1
|
Rauf A, Naeem M, Ramzan R, Cham A. Exploring physicochemical characteristics of cyclodextrin through M-polynomial indices. Sci Rep 2024; 14:20029. [PMID: 39198520 PMCID: PMC11358294 DOI: 10.1038/s41598-024-68775-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 07/29/2024] [Indexed: 09/01/2024] Open
Abstract
Cyclodextrin, a potent anti-tumor medication utilized predominantly in ovarian and breast cancer treatments, encounters significant challenges such as poor solubility, potential side effects, and resistance from tumor cells. Combining cyclodextrin with biocompatible substrates offers a promising strategy to address these obstacles. Understanding the atomic structure and physicochemical properties of cyclodextrin and its derivatives is essential for enhancing drug solubility, modification, targeted delivery, and controlled release. In this study, we investigate the topological indices of cyclodextrin using algebraic polynomials, specifically the degree-based M-polynomial and neighbor degree-based M-polynomial. By computing degree-based and neighbor degree-based topological indices, we aim to elucidate the structural characteristics of cyclodextrin and provide insights into its physicochemical behavior. The computed indices serve as predictive tools for assessing the health benefits and therapeutic efficacy of cyclodextrin-based formulations. In addition, we examined that the computed indices showed a significant relationship with the physicochemical characteristics of antiviral drugs. Graphical representations of the computed results further facilitate the visualization and interpretation of cyclodextrin's molecular structure, aiding researchers in designing novel drug delivery systems with improved pharmacological properties.
Collapse
Affiliation(s)
- Abdul Rauf
- Department of Mathematics, Air University Multan Campus, Multan, Pakistan
| | - Muhammad Naeem
- Department of Mathematics, School of Natural Sciences, National University of Sciences and Technology (NUST), Islamabad, Pakistan
| | - Rahila Ramzan
- Department of Mathematics, Air University Multan Campus, Multan, Pakistan
| | - Alhagie Cham
- School of Arts and Sciences, University of The Gambia, Banjul, The Gambia.
| |
Collapse
|
2
|
Pourali P, Neuhöferová E, Dzmitruk V, Svoboda M, Stodůlková E, Flieger M, Yahyaei B, Benson V. Bioproduced Nanoparticles Deliver Multiple Cargoes via Targeted Tumor Therapy In Vivo. ACS OMEGA 2024; 9:33789-33804. [PMID: 39130536 PMCID: PMC11307291 DOI: 10.1021/acsomega.4c03277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 07/01/2024] [Accepted: 07/10/2024] [Indexed: 08/13/2024]
Abstract
This study recognized biologically produced gold nanoparticles (AuNPs) as multiple cargo carriers with a perspective of drug delivery into specialized tumor cells in vivo. Paclitaxel (PTX), transferrin, and antimiR-135b were conjugated with AuNPs and their uptake by mouse tumor cells in an induced breast cancer model was investigated. Each of the above-mentioned molecules was conjugated to the AuNPs separately as well as simultaneously, loading efficiency of each cargo was assessed, and performance of the final product (FP) was judged. After tumor induction in BALB/c mice, sub-IC50 doses of FP as well as control AuNPs, PTX, and phosphate buffered saline were administered in vivo. Round AuNPs were prepared using Fusarium oxysporum and exhibited a size of 13 ± 1.3 nm and a zeta potential of -35.8 ± 1.3 mV. The cytotoxicity of individual conjugates and FP were tested by MTT assay in breast tumor cells 4T1 and nontumor fibroblasts NIH/3T3 cells. The conjugation of individual molecules with AuNPs was confirmed, and FP (size of 54 ± 14 nm and zeta potential of -31.9 ± 2.08 mV) showed higher 4T1-specific toxicity in vitro when compared to control conjugates. After in vivo application of the FP, transmission electron microscopy analyses proved the presence of AuNPs in the tumor cells. Hematoxylin and eosin staining of the tumor tissue revealed that the FP group exhibited the highest amounts of inflammatory, necrotic, and apoptotic cells in contrast to the control groups. Finally, qPCR results showed that FP could transfect and suppress miR-135b expression in vivo, confirming the tumor-targeting properties of FP. The capacity of biologically produced gold nanoparticles to conjugate with multiple decorative molecules while retaining their stability and effective intracellular uptake makes them a promising alternative strategy superior to current drug carriers.
Collapse
Affiliation(s)
- Parastoo Pourali
- Institute
of Microbiology, Czech Academy of Sciences, Praha 142 20, Czech Republic
| | - Eva Neuhöferová
- Institute
of Microbiology, Czech Academy of Sciences, Praha 142 20, Czech Republic
| | - Volha Dzmitruk
- Center
of Molecular Structure, Institute of Biotechnology,
Czech Academy of Sciences, Vestec 252 20, Czech Republic
| | - Milan Svoboda
- Institute
of Analytical Chemistry, Czech Academy of
Sciences, Brno 602 00, Czech Republic
| | - Eva Stodůlková
- Institute
of Microbiology, Czech Academy of Sciences, Praha 142 20, Czech Republic
| | - Miroslav Flieger
- Institute
of Microbiology, Czech Academy of Sciences, Praha 142 20, Czech Republic
| | - Behrooz Yahyaei
- Department
of Medical Sciences, Shahrood Branch, Islamic
Azad University, Shahrood 9WVM+5HC, Iran
- Department
of Medical Sciences, Biological Nanoparticles in Medicine Research
Center, Shahrood Branch, Islamic Azad University, Shahrood 9WVM+5HC, Iran
| | - Veronika Benson
- Institute
of Microbiology, Czech Academy of Sciences, Praha 142 20, Czech Republic
- Faculty of
Health Studies, Technical University of
Liberec, Liberec 46001, Czech Republic
| |
Collapse
|
3
|
Islam F, Shohag S, Uddin MJ, Islam MR, Nafady MH, Akter A, Mitra S, Roy A, Emran TB, Cavalu S. Exploring the Journey of Zinc Oxide Nanoparticles (ZnO-NPs) toward Biomedical Applications. MATERIALS (BASEL, SWITZERLAND) 2022; 15:2160. [PMID: 35329610 PMCID: PMC8951444 DOI: 10.3390/ma15062160] [Citation(s) in RCA: 82] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 02/28/2022] [Accepted: 03/03/2022] [Indexed: 12/24/2022]
Abstract
The field of nanotechnology is concerned with the creation and application of materials having a nanoscale spatial dimensioning. Having a considerable surface area to volume ratio, nanoparticles have particularly unique properties. Several chemical and physical strategies have been used to prepare zinc oxide nanoparticles (ZnO-NPs). Still, biological methods using green or natural routes in various underlying substances (e.g., plant extracts, enzymes, and microorganisms) can be more environmentally friendly and cost-effective than chemical and/or physical methods in the long run. ZnO-NPs are now being studied as antibacterial agents in nanoscale and microscale formulations. The purpose of this study is to analyze the prevalent traditional method of generating ZnO-NPs, as well as its harmful side effects, and how it might be addressed utilizing an eco-friendly green approach. The study's primary focus is on the potential biomedical applications of green synthesized ZnO-NPs. Biocompatibility and biomedical qualities have been improved in green-synthesized ZnO-NPs over their traditionally produced counterparts, making them excellent antibacterial and cancer-fighting drugs. Additionally, these ZnO-NPs are beneficial when combined with the healing processes of wounds and biosensing components to trace small portions of biomarkers linked with various disorders. It has also been discovered that ZnO-NPs can distribute and sense drugs. Green-synthesized ZnO-NPs are compared to traditionally synthesized ones in this review, which shows that they have outstanding potential as a potent biological agent, as well as related hazardous properties.
Collapse
Affiliation(s)
- Fahadul Islam
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka 1207, Bangladesh; (F.I.); (M.R.I.); (A.A.)
| | - Sheikh Shohag
- Department of Biochemistry and Molecular Biology, Faculty of Life Science, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh; (S.S.); (M.J.U.)
| | - Md. Jalal Uddin
- Department of Biochemistry and Molecular Biology, Faculty of Life Science, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh; (S.S.); (M.J.U.)
| | - Md. Rezaul Islam
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka 1207, Bangladesh; (F.I.); (M.R.I.); (A.A.)
| | - Mohamed H. Nafady
- Faculty of Applied Health Science Technology, Misr University for Science and Technology, Giza 12568, Egypt;
| | - Aklima Akter
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka 1207, Bangladesh; (F.I.); (M.R.I.); (A.A.)
| | - Saikat Mitra
- Department of Pharmacy, Faculty of Pharmacy, University of Dhaka, Dhaka 1000, Bangladesh;
| | - Arpita Roy
- Department of Biotechnology, School of Engineering & Technology, Sharda University, Greater Noida 201310, India;
| | - Talha Bin Emran
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka 1207, Bangladesh; (F.I.); (M.R.I.); (A.A.)
