunge Circular Road, Kolkata, West Bengal 700019 India
| | - Krishnendu Acharya
- Molecular and Applied Mycology and Plant Pathology Laboratory, Department of Botany, University of Calcutta, 35, Ballygunge Circular Road, Kolkata, West Bengal 700019 India
| |
Collapse
|
11
|
Kurrikoff K, Aphkhazava D, Langel Ü. The future of peptides in cancer treatment. Curr Opin Pharmacol 2019; 47:27-32. [DOI: 10.1016/j.coph.2019.01.008] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 01/25/2019] [Accepted: 01/31/2019] [Indexed: 12/25/2022]
|
12
|
Zhao K, Yang S, Geng J, Gong X, Gong W, Shen L, Ning B. Combination of anginex gene therapy and radiation decelerates the growth and pulmonary metastasis of human osteosarcoma xenografts. Cancer Med 2018; 7:2518-2529. [PMID: 29659181 PMCID: PMC6010866 DOI: 10.1002/cam4.1476] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 03/08/2018] [Accepted: 03/09/2018] [Indexed: 01/23/2023] Open
Abstract
Investigate whether rAAV-anginex gene therapy combined with radiotherapy could decrease growth and pulmonary metastasis of osteosarcoma in mice and examine the mechanisms involved in this therapeutic strategy. During in vitro experiment, multiple treatment regimes (rAAV-eGFP, radiotherapy, rAAV-anginex, combination therapy) were applied to determine effects on proliferation of endothelial cells (ECs) and G-292 osteosarcoma cells. During in vivo analysis, the same multiple treatment regimes were applied to osteosarcoma tumor-bearing mice. Use microcomputed tomography to evaluate tumor size. Eight weeks after tumor cell inoculation, immunohistochemistry was used to assess the therapeutic efficacy according to microvessel density (MVD), proliferating cell nuclear antigen (PCNA), and terminal-deoxynucleotidyl transferase-mediated nick-end labeling (TUNEL) assays. Metastasis of lungs was also evaluated by measuring number of metastatic nodules and wet weight of metastases. The proliferation of ECs and the tumor volumes in combination therapy group were inhibited more effectively than the other three groups at end point (P < 0.05). Cell clone assay showed anginex had radiosensitization effect on ECs. Immunohistochemistry showed tumors from mice treated with combination therapy exhibited the lowest MVD and proliferation rate, with highest apoptosis rate, as confirmed by IHC staining for CD34 and PCNA and TUNEL assays (P < 0.05). Combination therapy also induced the fewest metastatic nodules and lowest wet weights of the lungs (P < 0.05). rAAV-anginex combined with radiotherapy induced apoptosis of osteosarcoma cells and inhibited tumor growth and pulmonary metastasis on the experimental osteosarcoma models. We conclude that the primary mechanism of this process may be due to sensitizing effect of anginex to radiotherapy.
Collapse
Affiliation(s)
- Kai Zhao
- Jinan Central Hospital Affiliated to Shandong UniversityNo. 105, Jiefang RoadJinan250013ShandongChina
| | - Shang‐You Yang
- Department of Surgery, OrthopedicsUniversity of Kansas School of Medicine‐WichitaWichita67214Kansas
| | - Jun Geng
- Jinan Central Hospital Affiliated to Shandong UniversityNo. 105, Jiefang RoadJinan250013ShandongChina
| | - Xuan Gong
- Department of Surgery, OrthopedicsUniversity of Kansas School of Medicine‐WichitaWichita67214Kansas
| | - Weiming Gong
- Jinan Central Hospital Affiliated to Shandong UniversityNo. 105, Jiefang RoadJinan250013ShandongChina
| | - Lin Shen
- Jinan Central Hospital Affiliated to Shandong UniversityNo. 105, Jiefang RoadJinan250013ShandongChina
| | - Bin Ning
- Jinan Central Hospital Affiliated to Shandong UniversityNo. 105, Jiefang RoadJinan250013ShandongChina
| |
Collapse
|
13
|
Anti-tumor properties of anthocyanins from Lonicera caerulea 'Beilei' fruit on human hepatocellular carcinoma: In vitro and in vivo study. Biomed Pharmacother 2018; 104:520-529. [PMID: 29800916 DOI: 10.1016/j.biopha.2018.05.057] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Revised: 05/10/2018] [Accepted: 05/14/2018] [Indexed: 12/20/2022] Open
Abstract
In this study, the anthocyanin from Lonicera caerulea 'Beilei' fruit (ABL) was extracted and purified. The purified component (ABL-2) was then evaluated for its anti-tumor properties on human hepatoma cells (SMMC-7721) in vitro and the murine hepatoma cells (H22) in vivo. In vitro, ABL-2 not only significantly inhibited the growth of SMMC-7721 cells, but also remarkably blocked the cells' cycle in G2/M phase, inducing DNA damage and eventually leading to apoptosis. In vivo, ABL also killed tumor cells, inhibited tumor growth, and improved the survival status of H22 tumor-bearing mice. These effects were associated with an increase in the activities of antioxidase and a decrease in the level of lipid peroxidation, as evidenced by changes in SOD, GSH-Px, GSH, and MDA levels. In addition, ABL-2 also regulated the levels of immune cytokines including IL-2, IFN-γ, and TNF-α. These results revealed that ABL-2 exerts an effective anti-tumor effect by dynamically adjusting the REDOX balance and improving the immunoregulatory activity of H22 tumor-bearing mice. High performance liquid chromatography (HPLC) analysis revealed that cyanidin-3,5-diglucoside (8.16 mg/g), cyanidin-3-glucoside (387.60 mg/g), cyanidin-3-rutinoside (23.62 mg/g), and peonidin-3-glucoside (22.20 mg/g) were the main components in ABL-2, which may contribute to its anti-tumor activity.
Collapse
|
14
|
Dings RPM, Miller MC, Griffin RJ, Mayo KH. Galectins as Molecular Targets for Therapeutic Intervention. Int J Mol Sci 2018; 19:ijms19030905. [PMID: 29562695 PMCID: PMC5877766 DOI: 10.3390/ijms19030905] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 03/14/2018] [Accepted: 03/15/2018] [Indexed: 02/06/2023] Open
Abstract
Galectins are a family of small, highly conserved, molecular effectors that mediate various biological processes, including chemotaxis and angiogenesis, and that function by interacting with various cell surface glycoconjugates, usually targeting β-galactoside epitopes. Because of their significant involvement in various biological functions and pathologies, galectins have become a focus of therapeutic discovery for clinical intervention against cancer, among other pathological disorders. In this review, we focus on understanding galectin structure-function relationships, their mechanisms of action on the molecular level, and targeting them for therapeutic intervention against cancer.
