1
|
Kurmi BD, Patel P, Paliwal R, Paliwal SR. Molecular approaches for targeted drug delivery towards cancer: A concise review with respect to nanotechnology. J Drug Deliv Sci Technol 2020. [DOI: 10.1016/j.jddst.2020.101682] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
2
|
Lin CH, Wen HC, Chiang CC, Huang JS, Chen Y, Wang SK. Polyproline Tri-Helix Macrocycles as Nanosized Scaffolds to Control Ligand Patterns for Selective Protein Oligomer Interactions. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2019; 15:e1900561. [PMID: 30977296 DOI: 10.1002/smll.201900561] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Indexed: 06/09/2023]
Abstract
Multivalent ligand-receptor interactions play essential roles in biological recognition and signaling. As the receptor arrangement on the cell surface can alter the outcome of cell signaling and also provide spatial specificity for ligand binding, controlling the presentation of ligands has become a promising strategy to manipulate or selectively target protein receptors. The lack of adjustable universal tools to control ligand positions at the size of a few nanometers has prompted the development of polyproline tri-helix macrocycles as scaffolds to present ligands in designated patterns. Model lectin Helix pomatia agglutinin has shown selectivity toward the matching GalNAc ligand pattern matching its binding sites arrangement. The GalNAc pattern selectivity is also observed on intact asialoglycoprotein receptor oligomer on human hepatoma cells showing the pattern-selective interaction can be achieved not only on isolated protein oligomers but also the receptors arranged on the cell surface. As the scaffold design allows convenient creation of versatile ligand patterns, it can be expected as a promising tool to probe the arrangement of receptors on the cell surface and as nanomedicine to manipulate signaling or cell recognition.
Collapse
Affiliation(s)
- Cin-Hao Lin
- Department of Chemistry, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Hsin-Chuan Wen
- Department of Chemistry, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Cheng-Chin Chiang
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Jen-Sheng Huang
- Department of Chemistry, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Yunching Chen
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan
- Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Sheng-Kai Wang
- Department of Chemistry, National Tsing Hua University, Hsinchu, 30013, Taiwan
- Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu, 30013, Taiwan
| |
Collapse
|
3
|
Conibear AC, Thewes K, Groysbeck N, Becker CFW. Multifunctional Scaffolds for Assembling Cancer-Targeting Immune Stimulators Using Chemoselective Ligations. Front Chem 2019; 7:113. [PMID: 30895175 PMCID: PMC6414710 DOI: 10.3389/fchem.2019.00113] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 02/12/2019] [Indexed: 01/09/2023] Open
Abstract
Chemoselective ligations allow chemical biologists to functionalise proteins and peptides for biomedical applications and to probe biological processes. Coupled with solid phase peptide synthesis, chemoselective ligations enable not only the design of homogeneous proteins and peptides with desired natural and unnatural modifications in site-specific locations but also the design of new peptide and protein topologies. Although several well-established ligations are available, each method has its own advantages and disadvantages and they are seldom used in combination. Here we have applied copper-catalyzed azide-alkyne “click,” oxime, maleimide, and native chemical ligations to develop a modular synthesis of branched peptide and polymer constructs that act as cancer-targeting immune system engagers (ISErs) and functionalised them for detection in biological systems. We also note some potential advantages and pitfalls of these chemoselective ligations to consider when designing orthogonal ligation strategies. The modular synthesis and functionalization of ISErs facilitates optimisation of their activity and mechanism of action as potential cancer immunotherapies.
