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

Cell membrane coated nanoparticles: next-generation therapeutics

    Ashwin Narain

    Department of Biotechnology, National Institute of Technology, Warangal – 506004, TS, India

    ,
    Simran Asawa

    Department of Biotechnology, National Institute of Technology, Warangal – 506004, TS, India

    Warsaw University of Life Sciences, Warsaw, Poland

    ,
    Vikesh Chhabria

    *Author for correspondence:

    E-mail Address: VNChhabria@uclan.ac.uk

    School of Pharmacy & Biomedical Sciences, University of Central Lancashire, Preston, UK

    &
    Yogita Patil-Sen

    **Author for correspondence:

    E-mail Address: ypatil-sen@uclan.ac.uk

    School of Physical Sciences & Computing, University of Central Lancashire, Preston, UK

    Published Online:https://doi.org/10.2217/nnm-2017-0225

    Cell membrane coated nanoparticles (NPs) is a biomimetic strategy developed to engineer therapeutic devices consisting of a NP core coated with membrane derived from natural cells such as erythrocytes, white blood cells, cancer cells, stem cells, platelets or bacterial cells. These biomimetic NPs have gained a lot of attention recently owing to their cell surface mimetic features and tailored nanomaterial characteristics. They have shown strong potential in diagnostic and therapeutic applications including those in drug delivery, immune modulation, vaccination and detoxification. Herein we review the various types of cell membrane coated NPs reported in the literature and the unique strengths of these biomimetic NPs with an emphasis on how these bioinspired camouflage strategies have led to improved therapeutic efficacy. We also highlight the recent progress made by each platform in advancing healthcare and precis the major challenges associated with these NPs.