Collapse
Affiliation(s)
- Ruud P M Dings
- Department of Radiation Oncology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.
| | - Michelle C Miller
- Department of Biochemistry, Molecular Biology & Biophysics, University of Minnesota, Minneapolis, MN 55455, USA.
| | - Robert J Griffin
- Department of Radiation Oncology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.
| | - Kevin H Mayo
- Department of Biochemistry, Molecular Biology & Biophysics, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
15
|
Galectin Targeted Therapy in Oncology: Current Knowledge and Perspectives. Int J Mol Sci 2018; 19:ijms19010210. [PMID: 29320431 PMCID: PMC5796159 DOI: 10.3390/ijms19010210] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 12/23/2017] [Accepted: 12/28/2017] [Indexed: 12/13/2022] Open
Abstract
The incidence and mortality of cancer have increased over the past decades. Significant progress has been made in understanding the underpinnings of this disease and developing therapies. Despite this, cancer still remains a major therapeutic challenge. Current therapeutic research has targeted several aspects of the disease such as cancer development, growth, angiogenesis and metastases. Many molecular and cellular mechanisms remain unknown and current therapies have so far failed to meet their intended potential. Recent studies show that glycans, especially oligosaccharide chains, may play a role in carcinogenesis as recognition patterns for galectins. Galectins are members of the lectin family, which show high affinity for β-galactosides. The galectin–glycan conjugate plays a fundamental role in metastasis, angiogenesis, tumor immunity, proliferation and apoptosis. Galectins’ action is mediated by a structure containing at least one carbohydrate recognition domain (CRD). The potential prognostic value of galectins has been described in several neoplasms and helps clinicians predict disease outcome and determine therapeutic interventions. Currently, new therapeutic strategies involve the use of inhibitors such as competitive carbohydrates, small non-carbohydrate binding molecules and antibodies. This review outlines our current knowledge regarding the mechanism of action and potential therapy implications of galectins in cancer.
Collapse
|
16
|
Jenkins SV, Nima ZA, Vang KB, Kannarpady G, Nedosekin DA, Zharov VP, Griffin RJ, Biris AS, Dings RPM. Triple-negative breast cancer targeting and killing by EpCAM-directed, plasmonically active nanodrug systems. NPJ Precis Oncol 2017; 1:27. [PMID: 29872709 PMCID: PMC5871908 DOI: 10.1038/s41698-017-0030-1] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Revised: 06/29/2017] [Accepted: 07/19/2017] [Indexed: 11/16/2022] Open
Abstract
An ongoing need for new cancer therapeutics exists, especially ones that specifically home and target triple-negative breast cancer. Because triple-negative breast cancer express low or are devoid of estrogen, progesterone, or Her2/Neu receptors, another target must be used for advanced drug delivery strategies. Here, we engineered a nanodrug delivery system consisting of silver-coated gold nanorods (AuNR/Ag) targeting epithelial cell adhesion/activating molecule (EpCAM) and loaded with doxorubicin. This nanodrug system, AuNR/Ag/Dox-EpCAM, was found to specifically target EpCAM-expressing tumors compared to low EpCAM-expressing tumors. Namely, the nanodrug had an effective dose (ED50) of 3 μM in inhibiting 4T1 cell viability and an ED50 of 110 μM for MDA-MD-231 cells. Flow cytometry data indicated that 4T1 cells, on average, express two orders of magnitude more EpCAM than MDA-MD-231 cells, which correlates with our ED50 findings. Moreover, due to the silver coating, the AuNR/Ag can be detected simultaneously by surface-enhanced Raman spectroscopy and photoacoustic microscopy. Analysis by these imaging detection techniques as well as by inductively coupled plasma mass spectrometry showed that the targeted nanodrug system was taken up by EpCAM-expressing cells and tumors at significantly higher rates than untargeted nanoparticles (p < 0.05). Thus, this approach establishes a plasmonically active nanodrug theranostic for triple-negative breast cancer and, potentially, a delivery platform with improved multimodal imaging capability for other clinically relevant chemotherapeutics with dose-limiting toxicities, such as platinum-based or taxane-based therapies. Silver-coated gold nanorods deliver drugs to a difficult-to-treat breast cancer by targeting an over-expressed antigen on its surface. Ruud Dings and colleagues at the University of Arkansas in the USA loaded the chemotherapeutic drug doxorubicin onto silver-coated gold nanorods that were conjugated with an antibody that specifically targets an over-expressed antigen on many types of ‘triple-negative breast cancers’ (TNBCs). Unlike other breast cancers, TNBCs lack certain receptors, making them difficult to target with cancer therapies. The team found that one of the two TNBC cell lines studied over-expressed the epithelial antigen EpCAM 100 times more than the other. Their drug-loaded silver-coated gold nanorods specifically targeted the EpCAM over-expressing cells over the low-expressing ones. The nanorods’ coatings also allowed them to be easily detected by two different imaging techniques: surfaced-enhanced Raman spectroscopy and photoacoustic microscopy.
