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

Emerging advances in cationic liposomal cancer nanovaccines: opportunities and challenges

    Mohammad Z Ahmad

    *Author for correspondence:

    E-mail Address: zaki.manipal@hotmail.com

    Department of Pharmaceutics, College of Pharmacy, Najran University, Najran 66241, Kingdom of Saudi Arabia

    ,
    Javed Ahmad

    Department of Pharmaceutics, College of Pharmacy, Najran University, Najran 66241, Kingdom of Saudi Arabia

    ,
    Mohammed Y Alasmary

    Department of Internal Medicine, College of Medicine, Najran University Hospital, Najran 66241, Kingdom of Saudi Arabia

    ,
    Basel A Abdel-Wahab

    Department of Pharmacology, College of Pharmacy, Najran University, Najran 66241, Kingdom of Saudi Arabia

    Department of Medical Pharmacology, College of Medicine, Assiut University, Assiut 71111, Egypt

    ,
    Musarrat H Warsi

    Department of Pharmaceutics & Industrial Pharmacy, College of Pharmacy, Taif University, Taif-Al-Haweiah 21974, Kingdom of Saudi Arabia

    ,
    Anzarul Haque

    Department of Pharmacognosy, College of Pharmacy, Prince Sattam bin Abdulaziz University, Al-Kharj 11942, Kingdom of Saudi Arabia

    &
    Pramila Chaubey

    Department of Pharmaceutics, College of Pharmacy, Shaqra University, Al-Dawadmi 17431, Kingdom of Saudi Arabia

    Published Online:https://doi.org/10.2217/imt-2020-0258

    Advancements in the field of cancer therapeutics have witnessed a recent surge in the use of liposomes. The physicochemical characteristics of the liposomes and their components, including the lipid phase transition temperature, vesicular size and size distribution, surface properties, and route of administration, play a significant role in the modulation of the immune response as an adjuvant and for loaded antigen (Ag). Cationic liposomes, concerning their potential ability to amplify the immunogenicity of the loaded Ag/adjuvant, have received enormous interest as a promising vaccine delivery platform for cancer immunotherapy. In the present review, the physicochemical considerations for the development of Ag/adjuvant-loaded liposomes and the cationic liposomes’ effectiveness for promoting cancer immunotherapy have been summarized.