Collapse
Affiliation(s)
- Anne C Conibear
- Faculty of Chemistry, Institute of Biological Chemistry, University of Vienna, Vienna, Austria
| | - Karine Thewes
- Faculty of Chemistry, Institute of Biological Chemistry, University of Vienna, Vienna, Austria
| | - Nadja Groysbeck
- Faculty of Chemistry, Institute of Biological Chemistry, University of Vienna, Vienna, Austria
| | - Christian F W Becker
- Faculty of Chemistry, Institute of Biological Chemistry, University of Vienna, Vienna, Austria
| |
Collapse
|
4
|
Li J, Carney RP, Liu R, Fan J, Zhao S, Chen Y, Lam KS, Pan T. Microfluidic Print-to-Synthesis Platform for Efficient Preparation and Screening of Combinatorial Peptide Microarrays. Anal Chem 2018; 90:5833-5840. [PMID: 29633611 PMCID: PMC11870389 DOI: 10.1021/acs.analchem.8b00371] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
In this paper, we introduce a novel microfluidic combinatorial synthesis platform, referred to as Microfluidic Print-to-Synthesis (MPS), for custom high-throughput and automated synthesis of a large number of unique peptides in a microarray format. The MPS method utilizes standard Fmoc chemistry to link amino acids on a polyethylene glycol (PEG)-functionalized microdisc array. The resulting peptide microarrays permit rapid screening for interactions with molecular targets or live cells, with low nonspecific binding. Such combinatorial peptide microarrays can be reliably prepared at a spot size of 200 μm with 1 mm center-to-center distance, dimensions that require only minimal reagent consumption (less than 30 nL per spot per coupling reaction). The MPS platform has a scalable design for extended multiplexibility, allowing for 12 different building blocks and coupling reagents to be dispensed in one microfluidic cartridge in the current format, and could be further scaled up. As proof of concept for the MPS platform, we designed and constructed a focused tetrapeptide library featuring 2560 synthetic peptide sequences, capped at the N-terminus with 4-[( N'-2-methylphenyl)ureido]phenylacetic acid. We then used live human T lymphocyte Jurkat cells as a probe to screen the peptide microarrays for their interaction with α4β1 integrin overexpressed and activated on these cells. Unlike the one-bead-one-compound approach that requires subsequent decoding of positive beads, each spot in the MPS array is spatially addressable. Therefore, this platform is an ideal tool for rapid optimization of lead compounds found in nature or discovered from diverse combinatorial libraries, using either biochemical or cell-based assays.
Collapse
Affiliation(s)
- Jiannan Li
- Department of Biomedical Engineering, University of California, Davis, California 95765, United States
| | - Randy P. Carney
- Department of Biochemistry and Molecular Medicine, University of California, Davis, California 95765, United States
| | - Ruiwu Liu
- Department of Biochemistry and Molecular Medicine, University of California, Davis, California 95765, United States
| | - Jinzhen Fan
- Department of Biomedical Engineering, University of California, Davis, California 95765, United States
| | - Siwei Zhao
- Department of Biomedical Engineering, University of California, Davis, California 95765, United States
| | - Yan Chen
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, People’s Republic of China
| | - Kit S. Lam
- Department of Biochemistry and Molecular Medicine, University of California, Davis, California 95765, United States
| | - Tingrui Pan
- Department of Biomedical Engineering, University of California, Davis, California 95765, United States
| |
Collapse
|
5
|
Conibear AC, Pötgens AJG, Thewes K, Altdorf C, Hilzendeger C, Becker CFW. Synthetic Cancer-Targeting Innate Immune Stimulators Give Insights into Avidity Effects. Chembiochem 2018; 19:459-469. [PMID: 29230922 DOI: 10.1002/cbic.201700522] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Indexed: 11/08/2022]
Abstract
Multispecific and multivalent antibodies are seen as promising cancer therapeutics, and numerous antibody fragments and derivatives have been developed to exploit avidity effects that result in increased selectivity. Most of these multispecific and multivalent antibody strategies make use of recombinant expression of antigen-binding modules. In contrast, chemical synthesis and chemoselective ligations can be used to generate a variety of molecules with different numbers and combinations of binding moieties in a modular and homogeneous fashion. In this study we synthesized a series of targeted immune system engagers (ISErs) by using solid-phase peptide synthesis and chemoselective ligations. To explore avidity effects, we constructed molecules bearing different numbers and combinations of two "binder" peptides that target ephrin A2 and integrin α3 receptors and an "effector" peptide that binds to formyl peptide receptors and stimulates an immune response. We investigated various strategies for generating multivalent and multispecific targeted innate immune stimulators and studied their activities in terms of binding to cancer cells and stimulation of immune cells. This study gives insights into the influence that multivalency and receptor density have on avidity effects and is useful for the design of potential anticancer therapeutics.