Collapse
Affiliation(s)
- Samir V Jenkins
- 1Department of Radiation Oncology, University of Arkansas for Medical Sciences, Little Rock, AR USA
| | - Zeid A Nima
- 2Center for Integrative Nanotechnology Sciences, University of Arkansas at Little Rock, Little Rock, AR USA
| | - Kieng B Vang
- 2Center for Integrative Nanotechnology Sciences, University of Arkansas at Little Rock, Little Rock, AR USA
| | - Ganesh Kannarpady
- 2Center for Integrative Nanotechnology Sciences, University of Arkansas at Little Rock, Little Rock, AR USA
| | - Dmitry A Nedosekin
- 3The Phillips Classic Laser and Nanomedicine Laboratories, University of Arkansas for Medical Sciences, Little Rock, AR USA
| | - Vladimir P Zharov
- 3The Phillips Classic Laser and Nanomedicine Laboratories, University of Arkansas for Medical Sciences, Little Rock, AR USA
| | - Robert J Griffin
- 1Department of Radiation Oncology, University of Arkansas for Medical Sciences, Little Rock, AR USA
| | - Alexandru S Biris
- 2Center for Integrative Nanotechnology Sciences, University of Arkansas at Little Rock, Little Rock, AR USA
| | - Ruud P M Dings
- 1Department of Radiation Oncology, University of Arkansas for Medical Sciences, Little Rock, AR USA
| |
Collapse
|
17
|
Modifying Dendritic Cell Activation with Plasmonic Nano Vectors. Sci Rep 2017; 7:5513. [PMID: 28710434 PMCID: PMC5511287 DOI: 10.1038/s41598-017-04459-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Accepted: 05/03/2017] [Indexed: 11/23/2022] Open
Abstract
Dendritic cells (DCs) can acquire, process, and present antigens to T-cells to induce an immune response. For this reason, targeting cancer antigens to DCs in order to cause an immune response against cancer is an emerging area of nanomedicine that has the potential to redefine the way certain cancers are treated. The use of plasmonically active silver-coated gold nanorods (henceforth referred to as plasmonic nano vectors (PNVs)) as potential carriers for DC tumor vaccines has not been presented before. Effective carriers must be able to be phagocytized by DCs, present low toxicity, and induce the maturation of DCs—an early indication of an immune response. When we treated DCs with the PNVs, we found that the cell viability of DCs was unaffected, up to 200 μg/ml. Additionally, the PNVs associated with the DCs as they were phagocytized and they were found to reside within intracellular compartments such as endosomes. More importantly, the PNVs were able to induce expression of surface markers indicative of DC activation and maturation, i.e. CD40, CD86, and MHC class II. These results provide the first evidence that PNVs are promising carriers for DC-based vaccines and warrant further investigating for clinical use.
Collapse
|
18
|
Jenkins SV, Nedosekin DA, Miller EK, Zharov VP, Dings RPM, Chen J, Griffin RJ. Galectin-1-based tumour-targeting for gold nanostructure-mediated photothermal therapy. Int J Hyperthermia 2017; 34:19-29. [PMID: 28540812 DOI: 10.1080/02656736.2017.1317845] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Abstract
PURPOSE To demonstrate delivery of Au nanocages to cells using the galectin-1 binding peptide anginex (Ax) and to demonstrate the value of this targeting for selective in vitro photothermal cell killing. MATERIALS AND METHODS Au nanocages were synthesised, coated with polydopamine (PDA), and conjugated with Ax. Tumour and endothelial cell viability was measured with and without laser irradiation. Photoacoustic (PA) mapping and PA flow cytometry were used to confirm cell targeting in vitro and in tissue slices ex vivo. RESULTS Cell viability was maintained at ≥50% at 100 pM suggesting low toxicity of the nanocage alone. Combining the targeted construct (25 pM) with low power 808 nm laser irradiation for 10-20 min (a duration previously shown to induce rapid and sustained heating of Au nanocages [AuNC] in solution), resulted in over 50% killing of endothelial and tumour cells. In contrast, the untargeted construct combined with laser irradiation resulted in negligible cell killing. We estimate approximately 6 × 104 peptides were conjugated to each nanocage, which also resulted in inhibition of cell migration. Binding of the targeted nanocage reached a plateau after three hours, and cell association was 20-fold higher than non-targeted nanocages both in vitro and ex vivo on tumour tissue slices. A threefold increase in tumour accumulation was observed in preliminary in vivo studies. CONCLUSIONS These studies demonstrate Ax's potential as an effective targeting agent for Au-based theranostics to tumour and endothelial cells, enabling photothermal killing. This platform further suggests potential for multimodal in vivo therapy via next-generation drug-loaded nanocages.
Collapse
Affiliation(s)
| | - Dmitry A Nedosekin
- b Otolaryngology and Phillips Classic Laser and Nanomedicine Laboratories , University of Arkansas for Medical Sciences , Little Rock , AR , USA
| | - Emily K Miller
- c Department of Chemistry and Biochemistry , University of Arkansas , Fayetteville , AR , USA
| | - Vladimir P Zharov
- b Otolaryngology and Phillips Classic Laser and Nanomedicine Laboratories , University of Arkansas for Medical Sciences , Little Rock , AR , USA
| | | | - Jingyi Chen
- c Department of Chemistry and Biochemistry , University of Arkansas , Fayetteville , AR , USA
| | | |
Collapse
|
19
|
Seidi K, Jahanban-Esfahlan R, Zarghami N. Tumor rim cells: From resistance to vascular targeting agents to complete tumor ablation. Tumour Biol 2017; 39:1010428317691001. [DOI: 10.1177/1010428317691001] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Current vascular targeting strategies pursue two main goals: anti-angiogenesis agents aim to halt sprouting and the formation of new blood vessels, while vascular disrupting agents along with coaguligands seek to compromise blood circulation in the vessels. The ultimate goal of such therapies is to deprive tumor cells out of oxygen and nutrients long enough to succumb cancer cells to death. Most of vascular targeting agents presented promising therapeutic potential, but the final goal which is cure is rarely achieved. Nevertheless, in both preclinical and clinical settings, tumors tend to grow back, featuring a highly invasive, metastatic, and extremely resistant form. This review highlights the critical significance of tumor rim cells as the main factor, determining therapy success with vascular targeting agents. We present an overview of different single and combination treatments with vascular targeting agents that enable efficient targeting of tumor rim cells and long-lasting tumor cure. Understanding the nature of tumor rim cells, how they establish, how they manage to survive of vascular targeting agents, and how they contribute in tumor refractoriness, may open new avenues to the development of beneficial strategies, capable to eliminate residual rim cells, and enable tumor ablation once and forever.