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

    References

    • 1. World Health Organization. Cancer. (2020). www.who.int/news-room/fact-sheets/detail/cancer
    • 2. American Cancer Society. Cancer Facts & Figures 2019. (2019). www.cancer.org/content/dam/cancer-org/research/cancer-facts-and-statistics/annual-cancer-facts-and-figures/2019/cancer-facts-and-figures-2019.pdf
    • 3. Howard CJ, Charleston B, Stephens SA, Sopp P, Hope JC. The role of dendritic cells in shaping the immune response. Anim. Health. Res. Rev. 5(2), 191–195 (2004).
    • 4. Gao A, Hu XL, Saeed M, Chen BF, Li YP, Yu HJ. Overview of recent advances in liposomal nanoparticle-based cancer immunotherapy. Acta. Pharmacol. Sin. 40(9), 1129–1137 (2019).
    • 5. Mbongue J, Nicholas D, Firek A, Langridge W. The role of dendritic cells in tissue-specific autoimmunity. J. Immunol. Res. 2014, 857143–857143 (2014).
    • 6. Nesmiyanov P. British Society for Immunology. Antigen Processing and Presentation. (2020). www.immunology.org/public-information/bitesized-immunology/systems-and-processes/antigen-processing-and-presentation
    • 7. Jia J, Zhang Y, Xin Y, Jiang C, Yan B, Zhai S. Interactions between nanoparticles and dendritic cells: from the perspective of cancer immunotherapy. Front. Oncol. 8, 404 (2018). • Recent progress in targeting dendritic cells using nanoparticles as a drug-delivery carrier in cancer immunotherapy.
    • 8. Ahmad MZ, Ahmad J, Haque A, Alasmary M, Abdel-Wahab B, Akhter S. Emerging advances in synthetic cancer nano-vaccines: opportunities and challenges. Expert Rev. Vaccines 19(11), 1053–1071 (2020). •• Advancements in nanomedicine-based cancer nanovaccines.
    • 9. Grimaldi AM, Incoronato M, Salvatore M, Soricelli A. Nanoparticle-based strategies for cancer immunotherapy and immunodiagnostics. Nanomedicine (Lond.) 12(19), 2349–2365 (2017).
    • 10. Liu J, Zhang R, Xu ZP. Nanoparticle-based nanomedicines to promote cancer immunotherapy: recent advances and future directions. Small 15(32), e1900262 (2019).
    • 11. Yan S, Zhao P, Yu T, Gu N. Current applications and future prospects of nanotechnology in cancer immunotherapy. Cancer Biol. Med. 16(3), 486–497 (2019).
    • 12. Van Der Burg SH, Arens R, Ossendorp F, Van Hall T, Melief CJ. Vaccines for established cancer: overcoming the challenges posed by immune evasion. Nat. Rev. Cancer 16(4), 219–233 (2016).
    • 13. Schwendener RA. Liposomes as vaccine delivery systems: a review of the recent advances. Ther. Adv. Vaccines 2(6), 159–182 (2014).
    • 14. Blume G, Cevc G, Crommelin MD, Bakker-Woudenberg IA, Kluft C, Storm G. Specific targeting with poly(ethylene glycol)-modified liposomes: coupling of homing devices to the ends of the polymeric chains combines effective target binding with long circulation times. Biochem. Biophys. Acta. 1149(1), 180–184 (1993).
    • 15. Torchilin VP. Recent advances with liposomes as pharmaceutical carriers. Nat. Rev. Drug. Discov. 4(2), 145–160 (2005).
    • 16. Watson DS, Endsley AN, Huang L. Design considerations for liposomal vaccines: influence of formulation parameters on antibody and cell-mediated immune responses to liposome associated antigens. Vaccine 30(13), 2256–2272 (2012). •• Rational design of liposomal vaccines to elicit immune responses of a desired magnitude and quality.
    • 17. Henriksen-Lacey M, Devitt A, Perrie Y. The vesicle size of DDA:TDB liposomal adjuvants plays a role in the cell-mediated immune response but has no significant effect on antibody production. J. Control Rel. 154(2), 131–137 (2011).
    • 18. Evelyn Roopngam P. Liposome and polymer-based nanomaterials for vaccine applications. Nanomed. J. 6(1), 1–10 (2019).
    • 19. Shenzhen Institutes of Advanced Technology. CN102973506A (2011).
    • 20. National Taiwan Ocean University (NTOU). US20110182976A1 (2011).
    • 21. PDS Biotechnology Corporation. CN102137675A (2009).
    • 22. Inex Pharmaceuticals Corporation. JP2005530761A (2003).
    • 23. Curevac AG. ES2759910T3 (2020).
    • 24. Allison AC, Gregoriadis G. Liposomes as immunological adjuvants. In: Lymphocytes, Macrophages, and Cancer. Mathe GFlorentin ISimmler M-C (Eds). Springer Berlin Heidelberg, Berlin, Heidelberg, Germany, 58–64 (1976).
    • 25. Allison AG, Gregoriadis G. Liposomes as immunological adjuvants. Nature 252(5480), 252 (1974).
