1
|
Gulyaev IA, Sokol MB, Mollaeva MR, Klimenko MA, Yabbarov NG, Chirkina MV, Nikolskaya ED. Polymeric Drug Delivery Systems in Biomedicine. BIOCHEMISTRY. BIOKHIMIIA 2025; 90:S233-S262. [PMID: 40164161 DOI: 10.1134/s0006297924603976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 10/08/2024] [Accepted: 10/22/2024] [Indexed: 04/02/2025]
Abstract
Our review examines the key aspects of using polymeric carriers in biomedicine. The section "Polymers for Biomedicine" provides an overview of different types of polymers, their structural features and properties that determine their use as drug delivery vehicles. The section "Polymeric Carriers" characterizes the role of polymeric delivery systems in modern medicine. The main forms of polymeric carriers are described, as well as their key advantages for drug delivery. The section "Preclinical and Clinical Trials of Polymeric Drug Carriers" reviews the examples of clinical and preclinical studies of polymeric forms used for antitumor therapy, therapy for bacterial and infectious diseases. The final section "Targeted Drug Delivery Systems" is devoted to the discussion of approaches, as well as ligands that provide targeted drug delivery using polymeric carriers. We have paid special attention to modern approaches for increasing the efficacy of antibacterial therapy using vector molecules.
Collapse
Affiliation(s)
- Ivan A Gulyaev
- Emanuel Institute of Biochemical Physics of Russian Academy of Sciences, Moscow, 119334, Russia.
| | - Maria B Sokol
- Emanuel Institute of Biochemical Physics of Russian Academy of Sciences, Moscow, 119334, Russia
| | - Mariia R Mollaeva
- Emanuel Institute of Biochemical Physics of Russian Academy of Sciences, Moscow, 119334, Russia
| | - Maksim A Klimenko
- Emanuel Institute of Biochemical Physics of Russian Academy of Sciences, Moscow, 119334, Russia
| | - Nikita G Yabbarov
- Emanuel Institute of Biochemical Physics of Russian Academy of Sciences, Moscow, 119334, Russia
| | - Margarita V Chirkina
- Emanuel Institute of Biochemical Physics of Russian Academy of Sciences, Moscow, 119334, Russia
| | - Elena D Nikolskaya
- Emanuel Institute of Biochemical Physics of Russian Academy of Sciences, Moscow, 119334, Russia.
| |
Collapse
|
2
|
Shao Q, Zhang F, Li C, Yang Y, Liu S, Chen G, Fan B. Design of a prodrug photocage for cancer cells detection and anticancer drug release. Talanta 2024; 274:126002. [PMID: 38613948 DOI: 10.1016/j.talanta.2024.126002] [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: 12/27/2023] [Revised: 03/21/2024] [Accepted: 03/26/2024] [Indexed: 04/15/2024]
Abstract
Developing probes for simultaneous diagnosis and killing of cancer cells is crucial, yet challenging. This article presents the design and synthesis of a novel Rhodamine B fluorescence probe. The design strategy involves utilizing an anticancer drug (Melphalan) to bind with a fluorescent group (HRhod-OH), forming HRhod-MeL, which is non-fluorescent. However, when exposed to the high levels of reactive oxygen species (ROS) of cancer cells, HRhod-MeL transforms into a red-emitting Photocage (Rhod-MeL), and selectively accumulates in the mitochondria of cancer cells, where, when activated with green light (556 nm), anti-cancer drugs released. The Photocage improve the efficacy of anti-cancer drugs and enables the precise diagnosis and killing of cancer cells. Therefore, the prepared Photocage can detect cancer cells and release anticancer drugs in situ, which provides a new method for the development of prodrugs.
Collapse
Affiliation(s)
- Qianshan Shao
- School of Pharmacy, Hubei University of Science and Technology, No.88, Xianning avenue, XiananDistrict, Xianning, 437000, China
| | - Fei Zhang
- Hubei Provincial Key Laboratory of Radiation Chemistry and Functional Materials, Hubei University of Science and Technology, No.88, Xianning Avenue, Xianan District, Xianning, 437000, China
| | - Chunxiao Li
- School of Pharmacy, Hubei University of Science and Technology, No.88, Xianning avenue, XiananDistrict, Xianning, 437000, China
| | - Yuyu Yang
- School of Pharmacy, Hubei University of Science and Technology, No.88, Xianning avenue, XiananDistrict, Xianning, 437000, China
| | - Shihan Liu
- School of Pharmacy, Hubei University of Science and Technology, No.88, Xianning avenue, XiananDistrict, Xianning, 437000, China
| | - Guang Chen
- Shanxi Key Laboratory of Chemical Additives for Industry, College of Chemistry and Chemical Engineering, Shanxi University of Science and Technology, Xi'an, 710021, China.
| | - Baolei Fan
- School of Pharmacy, Hubei University of Science and Technology, No.88, Xianning avenue, XiananDistrict, Xianning, 437000, China; Hubei Provincial Key Laboratory of Radiation Chemistry and Functional Materials, Hubei University of Science and Technology, No.88, Xianning Avenue, Xianan District, Xianning, 437000, China.
| |
Collapse
|
3
|
Bhat MF, Prats Luján A, Saifuddin M, Fodran P, Poelarends GJ. Multigram-scale chemoenzymatic synthesis of diverse aminopolycarboxylic acids as potential metallo-β-lactamase inhibitors. Org Biomol Chem 2024; 22:491-495. [PMID: 38126753 PMCID: PMC10792612 DOI: 10.1039/d3ob01405c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Accepted: 12/12/2023] [Indexed: 12/23/2023]
Abstract
Toxin A, a precursor to naturally occurring aspergillomarasmine A, aspergillomarasmine B, lycomarasmine and related aminopolycarboxylic acids, was synthesized as the desired (2S,2'S)-diastereomer on a multigram-scale (>99% conversion, 82% isolated yield, dr > 95 : 5) from commercially available starting materials using the enzyme ethylenediamine-N,N'-disuccinic acid lyase. A single-step protection route of this chiral synthon was developed to aid N-sulfonylation/-alkylation and reductive amination at the terminal primary amine for easy derivatization, followed by global deprotection to give the corresponding toxin A derivatives, including lycomarasmine, in moderate to good yields (23-66%) and with high stereopurity (dr > 95 : 5). Furthermore, a chemoenzymatic route was developed to introduce a click handle on toxin A (yield 72%, dr > 95 : 5) and its cyclized congener for further analogue design. Finally, a chemoenzymatic route towards the synthesis of photocaged aspergillomarasmine B (yield 8%, dr > 95 : 5) was established, prompting further steps into smart prodrug design and precision delivery. These new synthetic methodologies have the prospective of facilitating research into the finding of more selective and potent metallo-β-lactamase (MBL) inhibitors, which are urgently needed to combat MBL-based infections.
Collapse
Affiliation(s)
- Mohammad Faizan Bhat
- Department of Chemical and Pharmaceutical Biology, Groningen Research Institute of Pharmacy, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands.
| | - Alejandro Prats Luján
- Department of Chemical and Pharmaceutical Biology, Groningen Research Institute of Pharmacy, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands.
| | - Mohammad Saifuddin
- Department of Chemical and Pharmaceutical Biology, Groningen Research Institute of Pharmacy, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands.
| | - Peter Fodran
- Department of Chemical and Pharmaceutical Biology, Groningen Research Institute of Pharmacy, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands.
| | - Gerrit J Poelarends
- Department of Chemical and Pharmaceutical Biology, Groningen Research Institute of Pharmacy, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands.
| |
Collapse
|
4
|
Kalyvas JT, Facal Marina P, Stachura DL, Horsley JR, Abell AD. Smart Wearable Patches Using Light-Controlled Activation and Delivery of Photoswitchable Antimicrobial Peptides. Chemistry 2023; 29:e202301487. [PMID: 37309073 DOI: 10.1002/chem.202301487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 06/07/2023] [Accepted: 06/09/2023] [Indexed: 06/14/2023]
Abstract
A novel strategy to treat Staphylococcus aureus (S. aureus) skin infections is presented, where UV light is used to facilitate concomitant light-controlled activation and delivery of an antimicrobial therapeutic agent. Specifically, a new photoswitchable gramicidin S analogue was immobilized onto a polymeric wearable patch via a photocleavable linker that undergoes photolysis at the same wavelength of light required for activation of the peptide. Unlike toxic gramicidin S, the liberated active photoswitchable peptide exhibits antimicrobial activity against S. aureus while being ostensibly non-haemolytic to red blood cells. Moreover, irradiation with visible light switches off the antimicrobial properties of the peptide within seconds, presenting an ideal strategy to regulate antibiotic activity for localized bacterial infections with the potential to mitigate resistance.