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

    References

    • 1 Fang RH, Jiang Y, Fang JC, Zhang L. Cell membrane-derived nanomaterials for biomedical applications. Biomaterials 128, 69–83 (2017). •• This paper has shown the therapeutic benefits of using cell membrane coated nanoparticles and their effect on a molecular scale.
    • 2 Surendiran A, Sandhiya S, Pradhan S, Adithan C. Novel applications of nanotechnology in medicine. Indian J. Med. Res. 130(6), 689–701 (2009).
    • 3 Shang L, Nienhaus K, Nienhaus GU. Engineered nanoparticles interacting with cells: size matters. J. Nanobiotechnol. 12(1), 5 (2014).
    • 4 Kalepu S, Nekkanti V. Insoluble drug delivery strategies: review of recent advances and business prospects. Acta Pharm. Sin. B 5(5), 442–453 (2015).
    • 5 Fahs S, Patil-Sen Y, Snape TJ. Foldamers as anticancer therapeutics: targeting protein-protein interactions and the cell membrane. ChemBioChem 16(13), 1840–1853 (2015).
    • 6 Fahs S, Rowther FB, Dennison SR, Patil-Sen Y, Warr T, Snape TJ. Development of a novel, multifunctional, membrane-interactive pyridinium salt with potent anticancer activity. Bioorg. Med. Chem. Lett. 24(15), 3430–3433 (2014).
    • 7 Patil-Sen Y, Dennison SR, Snape TJ. Functional foldamers that target bacterial membranes: the effect of charge, amphiphilicity and conformation. Biorg. Med. Chem. 24(18), 4241–4245 (2016).
    • 8 Fang RH, Hu C-MJ, Chen KNH et al. Lipid-insertion enables targeting functionalization of erythrocyte membrane-cloaked nanoparticles. Nanoscale 5(19), 8884–8888 (2013).
    • 9 Bose RJC, Arai Y, Ahn JC, Park H, Lee S-H. Influence of cationic lipid concentration on properties of lipid–polymer hybrid nanospheres for gene delivery. Int. J. Nanomed. 10, 5367–5382 (2015).
    • 10 Steichen SD, Caldorera-Moore M, Peppas NA. A review of current nanoparticle and targeting moieties for the delivery of cancer therapeutics. Eur. J. Pharm. Sci. 48(3), 416–427 (2013).
    • 11 Mout R, Moyano DF, Rana S, Rotello VM. Surface functionalization of nanoparticles for nanomedicine. Chem. Soc. Rev. 41(7), 2539–2544 (2012).
    • 12 Peer D, Karp JM, Hong S, Farokhzad OC, Margalit R, Langer R. Nanocarriers as an emerging platform for cancer therapy. Nat. Nanotechnol. 2(12), 751–760 (2007).
    • 13 Hofheinz R-D, Gnad-Vogt SU, Beyer U, Hochhaus A. Liposomal encapsulated anti-cancer drugs. Anti-Cancer Drugs 16(7), 691–707 (2005).
    • 14 Immordino ML, Dosio F, Cattel L. Stealth liposomes: review of the basic science, rationale, and clinical applications, existing and potential. Int. J. Nanomed. 1(3), 297–315 (2006).
    • 15 Kulkarni CV, Moinuddin Z, Patil-Sen Y, Littlefield R, Hood M. Lipid-hydrogel films for sustained drug release. Int. J. Pharm. 479(2), 416–421 (2015).
    • 16 Hans M, Lowman A. Biodegradable nanoparticles for drug delivery and targeting. Curr. Opin. Solid State Mater. Sci. 6(4), 319–327 (2002).
    • 17 Enlow EM, Luft JC, Napier ME, Desimone JM. Potent engineered PLGA nanoparticles by virtue of exceptionally high chemotherapeutic loadings. Nano Lett. 11(2), 808–813 (2011).
    • 18 Li Z, Barnes JC, Bosoy A, Stoddart JF, Zink JI. Mesoporous silica nanoparticles in biomedical applications. Chem. Soc. Rev. 41(7), 2590–2605 (2012).
    • 19 Gupta AK, Gupta M. Synthesis and surface engineering of iron oxide nanoparticles for biomedical applications. Biomaterials 26(18), 3995–4021 (2005).
    • 20 Dykman L, Khlebtsov N. Gold nanoparticles in biomedical applications: recent advances and perspectives. Chem. Soc. Rev. 41(6), 2256–2282 (2012).
    • 21 Patil-Sen Y, Tiddy GJT, Brezesinski G, Dewolf C. A monolayer phase behaviour study of phosphatidylinositol, phosphatidylinositol 4-monophosphate and their binary mixtures with distearoylphosphatidylethanolamine. PCCP 6(7), 1562–1565 (2004).
    • 22 Sharma A, Sharma US. Liposomes in drug delivery: progress and limitations. Int. J. Pharm. 154(2), 123–140 (1997).