Collapse
Affiliation(s)
- Khaled Seidi
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Rana Jahanban-Esfahlan
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nosratollah Zarghami
- Department of Clinical Biochemistry and Laboratory Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
20
|
Blanchard H, Bum-Erdene K, Bohari MH, Yu X. Galectin-1 inhibitors and their potential therapeutic applications: a patent review. Expert Opin Ther Pat 2016; 26:537-54. [PMID: 26950805 DOI: 10.1517/13543776.2016.1163338] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Galectins have affinity for β-galactosides. Human galectin-1 is ubiquitously expressed in the body and its expression level can be a marker in disease. Targeted inhibition of galectin-1 gives potential for treatment of inflammatory disorders and anti-cancer therapeutics. AREAS COVERED This review discusses progress in galectin-1 inhibitor discovery and development. Patent applications pertaining to galectin-1 inhibitors are categorised as monovalent- and multivalent-carbohydrate-based inhibitors, peptides- and peptidomimetics. Furthermore, the potential of galectin-1 protein as a therapeutic is discussed along with consideration of the unique challenges that galectin-1 presents, including its monomer-dimer equilibrium and oxidized and reduced forms, with regard to delivering an intact protein to a pathologically relevant site. EXPERT OPINION Significant evidence implicates galectin-1's involvement in cancer progression, inflammation, and host-pathogen interactions. Conserved sequence similarity of the carbohydrate-binding sites of different galectins makes design of specific antagonists (blocking agents/inhibitors of function) difficult. Key challenges pertaining to the therapeutic use of galectin-1 are its monomer-dimer equilibrium, its redox state, and delivery of intact galectin-1 to the desired site. Developing modified forms of galectin-1 has resulted in increased stability and functional potency. Gene and protein therapy approaches that deliver the protein toward the target are under exploration as is exploitation of different inhibitor scaffolds.
Collapse
Affiliation(s)
- Helen Blanchard
- a Institute for Glycomics , Griffith University , Gold Coast Campus , Queensland , Australia
| | - Khuchtumur Bum-Erdene
- a Institute for Glycomics , Griffith University , Gold Coast Campus , Queensland , Australia
| | | | - Xing Yu
- a Institute for Glycomics , Griffith University , Gold Coast Campus , Queensland , Australia
| |
Collapse
|
21
|
Abstract
Immune adjuvants have been used in cancer biotherapies to stimulate immune response to tumor cells. Despite their potential as anticancer reagents, there are several impediments to their use in clinical applications. In this study, we aim to modify the existing tuftsin structure and evaluate its antitumor activity in preclinical models. We synthesized a novel tuftsin derivative, namely, the T peptide (TP), by linking four tuftsin peptides, which showed enhanced stability in vivo. We then evaluated its anticancer activity in a postoperative residual tumor model in mice, where we surgically removed most of the primary tumor from the host, a procedure mimicking clinically postoperative patients. Despite the limited effect in intact solid tumors, TP strongly inhibited relapsed growth of residual tumors in postsurgical mice. Surgical resection of tumors accelerated residual tumor growth, but TP slowed down this process significantly. Interestingly, TP showed similar effects in human xenograft residual models. As an immunomodulator, TP could synergize the functions of macrophages, thus inhibiting the growth of cocultured tumor cells in vitro. Furthermore, TP could shift the macrophages to the tumor-suppressive M1 type and mobilize them to produce elevated cytotoxic TNF-α and NO. As a result, TP effectively prolonged the survival time of tumor-resected mice. Using the postoperative residual tumor models, we provide a body of evidence showing the antitumor activity of TP, which causes no obvious toxicity. Our study highlights the potential of TP as a postoperative adjuvant in cancer therapies.
Collapse
|
22
|
Wu D, Gao Y, Qi Y, Chen L, Ma Y, Li Y. Peptide-based cancer therapy: opportunity and challenge. Cancer Lett 2014; 351:13-22. [PMID: 24836189 DOI: 10.1016/j.canlet.2014.05.002] [Citation(s) in RCA: 212] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Revised: 03/31/2014] [Accepted: 05/01/2014] [Indexed: 01/01/2023]
Abstract
Cancer is one of the leading causes of death worldwide. Conventional cancer therapies mainly focus on mass cell killing without high specificity and often cause severe side effects and toxicities. Peptides are a novel class of anticancer agents that could specifically target cancer cells with lower toxicity to normal tissues, which will offer new opportunities for cancer prevention and treatment. Anticancer peptides face several therapeutic challenges. In this review, we present the sources and mechanisms of anticancer peptides and further discuss modification strategies to improve the anticancer effects of bioactive peptides.
Collapse
Affiliation(s)
- Dongdong Wu
- College of Medicine, Henan University, Kaifeng 475004, Henan, China
| | - Yanfeng Gao
- School of Life Science, Zhengzhou University, Zhengzhou 450001, Henan, China
| | - Yuanming Qi
- School of Life Science, Zhengzhou University, Zhengzhou 450001, Henan, China.
| | - Lixiang Chen
- School of Life Science, Zhengzhou University, Zhengzhou 450001, Henan, China
| | - Yuanfang Ma
- College of Medicine, Henan University, Kaifeng 475004, Henan, China
| | - Yanzhang Li
- College of Medicine, Henan University, Kaifeng 475004, Henan, China.
| |
Collapse
|
23
|
Bioactive metabolites from macrofungi: ethnopharmacology, biological activities and chemistry. FUNGAL DIVERS 2013. [DOI: 10.1007/s13225-013-0265-2] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
24
|
Dings RPM, Levine JI, Brown SG, Astorgues-Xerri L, MacDonald JR, Hoye TR, Raymond E, Mayo KH. Polycationic calixarene PTX013, a potent cytotoxic agent against tumors and drug resistant cancer. Invest New Drugs 2013; 31:1142-50. [PMID: 23392775 PMCID: PMC4242102 DOI: 10.1007/s10637-013-9932-0] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2012] [Accepted: 01/24/2013] [Indexed: 01/01/2023]
Abstract
Previously, we reported on the anti-tumor activities of two designed calix[4]arene-based topomimetics (PTX008 and PTX009) of the amphipathic, angiostatic peptide Anginex. Here, we chemically modified the hydrophobic and hydrophilic faces of PTX008 and PTX009, and discovered new calixarene compounds that are more potent, cytotoxic anti-tumor agents. One of them, PTX013, is particularly effective at inhibiting the growth of several human cancer cell lines, as well as drug resistant cancer cells. Mechanistically, PTX013 induces cell cycle arrest in sub-G1 and G0/G1 phases of e.g. SQ20B cells, a radio-resistant human head and neck carcinoma model. In the syngeneic B16F10 melanoma tumor mouse model, PTX013 (0.5 mg/Kg) inhibits tumor growth by about 50-fold better than parent PTX008. A preliminary pharmacodynamics study strongly suggests that PTX013 exhibits good in vivo exposure and a relatively long half-life. Overall, this research contributes to the discovery of novel therapeutics as potentially useful agents against cancer in the clinic.
