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

Surface modification potentials of cell membrane-based materials for targeted therapies: a chemotherapy-focused review

    Hongliang Duan

    Department of Thyroid Surgery, the Second Hospital of Jilin University, Changchun, 130000, China

    ,
    LiJuan Wang

    *Author for correspondence:

    E-mail Address: lijuan01@jlu.edu.cn

    Department of Endocrinology, the Second Hospital of Jilin University, Changchun, 130000, China

    ,
    Sen Wang

    Department of Thyroid Surgery, the Second Hospital of Jilin University, Changchun, 130000, China

    &
    Yangfang He

    Department of Endocrinology, the Second Hospital of Jilin University, Changchun, 130000, China

    Published Online:https://doi.org/10.2217/nnm-2023-0164

    Nanotechnology has significant potential for cancer management at all stages, including prevention, diagnosis and treatment. In therapeutic applications, nanoparticles (NPs) have biological stability, targeting and body-clearance issues. To overcome these difficulties, biomimetic or cell membrane-coating methods using immune cell membranes are advised. Macrophage or neutrophil cell membrane-coated NPs may impede cancer progression in malignant tissue. Immune cell surface proteins and their capacity to maintain activity after membrane extraction and NP coating determine NP functioning. Immune cell surface proteins may offer NPs higher cellular interactions, blood circulation, antigen recognition for targeting, progressive drug release and reduced in vivo toxicity. This article examines nano-based systems with immune cell membranes, their surface modification potential, and their application in cancer treatment.

    Plain language summary

    Nanoparticles (NPs) are small particles that range between 1 and 100 nanometres in size that are used to deliver substances that aid in the prevention, diagnosis and treatment of cancer. NPs are promising for therapeutic use but face challenges like stability, cancer targeting and clearance in the body. This article suggests that these challenges can be overcome using biomimetic methods. This involves coating NPs in cell membranes from immune cells. This has been demonstrated using two types of white blood cells, called macrophages and neutrophils. NPs coated in membranes derived from these cells have been shown to hinder cancer progression. How effective these coated NP cells are depends on what proteins from the surface of the immune cells are included and whether they remain active. These immune cell surface proteins allow coated NPs to have improved interactions with cells, circulate in the blood for longer, target proteins overexpressed on cancer cells and release drugs gradually. Biomimentic cell membrane coating also decreases cell membrane toxicity. The article examines NP-based systems using immune cell membranes, their potential for surface modification and their application in cancer treatment.