    • 23 Maurer N, Fenske DB, Cullis PR. Developments in liposomal drug delivery systems. Expert Opin. Biol. Ther. 1(6), 923–947 (2001).
    • 24 Ekimov A, Onushchenko A. Quantum size effect in three-dimensional microscopic semiconductor crystals. Pis'ma Zh. Eksp. Teor. Fiz. 34, 363–366 (1981).
    • 25 Resch-Genger U, Grabolle M, Cavaliere-Jaricot S, Nitschke R, Nann T. Quantum dots versus organic dyes as fluorescent labels. Nat. Methods 5(9), 763–775 (2008).
    • 26 Sinha N, Yeow JT. Carbon nanotubes for biomedical applications. IEEE Trans. NanoBioscience 4(2), 180–195 (2005).
    • 27 Gaunt NP, Patil-Sen Y, Baker MJ, Kulkarni CV. Carbon nanotubes for stabilization of nanostructured lipid particles. Nanoscale 7(3), 1090–1095 (2015).
    • 28 Patil-Sen Y, Sadeghpour A, Rappolt M, Kulkarni CV. Facile preparation of internally self-assembled lipid particles stabilized by carbon nanotubes. J. Vis. Exp. (108), 53489 (2016).
    • 29 Kulkarni M, Patil-Sen Y, Junkar I, Kulkarni CV, Lorenzetti M, Iglic A. Wettability studies of topologically distinct titanium surfaces. Colloid Surf. B 129, 47–53 (2015).
    • 30 Naskar J, Roy S, Joardar A, Das S, Banerjee A. Self-assembling dipeptide-based nontoxic vesicles as carriers for drugs and other biologically important molecules. Org. Biomol. Chem. 9(19), 6610–6615 (2011).
    • 31 Bawa R, Fung S-Y, Shiozaki A et al. Self-assembling peptide-based nanoparticles enhance cellular delivery of the hydrophobic anticancer drug ellipticine through caveolae-dependent endocytosis. Nanomed. Nanotechnol. Biol. Med. 8(5), 647–654 (2012).
    • 32 Majid A, Patil-Sen Y, Ahmed W, Sen T. Tunable self-assembled peptide structure: a novel approach to design dual-use biological agents. Materials Today 4(1), 32–40 (2017).
    • 33 Crist RM, Grossman JH, Patri AK et al. Common pitfalls in nanotechnology: lessons learned from NCI's Nanotechnology Characterization Laboratory. Integr. Biol. (Camb). 5(1), 66–73 (2013).
    • 34 Shvedova AA, Kagan VE, Fadeel B. Close encounters of the small kind: adverse effects of man-made materials interfacing with the nano-cosmos of biological systems. Annu. Rev. Pharmacol. Toxicol. 50, 63–88 (2010).
    • 35 Zhang L, Gu FX, Chan JM, Wang AZ, Langer RS, Farokhzad OC. Nanoparticles in medicine: therapeutic applications and developments. Clin. Pharmacol. Ther. 83(5), 761–769 (2008). •• Pioneers in the field of nanomedicine. Zhang L has pioneered the development of cell membrane coated nanoparticles. This paper is of significant interest as it outlines drug delivery systems used in 2008, its benefits and drawbacks with respect to their applications.
    • 36 Fox ME, Szoka FC, Fréchet JMJ. Soluble polymer carriers for the treatment of cancer: the importance of molecular architecture. Acc. Chem. Res. 42(8), 1141–1151 (2009).
    • 37 Barenholz YC. Doxil® – the first FDA-approved nano-drug: lessons learned. J. Control. Release 160(2), 117–134 (2012).
    • 38 Dawidczyk CM, Kim C, Park JH et al. State-of-the-art in design rules for drug delivery platforms: lessons learned from FDA-approved nanomedicines. J. Control. Release 187, 133–144 (2014).
    • 39 Maier-Hauff K, Ulrich F, Nestler D et al. Efficacy and safety of intratumoral thermotherapy using magnetic iron-oxide nanoparticles combined with external beam radiotherapy on patients with recurrent glioblastoma multiforme. J. Neurooncol. 103(2), 317–324 (2011).
    • 40 He H, Pham-Huy LA, Dramou P, Xiao D, Zuo P, Pham-Huy C. Carbon nanotubes: applications in pharmacy and medicine. Biomed Res. Int. 2013, 578290, 1–12 (2013).
    • 41 Gao Z, Fain HD, Rapoport N. Ultrasound-enhanced tumor targeting of polymeric micellar drug carriers. Mol. Pharm. 1(4), 317–330 (2004).
    • 42 Zhang H. Erythrocytes in nanomedicine: an optimal blend of natural and synthetic materials. Biomater. Sci. 4(7), 1024–1031 (2016).