Collapse
Affiliation(s)
- Ruud P. M. Dings
- Department of Biochemistry, Molecular Biology & Biophysics,
University of Minnesota, Minneapolis, MN 55455, USA
| | - Joseph I. Levine
- Department of Biochemistry, Molecular Biology & Biophysics,
University of Minnesota, Minneapolis, MN 55455, USA
- Department of Chemistry, University of Minnesota, Minneapolis, MN
55455, USA
| | - Susan G. Brown
- Department of Chemistry, University of Minnesota, Minneapolis, MN
55455, USA
| | - Lucile Astorgues-Xerri
- INSERM U728 and Department of Medical Oncology, Beaujon University
Hospital, (AP-HP – PRES Paris 7 Diderot), 100 bd du
Général Leclerc, 92110 Paris-Clichy, France
| | | | - Thomas R. Hoye
- Department of Chemistry, University of Minnesota, Minneapolis, MN
55455, USA
| | - Eric Raymond
- INSERM U728 and Department of Medical Oncology, Beaujon University
Hospital, (AP-HP – PRES Paris 7 Diderot), 100 bd du
Général Leclerc, 92110 Paris-Clichy, France
| | - Kevin H. Mayo
- Department of Biochemistry, Molecular Biology & Biophysics,
University of Minnesota, Minneapolis, MN 55455, USA
| |
Collapse
|
25
|
Astorgues-Xerri L, Riveiro ME, Tijeras-Raballand A, Serova M, Neuzillet C, Albert S, Raymond E, Faivre S. Unraveling galectin-1 as a novel therapeutic target for cancer. Cancer Treat Rev 2013; 40:307-19. [PMID: 23953240 DOI: 10.1016/j.ctrv.2013.07.007] [Citation(s) in RCA: 163] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2013] [Revised: 07/22/2013] [Accepted: 07/24/2013] [Indexed: 12/13/2022]
Abstract
Galectins belong to a family of carbohydrate-binding proteins with an affinity for β-galactosides. Galectin-1 is differentially expressed by various normal and pathologic tissues and displays a wide range of biological activities. In oncology, galectin-1 plays a pivotal role in tumor growth and in the multistep process of invasion, angiogenesis, and metastasis. Evidence indicates that galectin-1 exerts a variety of functions at different steps of tumor progression. Moreover, it has been demonstrated that galectin-1 cellular localization and galectin-1 binding partners depend on tumor localization and stage. Recently, galectin-1 overexpression has been extensively documented in several tumor types and/or in the stroma of cancer cells. Its expression is thought to reflect tumor aggressiveness in several tumor types. Galectin-1 has been identified as a promising drug target using synthetic and natural inhibitors. Preclinical data suggest that galectin-1 inhibition may lead to direct antiproliferative effects in cancer cells as well as antiangiogenic effects in tumors. We provide an up-to-date overview of available data on the role of galectin-1 in different molecular and biochemical pathways involved in human malignancies. One of the major challenges faced in targeting galectin-1 is the translation of current knowledge into the design and development of effective galectin-1 inhibitors in cancer therapy.
Collapse
|
26
|
Dings RPM, Kumar N, Miller MC, Loren M, Rangwala H, Hoye TR, Mayo KH. Structure-based optimization of angiostatic agent 6DBF7, an allosteric antagonist of galectin-1. J Pharmacol Exp Ther 2012; 344:589-99. [PMID: 23232447 DOI: 10.1124/jpet.112.199646] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Galectin-1 (gal-1), which binds β-galactoside groups on various cell surface receptors, is crucial to cell adhesion and migration, and is found to be elevated in several cancers. Previously, we reported on 6DBF7, a dibenzofuran (DBF)-based peptidomimetic of the gal-1 antagonist anginex. In the present study, we used a structure-based approach to optimize 6DBF7. Initial NMR studies showed that 6DBF7 binds to gal-1 on one side of the β-sandwich away from the lectin's carbohydrate binding site. Although an alanine scan of 6DBF7 showed that the two cationic groups (lysines) in the partial peptide are crucial to its angiostatic activity, it is the hydrophobic face of the amphipath that appears to interact directly with the surface of gal-1. Based on this structural information, we designed and tested additional DBF analogs. In particular, substitution of the C-terminal Asp for alanine and branched alkyl side chains (Val, Leu, Ile) for linear ones (Nle, Nva) rendered the greatest improvements in activity. Flow cytometry with gal-1(-/-) splenocytes showed that 6DBF7 and two of its more potent analogs (DB16 and DB21) can fully inhibit fluorescein isothiocyanate-gal-1 binding. Moreover, heteronuclear single-quantum coherence NMR titrations showed that the presence of DB16 decreases gal-1 affinity for lactose, indicating that the peptidomimetic targets gal-1 as a noncompetitive, allosteric inhibitor of glycan binding. Using tumor mouse models (B16F10 melanoma, LS174 lung, and MA148 ovarian), we found that DB21 inhibits tumor angiogenesis and tumor growth significantly better than 6DBF7, DB16, or anginex. DB21 is currently being developed further and holds promise for the management of human cancer in the clinic.