Collapse
Affiliation(s)
- John T Kalyvas
- School of Physics, Chemistry & Earth Sciences, The University of Adelaide Adelaide, South Australia, 5005, Australia
| | - Paula Facal Marina
- School of Physics, Chemistry & Earth Sciences, The University of Adelaide Adelaide, South Australia, 5005, Australia
- Flinders Institute for NanoScale Science and Technology College of Science and Engineering, Flinders University, South Australia, 5042, Australia
| | - Damian L Stachura
- School of Physics, Chemistry & Earth Sciences, The University of Adelaide Adelaide, South Australia, 5005, Australia
| | - John R Horsley
- School of Physics, Chemistry & Earth Sciences, The University of Adelaide Adelaide, South Australia, 5005, Australia
| | - Andrew D Abell
- School of Physics, Chemistry & Earth Sciences, The University of Adelaide Adelaide, South Australia, 5005, Australia
| |
Collapse
|
5
|
Sarabando SN, Palmeira A, Sousa ME, Faustino MAF, Monteiro CJP. Photomodulation Approaches to Overcome Antimicrobial Resistance. Pharmaceuticals (Basel) 2023; 16:682. [PMID: 37242465 PMCID: PMC10221556 DOI: 10.3390/ph16050682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 04/24/2023] [Accepted: 04/30/2023] [Indexed: 05/28/2023] Open
Abstract
Photopharmacology is an approach that aims to be an alternative to classical chemotherapy. Herein, the different classes of photoswitches and photocleavage compounds and their biological applications are described. Proteolysis targeting chimeras (PROTACs) containing azobenzene moieties (PHOTACs) and photocleavable protecting groups (photocaged PROTACs) are also mentioned. Furthermore, porphyrins are referenced as successful photoactive compounds in a clinical context, such as in the photodynamic therapy of tumours as well as preventing antimicrobial resistance, namely in bacteria. Porphyrins combining photoswitches and photocleavage systems are highlighted, taking advantage of both photopharmacology and photodynamic action. Finally, porphyrins with antibacterial activity are described, taking advantage of the synergistic effect of photodynamic treatment and antibiotic therapy to overcome bacterial resistance.
Collapse
Affiliation(s)
- Sofia N. Sarabando
- Laboratory of Organic and Pharmaceutical Chemistry, Chemical Sciences Department, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (S.N.S.); (A.P.)
- LAQV-Requimte and Department of Chemistry, University of Aveiro, 3010-193 Aveiro, Portugal;
| | - Andreia Palmeira
- Laboratory of Organic and Pharmaceutical Chemistry, Chemical Sciences Department, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (S.N.S.); (A.P.)
- CIIMAR—Interdisciplinary Centre of Marine and Environmental Research, 4450-208 Porto, Portugal
| | - Maria Emília Sousa
- Laboratory of Organic and Pharmaceutical Chemistry, Chemical Sciences Department, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (S.N.S.); (A.P.)
- CIIMAR—Interdisciplinary Centre of Marine and Environmental Research, 4450-208 Porto, Portugal
| | | | - Carlos J. P. Monteiro
- LAQV-Requimte and Department of Chemistry, University of Aveiro, 3010-193 Aveiro, Portugal;
| |
Collapse
|
6
|
Yu L, Shang Z, Jin Q, Chan SY, Hong W, Li N, Li P. Antibody-Antimicrobial Conjugates for Combating Antibiotic Resistance. Adv Healthc Mater 2023; 12:e2202207. [PMID: 36300640 DOI: 10.1002/adhm.202202207] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 10/19/2022] [Indexed: 02/03/2023]
Abstract
As the development of new antibiotics lags far behind the emergence of drug-resistant bacteria, alternative strategies to resolve this dilemma are urgently required. Antibody-drug conjugate is a promising therapeutic platform to delivering cytotoxic payloads precisely to target cells for efficient disease treatment. Antibody-antimicrobial conjugates (AACs) have recently attracted considerable interest from researchers as they can target bacteria in the target sites and improve the effectiveness of drugs (i.e., reduced drug dosage and adverse effects), abating the upsurge of antimicrobial resistance. In this review, the selection and progress of three essential blocks that compose the AACs: antibodies, antimicrobial payloads, and linkers are discussed. The commonly used conjugation strategies and the latest applications of AACs in recent years are also summarized. The challenges and opportunities of this booming technology are also discussed at the end of this review.
Collapse
Affiliation(s)
- Luofeng Yu
- Frontiers Science Center for Flexible Electronics (FSCFE), Xi'an Institute of Flexible Electronics (IFE), Xi'an Institute of Biomedical Materials and Engineering (IBME), Northwestern Polytechnical University (NPU), 127 West Youyi Road, Xi'an, 710072, China
| | - Zifang Shang
- Frontiers Science Center for Flexible Electronics (FSCFE), Xi'an Institute of Flexible Electronics (IFE), Xi'an Institute of Biomedical Materials and Engineering (IBME), Northwestern Polytechnical University (NPU), 127 West Youyi Road, Xi'an, 710072, China.,Institute of Pediatrics, Shenzhen Children's Hospital, Shenzhen, Guangdong Province, 518026, China.,CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology Chinese Academy of Sciences, Beijing, 100101, China
| | - Qizhe Jin
- Frontiers Science Center for Flexible Electronics (FSCFE), Xi'an Institute of Flexible Electronics (IFE), Xi'an Institute of Biomedical Materials and Engineering (IBME), Northwestern Polytechnical University (NPU), 127 West Youyi Road, Xi'an, 710072, China
| | - Siew Yin Chan
- Frontiers Science Center for Flexible Electronics (FSCFE), Xi'an Institute of Flexible Electronics (IFE), Xi'an Institute of Biomedical Materials and Engineering (IBME), Northwestern Polytechnical University (NPU), 127 West Youyi Road, Xi'an, 710072, China.,Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), 2 Fusionopolis Way, Innovis, #08-03, Singapore, 138634, Singapore
| | - Weilin Hong
- Frontiers Science Center for Flexible Electronics (FSCFE), Xi'an Institute of Flexible Electronics (IFE), Xi'an Institute of Biomedical Materials and Engineering (IBME), Northwestern Polytechnical University (NPU), 127 West Youyi Road, Xi'an, 710072, China
| | - Nan Li
- Frontiers Science Center for Flexible Electronics (FSCFE), Xi'an Institute of Flexible Electronics (IFE), Xi'an Institute of Biomedical Materials and Engineering (IBME), Northwestern Polytechnical University (NPU), 127 West Youyi Road, Xi'an, 710072, China
| | - Peng Li
- Frontiers Science Center for Flexible Electronics (FSCFE), Xi'an Institute of Flexible Electronics (IFE), Xi'an Institute of Biomedical Materials and Engineering (IBME), Northwestern Polytechnical University (NPU), 127 West Youyi Road, Xi'an, 710072, China
| |
Collapse
|
7
|
Nwabuife JC, Omolo CA, Govender T. Nano delivery systems to the rescue of ciprofloxacin against resistant bacteria "E. coli; P. aeruginosa; Saureus; and MRSA" and their infections. J Control Release 2022; 349:338-353. [PMID: 35820538 DOI: 10.1016/j.jconrel.2022.07.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 06/29/2022] [Accepted: 07/03/2022] [Indexed: 10/17/2022]
Abstract
Ciprofloxacin (CIP) a broad-spectrum antibiotic, is used extensively for the treatment of diverse infections and diseases of bacteria origin, and this includes infections caused by E. coli; P. aeruginosa; S. aureus; and MRSA. This extensive use of CIP has therefore led to an increase in resistance by these infection causing organisms. Nano delivery systems has recently proven to be a possible solution to resistance to these organisms. They have been applied as a strategy to improve the target specificity of CIP against infections and diseases caused by these organisms, thereby maximising the efficacy of CIP to overcome the resistance. Herein, we proffer a brief overview of the mechanisms of resistance; the causes of resistance; and the various approaches employed to overcome this resistance. The review then proceeds to critically evaluate various nano delivery systems including inorganic based nanoparticles; lipid-based nanoparticles; capsules, dendrimers, hydrogels, micelles, and polymeric nanoparticles; and others; that have been applied for the delivery of CIP against E. coli; P. aeruginosa; S. aureus; and MRSA infections. Finally, the review highlights future areas of research, for the optimisation of various nano delivery systems, to maximise the therapeutic efficacy of CIP against these organisms. This review confirms the potential of nano delivery systems, for addressing the challenges of resistance to caused by E. coli; P. aeruginosa; S. aureus; and MRSA to CIP.