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

    References

    • 1. Fröhlich E. The role of surface charge in cellular uptake and cytotoxicity of medical nanoparticles. Int. J. Nanomed. 2012(7), 5577–5591 (2012). • Cationic nanoparticles cause more pronounced cell disruption, while nonphagocytic cells preferentially uptake them. Understanding surface charge effects could improve the selectivity and efficiency of nanoparticles in drug delivery and imaging.
    • 2. Chen W, Qin M, Chen X et al. Combining photothermal therapy and immunotherapy against melanoma by polydopamine-coated Al2O3 nanoparticles. Theranostics 8(8), 2229 (2018).
    • 3. Jiang D, Xu M, Pei Y et al. Core-matched nanoassemblies for targeted co-delivery of chemotherapy and photosensitizer to treat drug-resistant cancer. Acta Biomaterialia 88, 406–421 (2019).
    • 4. Liang X-J, Chen C, Zhao Y et al. Circumventing tumor resistance to chemotherapy by nanotechnology. Multi-Drug Resist. Cancer 467–488 (2010).
    • 5. Patel NR, Pattni BS, Abouzeid AH et al. Nanopreparations to overcome multidrug resistance in cancer. Adv. Drug Deliv. Rev. 65(13–14), 1748–1762 (2013).
    • 6. Mamnoon B, Feng L, Froberg J et al. Hypoxia-responsive, polymeric nanocarriers for targeted drug delivery to estrogen receptor-positive breast cancer cell spheroids. Mol. Pharmaceut. 17(11), 4312–4322 (2020). • Fluorescence microscopic studies indicated increased cytosolic and nuclear internalization of targeted polymersomes compared with nontargeted ones. Cell viability and cytotoxicity studies on ER-positive MCF7 cells and 3D spheroid cultures revealed promising potential for targeted drug delivery in estrogen receptor-positive breast cancer therapy.
    • 7. Fan W, Yung B, Huang P et al. Nanotechnology for multimodal synergistic cancer therapy. Chem. Rev. 117(22), 13566–13638 (2017).
    • 8. Zeng Y, Li S, Zhang S et al. Cell membrane coated-nanoparticles for cancer immunotherapy. Acta Pharmaceutica Sinica B. 12(8), 3233–3254 (2022).
    • 9. Shi J, Kantoff PW, Wooster R et al. Cancer nanomedicine: progress, challenges and opportunities. Nat. Rev. Cancer 17(1), 20–37 (2017).
    • 10. Tao J, Diao L, Chen F et al. pH-sensitive nanoparticles codelivering docetaxel and dihydroartemisinin effectively treat breast cancer by enhancing reactive oxidative species-mediated mitochondrial apoptosis. Mol. Pharmaceut. 18(1), 74–86 (2020).
    • 11. Lee NK, Lee EJ, Kim S et al. Ferritin nanocage with intrinsically disordered proteins and affibody: a platform for tumor targeting with extended pharmacokinetics. J. Control. Rel. 267, 172–180 (2017).
    • 12. Steichen SD, Caldorera-Moore M, Peppas NA. A review of current nanoparticle and targeting moieties for the delivery of cancer therapeutics. Eur. J. Pharmaceut. Sci. 48(3), 416–427 (2013). • Tumor physiology and specific barriers within the tumor microenvironment and systemic delivery are also examined, emphasizing the potential benefits of nanoscale carriers in enhancing cancer therapeutics.
    • 13. Maeda H. Toward a full understanding of the EPR effect in primary and metastatic tumors as well as issues related to its heterogeneity. Adv. Drug Deliv. Rev. 91, 3–6 (2015).
    • 14. Dobrovolskaia MA, Shurin M, Shvedova AA. Current understanding of interactions between nanoparticles and the immune system. Toxicol. Appl. Pharmacol. 299, 78–89 (2016).
    • 15. Vivier E, Malissen B. Innate and adaptive immunity: specificities and signaling hierarchies revisited. Nat. Immunol. 6(1), 17–21 (2005).
    • 16. Kononenko V, Narat M, Drobne D. Nanoparticle interaction with the immune system. Arch. Industr. Hygiene Toxicol. 66(2), 97–108 (2015).