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong 4381, Bangladesh
| | - Simona Cavalu
- Faculty of Medicine and Pharmacy, University of Oradea, 400087 Oradea, Romania
| |
Collapse
|
4
|
Qidwai A, Annu, Nabi B, Kotta S, Narang JK, Baboota S, Ali J. Role of nanocarriers in photodynamic therapy. Photodiagnosis Photodyn Ther 2020; 30:101782. [DOI: 10.1016/j.pdpdt.2020.101782] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 04/08/2020] [Accepted: 04/13/2020] [Indexed: 12/15/2022]
|
5
|
Zhu W, Hao L, Liu X, Borrás-Hidalgo O, Zhang Y. Enhanced anti-proliferative efficacy of epothilone B loaded with Escherichia coli Nissle 1917 bacterial ghosts on the HeLa cells by mitochondrial pathway of apoptosis. Drug Dev Ind Pharm 2018. [DOI: 10.1080/03639045.2018.1449855] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Wenxing Zhu
- Shandong Provincial Key Lab of Microbial Engineering, School of Bioengineering, Qilu University of Technology, Jinan, P. R. China
| | - Lujiang Hao
- Shandong Provincial Key Lab of Microbial Engineering, School of Bioengineering, Qilu University of Technology, Jinan, P. R. China
| | - Xinli Liu
- Shandong Provincial Key Lab of Microbial Engineering, School of Bioengineering, Qilu University of Technology, Jinan, P. R. China
| | - Orlando Borrás-Hidalgo
- Shandong Provincial Key Lab of Microbial Engineering, School of Bioengineering, Qilu University of Technology, Jinan, P. R. China
| | - Yuyu Zhang
- Shandong Key Lab of Animal Disease Control and Breeding, Shandong Academy of Agricultural Sciences, Jinan, P. R. China
| |
Collapse
|
6
|
Dissanayake S, Denny WA, Gamage S, Sarojini V. Recent developments in anticancer drug delivery using cell penetrating and tumor targeting peptides. J Control Release 2017; 250:62-76. [DOI: 10.1016/j.jconrel.2017.02.006] [Citation(s) in RCA: 175] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2016] [Revised: 02/02/2017] [Accepted: 02/02/2017] [Indexed: 12/13/2022]
|
7
|
Delforoush M, Strese S, Wickström M, Larsson R, Enblad G, Gullbo J. In vitro and in vivo activity of melflufen (J1)in lymphoma. BMC Cancer 2016; 16:263. [PMID: 27044263 PMCID: PMC4820867 DOI: 10.1186/s12885-016-2299-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Accepted: 03/31/2016] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Melphalan has been used in the treatment of various hematologic malignancies for almost 60 years. Today it is part of standard therapy for multiple myeloma and also as part of myeloablative regimens in association with autologous allogenic stem cell transplantation. Melflufen (melphalan flufenamide ethyl ester, previously called J1) is an optimized derivative of melphalan providing targeted delivery of active metabolites to cells expressing aminopeptidases. The activity of melflufen has compared favorably with that of melphalan in a series of in vitro and in vivo experiments performed preferentially on different solid tumor models and multiple myeloma. Melflufen is currently being evaluated in a clinical phase I/II trial in relapsed or relapsed and refractory multiple myeloma. METHODS Cytotoxicity of melflufen was assayed in lymphoma cell lines and in primary tumor cells with the Fluorometric Microculture Cytotoxicity Assay and cell cycle analyses was performed in two of the cell lines. Melflufen was also investigated in a xenograft model with subcutaneous lymphoma cells inoculated in mice. RESULTS Melflufen showed activity with cytotoxic IC50-values in the submicromolar range (0.011-0.92 μM) in the cell lines, corresponding to a mean of 49-fold superiority (p < 0.001) in potency vs. melphalan. In the primary cultures melflufen yielded slightly lower IC50-values (2.7 nM to 0.55 μM) and an increased ratio vs. melphalan (range 13-455, average 108, p < 0.001). Treated cell lines exhibited a clear accumulation in the G2/M-phase of the cell cycle. Melflufen also showed significant activity and no, or minimal side effects in the xenografted animals. CONCLUSION This study confirms previous reports of a targeting related potency superiority of melflufen compared to that of melphalan. Melflufen was active in cell lines and primary cultures of lymphoma cells, as well as in a xenograft model in mice and appears to be a candidate for further evaluation in the treatment of this group of malignant diseases.
Collapse
Affiliation(s)
- Maryam Delforoush
- />Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala, Sweden
| | - Sara Strese
- />Department of Medical Sciences, Section of Clinical Pharmacology, Uppsala University Hospital, Uppsala, Sweden
| | - Malin Wickström
- />Department of Medical Sciences, Section of Clinical Pharmacology, Uppsala University Hospital, Uppsala, Sweden
- />Department of Women’s and Children’s Health, Karolinska Institutet, Childhood Cancer Research Unit, Stockholm, Sweden
| | - Rolf Larsson
- />Department of Medical Sciences, Section of Clinical Pharmacology, Uppsala University Hospital, Uppsala, Sweden
| | - Gunilla Enblad
- />Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala, Sweden
| | - Joachim Gullbo
- />Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala, Sweden
- />Department of Medical Sciences, Section of Clinical Pharmacology, Uppsala University Hospital, Uppsala, Sweden
| |
Collapse
|
8
|
Govindaraju K, Krishnamoorthy K, Alsagaby SA, Singaravelu G, Premanathan M. Green synthesis of silver nanoparticles for selective toxicity towards cancer cells. IET Nanobiotechnol 2015; 9:325-30. [DOI: 10.1049/iet-nbt.2015.0001] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Kasivelu Govindaraju
- Nanoscience DivisionDepartment of ZoologyThiruvalluvar UniversityVellore632004TamilnaduIndia
- Nanoscience DivisionCentre for Ocean ResearchSathyabama UniversityChennai 600119India
| | - Karthikeyan Krishnamoorthy
- Nanomaterials and System LabDepartment of Mechanical EngineeringJeju National UniversityJeju 690‐456Republic of Korea
| | - Suliman A. Alsagaby
- Biosciences Research CentreDepartment of Medical LaboratoryCollege of ScienceMajmaah UniversityZulfi 11932Saudi Arabia
| | - Ganesan Singaravelu
- Nanoscience DivisionDepartment of ZoologyThiruvalluvar UniversityVellore632004TamilnaduIndia
| | - Mariappan Premanathan
- Biosciences Research CentreDepartment of Medical LaboratoryCollege of ScienceMajmaah UniversityZulfi 11932Saudi Arabia
| |
Collapse
|
9
|
|
10
|
Abstract
Whilst small interfering (si) RNAs have emerged as a promising therapeutic modality for treating a diversity of human diseases, delivery constitutes the most serious obstacle to siRNA drug development. As the most used delivery agents can enter all cell types, specificity must be built into the delivery agents or directly attached to the siRNA molecules. The use of antibodies, peptides, Peptide-Fc fusions, aptamers, and other targeting ligands has now enabled efficient gene silencing in the desired cell populations/tissues in vitro and in vivo. The present review summarizes these current innovations, which are important for the design of safe therapeutic siRNAs.
Collapse
Affiliation(s)
- Mouldy Sioud
- Departments of Immunology, Institute for Cancer Research, The Norwegian Radium Hospital, Radiumhospitalet-Rikshospitalet University Hospital, Ullernchausseen 70, Montebello, 310, Oslo, Norway,
| |
Collapse
|
11
|
Kaittanis C, Shaffer TM, Ogirala A, Santra S, Perez JM, Chiosis G, Li Y, Josephson L, Grimm J. Environment-responsive nanophores for therapy and treatment monitoring via molecular MRI quenching. Nat Commun 2014; 5:3384. [PMID: 24594970 PMCID: PMC4108301 DOI: 10.1038/ncomms4384] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2013] [Accepted: 02/05/2014] [Indexed: 11/25/2022] Open
Abstract
The effective delivery of therapeutics to disease sites significantly contributes to drug efficacy, toxicity and clearance. Here we demonstrate that clinically approved iron oxide nanoparticles (Ferumoxytol) can be utilized to carry one or multiple drugs. These so called ‘nanophores’ retain their cargo within their polymeric coating through weak electrostatic interactions and release it in slightly acidic conditions (pH 6.8 and below). The loading of drugs increases the nanophores’ transverse T2 and longitudinal T1 NMR proton relaxation times, which is proportional to amount of carried cargo. Chemotherapy with translational nanophores is more effective than the free drug in vitro and in vivo, without subjecting the drugs or the carrier nanoparticle to any chemical modification. Evaluation of cargo incorporation and payload levels in vitro and in vivo can be assessed via benchtop magnetic relaxometers, common NMR instruments or MRI scanners.
