We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine
Research Article

Conformationally restricted, dipeptide-based, self-assembled nanoparticles for efficient vancomycin delivery

    Nitin Yadav

    Molecular Medicine Group, International Centre for Genetic Engineering & Biotechnology, Aruna Asaf Ali Marg, New Delhi, 110067, India

    Delhi Institute of Pharmaceutical Sciences & Research, Mehrauli-Badarpur Road, Sector-3, Pushpvihar, New Delhi, 110017, India

    ‡These authors contributed equally to this work

    Search for more papers by this author

    ,
    Utkarsh Kumar

    Molecular Medicine Group, International Centre for Genetic Engineering & Biotechnology, Aruna Asaf Ali Marg, New Delhi, 110067, India

    ‡These authors contributed equally to this work

    Search for more papers by this author

    &
    Virander Singh Chauhan

    *Author for correspondence:

    E-mail Address: virander@icgeb.res.in

    Molecular Medicine Group, International Centre for Genetic Engineering & Biotechnology, Aruna Asaf Ali Marg, New Delhi, 110067, India

    Published Online:https://doi.org/10.2217/nnm-2022-0144

    Aim: Emergence of vancomycin (Van) resistance, and usage of its higher dose and short half-life are posing a serious concern. Slow and sustained release of Van using a nanodelivery system may overcome these problems. Materials & methods: Arginine-α,β-dehydrophenylalanine (RΔF) was synthesized using solution-phase synthesis which self-assembled into nanospheres. Van was entrapped in the nanoparticles (NPs). In vitro and in vivo efficacy of Van-RΔF was determined using broth microdilution and the mouse thigh infection model, respectively. Results & conclusion: Van-RΔF NPs efficiently inhibited bacterial growth (Staphylococcus aureus), while Van alone showed limited growth inhibition in in vitro. Intravenous administration of Van-RΔF in mice with bacterial thigh infection showed enhanced efficacy (double) compared with Van alone, which indicates its high potential for further development.

    Plain language summary

    Currently, microbial infections have become the most prevalent threat to human health. In the past few decades, researchers have tried different strategies to deal with these infections by developing new antimicrobial agents and/or improving the efficacy of already available antimicrobial agents. Controlled delivery of antimicrobial agents using nanosized vehicle systems has shown great promise in antimicrobial therapy. The authors have developed an ultrashort, modified, peptide-based nanoparticle system that can load vancomycin and release it in a controlled and sustained manner. Unlike other polymer-based nanoparticles, these dipeptide-based nanoparticles are easy to synthesize and highly biocompatible in nature. Vancomycin delivered via these peptide-based nanoparticles showed higher antibacterial activity than the drug alone. These results clearly indicated the high potential of this nanoformulation for further development as a delivery vehicle system for efficient antimicrobial therapy.