Collapse
Affiliation(s)
- Joshua C Nwabuife
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban 4000, South Africa
| | - Calvin A Omolo
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban 4000, South Africa.; Department of Pharmaceutics, School of Pharmacy and Health Sciences, United States International University-Africa, P. O. Box 14634-00800, Nairobi, Kenya
| | - Thirumala Govender
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban 4000, South Africa..
| |
Collapse
|
8
|
Huang Y, Zou L, Wang J, Jin Q, Ji J. Stimuli-responsive nanoplatforms for antibacterial applications. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2022; 14:e1775. [PMID: 35142071 DOI: 10.1002/wnan.1775] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 12/22/2021] [Accepted: 12/23/2021] [Indexed: 12/13/2022]
Abstract
The continuously increasing bacterial resistance has become a big threat to public health worldwide, which makes it urgent to develop innovative antibacterial strategies. Nanotechnology-based drug delivery systems are considered as promising strategies in combating bacterial infections which are expected to improve the therapeutic efficacy and minimize the side effects. Unfortunately, the conventional nanodrug delivery systems always suffer from practical dilemmas, including incomplete and slow drug release, insufficient accumulation in infected sites, and weak biofilm penetration ability. Stimuli-responsive nanoplatforms are hence developed to overcome the disadvantages of conventional nanoparticles. In this review, we provide an extensive review of the recent progress of endogenous and exogenous stimuli-responsive nanoplatforms in the antibacterial area, including planktonic bacteria, intracellular bacteria, and bacterial biofilms. Taking advantage of the specific infected microenvironment (pH, enzyme, redox, and toxin), the mechanisms and strategies of the design of endogenous stimuli-responsive nanoplatforms are discussed, with an emphasis on how to improve the therapeutic efficacy and minimize side effects. How to realize controlled drug delivery using exogenous stimuli-responsive nanoplatforms especially light-responsive nanoparticles for improved antibacterial effects is another topic of this review. We especially highlight photothermal-triggered drug delivery systems by the combination of photothermal agents and thermo-responsive materials. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Infectious Disease Therapeutic Approaches and Drug Discovery > Emerging Technologies.
Collapse
Affiliation(s)
- Yue Huang
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization of Ministry of Education, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, China
| | - Lingyun Zou
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization of Ministry of Education, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, China
| | - Jing Wang
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization of Ministry of Education, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, China
| | - Qiao Jin
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization of Ministry of Education, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, China
| | - Jian Ji
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization of Ministry of Education, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, China
| |
Collapse
|
9
|
Stimuli-sensitive drug delivery systems for site-specific antibiotic release. Drug Discov Today 2022; 27:1698-1705. [DOI: 10.1016/j.drudis.2022.02.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 01/19/2022] [Accepted: 02/17/2022] [Indexed: 12/12/2022]
|
10
|
Shende P, Gupta S. Role of lipopolysaccharides in potential applications of nanocarrier systems. Curr Pharm Des 2021; 28:1000-1010. [PMID: 34818999 DOI: 10.2174/1381612827666211124094302] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 08/24/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Lipopolysaccharides (LPS) are considered the main molecular component in the outer membrane of gram-negative bacteria. The LPS molecule in the bacterial cell wall acts as a primary physical barrier and protects gram-negative bacteria from the surrounding environment. LPS (endotoxins) show immunomodulatory therapeutic properties as well as toxicity to the host cell, whereas potential applications encompass. OBJECTIVE This review article aims to describe the recent developments of lipopolysaccharides in nanocarrier systems for various applications such as vaccination, cancer chemotherapy and immune stimulants action. Different nanocarriers like cubosomes, niosomes, dendrimers and metal nanoparticles used in the delivery of actives are employed to decorate lipopolysaccharide molecules superficially. METHODS A narrative review of all the relevant papers known to the author was conducted. CONCLUSION Commercially available lipid nanoparticles contribute to many advances as promising nanocarriers in cancer therapy and are used as a vaccine adjuvant by improving the immune response due to their properties such as size, shape, biocompatibility, and biodegradability. Whereas lipopolysaccharide-decorated nanoparticles change the host's tolerability and increase the effectiveness of molecule in cancer immunotherapy. These nanoconjugate systems enhance overall immunogenic response and effectiveness in vaccine immunotherapy and targeted therapy, not only limited to humans application but also for poultry and aquaculture. Newer opportunities using lipopolysaccharides for the treatment and management of diseases with unique characteristics like the presence of lipoprotein that act as an alternative for bacterial infections over conventional dosage forms.
Collapse
Affiliation(s)
- Pravin Shende
- Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM'S NMIMS, V. L. Mehta Road, Vile Parle (W), Mumbai. India
| | - Shubham Gupta
- Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM'S NMIMS, V. L. Mehta Road, Vile Parle (W), Mumbai. India
| |
Collapse
|
11
|
Cannon J, Tang S, Choi SK. Caged Oxime Reactivators Designed for the Light Control of Acetylcholinesterase Reactivation †. Photochem Photobiol 2021; 98:334-346. [PMID: 34558680 DOI: 10.1111/php.13530] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 09/17/2021] [Accepted: 09/22/2021] [Indexed: 01/13/2023]
Abstract
Despite its promising role in the active control of biological functions by light, photocaging remains untested in acetylcholinesterase (AChE), a key enzyme in the cholinergic family. Here, we describe synthesis, photochemical properties and biochemical activities of two caged oxime compounds applied in the photocontrolled reactivation of the AChE inactivated by reactive organophosphate. Each of these consists of a photocleavable coumarin cage tethered to a known oxime reactivator for AChE that belongs in an either 2-(hydroxyimino)acetamide or pyridiniumaldoxime class. Of these, the first caged compound was able to successfully go through oxime uncaging upon irradiation at long-wavelength ultraviolet light (365 nm) or visible light (420 nm). It was further evaluated in AChE assays in vitro under variable light conditions to define its activity in the photocontrolled reactivation of paraoxon-inactivated AChE. This assay result showed its lack of activity in the dark but its induction of activity under light conditions only. In summary, this article reports a first class of light-activatable modulators for AChE and it offers assay methods and novel insights that help to achieve an effective design of caged compounds in the enzyme control.
Collapse
Affiliation(s)
- Jayme Cannon
- Michigan Nanotechnology Institute for Medicine and Biological Sciences, University of Michigan Medical School, Ann Arbor, Michigan, USA.,Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Shengzhuang Tang
- Michigan Nanotechnology Institute for Medicine and Biological Sciences, University of Michigan Medical School, Ann Arbor, Michigan, USA.,Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Seok Ki Choi
- Michigan Nanotechnology Institute for Medicine and Biological Sciences, University of Michigan Medical School, Ann Arbor, Michigan, USA.,Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
| |
Collapse
|
12
|
Targeted polymer-based antibiotic delivery system: A promising option for treating bacterial infections via macromolecular approaches. Prog Polym Sci 2021. [DOI: 10.1016/j.progpolymsci.2021.101389] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
13
|
Jiang G, Liu S, Yu T, Wu R, Ren Y, van der Mei HC, Liu J, Busscher HJ. PAMAM dendrimers with dual-conjugated vancomycin and Ag-nanoparticles do not induce bacterial resistance and kill vancomycin-resistant Staphylococci. Acta Biomater 2021; 123:230-243. [PMID: 33508504 DOI: 10.1016/j.actbio.2021.01.032] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Revised: 01/19/2021] [Accepted: 01/21/2021] [Indexed: 12/12/2022]
Abstract
The effective life-time of new antimicrobials until the appearance of the first resistant strains is steadily decreasing, which discourages incentives for commercialization required for clinical translation and application. Therefore, development of new antimicrobials should not only focus on better and better killing of antimicrobial-resistant strains, but as a paradigm shift on developing antimicrobials that prevent induction of resistance. Heterofunctionalized, poly-(amido-amine) (PAMAM) dendrimers with amide-conjugated vancomycin (Van) and incorporated Ag nanoparticles (AgNP) showed a 6-7 log reduction in colony-forming-units of a vancomycin-resistant Staphylococcus aureus strain in vitro, while not inducing resistance in a vancomycin-susceptible strain. Healing of a superficial wound in mice infected with the vancomycin-resistant S. aureus was significantly faster and more effective by irrigation with low-dose, dual-conjugated Van-PAMAM-AgNP dendrimer suspension than by irrigation with vancomycin in solution or a PAMAM-AgNP dendrimer suspension. Herewith, dual-conjugation of vancomycin together with AgNPs in heterofunctionalized PAMAM dendrimers fulfills the need for new, prolonged life-time antimicrobials killing resistant pathogens without inducing resistance in susceptible strains. Important for clinical translation, this better use of antibiotics can be achieved with currently approved and clinically applied antibiotics, provided suitable for amide-conjugation.