    • 43 Gupta N, Patel B, Ahsan F. Nano-engineered erythrocyte ghosts as inhalational carriers for delivery of fasudil: preparation and characterization. Pharm. Res. 31(6), 1553–1565 (2014).
    • 44 Hu C-MJ, Fang RH, Copp J, Luk BT, Zhang L. A biomimetic nanosponge that absorbs pore-forming toxins. Nat. Nano 8(5), 336–340 (2013). • This was the first paper to show that erythrocyte membranes could be used as targets to adsorb pore-forming toxins rather than a drug delivery system.
    • 45 Hwang J, Jeong Y, Park JM, Lee KH, Hong JW, Choi J. Biomimetics: forecasting the future of science, engineering, and medicine. Int. J. Nanomed. 10, 5701–5713 (2015).
    • 46 Dinndorf PA, Gootenberg J, Cohen MH, Keegan P, Pazdur R. FDA drug approval summary: pegaspargase (oncaspar) for the first-line treatment of children with acute lymphoblastic leukemia (ALL). Oncologist 12(8), 991–998 (2007).
    • 47 Oerlemans C, Bult W, Bos M, Storm G, Nijsen JFW, Hennink WE. Polymeric micelles in anticancer therapy: targeting, imaging and triggered release. Pharm. Res. 27(12), 2569–2589 (2010).
    • 48 Wang Y-XJ. Superparamagnetic iron oxide based MRI contrast agents: current status of clinical application. Quant. Imaging Med. Surg. 1(1), 35–40 (2011).
    • 49 Fang M, Peng C-W, Pang D-W, Li Y. Quantum dots for cancer research: current status, remaining issues, and future perspectives. Cancer Biol. Med. 9(3), 151–163 (2012).
    • 50 Alibolandi M, Abnous K, Sadeghi F, Hosseinkhani H, Ramezani M, Hadizadeh F. Folate receptor-targeted multimodal polymersomes for delivery of quantum dots and doxorubicin to breast adenocarcinoma: in vitro and in vivo evaluation. Int. J. Pharm. 500(1–2), 162–178 (2016).
    • 51 Parodi A, Quattrocchi N, Van De Ven AL et al. Synthetic nanoparticles functionalized with biomimetic leukocyte membranes possess cell-like functions. Nat. Nanotechnol. 8(1), 61–68 (2013).
    • 52 Antonelli A, Sfara C, Manuali E, Bruce IJ, Magnani M. Encapsulation of superparamagnetic nanoparticles into red blood cells as new carriers of MRI contrast agents. Nanomedicine 6(2), 211–223 (2011). • Pioneers in the field of erythrocyte drug carriers (nanoerythrosomes). This paper has shown tremendous diagnostic potential of nanoerythrosomes for MRI imaging.
    • 53 Wei X, Gao J, Fang RH et al. Nanoparticles camouflaged in platelet membrane coating as an antibody decoy for the treatment of immune thrombocytopenia. Biomaterials 111, 116–123 (2016).
    • 54 Fang RH, Hu CMJ, Luk BT et al. Cancer cell membrane-coated nanoparticles for anticancer vaccination and drug delivery. Nano Lett. 14(4), 2181–2188 (2014).
    • 55 Gao WW, Fang RH, Thamphiwatana S et al. Modulating antibacterial immunity via bacterial membrane-coated nanoparticles. Nano Lett. 15(2), 1403–1409 (2015).
    • 56 Lai PY, Huang RY, Lin SY, Lin YH, Chang CW. Biomimetic stem cell membrane-camouflaged iron oxide nanoparticles for theranostic applications. RSC Adv. 5(119), 98222–98230 (2015).
    • 57 Hu CMJ, Zhang L, Aryal S, Cheung C, Fang RH, Zhang LF. Erythrocyte membrane-camouflaged polymeric nanoparticles as a biomimetic delivery platform. Proc. Natl Acad. Sci. USA 108(27), 10980–10985 (2011).
    • 58 Hu CMJ, Fang RH, Luk BT et al. ‘Marker-of-self’ functionalization of nanoscale particles through a top-down cellular membrane coating approach. Nanoscale 5(7), 2664–2668 (2013).
    • 59 Rodriguez PL, Harada T, Christian DA, Pantano DA, Tsai RK, Discher DE. Minimal “self” peptides that inhibit phagocytic clearance and enhance delivery of nanoparticles. Science 339(6122), 971–975 (2013).
    • 60 Gao WW, Hu CMJ, Fang RH, Luk BT, Su J, Zhang LF. Surface functionalization of gold nanoparticles with red blood cell membranes. Adv. Mater. 25(26), 3549–3553 (2013).
    • 61 Rao L, Bu LL, Xu JH et al. Red blood cell membrane as a biomimetic nanocoating for prolonged circulation time and reduced accelerated blood clearance. Small 11(46), 6225–6236 (2015).