Collapse
Affiliation(s)
- Ruud P M Dings
- Department of Biochemistry, University of Minnesota, 321 Church Street, Minneapolis, MN 55455, USA
| | | | | | | | | | | | | |
Collapse
|
27
|
Kim HJ, Jeon HK, Cho YJ, Park YA, Choi JJ, Do IG, Song SY, Lee YY, Choi CH, Kim TJ, Bae DS, Lee JW, Kim BG. High galectin-1 expression correlates with poor prognosis and is involved in epithelial ovarian cancer proliferation and invasion. Eur J Cancer 2012; 48:1914-21. [DOI: 10.1016/j.ejca.2012.02.005] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2011] [Revised: 01/16/2012] [Accepted: 02/07/2012] [Indexed: 12/31/2022]
|
28
|
Griffin RJ, Koonce NA, Dings RPM, Siegel E, Moros EG, Bräuer-Krisch E, Corry PM. Microbeam radiation therapy alters vascular architecture and tumor oxygenation and is enhanced by a galectin-1 targeted anti-angiogenic peptide. Radiat Res 2012; 177:804-812. [PMID: 22607585 PMCID: PMC3391740 DOI: 10.1667/rr2784.1] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Abstract
In this study, we sought to determine the therapeutic potential of variably sized (50 μm or 500 μm wide, 14 mm tall) parallel microbeam radiation therapy (MRT) alone and in combination with a novel anti-angiogenic peptide, anginex, in mouse mammary carcinomas (4T1)--a moderately hypoxic and radioresistant tumor with propensity to metastasize. The fraction of total tumor volume that was directly irradiated was approximately 25% in each case, but the distance between segments irradiated by the planar microbeams (width of valley dose region) varied by an order of magnitude from 150-1500 μm corresponding to 200 μm and 2000 μm center-to-center inter-microbeam distances, respectively. We found that MRT administered in 50 μm beams at 150 Gy was most effective in delaying tumor growth. Furthermore, tumor growth delay induced by 50 μm beams at 150 Gy was virtually indistinguishable from the 500 μm beams at 150 Gy. Fifty-micrometer beams at the lower peak dose of 75 Gy induced growth delay intermediate between 150 Gy and untreated tumors, while 500 μm beams at 75 Gy were unable to alter tumor growth compared to untreated tumors. However, the addition of anginex treatment increased the relative tumor growth delay after 500 μm beams at 75 Gy most substantially out of the conditions tested. Anginex treatment of animals whose tumors received the 50 μm beams at 150 Gy also led to an improvement in growth delay from that induced by the comparable MRT alone. Immunohistochemical staining for CD31 (endothelial cells) and αSMA (smooth muscle pericyte-associated blood vessels as a measure of vessel normalization) indicated that vessel density was significantly decreased in all irradiated groups and pericyte staining was significantly increased in the irradiated groups on day 14 after irradiation. The addition of anginex treatment further decreased the mean vascular density in all combination treatment groups and further increased the amount of pericyte staining in these tumors. Finally, evidence of tumor hypoxia was found to decrease in tumors analyzed at 1-14 days after MRT in the groups receiving 150 Gy peak dose, but not 75 Gy peak dose. Our results suggest that tumor vascular damage induced by MRT at these potentially clinically acceptable peak entrance doses may provoke vascular normalization and may be exploited to improve tumor control using agents targeting angiogenesis.
Collapse
Affiliation(s)
- Robert J Griffin
- Department of Radiation Oncology, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205, USA.
| | | | | | | | | | | | | |
Collapse
|
29
|
Dings RPM, Miller MC, Nesmelova I, Astorgues-Xerri L, Kumar N, Serova M, Chen X, Raymond E, Hoye TR, Mayo KH. Antitumor agent calixarene 0118 targets human galectin-1 as an allosteric inhibitor of carbohydrate binding. J Med Chem 2012; 55:5121-9. [PMID: 22575017 DOI: 10.1021/jm300014q] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Calix[4]arene compound 0118 is an angiostatic agent that inhibits tumor growth in mice. Although 0118 is a topomimetic of galectin-1-targeting angiostatic amphipathic peptide Anginex, we had yet to prove that 0118 targets galectin-1. Galectin-1 is involved in pathological disorders like tumor endothelial cell adhesion and migration and therefore presents a relevant target for therapeutic intervention against cancer. Here, (15)N-(1)H HSQC NMR spectroscopy demonstrates that 0118 indeed targets galectin-1 at a site away from the lectin's carbohydrate binding site and thereby attenuates lactose binding to the lectin. Flow cytometry and agglutination assays show that 0118 attenuates binding of galectin-1 to cell surface glycans, and the inhibition of cell proliferation by 0118 is found to be correlated with the cellular expression of the lectin. In general, our data indicate that 0118 targets galectin-1 as an allosteric inhibitor of glycan/carbohydrate binding. This work contributes to the clinical development of antitumor calixarene compound 0118.
Collapse
Affiliation(s)
- Ruud P M Dings
- Department of Biochemistry, Molecular Biology & Biophysics, University of Minnesota Health Sciences Center, 6-155 Jackson Hall, 321 Church Street, Minneapolis, Minnesota 55455, United States
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Medicinal mushrooms in supportive cancer therapies: an approach to anti-cancer effects and putative mechanisms of action. FUNGAL DIVERS 2012. [DOI: 10.1007/s13225-012-0151-3] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
31
|
Belotti D, Foglieni C, Resovi A, Giavazzi R, Taraboletti G. Targeting angiogenesis with compounds from the extracellular matrix. Int J Biochem Cell Biol 2011; 43:1674-85. [DOI: 10.1016/j.biocel.2011.08.012] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2011] [Revised: 08/05/2011] [Accepted: 08/10/2011] [Indexed: 02/08/2023]
|
32
|
Liu D, Liu F, Liu Z, Wang L, Zhang N. Tumor Specific Delivery and Therapy by Double-Targeted Nanostructured Lipid Carriers with Anti-VEGFR-2 Antibody. Mol Pharm 2011; 8:2291-301. [DOI: 10.1021/mp200402e] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Donghua Liu
- School of Pharmaceutical Sciences, Shandong University, 44 Wenhua Xi Road, Ji’nan 250012, China
| | - Fengxi Liu
- School of Pharmaceutical Sciences, Shandong University, 44 Wenhua Xi Road, Ji’nan 250012, China
| | - Zhihong Liu
- School of Pharmaceutical Sciences, Shandong University, 44 Wenhua Xi Road, Ji’nan 250012, China
| | - Lili Wang
- School of Pharmaceutical Sciences, Shandong University, 44 Wenhua Xi Road, Ji’nan 250012, China
| | - Na Zhang
- School of Pharmaceutical Sciences, Shandong University, 44 Wenhua Xi Road, Ji’nan 250012, China
| |
Collapse
|
33
|
Dings RPM, Vang KB, Castermans K, Popescu F, Zhang Y, Oude Egbrink MGA, Mescher MF, Farrar MA, Griffioen AW, Mayo KH. Enhancement of T-cell-mediated antitumor response: angiostatic adjuvant to immunotherapy against cancer. Clin Cancer Res 2011; 17:3134-45. [PMID: 21252159 DOI: 10.1158/1078-0432.ccr-10-2443] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
PURPOSE Tumor-released proangiogenic factors suppress endothelial adhesion molecule (EAM) expression and prevent leukocyte extravasation into the tumor. This is one reason why immunotherapy has met with limited success in the clinic. We hypothesized that overcoming EAM suppression with angiogenesis inhibitors would increase leukocyte extravasation and subsequently enhance the effectiveness of cellular immunotherapy. EXPERIMENTAL DESIGN Intravital microscopy, multiple color flow cytometry, immunohistochemistry, and various tumor mouse (normal and T-cell deficient) models were used to investigate the temporal dynamics of cellular and molecular events that occur in the tumor microenvironment during tumor progression and angiostatic intervention. RESULTS We report that while EAM levels and T-cell infiltration are highly attenuated early on in tumor growth, angiostatic therapy modulates these effects. In tumor models with normal and T-cell-deficient mice, we show the active involvement of the adaptive immune system in cancer and differentiate antiangiogenic effects from antiangiogenic mediated enhancement of immunoextravasation. Our results indicate that a compromised immune response in tumors can be obviated by the use of antiangiogenic agents. Finally, with adoptive transfer studies in mice, we show that a phased combination of angiostatic therapy and T-cell transfer significantly (P < 0.0013) improves tumor growth inhibition. CONCLUSIONS This research contributes to understand the cellular mechanism of action of angiostatic agents and the immune response within the tumor microenvironment, in particular as a consequence of the temporal dynamics of EAM levels. Moreover, our results suggest that adjuvant therapy with angiogenesis inhibitors holds promise for cellular immunotherapy in the clinic.