Collapse
Affiliation(s)
- Charalambos Kaittanis
- Molecular Pharmacology and Chemistry Program, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, New York 10065, USA
| | - Travis M Shaffer
- 1] Molecular Pharmacology and Chemistry Program, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, New York 10065, USA [2] Department of Chemistry, Hunter College of the City University of New York, Graduate Center, New York, New York 10065, USA
| | - Anuja Ogirala
- Molecular Pharmacology and Chemistry Program, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, New York 10065, USA
| | - Santimukul Santra
- Department of Chemistry, Pittsburg State University, 1701 S Broadway Street, Pittsburg, Kansas 66762, USA
| | - J Manuel Perez
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, Florida 32826, USA
| | - Gabriela Chiosis
- Molecular Pharmacology and Chemistry Program, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, New York 10065, USA
| | - Yueming Li
- Molecular Pharmacology and Chemistry Program, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, New York 10065, USA
| | - Lee Josephson
- Center for Advanced Medical Imaging Sciences, Massachusetts General Hospital, Building 149, 13th Street, Charlestown, Massachusetts 02129, USA
| | - Jan Grimm
- Molecular Pharmacology and Chemistry Program, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, New York 10065, USA
| |
Collapse
|
12
|
Huhtala T, Weisell J, Rytkönen J, Närvänen A. Peptide labelling strategies for imaging agents. Methods Mol Biol 2014; 1088:171-183. [PMID: 24146404 DOI: 10.1007/978-1-62703-673-3_12] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
The main goal in modern biomedicine is to develop specific diagnostic and therapeutic agents for different diseases. Especially in cancer research tumor targeted molecules are the key factor in the development of new anti-tumor drugs. In addition, the early diagnosis of the disease is an important factor for a successful therapy. Synthetic peptides have been shown to be specific targeting agents for next generation diagnostic and therapeutic agents. Noninvasive in vivo imaging using targeting molecules provides modern method for the diagnosis of the pathological alterations like cancer. To evaluate the usefulness of a synthetic peptide for in vivo diagnostic purposes the preclinical biodistribution and targeting studies are essential. Today the widely used preclinical imaging modalities for the biodistribution and tissue alteration studies in experimental animals are single photon emission computed tomography (SPECT) and magnetic resonance imaging (MRI). Together with conventional histochemistry, the biodistribution and tissue/cell location can be determined. In this chapter we describe the conjugation and labelling methods of the peptides for histochemistry and for the molecular imaging with SPECT and MRI modalities.
Collapse
Affiliation(s)
- Tuulia Huhtala
- Department of Biotechnology and Molecular Medicine, University of Eastern Finland, Kuopio, Finland
| | | | | | | |
Collapse
|
13
|
Saxena R, Nanjan MJ. Elastin-like polypeptides and their applications in anticancer drug delivery systems: a review. Drug Deliv 2013; 22:156-67. [PMID: 24215207 DOI: 10.3109/10717544.2013.853210] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Elastin-like polypeptides (ELPs) are large molecular weight biopolymers. They have been widely studied as macromolecular carriers for targeted delivery of drugs. The aim of the present article is to review the available information on ELPs (including our recent investigations), their properties, drug delivery applications to tumor sites and future perspectives. This review also provides information on the use of short synthetic ELPs for making ELP-drug conjugates, for targeted delivery of anticancer drugs. In the present review we also focus on the point that short ELPs can also be used for targeting anticancer drugs to tumor sites as they behave similar to long ELPs regarding their capacity to undergo inverse temperature transition (ITT) behavior.
Collapse
Affiliation(s)
- Rubha Saxena
- TIFAC CORE HD, J.S.S. College of Pharmacy (Off Campus, JSS University, Mysore) , Ootacamund, Tamil Nadu , India
| | | |
Collapse
|
14
|
Strese S, Wickström M, Fuchs PF, Fryknäs M, Gerwins P, Dale T, Larsson R, Gullbo J. The novel alkylating prodrug melflufen (J1) inhibits angiogenesis in vitro and in vivo. Biochem Pharmacol 2013; 86:888-95. [PMID: 23933387 DOI: 10.1016/j.bcp.2013.07.026] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Revised: 07/25/2013] [Accepted: 07/25/2013] [Indexed: 10/26/2022]
Abstract
Aminopeptidase N (APN) has been reported to have a functional role in tumor angiogenesis and repeatedly reported to be over-expressed in human tumors. The melphalan-derived prodrug melphalan-flufenamide (melflufen, previously designated J1) can be activated by APN. This suggests that this alkylating prodrug may exert anti-angiogenic properties, which will possibly contribute to the anti-tumoral activity in vivo. This work presents a series of experiments designed to investigate this effect of melflufen. In a cytotoxicity assay we show that bovine endothelial cells were more than 200 times more sensitive to melflufen than to melphalan, in HUVEC cells the difference was more than 30-fold and accompanied by aminopetidase-mediated accumulation of intracellular melphalan. In the chicken embryo chorioallantoic membrane (CAM) assay it is indicated that both melflufen and melphalan inhibit vessel ingrowth. Two commercially available assays with human endothelial cells co-cultured with fibroblasts (TCS Cellworks AngioKit, and Essen GFP-AngioKit) also illustrate the superior anti-angiogenic effect of melflufen compared to melphalan. Finally, in a commercially available in vivo assay in mice (Cultrex DIVAA angio-reactor assay) melflufen displayed an anti-angiogenic effect, comparable to bevacizumab. In conclusion, this study demonstrates through all methods used, that melphalan-flufenamide besides being an alkylating agent also reveals anti-angiogenic effects in different preclinical models in vitro and in vivo.
Collapse
Affiliation(s)
- Sara Strese
- Clinical Pharmacology, Institution of Medical Sciences, Uppsala University, Akademiska Sjukhuset, 751 85 Uppsala, Sweden
| | | | | | | | | | | | | | | |
Collapse
|
15
|
Selim A, Al-Sunaidi A, Tabet N. Effect of the surface texture and crystallinity of ZnO nanoparticles on their toxicity. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2012. [DOI: 10.1016/j.msec.2012.07.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
16
|
Analogue-based drug discovery: Contributions to medicinal chemistry principles and drug design strategies. Microtubule stabilizers as a case in point (Special Topic Article). PURE APPL CHEM 2012. [DOI: 10.1351/pac-con-12-02-13] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The benefits of utilizing marketed drugs as starting points to discover new therapeutic agents have been well documented within the IUPAC series of books that bear the title Analogue-based Drug Discovery (ABDD). Not as clearly demonstrated, however, is that ABDD also contributes to the elaboration of new basic principles and alternative drug design strategies that are useful to the field of medicinal chemistry in general. After reviewing the ABDD programs that have evolved around the area of microtubule-stabilizing chemo-therapeutic agents, the present article delineates the associated research activities that additionally contributed to general strategies that can be useful for prodrug design, identifying pharmacophores, circumventing multidrug resistance (MDR), and achieving targeted drug distribution.
Collapse
|
17
|
Bajjuri KM, Liu Y, Liu C, Sinha SC. The legumain protease-activated auristatin prodrugs suppress tumor growth and metastasis without toxicity. ChemMedChem 2011; 6:54-9. [PMID: 21154805 DOI: 10.1002/cmdc.201000478] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Krishna Mohan Bajjuri
- Department of Molecular Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | | | | | | |
Collapse
|
18
|
Hu L, Wu X, Han J, Chen L, Vass SO, Browne P, Hall BS, Bot C, Gobalakrishnapillai V, Searle PF, Knox RJ, Wilkinson SR. Synthesis and structure-activity relationships of nitrobenzyl phosphoramide mustards as nitroreductase-activated prodrugs. Bioorg Med Chem Lett 2011; 21:3986-91. [PMID: 21620697 DOI: 10.1016/j.bmcl.2011.05.009] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2011] [Revised: 04/28/2011] [Accepted: 05/02/2011] [Indexed: 10/18/2022]
Abstract
A series of nitrobenzyl phosphoramide mustards and their analogs was designed and synthesized to explore their structure-activity relationships as substrates of nitroreductases from Escherichia coli and trypanosomes and as potential antiproliferative and antiparasitic agents. The position of the nitro group on the phenyl ring was important with the 4-nitrobenzyl phosphoramide mustard (1) offering the best combination of enzyme activity and antiproliferative effect against both mammalian and trypanosomatid cells. A preference was observed for halogen substitutions ortho to benzyl phosphoramide mustard but distinct differences were found in their SAR of substituted 4-nitrobenzyl phosphoramide mustards in E. coli nitroreductase-expressing cells and in trypanosomatids expressing endogenous nitroreductases.
Collapse
Affiliation(s)
- Longqin Hu
- Department of Medicinal Chemistry, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Preparation and characterization of polymeric pH-sensitive STEALTH® nanoparticles for tumor delivery of a lipophilic prodrug of paclitaxel. Int J Pharm 2011; 408:208-12. [DOI: 10.1016/j.ijpharm.2011.01.061] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2010] [Revised: 01/27/2011] [Accepted: 01/31/2011] [Indexed: 11/30/2022]
|
20
|
Premanathan M, Karthikeyan K, Jeyasubramanian K, Manivannan G. Selective toxicity of ZnO nanoparticles toward Gram-positive bacteria and cancer cells by apoptosis through lipid peroxidation. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2010; 7:184-92. [PMID: 21034861 DOI: 10.1016/j.nano.2010.10.001] [Citation(s) in RCA: 569] [Impact Index Per Article: 37.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2010] [Revised: 10/02/2010] [Accepted: 10/03/2010] [Indexed: 10/18/2022]
Abstract
UNLABELLED Nanoparticles are increasingly recognized for their utility in biological applications including nanomedicine. The present study investigated the toxicity of zinc oxide (ZnO) nanoparticles toward prokaryotic and eukaryotic cells. Cytotoxicity of ZnO to mammalian cells was studied using human myeloblastic leukemia cells (HL60) and normal peripheral blood mononuclear cells (PBMCs). Antibacterial activity of ZnO was also tested against the Gram-negative bacteria Escherichia coli and Pseudomonas aeruginosa, as well as the Gram-positive bacterium Staphylococcus aureus, and the effect was more pronounced with the Gram-positive than the Gram-negative bacteria. ZnO nanoparticles exhibited a preferential ability to kill cancerous HL60 cells as compared with normal PBMCs. The nanoparticles enhanced ultrasound-induced lipid peroxidation in the liposomal membrane. The work suggested two mechanisms underlying the toxicity of ZnO: (i) involvement of the generation of reactive oxygen species (ROS) and (ii) induction of apoptosis. The work also revealed potential utility of ZnO nanoparticles in the treatment of cancer, for their selective toxicity to cancer cells. FROM THE CLINICAL EDITOR The toxicity of zinc oxide to bacteria was related to the generation of reactive oxygen species and to the induction of apoptosis. Interestingly, these effects were differentially greater in human myeloblastic leukemia cells (HL60) than normal peripheral blood mononuclear cells.