Collapse
Affiliation(s)
- Guimei Jiang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, 199 Ren'ai Rd, Suzhou 215123, Jiangsu, P. R. China; University of Groningen and University Medical Center Groningen, Department of Biomedical Engineering, Antonius Deusinglaan 1, 9713 AV Groningen, Netherlands
| | - Sidi Liu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, 199 Ren'ai Rd, Suzhou 215123, Jiangsu, P. R. China; University of Groningen and University Medical Center Groningen, Department of Biomedical Engineering, Antonius Deusinglaan 1, 9713 AV Groningen, Netherlands
| | - Tianrong Yu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, 199 Ren'ai Rd, Suzhou 215123, Jiangsu, P. R. China; University of Groningen and University Medical Center Groningen, Department of Biomedical Engineering, Antonius Deusinglaan 1, 9713 AV Groningen, Netherlands
| | - Renfei Wu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, 199 Ren'ai Rd, Suzhou 215123, Jiangsu, P. R. China; University of Groningen and University Medical Center Groningen, Department of Biomedical Engineering, Antonius Deusinglaan 1, 9713 AV Groningen, Netherlands
| | - Yijin Ren
- University of Groningen and University Medical Center of Groningen, Department of Orthodontics, Hanzeplein 1, 9700 RB Groningen, Netherlands
| | - Henny C van der Mei
- University of Groningen and University Medical Center Groningen, Department of Biomedical Engineering, Antonius Deusinglaan 1, 9713 AV Groningen, Netherlands.
| | - Jian Liu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, 199 Ren'ai Rd, Suzhou 215123, Jiangsu, P. R. China.
| | - Henk J Busscher
- University of Groningen and University Medical Center Groningen, Department of Biomedical Engineering, Antonius Deusinglaan 1, 9713 AV Groningen, Netherlands
| |
Collapse
|
14
|
Ryan LS, Nakatsuka A, Lippert AR. Photoactivatable 1,2-dioxetane chemiluminophores. RESULTS IN CHEMISTRY 2021. [DOI: 10.1016/j.rechem.2021.100106] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
|
15
|
Wei T, Jiang L, Chen Y, Chen X. Recent Progress of Photocage Molecules and Materials. ACTA CHIMICA SINICA 2021. [DOI: 10.6023/a20080361] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
16
|
Shchelik IS, Sieber S, Gademann K. Green Algae as a Drug Delivery System for the Controlled Release of Antibiotics. Chemistry 2020; 26:16644-16648. [PMID: 32910832 PMCID: PMC7894466 DOI: 10.1002/chem.202003821] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Indexed: 12/15/2022]
Abstract
New strategies to efficiently treat bacterial infections are crucial to circumvent the increase of resistant strains and to mitigate side effects during treatment. Skin and soft tissue infections represent one of the areas suffering the most from these resistant strains. We developed a new drug delivery system composed of the green algae, Chlamydomonas reinhardtii, which is generally recognized as safe, to target specifically skin diseases. A two-step functionalization strategy was used to chemically modify the algae with the antibiotic vancomycin. Chlamydomonas reinhardtii was found to mask vancomycin and the insertion of a photocleavable linker was used for the release of the antibiotic. This living drug carrier was evaluated in presence of Bacillus subtilis and, only upon UVA1-mediated release, growth inhibition of bacteria was observed. These results represent one of the first examples of a living organism used as a drug delivery system for the release of an antibiotic by UVA1-irradiation.
Collapse
Affiliation(s)
- Inga S. Shchelik
- Department of ChemistryUniversity of ZurichWinterthurerstrasse 1908057ZurichSwitzerland
| | - Simon Sieber
- Department of ChemistryUniversity of ZurichWinterthurerstrasse 1908057ZurichSwitzerland
| | - Karl Gademann
- Department of ChemistryUniversity of ZurichWinterthurerstrasse 1908057ZurichSwitzerland
| |
Collapse
|
17
|
Choi SK. Photoactivation Strategies for Therapeutic Release in Nanodelivery Systems. ADVANCED THERAPEUTICS 2020. [DOI: 10.1002/adtp.202000117] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Seok Ki Choi
- Michigan Nanotechnology Institute for Medicine and Biological Sciences University of Michigan Medical School Ann Arbor MI 48109 USA
- Department of Internal Medicine University of Michigan Medical School Ann Arbor MI 48109 USA
| |
Collapse
|
18
|
Svenningsen SW, Frederiksen RF, Counil C, Ficker M, Leisner JJ, Christensen JB. Synthesis and Antimicrobial Properties of a Ciprofloxacin and PAMAM-dendrimer Conjugate. Molecules 2020; 25:molecules25061389. [PMID: 32197523 PMCID: PMC7146445 DOI: 10.3390/molecules25061389] [Citation(s) in RCA: 9] [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: 02/18/2020] [Revised: 03/13/2020] [Accepted: 03/17/2020] [Indexed: 02/01/2023] Open
Abstract
Infections caused by bacteria resistant to antibiotics are an increasing problem. Multivalent antibiotics could be a solution. In the present study, a covalent conjugate between Ciprofloxacin and a G0-PAMAM dendrimer has been synthesized and tested against clinically relevant Gram-positive and Gram-negative bacteria. The conjugate has antimicrobial activity and there is a positive dendritic effect compared to Ciprofloxacin itself.
Collapse
Affiliation(s)
- Søren Wedel Svenningsen
- Department of Chemistry, University of Copenhagen, Thorvaldsensvej 40, DK-1871 Frederiksberg C, Denmark; (S.W.S.); (C.C.); (M.F.)
| | - Rikki Franklin Frederiksen
- Department of Veterinary and Animal Sciences, Food Safety and Zoonoses, University of Copenhagen, Grønnegårdsvej 15, DK-1870 Frederiksberg C, Denmark; (R.F.F.); (J.J.L.)
| | - Claire Counil
- Department of Chemistry, University of Copenhagen, Thorvaldsensvej 40, DK-1871 Frederiksberg C, Denmark; (S.W.S.); (C.C.); (M.F.)
| | - Mario Ficker
- Department of Chemistry, University of Copenhagen, Thorvaldsensvej 40, DK-1871 Frederiksberg C, Denmark; (S.W.S.); (C.C.); (M.F.)
| | - Jørgen J. Leisner
- Department of Veterinary and Animal Sciences, Food Safety and Zoonoses, University of Copenhagen, Grønnegårdsvej 15, DK-1870 Frederiksberg C, Denmark; (R.F.F.); (J.J.L.)
| | - Jørn Bolstad Christensen
- Department of Chemistry, University of Copenhagen, Thorvaldsensvej 40, DK-1871 Frederiksberg C, Denmark; (S.W.S.); (C.C.); (M.F.)
- Correspondence: ; Tel.: +45-3533-2452
| |
Collapse
|
19
|
Tang S, Cannon J, Yang K, Krummel MF, Baker JR, Choi SK. Spacer-Mediated Control of Coumarin Uncaging for Photocaged Thymidine. J Org Chem 2020; 85:2945-2955. [PMID: 32020803 PMCID: PMC7293860 DOI: 10.1021/acs.joc.9b02617] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Despite its importance in the design of photocaged molecules, less attention is focused on linker chemistry than the cage itself. Here, we describe unique uncaging properties displayed by two coumarin-caged thymidine compounds, each conjugated with (2) or without (1) an extended, self-immolative spacer. Photolysis of 1 using long-wavelength UVA (365 nm) or visible (420, 455 nm) light led to the release of free thymidine along with the competitive generation of a thymidine-bearing recombination product. The occurrence of this undesired side reaction, which is previously unreported, was not present with the photolysis of 2, which released thymidine exclusively with higher quantum efficiency. We propose that the spatial separation between the cage and the substrate molecule conferred by the extended linker can play a critical role in circumventing this unproductive reaction. This report reinforces the importance of linker selection in the design of coumarin-caged oligonucleosides and other conjugates.