Collapse
Affiliation(s)
- Ruud P M Dings
- Department of Biochemistry, The Masonic Cancer Center, University of Minnesota, Minnesota 55455, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Dings RPM, Loren ML, Zhang Y, Mikkelson S, Mayo KH, Corry P, Griffin RJ. Tumour thermotolerance, a physiological phenomenon involving vessel normalisation. Int J Hyperthermia 2011; 27:42-52. [PMID: 21204622 DOI: 10.3109/02656736.2010.510495] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The purpose of this study was to delineate the mechanisms by which stromal components of cancer may induce tumour thermotolerance and exploit alterations in stromal and tumour physiology to enhance radiation therapy. The vascular thermoresponse was monitored by daily one-hour 41.5°C heatings in two murine solid tumour models, SCK murine mammary carcinoma and B16F10 melanoma. A transient increase was seen in overall tumour oxygenation for 2-3 days, followed by a progressive decline in tumour pO(2) upon continued daily heatings. Vascular thermotolerance was further studied by treating tumours with different heating strategies, i.e. (1) a single 60 min 41.5°C treatment; (2) two consecutive daily treatments of 41.5°C for 60 min; (3) a single 60 min 43°C treatment or (4) two days of 41.5°C for 60 min followed by treatment with 43°C for 60 min on the third day. Pre-heating tumours with mild temperature hyperthermia induced vascular thermotolerance, which was accompanied by evidence of vessel normalisation, i.e. a decrease in microvessel density and an increase in pericyte coverage. Rational scheduling of fractionated radiation during heat-induced increases in tumour oxygen levels rendered a significantly greater, synergistic, tumour growth inhibition. In vitro clonogenic survival responses of the individual cell types associated (endothelial cells, fibroblasts, pericytes and tumour cells) indicated only a direct cellular thermotolerance in endothelial cells. Overall, this suggests that tumour thermotolerance is a physiological phenomenon mediated through improvement of functional vasculature.
Collapse
Affiliation(s)
- Ruud P M Dings
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, MN, USA
| | | | | | | | | | | | | |
Collapse
|
35
|
Schilder RJ, Blessing JA, Shahin MS, Miller DS, Tewari KS, Muller CY, Warshal DP, McMeekin S, Rotmensch J. A Phase 2 Evaluation of Irofulven as Second-line Treatment of Recurrent or Persistent Intermediately Platinum-Sensitive Ovarian or Primary Peritoneal Cancer. Int J Gynecol Cancer 2010; 20:1137-41. [DOI: 10.1111/igc.0b013e3181e8df36] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
|
36
|
Dings RPM, Van Laar ES, Loren M, Webber J, Zhang Y, Waters SJ, Macdonald JR, Mayo KH. Inhibiting tumor growth by targeting tumor vasculature with galectin-1 antagonist anginex conjugated to the cytotoxic acylfulvene, 6-hydroxylpropylacylfulvene. Bioconjug Chem 2010; 21:20-7. [PMID: 20020769 DOI: 10.1021/bc900287y] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Targeted delivery of therapeutic drugs promises to become the norm to treat cancer. Here, we conjugated the cytotoxic agent 6-hydroxypropylacylfulvene (HPAF) to anginex, a peptide that targets galectin-1, which is highly expressed in endothelial cells of tumor vessels. In a human ovarian cancer model in mice, the conjugate inhibited tumor growth better than equivalent doses of either compound alone. Immunofluorescence on tumor tissue demonstrated that the conjugate, like parent anginex, selectively targeted tumor vasculature and inhibited tumor angiogenesis. Increased activity from the conjugate further suggests that HPAF retains at least some of its normal cytotoxic activity when linked to anginex. More importantly perhaps is the observation that the conjugate abrogates apparent systemic toxicity from treatment with HPAF. This work contributes to the development of tumor vascular targeting agents against cancer in the clinic.