    Papers of special note have been highlighted as: • of interest

    References

    • 1. Rodvold KA, McConeghy KW. Methicillin-resistant Staphylococcus aureus therapy: past, present, and future. Clin. Infect. Dis. 58(Suppl. 1), S20–S27 (2014).
    • 2. Global Antimicrobial Resistance and Use Surveillance System (GLASS) Report. WHO, Geneva, Switzerland (2021).
    • 3. Kadri SS. Key takeaways from the U.S. CDC's 2019 antibiotic resistance threats report for frontline providers. Crit. Care Med. 48(7), 939–945 (2020).
    • 4. Chambers HF, DeLeo FR. Waves of resistance: Staphylococcus aureus in the antibiotic era. Nat. Rev. Microbiol. 7(9), 629–641 (2009).
    • 5. Barber M. Methicillin-resistant staphylococci. J. Clin. Pathol. 14, 385 (1961).
    • 6. Hiramatsu K, Kayayama Y, Matsuo M et al. Vancomycin-intermediate resistance in Staphylococcus aureus. J. Glob. Antimicrob. Resist. 2, 213–224 (2014).
    • 7. Hussain S, Joo J, Kang J et al. Antibiotic-loaded nanoparticles targeted to the site of infection enhance antibacterial efficacy. Nat. Biomed. Eng. 2, 95–103 (2018). • In this publication, a vancomycin-loaded nanoparticle has been conjugated to a peptide sequence – CARGGLKSC – which improves its ability to target Staphylococcus aureus.
    • 8. Liu J, Wang W, Wang X et al. PL/vancomycin/nano-hydroxyapatite sustained-release material to treat infectious bone defect. Open Life Sci. 15, 92–107 (2020).
    • 9. He X, Zhou Z, Han Z et al. Mechanism of controlled release of vancomycin from crumpled graphene oxides. ACS Omega 4, 12252–12258 (2019).
    • 10. Vanamala K, Tatiparti K, Bhise K et al. Novel approaches for the treatment of methicillin-resistant Staphylococcus aureus: using nanoparticles to overcome multidrug resistance. Drug Discov. Today 26, 31–43 (2021).
    • 11. Sande L, Sanchez M, Montes J et al. Liposomal encapsulation of vancomycin improves killing of methicillin-resistant Staphylococcus aureus in a murine infection model. J. Antimicrob. Chemother. 67, 2191–2194 (2012).
    • 12. Lu Z, Zhang J, Yu Z et al. Vancomycin-hybrid bimetallic Au/Ag composite nanoparticles: preparation of the nanoparticles and characterization of the antibacterial activity. New J. Chem. 41, 5276–5279 (2017).
    • 13. Zhang J, Wang C, Wang J, Qu Y, Liu G. In vivo drug release and antibacterial properties of vancomycin loaded hydroxyapatite/chitosan composite. Drug Deliv. 19, 264–269 (2012). • The authors have shown that when a mesoporous hydroxyapatite/chitosan scaffold loaded with vancomycin was implanted at the site of infection, it could maintain a local concentration of vancomycin higher than its MIC for Staphylococcus aureus.
    • 14. Bhatia S. Natural polymers vs synthetic polymer. In: Natural Polymer Drug Delivery Systems. Springer, Cham, Switzerland 95–118 (2016).
    • 15. Thota CK, Yadav N, Chauhan VS. A novel highly stable and injectable hydrogel based on a conformationally restricted ultrashort peptide. Sci. Rep. 6(1), 1–12 (2016).
    • 16. Ni M, Zhuo S. Applications of self-assembling ultrashort peptides in bionanotechnology. RSC Adv. 9, 844–852 (2019).
    • 17. Melchionna M, Styan KE, Marchesan S. The unexpected advantages of using D-amino acids for peptide self-assembly into nanostructured hydrogels for medicine. Curr. Top. Med. Chem. 16, 2009 (2016).
    • 18. Khara JS, Priestman M, Uhía I et al. Unnatural amino acid analogues of membrane-active helical peptides with anti-mycobacterial activity and improved stability. J. Antimicrob. Chemother. 71, 2181–2191 (2016).
    • 19. Gupta M, Bagaria A, Mishra A et al. Self-assembly of a dipeptide-containing conformationally restricted dehydrophenylalanine residue to form ordered nanotubes. Adv. Mater. 19, 858–861 (2007).
    • 20. Panda JJ, Varshney A, Chauhan VS. Self-assembled nanoparticles based on modified cationic dipeptides and DNA: novel systems for gene delivery. J. Nanobiotechnol. 11(1), 1–13 (2013).
    • 21. Weng J, Tong HH, Chow SF. In vitro release study of the polymeric drug nanoparticles: development and validation of a novel method. Pharmaceutics 12(8), 732 (2020).
    • 22. Elyasi S, Khalili H, Dashti-Khavidaki S, Mohammadpour A. Vancomycin-induced nephrotoxicity: mechanism, incidence, risk factors and special populations. A literature review. Eur. J. Clin. Pharmacol. 68(9), 1243–1255 (2012).