Collapse
Affiliation(s)
- Shengzhuang Tang
- Michigan Nanotechnology Institute for Medicine and Biological Sciences, Ann Arbor, Michigan 48109, United States of America
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan 48109, United States of America
| | - Jayme Cannon
- Michigan Nanotechnology Institute for Medicine and Biological Sciences, Ann Arbor, Michigan 48109, United States of America
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan 48109, United States of America
| | - Kelly Yang
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan 48109, United States of America
| | - Matthew F. Krummel
- Department of Pathology, University of California, San Francisco, 513 Parnassus Ave, HSW512, San Francisco, California 94143, United States of America
| | - James R. Baker
- Michigan Nanotechnology Institute for Medicine and Biological Sciences, Ann Arbor, Michigan 48109, United States of America
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan 48109, United States of America
| | - Seok Ki Choi
- Michigan Nanotechnology Institute for Medicine and Biological Sciences, Ann Arbor, Michigan 48109, United States of America
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan 48109, United States of America
| |
Collapse
|
20
|
Cheng AV, Wuest WM. Signed, Sealed, Delivered: Conjugate and Prodrug Strategies as Targeted Delivery Vectors for Antibiotics. ACS Infect Dis 2019; 5:816-828. [PMID: 30969100 PMCID: PMC6570538 DOI: 10.1021/acsinfecdis.9b00019] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Innate and developed resistance mechanisms of bacteria to antibiotics are obstacles in the design of novel drugs. However, antibacterial prodrugs and conjugates have shown promise in circumventing resistance and tolerance mechanisms via directed delivery of antibiotics to the site of infection or to specific species or strains of bacteria. The selective targeting and increased permeability and accumulation of these prodrugs not only improves efficacy over unmodified drugs but also reduces off-target effects, toxicity, and development of resistance. Herein, we discuss some of these methods, including sideromycins, antibody-directed prodrugs, cell penetrating peptide conjugates, and codrugs.
Collapse
Affiliation(s)
- Ana V. Cheng
- Department of Chemistry, Emory University, 1515 Dickey Drive, Atlanta, Georgia 30322, United States
| | - William M. Wuest
- Department of Chemistry, Emory University, 1515 Dickey Drive, Atlanta, Georgia 30322, United States
- Emory Antibiotic Resistance Center, Emory School of Medicine, 201 Dowman Drive, Atlanta, Georgia 30322, United States
| |
Collapse
|
21
|
Kamaruzzaman NF, Tan LP, Hamdan RH, Choong SS, Wong WK, Gibson AJ, Chivu A, Pina MDF. Antimicrobial Polymers: The Potential Replacement of Existing Antibiotics? Int J Mol Sci 2019; 20:E2747. [PMID: 31167476 PMCID: PMC6600223 DOI: 10.3390/ijms20112747] [Citation(s) in RCA: 142] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 04/08/2019] [Accepted: 04/11/2019] [Indexed: 12/22/2022] Open
Abstract
Antimicrobial resistance is now considered a major global challenge; compromising medical advancements and our ability to treat infectious disease. Increased antimicrobial resistance has resulted in increased morbidity and mortality due to infectious diseases worldwide. The lack of discovery of novel compounds from natural products or new classes of antimicrobials, encouraged us to recycle discontinued antimicrobials that were previously removed from routine use due to their toxicity, e.g., colistin. Since the discovery of new classes of compounds is extremely expensive and has very little success, one strategy to overcome this issue could be the application of synthetic compounds that possess antimicrobial activities. Polymers with innate antimicrobial properties or that have the ability to be conjugated with other antimicrobial compounds create the possibility for replacement of antimicrobials either for the direct application as medicine or implanted on medical devices to control infection. Here, we provide the latest update on research related to antimicrobial polymers in the context of ESKAPE (Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, and Enterobacter spp.) pathogens. We summarise polymer subgroups: compounds containing natural peptides, halogens, phosphor and sulfo derivatives and phenol and benzoic derivatives, organometalic polymers, metal nanoparticles incorporated into polymeric carriers, dendrimers and polymer-based guanidine. We intend to enhance understanding in the field and promote further work on the development of polymer based antimicrobial compounds.
Collapse
Affiliation(s)
- Nor Fadhilah Kamaruzzaman
- Faculty of Veterinary Medicine, Locked bag 36, Universiti Malaysia Kelantan, Pengkalan Chepa 16100, Kelantan, Malaysia.
| | - Li Peng Tan
- Faculty of Veterinary Medicine, Locked bag 36, Universiti Malaysia Kelantan, Pengkalan Chepa 16100, Kelantan, Malaysia.
| | - Ruhil Hayati Hamdan
- Faculty of Veterinary Medicine, Locked bag 36, Universiti Malaysia Kelantan, Pengkalan Chepa 16100, Kelantan, Malaysia.
| | - Siew Shean Choong
- Faculty of Veterinary Medicine, Locked bag 36, Universiti Malaysia Kelantan, Pengkalan Chepa 16100, Kelantan, Malaysia.
| | - Weng Kin Wong
- School of Health Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Kelantan, Malaysia.
| | - Amanda Jane Gibson
- Royal Veterinary College, Pathobiology and Population Sciences, Hawkshead Lane, North Mymms, Hatfield AL9 7TA, UK.
| | - Alexandru Chivu
- UCL Centre for Nanotechnology and Regenerative Medicine, Division of Surgery & Interventional Science, University College London, London NW3 2PF, UK.
| | - Maria de Fatima Pina
- Medicines and Healthcare Regulatory Products Agency, 10 South Colonnade, Canary Wharf, London E14 4PU, UK.
| |
Collapse
|
22
|
Lu C, Su Z, Jing D, Jin S, Xie L, Li L, Zheng K. Intramolecular Reductive Cyclization of o-Nitroarenes via Biradical Recombination. Org Lett 2019; 21:1438-1443. [PMID: 30767542 DOI: 10.1021/acs.orglett.9b00191] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
A visible-light-induced/thiourea-mediated intramolecular cyclization of o-nitroarenes under mild conditions is realized for the first time, which provides an efficient and environmentally friendly way to access pharmaceutical relevant quinazolinone derivatives. The reaction can be easily extended to gram level by using a continuous-flow setup with high efficiency. Mechanistic investigation including control experiments, transient fluorescence, UV-vis spectra, and DFT calculations suggests that the formation of active biradical intermediates via intramolecular single electron transfer (SET) is key stage in the catalytic cycle.