Collapse
Affiliation(s)
- Mariappan Premanathan
- Department of Biotechnology, Mepco Schlenk Engineering College, Sivakasi, Tamil Nadu, India.
| | | | | | | |
Collapse
|
21
|
Wu Z, Shah A, Patel N, Yuan X. Development of methotrexate proline prodrug to overcome resistance by MDA-MB-231 cells. Bioorg Med Chem Lett 2010; 20:5108-12. [DOI: 10.1016/j.bmcl.2010.07.024] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2010] [Revised: 07/06/2010] [Accepted: 07/07/2010] [Indexed: 11/26/2022]
|
22
|
The alkylating prodrug J1 can be activated by aminopeptidase N, leading to a possible target directed release of melphalan. Biochem Pharmacol 2010; 79:1281-90. [PMID: 20067771 DOI: 10.1016/j.bcp.2009.12.022] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2009] [Revised: 12/23/2009] [Accepted: 12/29/2009] [Indexed: 01/09/2023]
Abstract
The alkylating prodrug of melphalan, J1 (melphalanyl-L-p-fluorophenylalanyl ethyl ester) is currently in early clinical trials. Preclinical studies have shown that J1-mediated cytotoxicity is dependent on hydrolytic activity of tumor cells. In this report we have analyzed potential peptidases and esterases of importance for release of free melphalan from J1. Exposure of tumor cell lines to J1 resulted in a significant increased level of free intracellular melphalan, at least tenfold at C(max), compared to exposure to melphalan at the same molar concentration. This efficient intracellular delivery could be inhibited in both magnitude and in time by bestatin, a broad spectrum inhibitor of the aminopeptidases, including the metalloproteinase aminopeptidase N (APN, EC 3.4.11.2.), and ebelactone A, an esterase inhibitor. These effects resulted, as expected, in decreased cytotoxic effects of J1. A specific role of APN in hydrolyzing J1 releasing free melphalan was demonstrated in vitro with pure APN enzyme. By using plasmid-based overexpression of APN or down regulation of endogenous APN with siRNA in different tumor cell lines we here confirm the involvement of APN in J1-mediated cytotoxic and apoptotic signaling. In conclusion, this study demonstrates a role of APN in the activation of the melphalan prodrug J1 and subsequently, its cytotoxicity. Given that APN is shown to be overexpressed in several solid tumors our data suggest that J1 may be activated in a tumor selective manner.
Collapse
|
23
|
Abstract
Whilst significant advances have been made in the delivery of nucleic acids to mammalian cells, most of the used strategies do not distinguish between normal and cancer cells. The same challenge is also facing radioactive- and chemo-therapies which are highly toxic and poorly tolerated due to limited tumor specificity. Regardless of the nature of the drug, there is a need for developing a technology platform which targets drugs only to tumors cells, leaving normal cells undamaged. Among the targeting strategies, receptor-targeted delivery provides an innovative strategy to selectively direct therapeutics to cancer cells. Receptor-binding ligands (e.g., peptides, antibodies, aptamers) can be incorporated into gene delivery vesicles or directly conjugated to siRNA in the hope in promoting their localization in target cell expressing the cognate receptors. The present chapter discusses the current progress made in the specific delivery of siRNAs.
Collapse
Affiliation(s)
- Mouldy Sioud
- Department of Immunology, Institute for Cancer Research, Radiumhospitalet-Rikshopitalet Universtity Hospital, Oslo, Norway
| |
Collapse
|
24
|
|
25
|
Abstract
Anticancer prodrugs designed to target specifically tumor cells should increase therapeutic effectiveness and decrease systemic side effects in the treatment of cancer. Over the last 20 years, significant advances have been made in the development of anticancer prodrugs through the incorporation of triggers for reductive activation. Reductively activated prodrugs have been designed to target hypoxic tumor tissues, which are known to overexpress several endogenous reductive enzymes. In addition, exogenous reductive enzymes can be delivered to tumor cells through fusion with tumor-specific antibodies or overexpressed in tumor cells through gene delivery approaches. Many anticancer prodrugs have been designed to use both the endogenous and exogenous reductive enzymes for target-specific activation and these prodrugs often contain functional groups such as quinones, nitroaromatics, N-oxides, and metal complexes. Although no new agents have been approved for clinical use, several reductively activated prodrugs are in various stages of clinical trial. This review mainly focuses on the medicinal chemistry aspects of various classes of reductively activated prodrugs including design principles, structure-activity relationships, and mechanisms of activation and release of active drug molecules.
Collapse
Affiliation(s)
- Yu Chen
- Department of Pharmaceutical Chemistry, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, 160 Frelinghuysen Road, Piscataway, NJ 08854, USA
| | | |
Collapse
|
26
|
Doxorubicin–polyphosphazene conjugate hydrogels for locally controlled delivery of cancer therapeutics. Biomaterials 2009; 30:4752-62. [DOI: 10.1016/j.biomaterials.2009.05.031] [Citation(s) in RCA: 102] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2009] [Accepted: 05/15/2009] [Indexed: 11/20/2022]
|
27
|
Juan TY, Roffler SR, Hou HS, Huang SM, Chen KC, Leu YL, Prijovich ZM, Yu CP, Wu CC, Sun GH, Cha TL. Antiangiogenesis targeting tumor microenvironment synergizes glucuronide prodrug antitumor activity. Clin Cancer Res 2009; 15:4600-11. [PMID: 19584154 DOI: 10.1158/1078-0432.ccr-09-0090] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE This study is aimed at investigating the in vivo antitumor activity of a novel cell-impermeable glucuronide prodrug, 9-aminocamptothecin glucuronide (9ACG), and elucidating the synergistically antitumor effects of antiangiogenesis therapy by targeting the tumor microenvironment. EXPERIMENTAL DESIGN We analyzed the antitumor effects of 9ACG alone or combined with antiangiogenic monoclonal antibody DC101 on human tumor xenografts by measuring tumor growth and mouse survival in BALB/c nu/nu nude and NOD/SCID mice. The drug delivery, immune response, and angiogenesis status in treated tumors were assessed by high performance liquid chromatography, immunohistochemistry, and immunofluorescence assays. RESULTS We developed a nontoxic and cell-impermeable glucuronide prodrug, 9ACG, which can only be activated by extracellular beta-glucuronidase to become severely toxic. 9ACG possesses potent antitumor activity against human tumor xenografts in BALB/c nu/nu nude mice but not for tumors implanted in NOD/SCID mice deficient in macrophages and neutrophils, suggesting that these cells play an important role in activating 9ACG in the tumor microenvironment. Most importantly, antiangiogenic monoclonal antibody DC101 potentiated single-dose 9ACG antitumor activity and prolonged survival of mice bearing resistant human colon tumor xenografts by providing strong beta-glucuronidase activity and prodrug delivery through enhancing inflammatory cell infiltration and normalizing tumor vessels in the tumor microenvironment. We also show that inflammatory cells (neutrophils) were highly infiltrated in advanced human colon cancer tissues compared with normal counterparts. CONCLUSIONS Our study provides in vivo evidence that 9ACG has potential for prodrug monotherapy or in combination with antiangiognesis treatment for tumors with infiltration of macrophage or neutrophil inflammatory cells.
Collapse
Affiliation(s)
- Ting-Yi Juan
- Divisions of Urology, Graduate Institute of Life Sciences, Institute of Biomedical Sciences, Academia Sinica, National Yang-Ming University,Taipei,Taiwan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Chun C, Lee SM, Kim SY, Yang HK, Song SC. Thermosensitive poly(organophosphazene)–paclitaxel conjugate gels for antitumor applications. Biomaterials 2009; 30:2349-60. [DOI: 10.1016/j.biomaterials.2008.12.083] [Citation(s) in RCA: 97] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2008] [Accepted: 12/29/2008] [Indexed: 10/21/2022]
|
29
|
Abstract
RNA interference (RNAi) is a natural mechanism for gene silencing that can be harnessed for the development of RNA-based drugs. Although synthetic small interfering RNA (siRNAs) can be delivered in vitro to virtually all cell types using lipid-based transfection agents or electroporation, efficient strategies for achieving either systemic or targeted delivery remains one of the major in vivo challenges. Among the targeting strategies, receptor-targeted delivery provides an innovative strategy to selectively direct therapeutics to cancer cells. Receptor-binding peptides can be incorporated into gene-delivery vesicles or directly conjugated to siRNAs in the hope of promoting their localization in target cells expressing the cognate receptors. This chapter discusses the current status of siRNA-targeting strategies using either peptides identified through iterative screening of random peptide phage libraries or naturally occurring peptides. Also, transcriptional targeting strategies and detailed protocols for the selection of cancer cell-binding peptide from random peptide libraries are described.