    • 23. Filippone EJ, Kraft WK, Farber JL. The nephrotoxicity of vancomycin. Clin. Pharm. Ther. 102(3), 459–469 (2017).
    • 24. Rybak MJ, Albrecht LM, Boike SC, Chandrasekar PH. Nephrotoxicity of vancomycin, alone and with an aminoglycoside. J. Antimicrob. Chemother. 25(4), 679–687 (1990).
    • 25. Sahin M, Cam H, Olgar S et al. Protective role of erdosteine on vancomycin-induced oxidative stress in rat liver. Mol. Cell. Biochem. 291(1), 155–160 (2006).
    • 26. Kucukler S, Darendelioğlu E, Caglayan C, Ayna A, Yıldırım S, Kandemir FM. Zingerone attenuates vancomycin-induced hepatotoxicity in rats through regulation of oxidative stress, inflammation and apoptosis. Life Sci. 259, 118382 (2020).
    • 27. Wu Q, Sabokroo N, Wang Y, Hashemian M, Karamollahi S, Kouhsari E. Systematic review and meta-analysis of the epidemiology of vancomycin-resistance Staphylococcus aureus isolates. Antimicrob. Resist. Infect. Control 10(1), 1–13 (2021).
    • 28. Gao P, Nie X, Zou M, Shi Y, Cheng G. Recent advances in materials for extended-release antibiotic delivery system. J. Antibiot. 64(9), 625–634 (2011).
    • 29. Tesauro D, Accardo A, Diaferia C et al. Peptide-based drug-delivery systems in biotechnological applications: recent advances and perspectives. Molecules 24(2), 351 (2019).
    • 30. Gentilucci L, De Marco R, Cerisoli L. Chemical modifications designed to improve peptide stability: incorporation of non-natural amino acids, pseudo-peptide bonds, and cyclization. Curr. Pharm. Des. 16(28), 3185–3203 (2010).
    • 31. Yadav N, Chauhan MK, Chauhan VS. Conformationally constrained dipeptide-based hydrogel as a platform for 3D cell growth and tissue engineering applications. Appl. Nanosci. 11, 2019–2031 (2021).
    • 32. Scriboni AB, Couto VM, Ribeiro LNM et al. Fusogenic liposomes increase the antimicrobial activity of vancomycin against Staphylococcus aureus biofilm. Front. Pharmacol. 10, 1401 (2019).
    • 33. Aldawsari HM, Hosny KM. Solid lipid nanoparticles of vancomycin loaded with ellagic acid as a tool for overcoming nephrotoxic side effects: preparation, characterization, and nephrotoxicity evaluation. J. Drug Deliv. Sci. Technol. 45, 76–80 (2018). • In this publication, solid lipid nanoparticles loaded with vancomycin and ellagic acid were able to reduce nephrotoxicity compared with vancomycin alone.
    • 34. Yang Z, Liu J, Gao J, Chen S, Huang G. Chitosan coated vancomycin hydrochloride liposomes: characterizations and evaluation. Int. J. Pharm. 495, 508–515 (2015).
    • 35. Zakeri-Milani P, Loveymi BD, Jelvehgari M, Valizadeh H. The characteristics and improved intestinal permeability of vancomycin PLGA-nanoparticles as colloidal drug delivery system. Colloid. Surf. B Biointerfaces 103, 174–181 (2013).
    • 36. Li D, Tang G, Yao H et al. Formulation of pH-responsive PEGylated nanoparticles with high drug loading capacity and programmable drug release for enhanced antibacterial activity. Bioact. Mater. 16, 47–56 (2022). • The publication talks about the self-assembling property of amphiphilic poly(ethylene glycol)-Schiff-vancomycin and vancomycin into a nanoparticle. The nanoparticle formulation was able to release the entrapped drug when subjected to pH change at the site of infection.
    • 37. Kalhapure RS, Jadhav M, Rambharose S et al. pH-responsive chitosan nanoparticles from a novel twin-chain anionic amphiphile for controlled and targeted delivery of vancomycin. Colloid. Surf. B Biointerfaces 158, 650–657 (2017).
    • 38. Albanese A, Tang PS, Chan WCW. The effect of nanoparticle size, shape, and surface chemistry on biological systems. 14(1), 1–16 (2012).
    • 39. Reygaert WC. An overview of the antimicrobial resistance mechanisms of bacteria. AIMS Microbiol. 4, 482 (2018).
    • 40. Cao M, Feng Y, Zhang Y, Kang W, Lian K, Ai L. Studies on the metabolism and degradation of vancomycin in simulated in vitro and aquatic environment by UHPLC-Triple-TOF-MS/MS. Sci. Rep. 8(1), 1–13 (2018).
    • 41. Ducharme MP, Slaughter RL, Edwards DJ. Vancomycin pharmacokinetics in a patient population: effect of age, gender, and body weight. Ther. Drug Monit. 16, 513–518 (1994).
    • 42. Fathi HA, Abdelkader A, AbdelKarim MS et al. Electrospun vancomycin-loaded nanofibers for management of methicillin-resistant Staphylococcus aureus-induced skin infections. Int. J. Pharm. 586, 119620 (2020).