Collapse
Affiliation(s)
- Anne C Conibear
- University of Vienna, Faculty of Chemistry, Institute of Biological Chemistry, Währinger Strasse 38, 1090, Vienna, Austria
| | - André J G Pötgens
- Syntab Therapeutics GmbH, ZBMT, Pauwelstrasse 17, 52074, Aachen, Germany
| | - Karine Thewes
- University of Vienna, Faculty of Chemistry, Institute of Biological Chemistry, Währinger Strasse 38, 1090, Vienna, Austria
| | - Claudia Altdorf
- Syntab Therapeutics GmbH, ZBMT, Pauwelstrasse 17, 52074, Aachen, Germany
| | | | - Christian F W Becker
- University of Vienna, Faculty of Chemistry, Institute of Biological Chemistry, Währinger Strasse 38, 1090, Vienna, Austria
| |
Collapse
|
6
|
Synthetic integrin-binding immune stimulators target cancer cells and prevent tumor formation. Sci Rep 2017; 7:17592. [PMID: 29242512 PMCID: PMC5730604 DOI: 10.1038/s41598-017-17627-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Accepted: 11/28/2017] [Indexed: 01/03/2023] Open
Abstract
Immuno-oncology approaches mainly utilize monoclonal antibodies or protein-based scaffolds that bind with high affinity to cancer cells and can generate an immune response. Peptides can also bind with high affinity to cancer cells and are intermediate in size between antibodies and small molecules. They are also synthetically accessible and therefore easily modified to optimize their stability, binding affinity and selectivity. Here we describe the design of immune system engagers (ISErs), a novel class of synthetic peptide-based compounds that bind specifically to cancer cells and stimulate the immune system. A prototype, Y9, targets integrin α3, which is overexpressed on several cancer cells, and activates the immune system via a formyl methionine-containing effector peptide. Injection of Y9 leads to immune cell infiltration into tissue and prevents tumor formation in a guinea pig model. The anti-tumor activity and synthetic accessibility of Y9 illustrate that ISErs could be applied to a wide variety of targets and diseases.
Collapse
|
7
|
Duro-Castano A, Gallon E, Decker C, Vicent MJ. Modulating angiogenesis with integrin-targeted nanomedicines. Adv Drug Deliv Rev 2017; 119:101-119. [PMID: 28502767 DOI: 10.1016/j.addr.2017.05.008] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2017] [Revised: 04/12/2017] [Accepted: 05/09/2017] [Indexed: 12/20/2022]
Abstract
Targeting angiogenesis-related pathologies, which include tumorigenesis and metastatic processes, has become an attractive strategy for the development of efficient guided nanomedicines. In this respect, integrins are cell-adhesion molecules involved in angiogenesis signaling pathways and are overexpressed in many angiogenic processes. Therefore, they represent specific biomarkers not only to monitor disease progression but also to rationally design targeted nanomedicines. Arginine-glycine-aspartic (RGD) containing peptides that bind to specific integrins have been widely utilized to provide ligand-mediated targeting capabilities to small molecules, peptides, proteins, and antibodies, as well as to drug/imaging agent-containing nanomedicines, with the final aim of maximizing their therapeutic index. Within this review, we aim to cover recent and relevant examples of different integrin-assisted nanosystems including polymeric nanoconstructs, liposomes, and inorganic nanoparticles applied in drug/gene therapy as well as imaging and theranostics. We will also critically address the overall benefits of integrin-targeting.
Collapse
Affiliation(s)
- Aroa Duro-Castano
- Centro de Investigación Príncipe Felipe, Polymer Therapeutics Lab., Av. Eduardo Primo Yúfera 3, E-46012 Valencia, Spain.
| | - Elena Gallon
- Centro de Investigación Príncipe Felipe, Polymer Therapeutics Lab., Av. Eduardo Primo Yúfera 3, E-46012 Valencia, Spain.
| | - Caitlin Decker
- Centro de Investigación Príncipe Felipe, Polymer Therapeutics Lab., Av. Eduardo Primo Yúfera 3, E-46012 Valencia, Spain.
| | - María J Vicent
- Centro de Investigación Príncipe Felipe, Polymer Therapeutics Lab., Av. Eduardo Primo Yúfera 3, E-46012 Valencia, Spain.
| |
Collapse
|
8
|
Gleeson EC, Wang ZJ, Robinson SD, Chhabra S, MacRaild CA, Jackson WR, Norton RS, Robinson AJ. Stereoselective synthesis and structural elucidation of dicarba peptides. Chem Commun (Camb) 2016; 52:4446-9. [PMID: 26892179 DOI: 10.1039/c5cc10540d] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A facile stereoselective synthesis of cis and trans unsaturated dicarba peptides has been established using preformed diaminosuberic acid derivatives as bridging units. In addition, characteristic spectral differences in the (13)C-NMR spectra of the cis- and trans-isomers show that the chemical shift of carbons in the Δ4,5-diaminosuberic acid residue can be used to assign stereochemistry in unsaturated dicarba peptides formed from ring closing metathesis of linear peptide sequences.