Collapse
Affiliation(s)
- Cong Lu
- Key Laboratory of Green Chemistry & Technology, Ministry of Education, College of Chemistry , Sichuan University , Chengdu 610064 , P. R. China
| | - Zhishan Su
- Key Laboratory of Green Chemistry & Technology, Ministry of Education, College of Chemistry , Sichuan University , Chengdu 610064 , P. R. China
| | - Dong Jing
- Key Laboratory of Green Chemistry & Technology, Ministry of Education, College of Chemistry , Sichuan University , Chengdu 610064 , P. R. China
| | - Songyang Jin
- Key Laboratory of Green Chemistry & Technology, Ministry of Education, College of Chemistry , Sichuan University , Chengdu 610064 , P. R. China
| | - Lijuan Xie
- Key Laboratory of Green Chemistry & Technology, Ministry of Education, College of Chemistry , Sichuan University , Chengdu 610064 , P. R. China
| | - Liangrui Li
- Key Laboratory of Green Chemistry & Technology, Ministry of Education, College of Chemistry , Sichuan University , Chengdu 610064 , P. R. China
| | - Ke Zheng
- Key Laboratory of Green Chemistry & Technology, Ministry of Education, College of Chemistry , Sichuan University , Chengdu 610064 , P. R. China
| |
Collapse
|
23
|
Pardeshi KA, Kumar TA, Ravikumar G, Shukla M, Kaul G, Chopra S, Chakrapani H. Targeted Antibacterial Activity Guided by Bacteria-Specific Nitroreductase Catalytic Activation to Produce Ciprofloxacin. Bioconjug Chem 2019; 30:751-759. [DOI: 10.1021/acs.bioconjchem.8b00887] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Kundansingh A. Pardeshi
- Department of Chemistry, Indian Institute of Science Education and Research Pune, Dr. Homi Bhabha Road, Pune-411 008, Maharashtra, India
| | - T. Anand Kumar
- Department of Chemistry, Indian Institute of Science Education and Research Pune, Dr. Homi Bhabha Road, Pune-411 008, Maharashtra, India
| | - Govindan Ravikumar
- Department of Chemistry, Indian Institute of Science Education and Research Pune, Dr. Homi Bhabha Road, Pune-411 008, Maharashtra, India
| | - Manjulika Shukla
- Division of Microbiology, CSIR-Central Drug Research Institute, Lucknow-226031, Uttar Pradesh, India
| | - Grace Kaul
- Division of Microbiology, CSIR-Central Drug Research Institute, Lucknow-226031, Uttar Pradesh, India
| | - Sidharth Chopra
- Division of Microbiology, CSIR-Central Drug Research Institute, Lucknow-226031, Uttar Pradesh, India
| | - Harinath Chakrapani
- Department of Chemistry, Indian Institute of Science Education and Research Pune, Dr. Homi Bhabha Road, Pune-411 008, Maharashtra, India
| |
Collapse
|
24
|
Paik P, Kumar KS, Modak MD, Kumar U K, Maity S. UCN-SiO 2-GO: a core shell and conjugate system for controlling delivery of doxorubicin by 980 nm NIR pulse. RSC Adv 2018; 8:37492-37502. [PMID: 35557801 PMCID: PMC9089400 DOI: 10.1039/c8ra07030j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 10/24/2018] [Indexed: 11/21/2022] Open
Abstract
Herein, graphene oxide (GO) has been attached with core-shell upconversion-silica (UCN-SiO2) nanoparticles (NPs) to form a GO-UCN-SiO2 hybrid nanocomposite and used for controlled drug delivery. The formation of the nanocomposite has been confirmed by various characterization techniques. To date, a number of reports are available on GO and its drug delivery applications, however, the synergic properties that arise due to the combination of GO, UCNPs and SiO2 can be used for controlled drug delivery. New composite UCN@SiO2-GO has been synthesized through a bio-conjugation approach and used for drug delivery applications to counter the lack of quantum efficiency of the upconversion process and control sustained release. A model anticancer drug (doxorubicin, DOX) has been loaded to UCNPs, UCN@SiO2 NPs and the UCN@SiO2-GO nanocomposite. The photosensitive release of DOX from the UCN@SiO2-GO nanocomposite has been studied with 980 nm NIR laser excitation and the results obtained for UCNPs and UCN@SiO2 NPs compared. It is revealed that the increase in the NIR laser irradiation time from 1 s to 30 s leads to an increase in the amount of DOX release in a controlled manner. In vitro studies using model cancer cell lines have been performed to check the effectiveness of our materials for controlled drug delivery and therapeutic applications. Obtained results showed that the designed UCN@SiO2-GO nanocomposite can be used for controlled delivery based therapeutic applications and for cancer treatment.
Collapse
Affiliation(s)
- Pradip Paik
- School of Biomedical Engineering, Indian Institute of Technology (BHU) Varanasi 221 005 UP India
- School of Engineering Sciences and Technology, University of Hyderabad Hyderabad-500046 Telangana India (+91)-(040) 2313 4457 (O)
| | - K Santhosh Kumar
- School of Engineering Sciences and Technology, University of Hyderabad Hyderabad-500046 Telangana India (+91)-(040) 2313 4457 (O)
| | - Monami Das Modak
- School of Engineering Sciences and Technology, University of Hyderabad Hyderabad-500046 Telangana India (+91)-(040) 2313 4457 (O)
| | - Koushi Kumar U
- School of Engineering Sciences and Technology, University of Hyderabad Hyderabad-500046 Telangana India (+91)-(040) 2313 4457 (O)
| | - Somedutta Maity
- School of Biomedical Engineering, Indian Institute of Technology (BHU) Varanasi 221 005 UP India
- School of Engineering Sciences and Technology, University of Hyderabad Hyderabad-500046 Telangana India (+91)-(040) 2313 4457 (O)
| |
Collapse
|
25
|
Gulla S, Kotcherlakota R, Nimushakavi S, Nimmu NV, Khalid S, Patra CR, Chaudhuri A. Au-CGKRK Nanoconjugates for Combating Cancer through T-Cell-Driven Therapeutic RNA Interference. ACS OMEGA 2018; 3:8663-8676. [PMID: 31458997 PMCID: PMC6644890 DOI: 10.1021/acsomega.8b01051] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Accepted: 07/23/2018] [Indexed: 05/22/2023]
Abstract
Numerous prior studies on fighting cancer have been based on using inhibitors of JAK-STAT pathway (signal transducer and activator of transcription 3 (STAT3) inhibitor in particular), a signaling pathway responsible for progression of many types of cancer cells. However, recent studies have shown that STAT3 activation leads to upregulation of program death receptor-ligand 1 (PD-L1, an immune checkpoint protein that plays a major role behind evasion of immune systems by growing tumors) expression levels in tumor cells, leading to enhanced immune suppression. This is why global efforts are being witnessed in combating cancer through use of immune checkpoint inhibitors. Herein, we report on the design, synthesis, physicochemical characterizations, and bioactivity evaluation of novel tumor- and tumor-vasculature-targeting noncytotoxic Au-CGKRK nanoconjugates (17-80 nm) for combating tumor. Using a syngeneic mouse tumor model, we show that intraperitoneal (i.p.) administration of the Au-CGKRK nanoparticles (NPs) complexed with both PD-L1siRNA (the immune checkpoint inhibitor) and STAT3siRNA (the JAK-STAT pathway inhibitor) results in significant (>70%) enhancement in overall survivability (OS) in melanoma-bearing mice (n = 5) when compared to the OS in the untreated mice group. The expression levels of CD8 and CD4 proteins in the tumor lysates of differently treated mice groups (by Western blotting) are consistent with the observed OS enhancement being a T-cell-driven process. Biodistribution study using near-infrared dye-loaded Au-CGKRK nanoconjugates revealed selective accumulation of the dye in mouse tumor. Notably, the overall survival benefits were significantly less (∼35%) when melanoma-bearing mice were treated (i.p.) with Au-CGKRK NPs complexed with only PD-L1siRNA or with STAT3siRNA alone. The presently described Au-CGKRK nanoconjugates are expected to find future use in therapeutic RNA-interference-based cancer immunotherapy.
Collapse
Affiliation(s)
- Suresh
Kumar Gulla
- Division
of Applied Biology and Analytical & Mass Chemistry Division, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, Telangana, India
- Academy
of Scientific and Innovative Research, CSIR
Campus, CSIR Road, Taramani, Chennai 600113, Tamil Nadu, India
| | - Rajesh Kotcherlakota
- Division
of Applied Biology and Analytical & Mass Chemistry Division, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, Telangana, India
- Academy
of Scientific and Innovative Research, CSIR
Campus, CSIR Road, Taramani, Chennai 600113, Tamil Nadu, India
| | - Sahithi Nimushakavi
- Division
of Applied Biology and Analytical & Mass Chemistry Division, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, Telangana, India
- Academy
of Scientific and Innovative Research, CSIR
Campus, CSIR Road, Taramani, Chennai 600113, Tamil Nadu, India
| | - Narendra Varma Nimmu
- Division
of Applied Biology and Analytical & Mass Chemistry Division, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, Telangana, India
| | - Sara Khalid
- Division
of Applied Biology and Analytical & Mass Chemistry Division, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, Telangana, India
| | - Chitta Ranjan Patra
- Division
of Applied Biology and Analytical & Mass Chemistry Division, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, Telangana, India
- Academy
of Scientific and Innovative Research, CSIR
Campus, CSIR Road, Taramani, Chennai 600113, Tamil Nadu, India
| | - Arabinda Chaudhuri
- Division
of Applied Biology and Analytical & Mass Chemistry Division, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, Telangana, India
- Academy
of Scientific and Innovative Research, CSIR
Campus, CSIR Road, Taramani, Chennai 600113, Tamil Nadu, India
| |
Collapse
|
26
|
Peterson E, Joseph C, Peterson H, Bouwman R, Tang S, Cannon J, Sinniah K, Choi SK. Measuring the Adhesion Forces for the Multivalent Binding of Vancomycin-Conjugated Dendrimer to Bacterial Cell-Wall Peptide. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2018; 34:7135-7146. [PMID: 29792710 DOI: 10.1021/acs.langmuir.8b01137] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
Multivalent ligand-receptor interaction provides the fundamental basis for the hypothetical notion that high binding avidity relates to the strong force of adhesion. Despite its increasing importance in the design of targeted nanoconjugates, an understanding of the physical forces underlying the multivalent interaction remains a subject of urgent investigation. In this study, we designed three vancomycin (Van)-conjugated dendrimers G5(Van) n ( n = mean valency = 0, 1, 4) for bacterial targeting with generation 5 (G5) poly(amidoamine) dendrimer as a multivalent scaffold and evaluated both their binding avidity and physical force of adhesion to a bacterial model surface by employing surface plasmon resonance (SPR) spectroscopy and atomic force microscopy. The SPR experiment for these conjugates was performed in a biosensor chip surface immobilized with a bacterial cell-wall peptide Lys-d-Ala-d-Ala. Of these, G5(Van)4 bound most tightly with a KD of 0.34 nM, which represents an increase in avidity by 2 or 3 orders of magnitude relative to a monovalent conjugate G5(Van)1 or free vancomycin, respectively. By single-molecule force spectroscopy, we measured the adhesion force between G5(Van) n and the same cell-wall peptide immobilized on the surface. The distribution of adhesion forces increased in proportion to vancomycin valency with the mean force of 134 pN at n = 4 greater than 96 pN at n = 1 at a loading rate of 5200 pN/s. In summary, our results are strongly supportive of the positive correlation between the avidity and adhesion force in the multivalent interaction of vancomycin nanoconjugates.