    • 17. Dobrovolskaia MA, Mcneil SE. Handbook Of Immunological Properties Of Engineered Nanomaterials (In 3 Volumes). World Scientific, 5 Toh Tuck Link, Singapore, 6 (2016).
    • 18. Dwivedi PD, Misra A, Shanker R et al. Are nanomaterials a threat to the immune system? Nanotoxicology 3(1), 19–26 (2009).
    • 19. Lucarelli M, Gatti AM, Savarino G et al. Innate defence functions of macrophages can be biased by nano-sized ceramic and metallic particles. Eur. Cytokine Network 15(4), 339–346 (2004).
    • 20. Trickler WJ, Lantz SM, Murdock RC et al. Silver nanoparticle induced blood-brain barrier inflammation and increased permeability in primary rat brain microvessel endothelial cells. Toxicol. Sci. 118(1), 160–170 (2010).
    • 21. Park E-J, Kim H, Kim Y et al. Inflammatory responses may be induced by a single intratracheal instillation of iron nanoparticles in mice. Toxicology 275(1–3), 65–71 (2010).
    • 22. Nishimori H, Kondoh M, Isoda K et al. Silica nanoparticles as hepatotoxicants. Eur. J. Pharmaceut. Biopharmaceut. 72(3), 496–501 (2009).
    • 23. Pedata P, Petrarca C, Garzillo EM et al. Immunotoxicological impact of occupational and environmental nanoparticles exposure: The influence of physical, chemical, and combined characteristics of the particles. Int. J. Immunopathol. Pharmacol. 29(3), 343–353 (2016).
    • 24. Astete CE, Sabliov CM. Synthesis and characterization of PLGA nanoparticles. J. Biomat. Sci. Polymer Ed. 17(3), 247–289 (2006).
    • 25. Salykina YF, Zherdeva VV, Dezhurov SV et al. Biodistribution and clearance of quantum dots in small animals. In: Saratov Fall Meeting 2010: Optical Technologies in Biophysics and Medicine Xii. SPIE, 46–55 (2011).
    • 26. Shi Kam NW, Jessop TC, Wender PA et al. Nanotube molecular transporters: internalization of carbon nanotube−protein conjugates into mammalian cells. J. Am. Chem. Soc. 126(22), 6850–6851 (2004).
    • 27. Singh N, Jenkins GJ, Asadi R et al. Potential toxicity of superparamagnetic iron oxide nanoparticles (SPION). Nano Rev. 1(1), 5358 (2010).
    • 28. Patil RM, Thorat ND, Shete PB et al. Comprehensive cytotoxicity studies of superparamagnetic iron oxide nanoparticles. Biochem. Biophys. Rep. 13, 63–72 (2018).
    • 29. Fratoddi I, Venditti I, Cametti C et al. How toxic are gold nanoparticles? The state-of-the-art. Nano Res. 8, 1771–1799 (2015).
    • 30. Palmerston Mendes L, Pan J, Torchilin VP. Dendrimers as nanocarriers for nucleic acid and drug delivery in cancer therapy. Molecules 22(9), 1401 (2017).
    • 31. Liu F, Huang H, Gong Y et al. Evaluation of in vitro toxicity of polymeric micelles to human endothelial cells under different conditions. Chemico-Biol. Interact. 263, 46–54 (2017).
    • 32. Roursgaard M, Knudsen KB, Northeved H et al. In vitro toxicity of cationic micelles and liposomes in cultured human hepatocyte (HepG2) and lung epithelial (A549) cell lines. Toxicol. In Vitro 36, 164–171 (2016).
    • 33. Silva M, Santos AC, Conde J et al. Biomimetic cancer cell membrane-coated nanosystems as next-generation cancer therapies. Expert Opin. Drug Deliv. 17(11), 1515–1518 (2020).
    • 34. Asri R, Harun W, Samykano M et al. Corrosion and surface modification on biocompatible metals: a review. Mat. Sci. Eng. C 77, 1261–1274 (2017).
    • 35. Janib SM, Moses AS, Mackay JA. Imaging and drug delivery using theranostic nanoparticles. Adv. Drug Deliv. Rev. 62(11), 1052–1063 (2010).
    • 36. Liu Y, Hardie J, Zhang X, Rotello VM. Effects of engineered nanoparticles on the innate immune system. Semin. Immunol. 34, 25–32 (2017).
    • 37. Qiao Z-Y, Zhao W-J, Gao Y-J et al. Reconfigurable peptide nanotherapeutics at tumor microenvironmental pH. ACS Appl. Mater. Interface 9(36), 30426–30436 (2017).
    • 38. Malachowski T, Hassel A. Engineering nanoparticles to overcome immunological barriers for enhanced drug delivery. Eng. Regen. 1, 35–50 (2020).