Collapse
|
30
|
Samor C, Guerrini A, Varchi G, Beretta GL, Fontana G, Bombardelli E, Carenini N, Zunino F, Bertucci C, Fiori J, Battaglia A. The Role of Polyamine Architecture on the Pharmacological Activity of Open Lactone Camptothecin−Polyamine Conjugates. Bioconjug Chem 2008; 19:2270-9. [DOI: 10.1021/bc800033r] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Affiliation(s)
- Cristian Samor
- Istituto CNR per la Sintesi Organica e Fotoreattività “I.S.O.F.”, Area della Ricerca di Bologna, Via Gobetti 101, 40129 Bologna, Italy, Dipartimento di Oncologia Sperimentale e Laboratori, Fondazione IRCCS Istituto Nazionale Tumori, 20133 Milano, Italy, Indena SPA, viale Ortles 12, 20139 Milano, Italy, and Dipartimento di Scienze Farmaceutiche, Università di Bologna, Via Belmeloro 6, 40126 Bologna, Italy
| | - Andrea Guerrini
- Istituto CNR per la Sintesi Organica e Fotoreattività “I.S.O.F.”, Area della Ricerca di Bologna, Via Gobetti 101, 40129 Bologna, Italy, Dipartimento di Oncologia Sperimentale e Laboratori, Fondazione IRCCS Istituto Nazionale Tumori, 20133 Milano, Italy, Indena SPA, viale Ortles 12, 20139 Milano, Italy, and Dipartimento di Scienze Farmaceutiche, Università di Bologna, Via Belmeloro 6, 40126 Bologna, Italy
| | - Greta Varchi
- Istituto CNR per la Sintesi Organica e Fotoreattività “I.S.O.F.”, Area della Ricerca di Bologna, Via Gobetti 101, 40129 Bologna, Italy, Dipartimento di Oncologia Sperimentale e Laboratori, Fondazione IRCCS Istituto Nazionale Tumori, 20133 Milano, Italy, Indena SPA, viale Ortles 12, 20139 Milano, Italy, and Dipartimento di Scienze Farmaceutiche, Università di Bologna, Via Belmeloro 6, 40126 Bologna, Italy
| | - Giovanni Luca Beretta
- Istituto CNR per la Sintesi Organica e Fotoreattività “I.S.O.F.”, Area della Ricerca di Bologna, Via Gobetti 101, 40129 Bologna, Italy, Dipartimento di Oncologia Sperimentale e Laboratori, Fondazione IRCCS Istituto Nazionale Tumori, 20133 Milano, Italy, Indena SPA, viale Ortles 12, 20139 Milano, Italy, and Dipartimento di Scienze Farmaceutiche, Università di Bologna, Via Belmeloro 6, 40126 Bologna, Italy
| | - Gabriele Fontana
- Istituto CNR per la Sintesi Organica e Fotoreattività “I.S.O.F.”, Area della Ricerca di Bologna, Via Gobetti 101, 40129 Bologna, Italy, Dipartimento di Oncologia Sperimentale e Laboratori, Fondazione IRCCS Istituto Nazionale Tumori, 20133 Milano, Italy, Indena SPA, viale Ortles 12, 20139 Milano, Italy, and Dipartimento di Scienze Farmaceutiche, Università di Bologna, Via Belmeloro 6, 40126 Bologna, Italy
| | - Ezio Bombardelli
- Istituto CNR per la Sintesi Organica e Fotoreattività “I.S.O.F.”, Area della Ricerca di Bologna, Via Gobetti 101, 40129 Bologna, Italy, Dipartimento di Oncologia Sperimentale e Laboratori, Fondazione IRCCS Istituto Nazionale Tumori, 20133 Milano, Italy, Indena SPA, viale Ortles 12, 20139 Milano, Italy, and Dipartimento di Scienze Farmaceutiche, Università di Bologna, Via Belmeloro 6, 40126 Bologna, Italy
| | - Nives Carenini
- Istituto CNR per la Sintesi Organica e Fotoreattività “I.S.O.F.”, Area della Ricerca di Bologna, Via Gobetti 101, 40129 Bologna, Italy, Dipartimento di Oncologia Sperimentale e Laboratori, Fondazione IRCCS Istituto Nazionale Tumori, 20133 Milano, Italy, Indena SPA, viale Ortles 12, 20139 Milano, Italy, and Dipartimento di Scienze Farmaceutiche, Università di Bologna, Via Belmeloro 6, 40126 Bologna, Italy
| | - Franco Zunino
- Istituto CNR per la Sintesi Organica e Fotoreattività “I.S.O.F.”, Area della Ricerca di Bologna, Via Gobetti 101, 40129 Bologna, Italy, Dipartimento di Oncologia Sperimentale e Laboratori, Fondazione IRCCS Istituto Nazionale Tumori, 20133 Milano, Italy, Indena SPA, viale Ortles 12, 20139 Milano, Italy, and Dipartimento di Scienze Farmaceutiche, Università di Bologna, Via Belmeloro 6, 40126 Bologna, Italy
| | - Carlo Bertucci
- Istituto CNR per la Sintesi Organica e Fotoreattività “I.S.O.F.”, Area della Ricerca di Bologna, Via Gobetti 101, 40129 Bologna, Italy, Dipartimento di Oncologia Sperimentale e Laboratori, Fondazione IRCCS Istituto Nazionale Tumori, 20133 Milano, Italy, Indena SPA, viale Ortles 12, 20139 Milano, Italy, and Dipartimento di Scienze Farmaceutiche, Università di Bologna, Via Belmeloro 6, 40126 Bologna, Italy
| | - Jessica Fiori
- Istituto CNR per la Sintesi Organica e Fotoreattività “I.S.O.F.”, Area della Ricerca di Bologna, Via Gobetti 101, 40129 Bologna, Italy, Dipartimento di Oncologia Sperimentale e Laboratori, Fondazione IRCCS Istituto Nazionale Tumori, 20133 Milano, Italy, Indena SPA, viale Ortles 12, 20139 Milano, Italy, and Dipartimento di Scienze Farmaceutiche, Università di Bologna, Via Belmeloro 6, 40126 Bologna, Italy
| | - Arturo Battaglia
- Istituto CNR per la Sintesi Organica e Fotoreattività “I.S.O.F.”, Area della Ricerca di Bologna, Via Gobetti 101, 40129 Bologna, Italy, Dipartimento di Oncologia Sperimentale e Laboratori, Fondazione IRCCS Istituto Nazionale Tumori, 20133 Milano, Italy, Indena SPA, viale Ortles 12, 20139 Milano, Italy, and Dipartimento di Scienze Farmaceutiche, Università di Bologna, Via Belmeloro 6, 40126 Bologna, Italy
| |
Collapse
|
31
|
Hanley C, Layne J, Punnoose A, Reddy KM, Coombs I, Coombs A, Feris K, Wingett D. Preferential killing of cancer cells and activated human T cells using ZnO nanoparticles. NANOTECHNOLOGY 2008; 19:295103. [PMID: 18836572 PMCID: PMC2558672 DOI: 10.1088/0957-4484/19/29/295103] [Citation(s) in RCA: 409] [Impact Index Per Article: 24.1] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/14/2023]
Abstract
Nanoparticles are increasingly being recognized for their potential utility in biological applications including nanomedicine. Here we examine the response of normal human cells to ZnO nanoparticles under different signaling environments and compare it to the response of cancerous cells. ZnO nanoparticles exhibit a strong preferential ability to kill cancerous T cells ( approximately 28-35x) compared to normal cells. Interestingly, the activation state of the cell contributes toward nanoparticle toxicity, as resting T cells display a relative resistance while cells stimulated through the T cell receptor and CD28 costimulatory pathway show greater toxicity in direct relation to the level of activation. Mechanisms of toxicity appear to involve the generation of reactive oxygen species, with cancerous T cells producing higher inducible levels than normal T cells. In addition, nanoparticles were found to induce apoptosis and the inhibition of reactive oxygen species was found to be protective against nanoparticle induced cell death. The novel findings of cell selective toxicity, towards potential disease causing cells, indicate a potential utility of ZnO nanoparticles in the treatment of cancer and/or autoimmunity.
Collapse
Affiliation(s)
- Cory Hanley
- Department of Biological Sciences, Boise State University, Boise, ID 83725, USA
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Vicent MJ, Dieudonné L, Carbajo RJ, Pineda-Lucena A. Polymer conjugates as therapeutics: future trends, challenges and opportunities. Expert Opin Drug Deliv 2008; 5:593-614. [DOI: 10.1517/17425247.5.5.593] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
33
|
Yu L, Zhang ZF, Jing CX, Wu FL. Intraperitoneal administration of gonadotropin-releasing hormone-PE40 induces castration in male rats. World J Gastroenterol 2008; 14:2106-9. [PMID: 18395915 PMCID: PMC2701535 DOI: 10.3748/wjg.14.2106] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To evaluate the long-term effects of gonadotropin-releasing hormone (GnRH)-based vaccine on levels of GnRH antibody and testosterone, and vaccine-induced immunocastration on sexual behavior of male rats.