    • 62 Li LL, Xu JH, Qi GB, Zhao XZ, Yu FQ, Wang H. Core-shell supramolecular gelatin nanoparticles for adaptive and “on-demand” antibiotic delivery. ACS Nano 8(5), 4975–4983 (2014).
    • 63 Piao JG, Wang LM, Gao F, You YZ, Xiong YJ, Yang LH. Erythrocyte membrane is an alternative coating to polyethylene glycol for prolonging the circulation lifetime of gold nanocages for photothermal therapy. ACS Nano 8(10), 10414–10425 (2014).
    • 64 Rao L, Xu JH, Cai B et al. Synthetic nanoparticles camouflaged with biomimetic erythrocyte membranes for reduced reticuloendothelial system uptake. Nanotechnology 27(8), 085106 (2016).
    • 65 Wang ZJ, Li J, Cho J, Malik AB. Prevention of vascular inflammation by nanoparticle targeting of adherent neutrophils. Nat. Nanotechnol. 9(3), 204–210 (2014).
    • 66 Baggiolini M. Chemokines and leukocyte traffic. Nature 392(6676), 565–568 (1998).
    • 67 Springer TA. Traffic signals for lymphocyte recirculation and leukocyte emigration – the multistep paradigm. Cell 76(2), 301–314 (1994).
    • 68 Xuan M, Shao J, Dai L, He Q, Li J. Macrophage cell membrane camouflaged mesoporous silica nanocapsules for in vivo cancer therapy. Adv. Healthc. Mater. 4(11), 1645–1652 (2015).
    • 69 Gao C, Wu Z, Lin Z, Lin X, He Q. Polymeric capsule-cushioned leukocyte cell membrane vesicles as a biomimetic delivery platform. Nanoscale 8(6), 3548–3554 (2016).
    • 70 Xuan MJ, Shao JX, Dai LR, Li JB, He Q. Macrophage cell membrane camouflaged Au nanoshells for in vivo prolonged circulation life and enhanced cancer photothermal therapy. ACS Appl. Mater. Interfaces 8(15), 9610–9618 (2016).
    • 71 Krishnamurthy S, Gnanasammandhan MK, Xie C, Huang K, Cui MY, Chan JM. Monocyte cell membrane-derived nanoghosts for targeted cancer therapy. Nanoscale 8(13), 6981–6985 (2016).
    • 72 Osta WA, Chen Y, Mikhitarian K et al. EpCAM is overexpressed in breast cancer and is a potential target for breast cancer gene therapy. Cancer Res. 64(16), 5818–5824 (2004).
    • 73 Zhu JY, Zheng DW, Zhang MK et al. Preferential cancer cell self-recognition and tumor self-targeting by coating nanoparticles with homotypic cancer cell membranes. Nano Lett. 16(9), 5895–5901 (2016). • This paper is of considerable interest as it shows that functionalization with cancer cell membrane allows specific cancer cell targeting and internalization.
    • 74 Sun HP, Su JH, Meng QS et al. Cancer-cell-biomimetic nanoparticles for targeted therapy of homotypic tumors. Adv. Mater. 28(43), 9581–9588 (2016).
    • 75 Uccelli A, Moretta L, Pistoia V. Mesenchymal stem cells in health and disease. Nat. Rev. Immunol. 8(9), 726–736 (2008).
    • 76 Majumdar MK, Keane-Moore M, Buyaner D et al. Characterization and functionality of cell surface molecules on human mesenchymal stem cells. J. Biomed. Sci. 10(2), 228–241 (2003).
    • 77 De Miguel MP, Fuentes-Julian S, Blazquez-Martinez A et al. Immunosuppressive properties of mesenchymal stem cells: advances and applications. Curr. Mol. Med. 12(5), 574–591 (2012).
    • 78 Gao CY, Lin ZH, Jurado-Sanchez B, Lin XK, Wu ZG, He Q. Stem cell membrane-coated nanogels for highly efficient in vivo tumor targeted drug delivery. Small 12(30), 4056–4062 (2016).
    • 79 George D. Platelet-derived growth factor receptors: a therapeutic target in solid tumors. Semin. Oncol. 28(5 Suppl. 17), 27–33 (2001).
    • 80 Farokhzad OC, Langer R. Impact of nanotechnology on drug delivery. ACS Nano 3(1), 16–20 (2009).
    • 81 Hu CMJ, Fang RH, Wang KC et al. Nanoparticle biointerfacing by platelet membrane cloaking. Nature 526(7571), 118–121 (2015).
    • 82 Hu QY, Sun WJ, Qian CG, Wang C, Bomba HN, Gu Z. Anticancer platelet-mimicking nanovehicles. Adv. Mater. 27(44), 7043–7050 (2015). • This paper is of interest as it shows the anticancer effect of coating nanoparticle cores with platelet membranes.