    • 39. Fadeel B. Hide and seek: Nanomaterial interactions with the immune system. Front. Immunol. 10, 133 (2019).
    • 40. Sung H-J, Luk A, Murthy NS et al. Poly(ethylene glycol) as a sensitive regulator of cell survival fate on polymeric biomaterials: the interplay of cell adhesion and pro-oxidant signaling mechanisms. Soft Matter 6(20), 5196–5205 (2010).
    • 41. Mahmoudi M, Serpooshan V, Laurent S. Engineered nanoparticles for biomolecular imaging. Nanoscale 3(8), 3007–3026 (2011).
    • 42. Gulati NM, Stewart PL, Steinmetz NF. Bioinspired shielding strategies for nanoparticle drug delivery applications. Mol. Pharmaceut. 15(8), 2900–2909 (2018).
    • 43. Bellido E, Hidalgo T, Lozano MV et al. Heparin-engineered mesoporous iron metal-organic framework nanoparticles: toward stealth drug nanocarriers. Adv. Healthcare Mat. 4(8), 1246–1257 (2015).
    • 44. Wilson DR, Zhang N, Silvers AL et al. Synthesis and evaluation of cyclosporine A-loaded polysialic acid–polycaprolactone micelles for rheumatoid arthritis. Eur. J. Pharmaceut. Sci. 51, 146–156 (2014).
    • 45. Thasneem Y, Sajeesh S, Sharma CP. Glucosylated polymeric nanoparticles: a sweetened approach against blood compatibility paradox. Colloid. Surf. B. Biointerface 108, 337–344 (2013).
    • 46. Thomas SN, Van Der Vlies AJ, O'Neil CP et al. Engineering complement activation on polypropylene sulfide vaccine nanoparticles. Biomaterials 32(8), 2194–2203 (2011).
    • 47. Gulati N, Pitek A, Czapar A et al. The in vivo fates of plant viral nanoparticles camouflaged using self-proteins: overcoming immune recognition. J. Mater. Chem. B. 6(15), 2204–2216 (2018).
    • 48. Spicer CD, Jumeaux C, Gupta B et al. Peptide and protein nanoparticle conjugates: versatile platforms for biomedical applications. Chem. Soc. Rev. 47(10), 3574–3620 (2018).
    • 49. Nowinski AK, White AD, Keefe AJ et al. Biologically inspired stealth peptide-capped gold nanoparticles. Langmuir 30(7), 1864–1870 (2014).
    • 50. Schwarze SR, Ho A, Vocero-Akbani A et al. In vivo protein transduction: delivery of a biologically active protein into the mouse. Science 285(5433), 1569–1572 (1999).
    • 51. Grabrucker AM, Ruozi B, Belletti D et al. Nanoparticle transport across the blood–brain barrier. Tissue Barrier 4(1), e1153568 (2016).
    • 52. Rao L, Xu J-H, Cai B et al. Synthetic nanoparticles camouflaged with biomimetic erythrocyte membranes for reduced reticuloendothelial system uptake. Nanotechnology 27(8), 085106 (2016).
    • 53. Szeto GL, Lavik EB. Materials design at the interface of nanoparticles and innate immunity. J. Mater. Chem. B. 4(9), 1610–1618 (2016).
    • 54. 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).
    • 55. Hu C-MJ, Fang RH, Wang K-C et al. Nanoparticle biointerfacing by platelet membrane cloaking. Nature 526(7571), 118–121 (2015).
    • 56. Fang RH, Hu C-MJ, Luk BT et al. Cancer cell membrane-coated nanoparticles for anticancer vaccination and drug delivery. Nano Lett. 14(4), 2181–2188 (2014).
    • 57. Vijayan V, Uthaman S, Park I-K. Cell membrane-camouflaged nanoparticles: a promising biomimetic strategy for cancer theragnostics. Polymers 10(9), 983 (2018).
    • 58. Zhai Y, Su J, Ran W et al. Preparation and application of cell membrane-camouflaged nanoparticles for cancer therapy. Theranostics 7(10), 2575 (2017).
    • 59. Hu C-MJ, Zhang L, Aryal S et al. Erythrocyte membrane-camouflaged polymeric nanoparticles as a biomimetic delivery platform. Proc. Natl Acad. Sci. USA 108(27), 10980–10985 (2011).
    • 60. Gao F, Xu L, Yang B et al. Kill the real with the fake: eliminate intracellular Staphylococcus aureus using nanoparticle coated with its extracellular vesicle membrane as active-targeting drug carrier. ACS Infect. Dis. 5(2), 218–227 (2018).