Collapse
Affiliation(s)
- Elizabeth Peterson
- Department of Chemistry & Biochemistry , Calvin College , Grand Rapids , Michigan 49546 , United States
| | | | - Hannah Peterson
- Department of Chemistry & Biochemistry , Calvin College , Grand Rapids , Michigan 49546 , United States
| | - Rachael Bouwman
- Department of Chemistry & Biochemistry , Calvin College , Grand Rapids , Michigan 49546 , United States
| | | | | | - Kumar Sinniah
- Department of Chemistry & Biochemistry , Calvin College , Grand Rapids , Michigan 49546 , United States
| | | |
Collapse
|
27
|
Bowden S, Joseph C, Tang S, Cannon J, Francis E, Zhou M, Baker JR, Choi SK. Oritavancin Retains a High Affinity for a Vancomycin-Resistant Cell-Wall Precursor via Its Bivalent Motifs of Interaction. Biochemistry 2018; 57:2723-2732. [PMID: 29651842 DOI: 10.1021/acs.biochem.8b00187] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Despite its potent antibacterial activities against drug-resistant Gram-positive pathogens, oritavancin remains partially understood with respect to its primary mode of hydrogen bond interaction with a cell-wall peptide regarding the role of its lipophilic 4'-chlorobiphenyl moiety. Here we report a surface plasmon resonance (SPR) study performed in two cell-wall model surfaces, each prepared by immobilization with a vancomycin-susceptible Lys-d-Ala-d-Ala or vancomycin-resistant Lys-d-Ala-d-Lac peptide. Analysis of binding kinetics performed on the peptide surface showed that oritavancin bound ∼100-1000-fold more tightly than vancomycin on each model surface. Ligand competition experiments conducted by SPR and fluorescence spectroscopy provided evidence that such affinity enhancement can be attributed to its 4'-chlorobiphenyl moiety, possibly through a hydrophobic interaction that led to a gain of free energy with a contribution from enthalpy as suggested by a variable-temperature SPR experiment. On the basis of these findings, we propose a model for the bivalent motifs of interaction of oritavancin with cell-wall peptides, by which the drug molecule can retain a strong interaction even with the vancomycin-resistant peptide. In summary, this study advances our understanding of oritavancin and offers new insight into the significance of bivalent motifs in the design of glycopeptide antibiotics.
Collapse
|
28
|
Xu F, Hu M, Liu C, Choi SK. Yolk-structured multifunctional up-conversion nanoparticles for synergistic photodynamic-sonodynamic antibacterial resistance therapy. Biomater Sci 2018; 5:678-685. [PMID: 28280817 DOI: 10.1039/c7bm00030h] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The worldwide increase in bacterial antibiotic resistance has led to a search for alternative antibacterial therapies. The present study reports the development of yolk-structured multifunctional up-conversion nanoparticles (UCNPs) that combine photodynamic and sonodynamic therapy for effective killing of antibiotic-resistant bacteria. The multifunctional nanoparticles (NPs) were achieved by enclosing hematoporphyrin monomethyl ether (HMME) into its yolk-structured up-conversion core and covalently linked rose bengal (RB) on its silica (SiO2) shell. Excitation of UCNPs with near-infrared (NIR) light that has improved penetration depth for photodynamic therapy (PDT) enabled the activation of HMME and RB and thus the generation of singlet oxygen (1O2). The SiO2 layer, which improved the biocompatibility of the UCNPs, surrounded the yolk structure, with a cavity space which had a high efficiency of loading photosensitizers. Synergistic PDT and sonodynamic therapy (SDT) improved the photosensitizer utilization rate. As a result, a greater inhibition rate was observed when antibiotic-resistant bacteria were treated with a combined therapy (100%) compared with either the PDT (74.2%) or SDT (70%) alone. Our data indicate that the multifunctional NPs developed in this study have the potential for use in the clinical synergistic PDT-SDT treatment of infectious diseases caused by antibiotic-resistant bacteria.
Collapse
Affiliation(s)
- Feiya Xu
- Department of Chemistry, School of Science, Xi'an Jiaotong University, Xi'an 710049, China.
| | - Min Hu
- Department of Chemistry, School of Science, Xi'an Jiaotong University, Xi'an 710049, China. and State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan University, Changsha 410082, P.R. China
| | - Chengcheng Liu
- Department of Pathogenic Microbiology & Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an 710061, China.
| | - Seok Ki Choi
- Department of Internal Medicine, Michigan Nanotechnology Institute for Medicine and Biological Sciences, University of Michigan, Ann Arbor, MI 48109-1055, USA
| |
Collapse
|
29
|
Kumari P, Kulkarni A, Sharma AK, Chakrapani H. Visible-Light Controlled Release of a Fluoroquinolone Antibiotic for Antimicrobial Photopharmacology. ACS OMEGA 2018; 3:2155-2160. [PMID: 30023825 PMCID: PMC6045474 DOI: 10.1021/acsomega.7b01906] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Accepted: 02/08/2018] [Indexed: 06/01/2023]
Abstract
Owing to the dwindling arsenal of antibiotics, new methodologies for their effective and localized delivery are necessary. The use of optical control over delivery of drugs, also known as photopharmacology, has emerged as an important option for the spatiotemporally controlled generation of drugs and bioactive molecules. In the field of antimicrobial photopharmacology, most strategies utilize ultraviolet light for triggering release of the antibiotic. The use of such short wavelength light may have limitations such as phototoxicity. Here, a small molecule that is activated by visible light to release a fluoroquinolone, a broad-spectrum antibiotic, is reported. A boron-dipyrromethene, which is sensitive to cleavage at 470 nm, was used, and levofloxacin was used as a model fluoroquinolone. BDP-Levo was found to undergo cleavage in the presence of visible light to release the active antibiotic. Using growth inhibitory studies in Gram-positive as well as Gram-negative bacteria, the efficacy of BDP-Levo is demonstrated. Together, our study demonstrates that visible light can be used for optical control over antibiotic release and lays the foundation for visible-light-mediated antimicrobial photopharmacology.