Collapse
Affiliation(s)
- Ruud P M Dings
- Department of Biochemistry, University of Minnesota, Minnesota, USA
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Wu D, Gao Y, Chen L, Qi Y, Kang Q, Wang H, Zhu L, Ye Y, Zhai M. Anti-tumor effects of a novel chimeric peptide on S180 and H22 xenografts bearing nude mice. Peptides 2010; 31:850-64. [PMID: 20132854 DOI: 10.1016/j.peptides.2010.01.007] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2009] [Revised: 01/23/2010] [Accepted: 01/25/2010] [Indexed: 12/23/2022]
Abstract
In recent years, many endogenous peptides have been identified by screening combinatory phage display peptide library, which play important roles in the process of angiogenesis. A heptapeptide, ATWLPPR, binds specifically to NRP-1 and selectively inhibits VEGF165 binding to VEGFR-2. Another heptapeptide, NLLMAAS, blocks both Ang-1 and Ang-2 binding to Tie-2 in a dose-dependent manner. In the present study, we aimed to connect ATWLPPR (V1) with NLLMAAS (V2) via a flexible linker, Ala-Ala, to reconstruct a novel peptide ATWLPPRAANLLMAAS (V3). We firstly investigated the anti-tumor and anti-angiogenic effects of peptide V3 on sarcoma S180 and hepatoma H22 bearing BALB/c nude mice. Mice were continuously subcutaneously administrated with normal saline, V1 (320microg/kg/d), V2 (320microg/kg/d), V1+V2 (320microg/kg/d), and V3 (160, 320 and 480microg/kg/d), for 7 days. Treatment with peptide V3 could significantly reduce the tumor weight and volume. Pathological examination showed that the tumors treated with peptide V3 had a larger region of necrosis than that of peptide V1, V2, and V1+V2 at the same dose. A significant decrease of microvessel density (MVD) in a dose-dependent manner was observed in each group of peptide V3. The results of pathological examination on normal tissue, lung, heart, liver, spleen, kidney and white blood cells showed that peptide V3 might have no significant toxicity. In conclusion, our results demonstrated that peptide V3 could be more effective on inhibiting tumor growth and angiogenesis than that of V1, V2, and V1+V2. Peptide V3 could be considered as a novel chimeric peptide with potent anti-tumor activity.
Collapse
Affiliation(s)
- Dongdong Wu
- Department of Bioengineering, Zhengzhou University, Science Road 100, Zhengzhou 450001, China
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Siegel DS, Piizzi G, Piersanti G, Movassaghi M. Enantioselective total synthesis of (-)-acylfulvene and (-)-irofulven. J Org Chem 2009; 74:9292-304. [PMID: 19938810 PMCID: PMC2805080 DOI: 10.1021/jo901926z] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
We report our full account of the enantioselective total synthesis of (-)-acylfulvene (1) and (-)-irofulven (2), which features metathesis reactions for the rapid assembly of the molecular framework of these antitumor agents. We discuss (1) the application of an Evans Cu-catalyzed aldol addition reaction using a strained cyclopropyl ketenethioacetal, (2) an efficient enyne ring-closing metathesis cascade reaction in a challenging setting, (3) the reagent IPNBSH for a late-stage reductive allylic transposition reaction, and (4) the final RCM/dehydrogenation sequence for the formation of (-)-acylfulvene (1) and (-)-irofulven (2).
Collapse
Affiliation(s)
- Dustin S Siegel
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | | | | | | |
Collapse
|
39
|
Movassaghi M, Piizzi G, Siegel DS, Piersanti G. Observations in the Synthesis of the Core of the Antitumor Illudins via an Enyne Ring Closing Metathesis Cascade. Tetrahedron Lett 2009; 50:5489-5492. [PMID: 20160945 PMCID: PMC2766079 DOI: 10.1016/j.tetlet.2009.07.102] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Observations concerning the synthesis of the core spirocyclic AB-ring system of illudins using an enyne ring closing metathesis (EYRCM) cascade are discussed. Substituent effects, in addition to optimization of the reaction conditions and the olefin tether for the key EYRCM reaction, are examined.
Collapse
Affiliation(s)
- Mohammad Movassaghi
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Grazia Piizzi
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Dustin S. Siegel
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Giovanni Piersanti
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| |
Collapse
|
40
|
Abstract
Cancer therapy is in the midst of a major paradigm shift. Traditionally, cancer treatments have focused on tumour cells. However, studies over the past few decades have demonstrated that cancer is a vastly complex entity with multiple components affecting a tumour's growth, invasion and metastasis. These components, collectively termed the 'tumour microenvironment', include endothelial cells, pericytes, fibroblasts, inflammatory cells, leucocytes and elements of the extracellular matrix (ECM). Biological agents that target components of the tumour microenvironment may provide an interesting alternative to traditional tumour cell-directed therapy. Because of the complexity of the tumour milieu, the most beneficial therapy will likely involve the combination of one or more agents directed at this new target. This review highlights recent preclinical and clinical studies involving agents that target tumour vasculature, leucocytes, pericytes, cancer-associated fibroblasts and ECM components. We pay particular attention to combination therapies targeting multiple components of the tumour microenvironment, and aim to demonstrate that this strategy holds promise for the future of cancer treatment.
Collapse
Affiliation(s)
- E Hanna
- Tumour Angiogenesis Section, Surgery Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | |
Collapse
|
41
|
Abstract
Cationic peptides, known to disrupt bacterial membranes, are being developed as promising agents for therapeutic intervention against infectious disease. In the present study, we investigate structure-activity relationships in the bacterial membrane disruptor betapep-25, a peptide 33-mer. For insight into which amino acid residues are functionally important, we synthesized alanine-scanning variants of betapep-25 and assessed their ability to kill bacteria (Escherichia coli, Pseudomonas aeruginosa and Staphylococcus aureus) and to neutralize LPS (lipopolysaccharide). Activity profiles were found to vary with the bacterial strain examined. Specific cationic and smaller hydrophobic alkyl residues were crucial to optimal bactericidal activity against the Gram-negative bacteria, whereas larger hydrophobic and cationic residues mediated optimal activity against Gram-positive Staph. aureus. Lysine-substituted norleucine (n-butyl group) variants demonstrated that both charge and alkyl chain length mediate optimal activity. In terms of LPS neutralization, activity profiles were essentially the same against four species of LPS (E. coli 055 and 0111, Salmonella enterica serotype Typhimurium and Klebsiella pneumoniae), and different for two others (Ps. aeruginosa and Serratia marcescens), with specific hydrophobic, cationic and, surprisingly, anionic residues being functionally important. Furthermore, disulfide-bridged analogues demonstrated that an anti parallel beta-sheet structure is the bioactive conformation of betapep-25 in terms of its bactericidal, but not LPS endotoxin neutralizing, activity. Moreover, betapep-25 variants, like the parent peptide, do not lyse eukaryotic cells. This research contributes to the development and design of novel antibiotics.
Collapse
|