    • 61. Cao H, Dan Z, He X et al. Liposomes coated with isolated macrophage membrane can target lung metastasis of breast cancer. ACS Nano 10(8), 7738–7748 (2016).
    • 62. Wolfram J, Zhu M, Yang Y et al. Safety of nanoparticles in medicine. Curr. Drug Target. 16(14), 1671–1681 (2015).
    • 63. Ren H, Liu J, Li Y et al. Oxygen self-enriched nanoparticles functionalized with erythrocyte membranes for long circulation and enhanced phototherapy. Acta Biomater. 59, 269–282 (2017).
    • 64. Pereverzeva E, Treschalin I, Treschalin M et al. Toxicological study of doxorubicin-loaded PLGA nanoparticles for the treatment of glioblastoma. Int. J. Pharmaceut. 554, 161–178 (2019).
    • 65. Sun H, Su J, Meng Q et al. Cancer cell membrane-coated gold nanocages with hyperthermia-triggered drug release and homotypic target inhibit growth and metastasis of breast cancer. Adv. Funct. Mater. 27(3), 1604300 (2017).
    • 66. Zhen X, Cheng P, Pu K. Recent advances in cell membrane–camouflaged nanoparticles for cancer phototherapy. Small 15(1), 1804105 (2019).
    • 67. Ren X, Zheng R, Fang X et al. Red blood cell membrane camouflaged magnetic nanoclusters for imaging-guided photothermal therapy. Biomaterials 92, 13–24 (2016).
    • 68. Rao L, Meng Q-F, Bu L-L et al. Erythrocyte membrane-coated upconversion nanoparticles with minimal protein adsorption for enhanced tumor imaging. ACS Appl. Mater. Interface 9(3), 2159–2168 (2017).
    • 69. 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).
    • 70. Xu L, Wu S, Wang J. Cancer cell membrane-coated nanocarriers for homologous target inhibiting the growth of hepatocellular carcinoma. J. Bioactive Compat. Polymer. 34(1), 58–71 (2019).
    • 71. Rao L, Cai B, Bu L-L et al. Microfluidic electroporation-facilitated synthesis of erythrocyte membrane-coated magnetic nanoparticles for enhanced imaging-guided cancer therapy. ACS Nano 11(4), 3496–3505 (2017).
    • 72. Luk BT, Hu C-MJ, Fang RH et al. Interfacial interactions between natural RBC membranes and synthetic polymeric nanoparticles. Nanoscale 6(5), 2730–2737 (2014).
    • 73. Xia Q, Zhang Y, Li Z et al. Red blood cell membrane-camouflaged nanoparticles: a novel drug delivery system for antitumor application. Acta Pharmaceutica Sinica B. 9(4), 675–689 (2019).
    • 74. Wang Q, Cheng H, Peng H et al. Non-genetic engineering of cells for drug delivery and cell-based therapy. Adv. Drug Deliv. Rev. 91, 125–140 (2015).
    • 75. Hymel D, Peterson BR. Synthetic cell surface receptors for delivery of therapeutics and probes. Adv. Drug Deliv. Rev. 64(9), 797–810 (2012).
    • 76. Tian H, Luo Z, Liu L et al. Cancer cell membrane-biomimetic oxygen nanocarrier for breaking hypoxia-induced chemoresistance. Adv. Funct. Mater. 27(38), 1703197 (2017).
    • 77. Liddy N, Bossi G, Adams KJ et al. Monoclonal TCR-redirected tumor cell killing. Nat. Med. 18(6), 980–987 (2012).
    • 78. Jing L, Qu H, Wu D et al. Platelet-camouflaged nanococktail: simultaneous inhibition of drug-resistant tumor growth and metastasis via a cancer cells and tumor vasculature dual-targeting strategy. Theranostics 8(10), 2683 (2018).
    • 79. Zhou H, Fan Z, Lemons PK et al. A facile approach to functionalize cell membrane-coated nanoparticles. Theranostics 6(7), 1012 (2016).
    • 80. Smarr CB, Hsu C-L, Byrne AJ et al. Antigen-fixed leukocytes tolerize Th2 responses in mouse models of allergy. J. Immunol. 187(10), 5090–5098 (2011).
    • 81. Patel RB, Ye M, Carlson PM et al. Development of an in situ cancer vaccine via combinational radiation and bacterial-membrane-coated nanoparticles. Adv. Mat. 31(43), 1902626 (2019).