Collapse
Affiliation(s)
- Pooja Kumari
- Department of Chemistry, Indian Institute of Science Education and Research
Pune, Dr. Homi Bhabha Road, Pune 411008, Maharashtra, India
| | - Amogh Kulkarni
- Department of Chemistry, Indian Institute of Science Education and Research
Pune, Dr. Homi Bhabha Road, Pune 411008, Maharashtra, India
| | - Ajay Kumar Sharma
- Department of Chemistry, Indian Institute of Science Education and Research
Pune, Dr. Homi Bhabha Road, Pune 411008, Maharashtra, India
| | - Harinath Chakrapani
- Department of Chemistry, Indian Institute of Science Education and Research
Pune, Dr. Homi Bhabha Road, Pune 411008, Maharashtra, India
| |
Collapse
|
30
|
Wong PT, Tang S, Cannon J, Chen D, Sun R, Lee J, Phan J, Tao K, Sun K, Chen B, Baker JR, Choi SK. Photocontrolled Release of Doxorubicin Conjugated through a Thioacetal Photocage in Folate-Targeted Nanodelivery Systems. Bioconjug Chem 2017; 28:3016-3028. [PMID: 29148732 DOI: 10.1021/acs.bioconjchem.7b00614] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Despite their proven ability for precise and targeted release, nanoplatform systems for photocontrolled delivery often face formidable synthetic challenges, in part due to the paucity of advanced linker strategies. Here, we report on a novel linker strategy using a thioacetal ortho-nitrobenzaldehyde (TNB) cage, demonstrating its application for delivery of doxorubicin (Dox) in two nanoscale systems. This photocleavable linker, TNB(OH), which presents two identical arms, each terminated with a hydroxyl functionality, was prepared in a single step from 6-nitroveratraldehyde. TNB(OH) was used to cross-link Dox to a folate receptor (FAR)-targeting poly(amidoamine) dendrimer conjugate G5(FA)n=5.4(Dox)m=5.1, and also used to prepare an upconversion nanocrystal (UCN) conjugate, UCN-PPIX@(Dox)(G5FA), a larger core/shell nanostructure. In this core/shell nanostructure, the UCN core emits UV and visible light luminescence upon near-infrared (NIR) excitation, allowing for the photocleavage of the TNB linker as well as the photostimulation of protoporphyrin IX (PPIX) coupled as a cytotoxic photosensitizer. Drug-release experiments performed in aqueous solutions with long-wavelength ultraviolet A (UVA) light showed that Dox release occurred rapidly from its TNB linked form or from its dendrimer conjugated form with comparable decay kinetics. Cellular toxicity studies in FAR-overexpressing KB carcinoma cells demonstrated that each nanoconjugate lacked intrinsic cytotoxicity until exposed to UVA or NIR (980 nm) (for the UCN nanoconjugate), which resulted in induction of potent cytotoxicity. In summary, this new TNB strategy offers synthetic convenience in drug conjugation chemistry with the ability for the temporal control of drug activation at the delivery site.
Collapse
Affiliation(s)
| | | | | | - Dexin Chen
- State Key Laboratory of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiao Tong University , Shanghai 200240, People's Republic of China
| | | | | | | | - Ke Tao
- State Key Laboratory of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiao Tong University , Shanghai 200240, People's Republic of China
| | - Kang Sun
- State Key Laboratory of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiao Tong University , Shanghai 200240, People's Republic of China
| | - Biqiong Chen
- School of Mechanical and Aerospace Engineering, Queen's University Belfast , Stranmillis Road, Belfast BT9 5AH, United Kingdom
| | | | | |
Collapse
|
31
|
Alibolandi M, Taghdisi SM, Ramezani P, Hosseini Shamili F, Farzad SA, Abnous K, Ramezani M. Smart AS1411-aptamer conjugated pegylated PAMAM dendrimer for the superior delivery of camptothecin to colon adenocarcinoma in vitro and in vivo. Int J Pharm 2017; 519:352-364. [PMID: 28126548 DOI: 10.1016/j.ijpharm.2017.01.044] [Citation(s) in RCA: 105] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 01/17/2017] [Accepted: 01/21/2017] [Indexed: 12/17/2022]
Abstract
In the current study camptothecin-loaded pegylated PAMAM dendrimer were synthesized and were functionalized with AS1411 anti-nucleolin aptamers for site-specific targeting against colorectal cancer cells which over expresses nucleolin receptors. The morphological properties and size dispersity of the prepared nanoparticles were evaluated using transmission electron microscope (TEM) and DLS. The drug-loading content and encapsulation efficiency were obtained 8.1% and 93.67% respectively. The in vitro release of camptothecin from the formulation was provided the sustained release of encapsulated camptothecin during 4days. Comparative in vitro cytotoxicity experiments demonstrated that the targeted camptothecin loaded-pegylated dendrimers had higher antiproliferation activity, towards nucleolin-positive HT29 and C26 colorectal cancer cells than nucleolin-negative CHO cell line. Fluorscence microscopy and flow cytometry also confirmed the enhanced cellular uptake of AS1411 targeted pegylated-dendrimer. In vivo study in C26 tumor-bearing BALB/C mice revealed that the AS1411-functionalized camptothecin loaded pegylated dendrimers improved antitumor activity and survival rate of the encapsulated camptothecin. Conjugation of AS1411 aptamer to the camptothecin loaded-pegylated dendrimer surface provides site-specific delivery of camptothecin, inhibit C26 tumor growth in vivo and significantly decrease systemic toxicity. These results suggested that the new nucleolin-targeted pegylated PAMAM dendrimer as a delivery system for camptothecin have the potential for the treatment of nucleolin-overexpressed colorectal cancer.
Collapse
Affiliation(s)
- Mona Alibolandi
- Pharmaceutical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Mohammad Taghdisi
- Targeted Drug Delivery Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Pouria Ramezani
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fazileh Hosseini Shamili
- Department of Immunology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sara Amel Farzad
- Pharmaceutical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Khalil Abnous
- Pharmaceutical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Mohammad Ramezani
- Pharmaceutical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Nanotechnology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
32
|
Wong PT, Tang S, Cannon J, Mukherjee J, Isham D, Gam K, Payne M, Yanik SA, Baker JR, Choi SK. A Thioacetal Photocage Designed for Dual Release: Application in the Quantitation of Therapeutic Release by Synchronous Reporter Decaging. Chembiochem 2017; 18:126-135. [PMID: 27902870 PMCID: PMC5213739 DOI: 10.1002/cbic.201600494] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Indexed: 12/24/2022]
Abstract
Despite the immense potential of existing photocaging technology, its application is limited by the paucity of advanced caging tools. Here, we report on the design of a novel thioacetal ortho-nitrobenzaldehyde (TNB) dual arm photocage that enabled control of the simultaneous release of two payloads linked to a single TNB unit. By using this cage, which was prepared in a single step from commercial 6-nitroverataldehyde, three drug-fluorophore conjugates were synthesized: Taxol-TNB-fluorescein, Taxol-TNB-coumarin, and doxorubicin-TNB-coumarin, and long-wavelength UVA light-triggered release experiments demonstrated that dual payload release occurred with rapid decay kinetics for each conjugate. In cell-based assays performed in vitro, dual release could also be controlled by UV exposure, resulting in increased cellular fluorescence and cytotoxicity with potency equal to that of unmodified drug towards the KB carcinoma cell line. The extent of such dual release was quantifiable by reporter fluorescence measured in situ and was found to correlate with the extent of cytotoxicity. Thus, this novel dual arm cage strategy provides a valuable tool that enables both active control and real-time monitoring of drug activation at the delivery site.
Collapse
Affiliation(s)
- Pamela T Wong
- Michigan Nanotechnology Institute for Medicine and Biological Sciences, University of Michigan Medical School, 1150 W. Medical Ctr. Drive, Ann Arbor, MI, 48109, USA
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Shengzhuang Tang
- Michigan Nanotechnology Institute for Medicine and Biological Sciences, University of Michigan Medical School, 1150 W. Medical Ctr. Drive, Ann Arbor, MI, 48109, USA
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Jayme Cannon
- Michigan Nanotechnology Institute for Medicine and Biological Sciences, University of Michigan Medical School, 1150 W. Medical Ctr. Drive, Ann Arbor, MI, 48109, USA
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Jhindan Mukherjee
- Michigan Nanotechnology Institute for Medicine and Biological Sciences, University of Michigan Medical School, 1150 W. Medical Ctr. Drive, Ann Arbor, MI, 48109, USA
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Danielle Isham
- Michigan Nanotechnology Institute for Medicine and Biological Sciences, University of Michigan Medical School, 1150 W. Medical Ctr. Drive, Ann Arbor, MI, 48109, USA
| | - Kristina Gam
- Michigan Nanotechnology Institute for Medicine and Biological Sciences, University of Michigan Medical School, 1150 W. Medical Ctr. Drive, Ann Arbor, MI, 48109, USA
| | - Michael Payne
- Michigan Nanotechnology Institute for Medicine and Biological Sciences, University of Michigan Medical School, 1150 W. Medical Ctr. Drive, Ann Arbor, MI, 48109, USA
| | - Sean A Yanik
- Michigan Nanotechnology Institute for Medicine and Biological Sciences, University of Michigan Medical School, 1150 W. Medical Ctr. Drive, Ann Arbor, MI, 48109, USA
| | - James R Baker
- Michigan Nanotechnology Institute for Medicine and Biological Sciences, University of Michigan Medical School, 1150 W. Medical Ctr. Drive, Ann Arbor, MI, 48109, USA
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Seok Ki Choi
- Michigan Nanotechnology Institute for Medicine and Biological Sciences, University of Michigan Medical School, 1150 W. Medical Ctr. Drive, Ann Arbor, MI, 48109, USA
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| |
Collapse
|