    • 43. Qi G, Li L, Yu F, Wang H. Vancomycin-modified mesoporous silica nanoparticles for selective recognition and killing of pathogenic Gram-positive bacteria over macrophage-like cells. ACS Appl. Mater. Interfaces 5, 10874–10881 (2013).
    • 44. Xu J, Xu B, Shou D, Xia X, Hu Y. Preparation and evaluation of vancomycin-loaded N-trimethyl chitosan nanoparticles. Polymers 7, 1850–1870 (2015). • The authors have tested the antibacterial efficacy of N-trimethyl chitosan-based nanoparticles that are loaded with vancomycin. The formulation was effective against Staphylococcus aureus and promoted osteoblast proliferation.
    • 45. Maji R, Omolo CA, Jaglal Y et al. A transferosome-loaded bigel for enhanced transdermal delivery and antibacterial activity of vancomycin hydrochloride. Int. J. Pharm. 607, 120990 (2021).
    • 46. Yu X, Pan Q, Zheng Z et al. pH-responsive and porous vancomycin-loaded PLGA microspheres: evidence of controlled and sustained release for localized inflammation inhibition in vitro. RSC Adv. 8(65), 37424–37432 (2018).
    • 47. Hassani Besheli N, Mottaghitalab F, Eslami M et al. Sustainable release of vancomycin from silk fibroin nanoparticles for treating severe bone infection in rat tibia osteomyelitis model. ACS Appl. Mater. Interfaces 9(6), 5128–5138 (2017). • In this study, vancomycin was encapsulated into silk fibroin nanoparticles, and these were entrapped in silk scaffolds. This scaffold proved effective against methicillin-resistant S. aureus in a severe osteomyelitis rat model.
    • 48. Ahadi F, Khorshidi S, Karkhaneh A. A hydrogel/fiber scaffold based on silk fibroin/oxidized pectin with sustainable release of vancomycin hydrochloride. Eur. Polym. J. 118, 265–274 (2019).
    • 49. Liu J, Wang Z, Li F, Gao J, Wang L, Huang G. Liposomes for systematic delivery of vancomycin hydrochloride to decrease nephrotoxicity: characterization and evaluation. Asian J. Pharm. Sci. 10(3), 212–222 (2015). • In this publication, the authors have shown encapsulation of vancomycin in liposomes and achieved sustained delivery for longer durations to increase its antibacterial efficacy and decrease toxicity.
    • 50. Cai W, Liu J, Zheng L et al. Study on the anti-infection ability of vancomycin cationic liposome combined with polylactide fracture internal fixator. Int. J. Biol. Macromol. 167, 834–844 (2021).
    • 51. Weng W, Nie W, Zhou Q et al. Controlled release of vancomycin from 3D porous graphene-based composites for dual-purpose treatment of infected bone defects. RSC Adv. 7(5), 2753–2765 (2017).
    • 52. Gudmundsson S, Einarsson S, Erlendsdottir H, Moffat J, Bayer W, Craig WA. The post-antibiotic effect of antimicrobial combinations in a neutropenic murine thigh infection model. J. Antimicrob. Chemother. 31(Suppl. D), 177–191 (1993).
    • 53. Rodriguez CA, Agudelo M, Zuluaga AF, Vesga O. Generic vancomycin enriches resistant subpopulations of Staphylococcus aureus after exposure in a neutropenic mouse thigh infection model. Antimicrob. Agents Chemother. 56(1), 243–247 (2012).
    • 54. Ma K, Dong P, Liang M et al. Facile assembly of multifunctional antibacterial nanoplatform leveraging synergistic sensitization between silver nanostructure and vancomycin. ACS Appl. Mater. Interfaces 12(6), 6955–6965 (2020). • In this study, polydopamine nanoparticles were used to combine vancomycin and silver nanoparticles together to achieve a synergistic bactericidal activity on S. aureus and Escherichia coli strains.
    • 55. Böttger R, Hoffmann R, Knappe D. Differential stability of therapeutic peptides with different proteolytic cleavage sites in blood, plasma and serum. PLOS ONE 12(6), e0178943 (2017).
    • 56. Gorris HH, Bade S, RöCkendorf N et al. Rapid profiling of peptide stability in proteolytic environments. Anal. Chem. 81(4), 1580–1586 (2009).
    • 57. Bruno BJ, Miller GD, Lim CS. Basics and recent advances in peptide and protein drug delivery. Ther. Deliv. 4(11), 1443–1467 (2013).
    • 58. Otvos L Jr, Wade JD. Current challenges in peptide-based drug discovery. Front. Chem. 2, 62 (2014).
    • 59. Di L. Strategic approaches to optimizing peptide ADME properties. AAPS J. 17(1), 134–143 (2015).
    • 60. Lau JL, Dunn MK. Therapeutic peptides: historical perspectives, current development trends, and future directions. Bioorg. Med. Chem. Lett. 26(10), 2700–2707 (2018).