    • 82. Cheng H, Byrska-Bishop M, Zhang CT et al. Stem cell membrane engineering for cell rolling using peptide conjugation and tuning of cell–selectin interaction kinetics. Biomaterials 33(20), 5004–5012 (2012).
    • 83. Stephan MT, Moon JJ, Um SH et al. Therapeutic cell engineering with surface-conjugated synthetic nanoparticles. Nat. Med. 16(9), 1035–1041 (2010).
    • 84. Zhang Q, Wei W, Wang P et al. Biomimetic magnetosomes as versatile artificial antigen-presenting cells to potentiate T-cell-based anticancer therapy. ACS Nano 11(11), 10724–10732 (2017).
    • 85. Sackstein R, Merzaban JS, Cain DW et al. Ex vivo glycan engineering of CD44 programs human multipotent mesenchymal stromal cell trafficking to bone. Nat. Med. 14(2), 181–187 (2008).
    • 86. Ma W, Zhu D, Li J et al. Coating biomimetic nanoparticles with chimeric antigen receptor T cell-membrane provides high specificity for hepatocellular carcinoma photothermal therapy treatment. Theranostics 10(3), 1281 (2020).
    • 87. Bose RJ, Kim BJ, Arai Y et al. Bioengineered stem cell membrane functionalized nanocarriers for therapeutic targeting of severe hindlimb ischemia. Biomaterials 185, 360–370 (2018).
    • 88. Shao J, Abdelghani M, Shen G et al. Erythrocyte membrane modified janus polymeric motors for thrombus therapy. ACS Nano 12(5), 4877–4885 (2018).
    • 89. Li J, Ai Y, Wang L et al. Targeted drug delivery to circulating tumor cells via platelet membrane-functionalized particles. Biomaterials 76, 52–65 (2016).
    • 90. Dehaini D, Wei X, Fang RH et al. Erythrocyte–platelet hybrid membrane coating for enhanced nanoparticle functionalization. Adv. Mater. 29(16), 1606209 (2017).
    • 91. Prince ME, Ailles LE. Cancer stem cells in head and neck squamous cell cancer. J. Clin. Oncol. 26(17), 2871–2875 (2008).
    • 92. Jiang Q, Liu Y, Guo R et al. Erythrocyte-cancer hybrid membrane-camouflaged melanin nanoparticles for enhancing photothermal therapy efficacy in tumors. Biomaterials 192, 292–308 (2019).
    • 93. Xin Y, Huang Q, Tang J-Q et al. Nanoscale drug delivery for targeted chemotherapy. Cancer Lett. 379(1), 24–31 (2016).
    • 94. Wilhelm S, Tavares AJ, Dai Q et al. Analysis of nanoparticle delivery to tumours. Nat. Rev. Mater. 1(5), 1–12 (2016).
    • 95. Li S-Y, Cheng H, Xie B-R et al. Cancer cell membrane camouflaged cascade bioreactor for cancer targeted starvation and photodynamic therapy. ACS Nano 11(7), 7006–7018 (2017).
    • 96. Zhu J-Y, Zheng D-W, Zhang M-K 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).
    • 97. Gdowski A-S, Lampe J-B, Lin V-J et al. Bioinspired nanoparticles engineered for enhanced delivery to the bone. ACS Appl. Nano Mater. 2(10), 6249–6257 (2019).
    • 98. Chowdhury MR, Schumann C, Bhakta-Guha D et al. Cancer nanotheranostics: strategies, promises and impediments. Biomed. Pharmacother. 84, 291–304 (2016).
    • 99. Imran M, Shah MR, Ullah F et al. Double-tailed acyl glycoside niosomal nanocarrier for enhanced oral bioavailability of cefixime. Artific. Cell. Nanomed. Biotechnol. 45(7), 1440–1451 (2017).
    • 100. Chen Z, Zhao P, Luo Z et al. Cancer cell membrane–biomimetic nanoparticles for homologous-targeting dual-modal imaging and photothermal therapy. ACS Nano 10(11), 10049–10057 (2016).
    • 101. Hussain Z, Rahim MA, Jan N et al. Cell membrane cloaked nanomedicines for bio-imaging and immunotherapy of cancer: improved pharmacokinetics, cell internalization and anticancer efficacy. J. Control. Rel. 335, 130–157 (2021). •• The adaptation of cell membrane-cloaked nanomedicines revolutionizes drug-delivery strategies by optimizing plasma circulation time, enhancing permeation into the cancerous microenvironment, evading immune response and enabling cell–cell communication via cell membrane markers.