METHODS: The rats were treated with GnRH-PE40 intraperitoneally every other day for 12 wk. GnRH antibody and testosterone level in rat blood were determined by ELISA and radioimmunoassay, respectively. Morphological changes in testes and sexual behavior of rats were evaluated.
RESULTS: GnRH-PE40 induced a high production in GnRH antibody, decreased the serum testosterone level, testis atrophy and sexual function in rats.
CONCLUSION: Intraperitoneal administration of GnRH-PE40 produces structural and functional castration of male rat reproductive system by inducing anti-GnRH antibody.
Collapse
|
34
|
Atkinson JM, Siller CS, Gill JH. Tumour endoproteases: the cutting edge of cancer drug delivery? Br J Pharmacol 2008; 153:1344-52. [PMID: 18204490 PMCID: PMC2437906 DOI: 10.1038/sj.bjp.0707657] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2007] [Revised: 11/09/2007] [Accepted: 11/27/2007] [Indexed: 01/06/2023] Open
Abstract
Despite progression in anticancer drug development and improvements in the clinical utilization of therapies, current treatment regimes are still dependent upon the use of systemic antiproliferative cytotoxic agents. Although these agents are unquestionably potent, their efficacy is limited by toxicity towards 'normal' cells and a lack of tumour selective targeting, resulting in a therapeutic index which is modest at best. Consequently, the development of more tumour selective cancer treatments, with better discrimination between tumour and normal cells is unequivocally an important goal for cancer drug discovery. One such strategy is to exploit the tumour phenotype as a mechanism for tumour-selective delivery of potent therapeutics. An exciting approach in this area is to develop anticancer therapeutics as prodrugs, which are non-toxic until activated by enzymes localized specifically in the tumour. Enzymes suitable for tumour-activated prodrug development must have increased activity in the tumour relative to non-diseased tissue and an ability to activate the prodrug to its active form. One class of enzyme satisfying these criteria are the tumour endoproteases, particularly the serine- and metallo-proteases. These proteolytic enzymes are essential for tumour angiogenesis, invasion and metastasis, the major defining features of malignancy. This review describes the concept behind development of tumour-endoprotease activated prodrugs and discusses the various studies to date that have demonstrated the huge potential of this approach for improvement of cancer therapy.
Collapse
Affiliation(s)
- J M Atkinson
- Institute of Cancer Therapeutics, University of Bradford Bradford, UK
| | - C S Siller
- Institute of Cancer Therapeutics, University of Bradford Bradford, UK
| | - J H Gill
- Institute of Cancer Therapeutics, University of Bradford Bradford, UK
| |
Collapse
|
35
|
Hwang J, Li RC, Maynard HD. Well-defined polymers with activated ester and protected aldehyde side chains for bio-functionalization. J Control Release 2007; 122:279-86. [PMID: 17599628 DOI: 10.1016/j.jconrel.2007.04.010] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2007] [Revised: 04/04/2007] [Accepted: 04/09/2007] [Indexed: 10/23/2022]
Abstract
Polymers with reactive side chains and narrow molecular weight distributions were synthesized by reversible addition-fragmentation chain transfer (RAFT) polymerization, and the potential to utilize these polymers to prepare drug carriers was demonstrated. p-Nitrophenyl methacrylate (NPMA) and diethoxypropyl methacrylate (DEPMA) were polymerized utilizing cumyl dithiobenzoate (CDB) as the chain transfer agent and azobisisobutyronitrile (AIBN) as the initiator to high conversions (> or = 86%). The resulting pNPMA and pDEPMA had narrow molecular weight distributions (polydispersity indices < 1.3). The ability to functionalize these polymers was confirmed. For pNPMA, up to 86% of the side chains were substituted with the amino acid, glycine methyl ester. The side chains of pDEPMA were hydrolyzed to aldehydes and reaction with O-benzylhydroxylamine and O-methylhydroxylamine to form stable oxime bond conjugates was demonstrated. The percent substitution depended on the feed ratios. Conjugation of an aminooxy-functionalized RGD peptide was also demonstrated.
Collapse
Affiliation(s)
- Jungyeon Hwang
- Department of Chemistry and Biochemistry and California NanoSystems Institute, University of California, Los Angeles, 607 Charles E. Young Drive East, Los Angeles, CA 90095-1569, USA
| | | | | |
Collapse
|
36
|
Mittal S, Song X, Vig BS, Amidon GL. Proline Prodrug of Melphalan Targeted to Prolidase, a Prodrug Activating Enzyme Overexpressed in Melanoma. Pharm Res 2007; 24:1290-8. [PMID: 17377743 DOI: 10.1007/s11095-007-9249-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2006] [Accepted: 01/22/2007] [Indexed: 10/23/2022]
Abstract
PURPOSE To determine the bioactivation and uptake of prolidase-targeted proline prodrugs of melphalan in six cancer cell lines with variable prolidase expression and to evaluate prolidase-dependence of prodrug cytotoxicity in the cell lines compared to that of the parent drug, melphalan. MATERIALS AND METHODS Hydrolysis, cell uptake, and cell proliferation studies of melphalan and the L: - and D: -proline prodrugs of melphalan, prophalan-L: and prophalan-D: , respectively, were conducted in the cancer cell lines using established procedures. RESULTS The bioactivation of prophalan-L: in the cancer cell lines exhibited high correlation with their prolidase expression levels (r (2) = 0.86). There were no significant differences in uptake of melphalan and its prodrugs. The cytotoxicity of prophalan-L: (GI(50)) in cancer cells also showed high correlation with prolidase expression (r (2) = 0.88), while prophalan-D: was ineffective at comparable concentrations. A prolidase targeting index (ratio of melphalan to prophalan-L: cytotoxicity normalized to their uptake) was computed and showed high correlation with prolidase expression (r (2) = 0.82). CONCLUSIONS The data corroborates the specificity of prophalan-L: activation by prolidase as well as prolidase-targeted cytotoxicity of prophalan-L: in cancer cell lines. Hence, prophalan-L: , a stable prodrug of melphalan, exhibits potential for efficiently targeting melanoma with reduced systemic toxicity.
Collapse
Affiliation(s)
- Sachin Mittal
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA
| | | | | | | |
Collapse
|
37
|
Skwarczynski M, Hayashi Y, Kiso Y. Paclitaxel Prodrugs: Toward Smarter Delivery of Anticancer Agents. J Med Chem 2006; 49:7253-69. [PMID: 17149855 DOI: 10.1021/jm0602155] [Citation(s) in RCA: 142] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Mariusz Skwarczynski
- Department of Medicinal Chemistry, Center for Frontier Research in Medicinal Science, 21st Century COE Program, Kyoto Pharmaceutical University, Yamashina-ku, Kyoto, 607-8412, Japan
| | | | | |
Collapse
|
38
|
Skwarczynski M, Noguchi M, Hirota S, Sohma Y, Kimura T, Hayashi Y, Kiso Y. Development of first photoresponsive prodrug of paclitaxel. Bioorg Med Chem Lett 2006; 16:4492-6. [PMID: 16806915 DOI: 10.1016/j.bmcl.2006.06.030] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2006] [Accepted: 06/09/2006] [Indexed: 10/24/2022]
Abstract
A prodrug of paclitaxel which has a coumarin derivative conjugated to the amino acid moiety of isotaxel (O-acyl isoform of paclitaxel) has been synthesized. The prodrug was selectively converted to isotaxel by visible light irradiation (430 nm) with the cleavage of coumarin. Finally, paclitaxel was released by subsequent spontaneous O-N intramolecular acyl migration.
Collapse
Affiliation(s)
- Mariusz Skwarczynski
- Department of Medicinal Chemistry, Center for Frontier Research in Medicinal Science and 21st Century COE Program, Kyoto Pharmaceutical University, Yamashina-ku, Japan
| | | | | | | | | | | | | |
Collapse
|
39
|
Abstract
The transfer of polymer-protein conjugates into routine clinical use, and the clinical development of polymer-anticancer-drug conjugates, both as single agents and as components of combination therapy, is establishing polymer therapeutics as one of the first classes of anticancer nanomedicines. There is growing optimism that ever more sophisticated polymer-based vectors will be a significant addition to the armoury currently used for cancer therapy.
Collapse
Affiliation(s)
- Ruth Duncan
- Centre for Polymer Therapeutics, Welsh School of Pharmacy, Cardiff University, King Edward VII Avenue, Cardiff, CF10 3XF, UK.
| |
Collapse
|
40
|
|
41
|
Abstract
Polymer therapeutics encompass polymer-protein conjugates, drug-polymer conjugates, and supramolecular drug-delivery systems. Numerous polymer-protein conjugates with improved stability and pharmacokinetic properties have been developed, for example, by anchoring enzymes or biologically relevant proteins to polyethylene glycol components (PEGylation). Several polymer-protein conjugates have received market approval, for example the PEGylated form of adenosine deaminase. Coupling low-molecular-weight anticancer drugs to high-molecular-weight polymers through a cleavable linker is an effective method for improving the therapeutic index of clinically established agents, and the first candidates have been evaluated in clinical trials, including, N-(2-hydroxypropyl)methacrylamide conjugates of doxorubicin, camptothecin, paclitaxel, and platinum(II) complexes. Another class of polymer therapeutics are drug-delivery systems based on well-defined multivalent and dendritic polymers. These include polyanionic polymers for the inhibition of virus attachment, polycationic complexes with DNA or RNA (polyplexes), and dendritic core-shell architectures for the encapsulation of drugs. In this Review an overview of polymer therapeutics is presented with a focus on concepts and examples that characterize the salient features of the drug-delivery systems.