    • 83 Acevedo R, Fernandez S, Zayas C et al. Bacterial outer membrane vesicles and vaccine applications. Front. Immunol. 5, 121–126 (2014).
    • 84 Langemann T, Koller VJ, Muhammad A, Kudela P, Mayr UB, Lubitz W. The bacterial ghost platform system: production and applications. Bioeng. Bugs 1(5), 326–336 (2010).
    • 85 Dehaini D, Wei X, Fang RH et al. Erythrocyte-platelet hybrid membrane coating for enhanced nanoparticle functionalization. Adv. Mater. 29(16), 1606209 (2017).
    • 86 Rao L, Meng QF, Bu LL et al. Erythrocyte membrane-coated upconversion nanoparticles with minimal protein adsorption for enhanced tumor imaging. ACS Appl. Mater. Interfaces 9(3), 2159–2168 (2017).
    • 87 Luk BT, Fang RH, Hu C-MJ et al. Safe and immunocompatible nanocarriers cloaked in RBC membranes for drug delivery to treat solid tumors. Theranostics 6(7), 1004–1011 (2016).
    • 88 Palomba R, Parodi A, Evangelopoulos M et al. Biomimetic carriers mimicking leukocyte plasma membrane to increase tumor vasculature permeability. Sci. Rep. 6, 1–11 (2016).
    • 89 Chhabria V, Beeton S. Development of nanosponges from erythrocyte ghosts for removal of streptolysin-O from mammalian blood. Nanomedicine 11(21), 2797–2807 (2016).
    • 90 Albanese A, Tang PS, Chan WCW. The effect of nanoparticle size, shape, and surface chemistry on biological systems. In: Annual Review of Biomedical Engineering, Vol. 14. Yarmush ML (Ed.). Institute of Biomaterials and Biomedical Engineering; Terrence Donnelly Center for Cellular and Biomolecular Research; Materials Science and Engineering; Chemical Engineering; and Chemistry, University of Toronto, Toronto, ON, Canada, 1–16 (2012).
    • 91 Luk BT, Jack Hu C-M, Fang RH et al. Interfacial interactions between natural RBC membranes and synthetic polymeric nanoparticles. Nanoscale 6(5), 2730–2737 (2014).
    • 92 Meyer RA, Sunshine JC, Green JJ. Biomimetic particles as therapeutics. Trends Biotechnol. 33(9), 514–524 (2015).
    • 93 Zhang L, Li R, Chen H et al. Human cytotoxic T-lymphocyte membrane-camouflaged nanoparticles combined with low-dose irradiation: a new approach to enhance drug targeting in gastric cancer. Int. J. Nanomed. 12, 2129 (2017).
    • 94 Rao L, Bu LL, Cai B et al. Cancer cell membrane-coated upconversion nanoprobes for highly specific tumor imaging. Adv. Mater. 28(18), 3460–3466 (2016).
    • 95 Chen Z, Zhao PF, Luo ZY et al. Cancer cell membrane-biomimetic nanoparticles for homologous-targeting dual-modal imaging and photothermal therapy. ACS Nano 10(11), 10049–10057 (2016).
    • 96 Gao WW, Hu CMJ, Fang RH, Zhang LF. Liposome-like nanostructures for drug delivery. J. Mater. Chem. B 1(48), 6569–6585 (2013).
    • 97 He WP, Frueh J, Wu ZW, He Q. How leucocyte cell membrane modified janus microcapsules are phagocytosed by cancer cells. ACS Appl. Mater. Interfaces 8(7), 4407–4415 (2016).
    • 98 Rao L, He Z, Meng QF et al. Effective cancer targeting and imaging using macrophage membrane-camouflaged upconversion nanoparticles. J. Biomed. Mater. Res. A 105(2), 521–530 (2017).
    • 99 Evangelopoulos M, Parodi A, Martinez JO et al. Cell source determines the immunological impact of biomimetic nanoparticles. Biomaterials 82, 168–177 (2016).
    • 100 Bosch F, Rosich L. The contributions of Paul Ehrlich to pharmacology: a tribute on the occasion of the centenary of his nobel prize. Pharmacology 82(3), 171–179 (2008).
    • 101 Luk BT, Zhang LF. Cell membrane-camouflaged nanoparticles for drug delivery. J. Control. Release 220, 600–607 (2015).
    • 102 Tan SW, Wu TT, Zhang D, Zhang ZP. Cell or cell membrane-based drug delivery systems. Theranostics 5(8), 863–881 (2015).
    • 103 Anselmo AC, Modery-Pawlowski CL, Menegatti S et al. Platelet-like nanoparticles: mimicking shape, flexibility, and surface biology of platelets to target vascular injuries. ACS Nano 8(11), 11243–11253 (2014).