    • 102. Seledtsov V, Goncharov A, Seledtsova G. Clinically feasible approaches to potentiating cancer cell-based immunotherapies. Hum. Vaccin. Immunotherapeut. 11(4), 851–869 (2015).
    • 103. Fontana F, Shahbazi MA, Liu D et al. Multistaged nanovaccines based on porous silicon@ acetalated dextran@ cancer cell membrane for cancer immunotherapy. Adv. Mater. 29(7), 1603239 (2017).
    • 104. Jin J, Chang D, Chatterjee S et al. Cancer cell membrane coated biomimetic nanoparticles: synthesis, characterization, and functionality. Cancer Res. 77(Suppl. 13), 2198 (2017).
    • 105. Wang H, Liu Y, He R et al. Cell membrane biomimetic nanoparticles for inflammation and cancer targeting in drug delivery. Biomat. Sci. 8(2), 552–568 (2020).
    • 106. Li R, He Y, Zhang S et al. Cell membrane-based nanoparticles: a new biomimetic platform for tumor diagnosis and treatment. Acta Pharmaceutica Sinica B. 8(1), 14–22 (2018).
    • 107. Bhattacharyya S, Ghosh SS. Transmembrane TNFα-expressed macrophage membrane-coated chitosan nanoparticles as cancer therapeutics. ACS Omega 5(3), 1572–1580 (2020).
    • 108. Fontana F, Albertini S, Correia A et al. Bioengineered porous silicon nanoparticles@ macrophages cell membrane as composite platforms for rheumatoid arthritis. Adv. Funct. Mater. 28(22), 1801355 (2018).
    • 109. Zhang Y, Cai K, Li C et al. Macrophage-membrane-coated nanoparticles for tumor-targeted chemotherapy. Nano Lett. 18(3), 1908–1915 (2018).
    • 110. Meng Q-F, Rao L, Zan M et al. Macrophage membrane-coated iron oxide nanoparticles for enhanced photothermal tumor therapy. Nanotechnology 29(13), 134004 (2018).
    • 111. Cheng S, Xu C, Jin Y et al. Artificial mini dendritic cells boost T cell-based immunotherapy for ovarian cancer. Adv. Sci. 7(7), 1903301 (2020).
    • 112. Gong P, Wang Y, Zhang P et al. Immunocyte membrane-coated nanoparticles for cancer immunotherapy. Cancers 2021 13(1), 77 (2020).
    • 113. Jakubzick CV, Randolph GJ, Henson PM. Monocyte differentiation and antigen-presenting functions. Nat. Rev. Immunol. 17(6), 349–362 (2017).
    • 114. Zhang C, Zhang J, Shi G et al. A light responsive nanoparticle-based delivery system using pheophorbide a graft polyethylenimine for dendritic cell-based cancer immunotherapy. Mol. Pharmaceut. 14(5), 1760–1770 (2017).
    • 115. Pitchaimani A, Nguyen TDT, Aryal S. Natural killer cell membrane infused biomimetic liposomes for targeted tumor therapy. Biomaterials 160, 124–137 (2018).
    • 116. Wu L, Zhang F, Wei Z et al. Magnetic delivery of Fe3O4@ polydopamine nanoparticle-loaded natural killer cells suggest a promising anticancer treatment. Biomater. Sci. 6(10), 2714–2725 (2018).
    • 117. Deng G, Sun Z, Li S et al. Cell-membrane immunotherapy based on natural killer cell membrane coated nanoparticles for the effective inhibition of primary and abscopal tumor growth. ACS Nano 12(12), 12096–12108 (2018).
    • 118. Ricchi P, Zarrilli R, Di Palma A, Acquaviva A. Nonsteroidal anti-inflammatory drugs in colorectal cancer: from prevention to therapy. Br. J. Cancer 88(6), 803–807 (2003).
    • 119. Cao X, Hu Y, Luo S et al. Neutrophil-mimicking therapeutic nanoparticles for targeted chemotherapy of pancreatic carcinoma. Acta Pharmaceutica Sinica B. 9(3), 575–589 (2019).
    • 120. Kang T, Zhu Q, Wei D et al. Nanoparticles coated with neutrophil membranes can effectively treat cancer metastasis. ACS Nano 11(2), 1397–1411 (2017).
    • 121. He Z, Zhang Y, Feng N. Cell membrane-coated nanosized active targeted drug delivery systems homing to tumor cells: a review. Mat. Sci. Eng. C 106, 110298 (2020).
    • 122. Wang K, Lei Y, Xia D et al. Neutrophil membranes coated, antibiotic agent loaded nanoparticles targeting to the lung inflammation. Colloid. Surf. B. Biointerface 188, 110755 (2020).