Collapse
Affiliation(s)
- Ellen C Gleeson
- School of Chemistry, Monash University, Clayton 3800, Victoria, Australia.
| | - Zhen J Wang
- School of Chemistry, Monash University, Clayton 3800, Victoria, Australia.
| | - Samuel D Robinson
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville 3052, Victoria, Australia
| | - Sandeep Chhabra
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville 3052, Victoria, Australia
| | - Christopher A MacRaild
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville 3052, Victoria, Australia
| | - W Roy Jackson
- School of Chemistry, Monash University, Clayton 3800, Victoria, Australia.
| | - Raymond S Norton
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville 3052, Victoria, Australia
| | - Andrea J Robinson
- School of Chemistry, Monash University, Clayton 3800, Victoria, Australia.
| |
Collapse
|
9
|
Abstract
Unusual amino acids are fundamental building blocks of modern medicinal chemistry. The combination of readily functionalized amine and carboxyl groups attached to a chiral central core along with one or two potentially diverse side chains provides a unique three-dimensional structure with a high degree of functionality. This makes them invaluable as starting materials for syntheses of complex molecules, highly diverse elements for SAR campaigns, integral components of peptidomimetic drugs, and potential drugs on their own. This Perspective highlights the diversity of unnatural amino acid structures found in hit-to-lead and lead optimization campaigns and clinical stage and approved drugs, reflecting their increasingly important role in medicinal chemistry.
Collapse
Affiliation(s)
- Mark A T Blaskovich
- Institute for Molecular Bioscience, The University of Queensland , Brisbane, Queensland Australia 4072
| |
Collapse
|
10
|
|
11
|
Discovery and characterization of a high-affinity and high-specificity peptide ligand LXY30 for in vivo targeting of α3 integrin-expressing human tumors. EJNMMI Res 2016; 6:18. [PMID: 26922417 PMCID: PMC4769701 DOI: 10.1186/s13550-016-0165-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Accepted: 01/19/2016] [Indexed: 12/20/2022] Open
Abstract
Background α3β1 integrin is overexpressed in several types of human cancer and is associated with poor prognosis, metastasis, and resistance to cancer treatment. We previously identified a cyclic peptide ligand LXY1 that specifically binds to the α3β1 integrin on human glioblastoma U-87MG cells. Here, we optimized LXY1 through one-bead one-compound combinatorial library screening and site-specific modifications to improve its in vivo binding property. Methods Three bead libraries were synthesized and whole-cell binding assays were performed. The binding capacity of individual peptide ligands against different tumor cells was determined by flow cytometry and confirmed by optical imaging. A complex joining biotinylated ligand with streptavidin-Cy5.5 was used for in vivo target imaging in both subcutaneous and orthotopic U-87MG xenograft mouse models. Results LXY30, a cyclic peptide with the sequence cdG-Phe(3,5-diF)-G-Hyp-NcR, emerged as the most potent and selective ligand for the α3 subunit of α3β1 integrin with improved in vitro and in vivo tumor-targeting effects compared to LXY1 in U-87MG cells. LXY30 is considerably stable in plasma as demonstrated in an in vitro stability study in 90 % human plasma. LXY30 also binds to several other known α3β1 integrin-expressing glioblastoma, lung, and breast cancer cell lines with various affinities. Conclusions Our data support further investigating the role of LXY30 as a human tumor-targeting peptide ligand for systemic and intracranial delivery of imaging agents and cancer therapeutics. Electronic supplementary material The online version of this article (doi:10.1186/s13550-016-0165-z) contains supplementary material, which is available to authorized users.
Collapse
|
12
|
Dai W, Yang F, Ma L, Fan Y, He B, He Q, Wang X, Zhang H, Zhang Q. Combined mTOR inhibitor rapamycin and doxorubicin-loaded cyclic octapeptide modified liposomes for targeting integrin α3 in triple-negative breast cancer. Biomaterials 2014; 35:5347-5358. [DOI: 10.1016/j.biomaterials.2014.03.036] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2014] [Accepted: 03/14/2014] [Indexed: 11/25/2022]
|
13
|
Abstract
PEG-dendritic block copolymer (telodendrimer) is a unique class of polymers with well-defined structures and tunable aggregation properties, which have been recently developed as novel micelle-based nanocarriers. This new class of nanocarrier is highly versatile, robust, multifunctional and has many unique properties for drug delivery that are superior to most other nanocarriers reported in the literature. Reversible crosslinking of micelles is able to minimize the premature drug release during circulation. These crosslinks can be reversed with endogenous and/or exogenous stimuli. To further facilitate the precise delivery of nanoparticle drugs to cancer cells, the nanoparticles surface can be decorated with ovarian cancer targeting ligands. This review is focused on the various strategies used for the design, preparation, pharmacokinetic, biodistribution and preclinical therapeutic applications of telodendrimer-based nanocarriers for drug delivery in the treatment of ovarian cancer. Lastly, future perspectives for the development of ovarian cancer-targeting telodendrimer nanotherapeutics are also explored.
Collapse
|