Collapse
Affiliation(s)
- Rainer Haag
- Organic and Macromolecular Chemistry, Department of Chemistry and Biochemistry, Freie Universität Berlin, Takustrasse 3, 14195 Berlin, Germany
| | | |
Collapse
|
42
|
Abstract
The intricate problems associated with the delivery and various unnecessary in vivo transitions of proteins and drugs needs to be tackled soon to be able to exploit the myriad of putative therapeutics created by the biotechnology boom. Nanomedicine is one of the most promising applications of nanotechnology in the field of medicine. It has been defined as the monitoring, repair, construction and control of human biological systems at the molecular level using engineered nanodevices and nanostructures. These nanostructured medicines will eventually turn the world of drug delivery upside down. PEGylation (i.e. the attachment of polyethylene glycol to proteins and drugs) is an upcoming methodology for drug development and it has the potential to revolutionise medicine by drastically improving the pharmacokinetic and pharmacodynamic properties of the administered drug. This article provides a total strategy for improving the therapeutic efficacy of various biotechnological products in drug delivery. This article also presents an extensive analysis of most of the PEGylated proteins, peptides and drugs, together with extensive clinical data. Nanomedicines and PEGylation, the latest offshoots of nanotechnology will definitely pave a way in the field of drug delivery where targeted delivery, formulation, in vivo stability and retention are the major challenges.
Collapse
Affiliation(s)
- Suphiya Parveen
- Laboratory of Nanomedicine, Institute of Life Sciences, Chandrasekharpur, Bhubaneswar, Orissa, India
| | | |
Collapse
|
43
|
Abstract
Work on cytotoxic analogs of luteinizing hormone-releasing hormone (LH-RH), somatostatin and bombesin, designed for targeting chemotherapy to peptide receptors on various cancers, is reviewed here as the project is at advanced stages of development and clinical trials are pending. Cytotoxic analogs of LH-RH, AN-152 and AN-207, containing doxorubicin (DOX) or 2-pyrrolino-DOX (AN-201), respectively, target LH-RH receptors and can be used for the treatment of prostatic, breast, ovarian and endometrial cancers and melanomas. AN-201 was also incorporated into the cytotoxic analog of somatostatin, AN-238, which can be targeted to receptors for somatostatin in prostatic, renal, mammary, ovarian, gastric, colorectal and pancreatic cancers as well as glioblastomas and lung cancers, suppressing the growth of these tumors and their metastases. A cytotoxic analog of bombesin AN-215, containing 2-pyrrolino-DOX, was likewise synthesized and successfully tested in experimental models of prostate cancer, small cell lung carcinoma, gastrointestinal cancers and brain tumors expressing receptors for bombesin/gastrin-releasing peptide. This new class of targeted cytotoxic peptide analogs might provide a more effective therapy for various cancers.
Collapse
Affiliation(s)
- Andrew V Schally
- Endocrine, Polypeptide and Cancer Institute, Veterans Affairs Medical Center and Department of Medicine, Tulane University School of Medicine, New Orleans, LA 70112, USA.
| | | |
Collapse
|
44
|
Rooseboom M, Commandeur JNM, Vermeulen NPE. Enzyme-catalyzed activation of anticancer prodrugs. Pharmacol Rev 2004; 56:53-102. [PMID: 15001663 DOI: 10.1124/pr.56.1.3] [Citation(s) in RCA: 345] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The rationale fo the development of prodrugs relies upon delivery of higher concentrations of a drug to target cells compared to administration of the drug itself. In the last decades, numerous prodrugs that are enzymatically activated into anti-cancer agents have been developed. This review describes the most important enzymes involved in prodrug activation notably with respect to tissue distribution, up-regulation in tumor cells and turnover rates. The following endogenous enzymes are discussed: aldehyde oxidase, amino acid oxidase, cytochrome P450 reductase, DT-diaphorase, cytochrome P450, tyrosinase, thymidylate synthase, thymidine phosphorylase, glutathione S-transferase, deoxycytidine kinase, carboxylesterase, alkaline phosphatase, beta-glucuronidase and cysteine conjugate beta-lyase. In relation to each of these enzymes, several prodrugs are discussed regarding organ- or tumor-selective activation of clinically relevant prodrugs of 5-fluorouracil, axazaphosphorines (cyclophosphamide, ifosfamide, and trofosfamide), paclitaxel, etoposide, anthracyclines (doxorubicin, daunorubicin, epirubicin), mercaptopurine, thioguanine, cisplatin, melphalan, and other important prodrugs such as menadione, mitomycin C, tirapazamine, 5-(aziridin-1-yl)-2,4-dinitrobenzamide, ganciclovir, irinotecan, dacarbazine, and amifostine. In addition to endogenous enzymes, a number of nonendogenous enzymes, used in antibody-, gene-, and virus-directed enzyme prodrug therapies, are described. It is concluded that the development of prodrugs has been relatively successful; however, all prodrugs lack a complete selectivity. Therefore, more work is needed to explore the differences between tumor and nontumor cells and to develop optimal substrates in terms of substrate affinity and enzyme turnover rates fo prodrug-activating enzymes resulting in more rapid and selective cleavage of the prodrug inside the tumor cells.
Collapse
Affiliation(s)
- Martijn Rooseboom
- Leiden/Amsterdam Center for Drug Research (L.A.C.D.R.), Division of Molecular Toxicology, Department of Pharmacochemistry, Vrije Universiteit Amsterdam, De Boelelaan 1083, Amsterdam, The Netherlands.
| | | | | |
Collapse
|
45
|
Paukner S, Kohl G, Lubitz W. Bacterial ghosts as novel advanced drug delivery systems: antiproliferative activity of loaded doxorubicin in human Caco-2 cells. J Control Release 2004; 94:63-74. [PMID: 14684272 DOI: 10.1016/j.jconrel.2003.09.010] [Citation(s) in RCA: 77] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Systemic application of anticancer drugs often causes severe toxic side effects. To reduce the undesired effects, advanced drug delivery systems are needed which are based on specific cell targeting vehicles. In this study, bacterial ghosts from Mannheimia haemolytica were used for site-specific delivery of doxorubicin (DOX) to human colorectal adenocarcinoma cells (Caco-2). Bacterial ghosts are non-denatured envelopes of Gram-negative bacteria with fully intact surface structures for specific attachment to mammalian cells. The in vitro release profile of DOX-ghosts demonstrated that the loaded drug was non-covalently associated with the bacterial ghosts and that the drug delivery vehicles themselves represent a slow release system. Adherence studies showed that the M. haemolytica ghosts more efficiently than E. coli ghosts targeted the Caco-2 cells and released the loaded DOX within the cells. Cytotoxicity assays revealed that the DOX-ghosts exhibited potent antiproliferative activities on Caco-2 cells as the DOX associated with ghosts was two magnitude of orders more cytotoxic than free DOX provided in the medium at the same concentrations. Notably, a significant reduction in the cell viability was measured with DOX-ghosts at low DOX concentrations, which had no inhibitory effect when applied as free DOX after incubation for 16 h or when applied at higher concentrations for only 10 min to the cells. As the higher antiproliferative effects of DOX on Caco-2 cells were mediated by the specific drug targeting properties of the bacterial ghosts, the bacterial ghost system represents a novel platform for advanced drug delivery.
Collapse
Affiliation(s)
- Susanne Paukner
- Institute for Microbiology and Genetics, Vienna Biocenter, University of Vienna, Vienna, Austria
| | | | | |
Collapse
|
46
|
Abstract
As we enter the twenty-first century, research at the interface of polymer chemistry and the biomedical sciences has given rise to the first nano-sized (5-100 nm) polymer-based pharmaceuticals, the 'polymer therapeutics'. Polymer therapeutics include rationally designed macromolecular drugs, polymer-drug and polymer-protein conjugates, polymeric micelles containing covalently bound drug, and polyplexes for DNA delivery. The successful clinical application of polymer-protein conjugates, and promising clinical results arising from trials with polymer-anticancer-drug conjugates, bode well for the future design and development of the ever more sophisticated bio-nanotechnologies that are needed to realize the full potential of the post-genomic age.