    • 123. Gao H, Priebe W, Glod J et al. Activation of signal transducers and activators of transcription 3 and focal adhesion kinase by stromal cell-derived factor 1 is required for migration of human mesenchymal stem cells in response to tumor cell-conditioned medium. Stem Cells 27(4), 857–865 (2009).
    • 124. Khosravi N, Pishavar E, Baradaran B et al. Stem cell membrane, stem cell-derived exosomes and hybrid stem cell camouflaged nanoparticles: a promising biomimetic nanoplatforms for cancer theranostics. J. Control. Rel. 348, 706–722 (2022).
    • 125. Ahn JO, Lee HW, Seo KW et al. Anti-tumor effect of adipose tissue derived-mesenchymal stem cells expressing interferon-β and treatment with cisplatin in a xenograft mouse model for canine melanoma. PLOS ONE 8(9), e74897 (2013).
    • 126. Li Y-S, Wu H-H, Jiang X-C et al. Active stealth and self-positioning biomimetic vehicles achieved effective antitumor therapy. J. Control. Rel. 335, 515–526 (2021).
    • 127. Zhou D, Chen Y, Bu W et al. Modification of metal-organic framework nanoparticles using dental pulp mesenchymal stem cell membranes to target oral squamous cell carcinoma. J. Colloid Interf. Sci. 601, 650–660 (2021).
    • 128. Mu X, Zhang M, Wei A et al. Doxorubicin and PD-L1 siRNA co-delivery with stem cell membrane-coated polydopamine nanoparticles for the targeted chemoimmunotherapy of PCa bone metastases. Nanoscale 13(19), 8998–9008 (2021).
    • 129. Olsson M, Bruhns P, Frazier WA et al. Platelet homeostasis is regulated by platelet expression of CD47 under normal conditions and in passive immune thrombocytopenia. Blood 105(9), 3577–3582 (2005).
    • 130. Lievens D, Von Hundelshausen P. Platelets in atherosclerosis. Thromb. Haemostasis 106(11), 827–838 (2011).
    • 131. Chi C, Li F, Liu H et al. Docetaxel-loaded biomimetic nanoparticles for targeted lung cancer therapy in vivo. J. Nanoparticle Res. 21, 1–10 (2019).
    • 132. Zhuang J, Gong H, Zhou J et al. Targeted gene silencing in vivo by platelet membrane–coated metal-organic framework nanoparticles. Sci. Adv. 6(13), eaaz6108 (2020).
    • 133. Shibu ES, Hamada M, Murase N et al. Nanomaterials formulations for photothermal and photodynamic therapy of cancer. J. Photochem. Photobiol. C: Photochem. Rev. 15, 53–72 (2013).
    • 134. Xu L, Gao F, Fan F et al. Platelet membrane coating coupled with solar irradiation endows a photodynamic nanosystem with both improved antitumor efficacy and undetectable skin damage. Biomaterials 159, 59–67 (2018).
    • 135. Hu Q, Sun W, Qian C et al. Anticancer platelet-mimicking nanovehicles. Adv. Mater. 27(44), 7043 (2015).
    • 136. Oikonomou E, Pintzas A. The TRAIL of oncogenes to apoptosis. Biofactors 39(4), 343–354 (2013).
    • 137. Wang H, Bremner DH, Wu K et al. Platelet membrane biomimetic bufalin-loaded hollow MnO2 nanoparticles for MRI-guided chemo-chemodynamic combined therapy of cancer. Chem. Eng. J. 382, 122848 (2020).
    • 138. Pisano S, Pierini I, Gu J et al. Immune (cell) derived exosome mimetics (IDEM) as a treatment for ovarian cancer. Front. Cell Develop. Biol. 8, 553576 (2020).
    • 139. Li B, Wang F, Gui L et al. The potential of biomimetic nanoparticles for tumor-targeted drug delivery. Nanomedicine 13(16), 2099–2118 (2018).
    • 140. Susa F, Limongi T, Dumontel B et al. Engineered extracellular vesicles as a reliable tool in cancer nanomedicine. Cancers 11(12), 1979 (2019).
    • 141. Jin K, Luo Z, Zhang B et al. Biomimetic nanoparticles for inflammation targeting. Acta Pharmaceutica Sinica B. 8(1), 23–33 (2018).
    • 142. Xia Y, Rao L, Yao H et al. Engineering macrophages for cancer immunotherapy and drug delivery. Adv. Mater. 32(40), 2002054 (2020).