Collapse
Affiliation(s)
- Ruth Duncan
- Centre for Polymer Therapeutics, Welsh School of Pharmacy, Cardiff University, King Edward VII Avenue, Cardiff CF10 3XF, UK.
| |
Collapse
|
47
|
de Groot FMH, Busscher GF, Aben RWM, Scheeren HW. Novel 20-carbonate linked prodrugs of camptothecin and 9-aminocamptothecin designed for activation by tumour-associated plasmin. Bioorg Med Chem Lett 2002; 12:2371-6. [PMID: 12161136 DOI: 10.1016/s0960-894x(02)00388-8] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The first prodrugs of camptothecin and 9-aminocamptothecin that are activated by the tumour-associated protease plasmin are reported. The tripartate prodrugs consist of a tripeptide sequence recognised by plasmin, which is linked to the 20-hydroxyl group of the camptothecins via a 1,6-elimination spacer. After selective N-protection of 9-aminocamptothecin with an Aloc group, the promoiety (tripeptide-spacer conjugate) was linked to camptothecin or 9-Aloc-9-aminocamptothecin via a 20-carbonate linkage by reacting parent drugs with the p-nitrophenyl carbonate activated promoiety in the presence of DMAP. Both prodrugs showed to be stable in buffer solution and both parent drugs were released upon incubation in the presence of plasmin. Furthermore, the prodrugs showed an average 10-fold decreased cytotoxicity with respect to their parent drugs upon incubation in seven human tumour cell lines.
Collapse
Affiliation(s)
- Franciscus M H de Groot
- Department of Organic Chemistry, NSR-Center for Molecular Structure, Design and Synthesis, University of Nijmegen, The Netherlands
| | | | | | | |
Collapse
|
48
|
Staerk D, Lykkeberg AK, Christensen J, Budnik BA, Abe F, Jaroszewski JW. In vitro cytotoxic activity of phenanthroindolizidine alkaloids from Cynanchum vincetoxicum and Tylophora tanakae against drug-sensitive and multidrug-resistant cancer cells. JOURNAL OF NATURAL PRODUCTS 2002; 65:1299-1302. [PMID: 12350151 DOI: 10.1021/np0106384] [Citation(s) in RCA: 98] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
Two known phenanthroindolizidine alkaloids, (-)-(R)-13aalpha-antofine (1) and (-)-(R)-13aalpha-6-O-desmethylantofine (2), and two new natural products, (-)-(R)-13aalpha-secoantofine (3) and (-)-(R)-13aalpha-6-O-desmethylsecoantofine (4), were isolated from Cynanchum vincetoxicum. The structures of all compounds were established by means of NMR methods including COSY, NOESY, HSQC, and HMBC experiments, supported by HRMS and optical rotation data. Cytotoxic activity of the isolated alkaloids, and of three other alkaloids previously isolated from Tylophora tanakae, (-)-(R)-13aalpha-tylophorine (5), (-)-(R)-13aalpha-7-O-desmethyltylophorine (6), and (+)-(S)-13abeta-isotylocrebrine (7), was assessed in vitro using a drug-sensitive KB-3-1 and a multidrug-resistant KB-V1 cancer cell line. Structure-activity relationships in this series of alkaloids are discussed. The IC(50) values of some of the alkaloids are in the low nanomolar range, being thus comparable to the activity of clinically used cytotoxic drugs. Previously reported adverse side effects of these alkaloids could possibly be overcome by modern tissue-specific drug targeting techniques.
Collapse
MESH Headings
- Alkaloids/chemistry
- Alkaloids/isolation & purification
- Alkaloids/pharmacology
- Antineoplastic Agents, Phytogenic/chemistry
- Antineoplastic Agents, Phytogenic/isolation & purification
- Antineoplastic Agents, Phytogenic/pharmacology
- Cynanchum/chemistry
- Denmark
- Drug Resistance, Neoplasm
- Drug Screening Assays, Antitumor
- Female
- Humans
- Indoles
- Inhibitory Concentration 50
- Isoquinolines/chemistry
- Isoquinolines/isolation & purification
- Isoquinolines/pharmacology
- Molecular Conformation
- Molecular Structure
- Nuclear Magnetic Resonance, Biomolecular
- Phenanthrenes
- Plant Leaves/chemistry
- Plants, Medicinal/chemistry
- Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
- Stereoisomerism
- Structure-Activity Relationship
- Tumor Cells, Cultured
- Uterine Cervical Neoplasms
- Vinblastine/pharmacology
Collapse
Affiliation(s)
- Dan Staerk
- Department of Medicinal Chemistry, Royal Danish School of Pharmacy, Universitetsparken 2, DK-2100 Copenhagen, Denmark.
| | | | | | | | | | | |
Collapse
|
49
|
Wüest T, Gerlach E, Banerjee D, Gerspach J, Moosmayer D, Pfizenmaier K. TNF-Selectokine: a novel prodrug generated for tumor targeting and site-specific activation of tumor necrosis factor. Oncogene 2002; 21:4257-65. [PMID: 12082613 DOI: 10.1038/sj.onc.1205193] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2001] [Accepted: 11/07/2001] [Indexed: 11/09/2022]
Abstract
We describe a TNF fusion protein designated TNF-Selectokine, which is a homo-trimeric molecule comprised of a single chain antibody (scFv) targeting module, a trimerization domain and TNF. TNF-Selectokine exerts high bioactivity towards the targeted and adjacent, antigen negative cells. Membrane targeting dependent immobilization of the TNF-Selectokine induced cell death in TNFR1 and TNFR2 dependent manner, thus cell bound TNF-Selectokine mimicks membrane TNF. To restrict TNF activity to the tumor, a prototype of a TNF-Selectokine prodrug was constructed by insertion of a TNFR1 fragment, separated from TNF by a protease-sensitive linker. The prodrug exerts minimal TNF activity, but can be activated in vitro several thousand-fold by proteolytic digest, showing the principal feasibility of this approach. Choice of cleavage site(s) recognized by protease(s) typically associated with a given carcinoma should allow high dose systemic application of the respective TNF prodrug that unveils its specific bioactivity only in targeted tissues.
Collapse
MESH Headings
- Adenocarcinoma/pathology
- Amino Acid Sequence
- Animals
- Antigen-Antibody Reactions
- Antigens, CD/drug effects
- Antigens, CD/genetics
- Antineoplastic Agents/chemistry
- Antineoplastic Agents/metabolism
- Antineoplastic Agents/pharmacology
- Apoptosis/drug effects
- Binding Sites
- Biotransformation
- CHO Cells
- Coculture Techniques
- Colonic Neoplasms/pathology
- Cricetinae
- Cricetulus
- Drug Design
- Humans
- Immunoglobulin Fragments/chemistry
- Immunoglobulin Fragments/genetics
- Immunoglobulin Fragments/metabolism
- Immunoglobulin Fragments/pharmacology
- Models, Molecular
- Molecular Sequence Data
- Prodrugs/metabolism
- Prodrugs/pharmacology
- Protein Conformation
- Protein Structure, Tertiary
- Receptors, Tumor Necrosis Factor/drug effects
- Receptors, Tumor Necrosis Factor/genetics
- Receptors, Tumor Necrosis Factor, Type I
- Receptors, Tumor Necrosis Factor, Type II
- Recombinant Fusion Proteins/genetics
- Recombinant Fusion Proteins/metabolism
- Recombinant Fusion Proteins/pharmacology
- Rhabdomyosarcoma/pathology
- Single-Chain Antibodies
- Tenascin/chemistry
- Tenascin/genetics
- Trypsin/metabolism
- Trypsin/pharmacology
- Tumor Cells, Cultured/metabolism
- Tumor Cells, Cultured/pathology
- Tumor Necrosis Factor-alpha/chemistry
- Tumor Necrosis Factor-alpha/metabolism
- Tumor Necrosis Factor-alpha/pharmacology
Collapse
Affiliation(s)
- Thomas Wüest
- Institute of Cell Biology and Immunology, University of Stuttgart, Germany
| | | | | | | | | | | |
Collapse
|
50
|
de Groot FM, Loos WJ, Koekkoek R, van Berkom LW, Busscher GF, Seelen AE, Albrecht C, de Bruijn P, Scheeren HW. Elongated multiple electronic cascade and cyclization spacer systems in activatible anticancer prodrugs for enhanced drug release. J Org Chem 2001; 66:8815-30. [PMID: 11749612 DOI: 10.1021/jo0158884] [Citation(s) in RCA: 90] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The design and synthesis of several novel elongated self-elimination spacer systems for application in prodrugs is described. These elongated spacer systems can be incorporated between a cleavable specifier and the parent drug. Naphthalene- and biphenyl-containing spacers were synthesized but did not eliminate. Prodrugs of the anticancer agents doxorubicin and paclitaxel are reported that contain two or three electronic cascade spacers. A novel catalytic application of HOBt was found for the synthesis of N-aryl carbamates through reacting a 4-nitrophenyl carbonate with an aniline derivative, to connect the 1,6-elimination spacers via a carbamate linkage. In addition, a double spacer-containing paclitaxel prodrug was synthesized, comprising a 1,6-elimination spacer and a bis-amine linker connected to paclitaxel via a 2'-carbamate linkage. Prodrugs in which the novel spacer systems were incorporated between a specific tripeptide specifier and the parent drug doxorubicin or paclitaxel proved to be significantly faster activated by plasmin in comparison with prodrugs containing conventional spacer systems. It is expected that the generally applicable novel spacer systems reported herein will contribute to future development of improved enzymatically activated prodrugs.
Collapse
Affiliation(s)
- F M de Groot
- Department of Organic Chemistry, NSR-Center for Molecular Structure, Design and Synthesis, University of Nijmegen, Toernooiveld 1, 6525 ED Nijmegen, The Netherlands
| | | | | | | | | | | | | | | | | |
Collapse
|