    • 26. Shao K, Singha S, Clemente-Casares X, Tsai S, Yang Y, Santamaria P. Nanoparticle-based immunotherapy for cancer. ACS Nano. 9(1), 16–30 (2015).
    • 27. Mahjub R, Jatana S, Lee SE et al. Recent advances in applying nanotechnologies for cancer immunotherapy. J. Control. Rel. 288, 239–263 (2018). •• Advancement in nanotechnology for immunotherapy against cancer.
    • 28. Kwong B, Gai SA, Elkhader J, Wittrup KD, Irvine DJ. Localized immunotherapy via liposome-anchored anti-CD137 + IL-2 prevents lethal toxicity and elicits local and systemic antitumor immunity. Cancer Res. 73(5), 1547 (2013).
    • 29. Pardoll DM. Paracrine cytokine adjuvants in cancer immunotherapy. Annu. Rev. Immunol. 13(1), 399–415 (1995).
    • 30. Chen WC, Huang L. Non-viral vector as vaccine carrier. Adv. Genet. 54, 315–337 (2005).
    • 31. Zamani P, Momtazi-Borojeni AA, Nik ME, Oskuee RK, Sahebkar A. Nanoliposomes as the adjuvant delivery systems in cancer immunotherapy. J. Cell. Physiol. 233(7), 5189–5199 (2018). • Properties of nanoliposomes as delivery systems for cancer immunotherapy.
    • 32. Liang MT, Davies NM, Toth I. Encapsulation of lipopeptides within liposomes: effect of number of lipid chains, chain length and method of liposome preparation. Int. J. Pharm. 301(1–2), 247–254 (2005).
    • 33. Dancey GF, Yasuda T, Kinsky SC. Effect of liposomal model membrane composition on immunogenicity. J. Immunol. 120(4), 1109–1113 (1978).
    • 34. Nakano Y, Mori M, Nishinohara S et al. Surface-linked liposomal antigen induces ige-selective unresponsiveness regardless of the lipid components of liposomes. Bioconjug. Chem. 12(3), 391–395 (2001).
    • 35. Mazumdar T, Anam K, Ali N. Influence of phospholipid composition on the adjuvanticity and protective efficacy of liposome-encapsulated Leishmania donovani antigens. J. Parasitol. 91(2), 269–274 (2005).
    • 36. Christensen D, Henriksen-Lacey M, Kamath AT et al. A cationic vaccine adjuvant based on a saturated quaternary ammonium lipid have different in vivo distribution kinetics and display a distinct CD4 T cell-inducing capacity compared to its unsaturated analog. J. Control. Rel. 160(3), 468–476 (2012).
    • 37. Badiee A, Khamesipour A, Samiei A et al. The role of liposome size on the type of immune response induced in BALB/c mice against leishmaniasis: rgp63 as a model antigen. Exp. Parasitol. 132(4), 403–409 (2012).
    • 38. Bachmann MF, Jennings GT. Vaccine delivery: a matter of size, geometry, kinetics and molecular patterns. Nat. Rev. Immunol. 10(11), 787–796 (2010).
    • 39. Reddy ST, Van Der Vlies AJ, Simeoni E et al. Exploiting lymphatic transport and complement activation in nanoparticle vaccines. Nat. Biotechnol. 25(10), 1159–1164 (2007).
    • 40. Carstens MG, Camps MG, Henriksen-Lacey M et al. Effect of vesicle size on tissue localization and immunogenicity of liposomal DNA vaccines. Vaccine 29(29–30), 4761–4770 (2011).
    • 41. Manolova V, Flace A, Bauer M, Schwarz K, Saudan P, Bachmann MF. Nanoparticles target distinct dendritic cell populations according to their size. Eur. J. Immunol. 38(5), 1404–1413 (2008).
    • 42. Kaminskas LM, Porter CJ. Targeting the lymphatics using dendritic polymers (dendrimers). Adv. Drug Deliv. Rev. 63(10–11), 890–900 (2011).
    • 43. Kourtis IC, Hirosue S, De Titta A et al. Peripherally administered nanoparticles target monocytic myeloid cells, secondary lymphoid organs and tumors in mice. PLoS ONE 8(4), e61646 (2013).
    • 44. Brewer JM, Tetley L, Richmond J, Liew FY, Alexander J. Lipid vesicle size determines the Th1 or Th2 response to entrapped antigen. J. Immunol. 161(8), 4000–4007 (1998). • Novel mechanism by which the balance of the Th1/Th2 response to an Ag can be influenced.
    • 45. Oyewumi MO, Kumar A, Cui Z. Nano-microparticles as immune adjuvants: correlating particle sizes and the resultant immune responses. Expert Rev. Vaccines 9(9), 1095–1107 (2010).
    • 46. Conniot J, Silva JM, Fernandes JG et al. Cancer immunotherapy: nanodelivery approaches for immune cell targeting and tracking. Front. Chem. 2, 105 (2014). • Nanodelivery strategies for cancer immunotherapy, cancer targeting mechanisms and nanocarrier functionalization.
    • 47. Perrie Y, Crofts F, Devitt A, Griffiths HR, Kastner E, Nadella V. Designing liposomal adjuvants for the next generation of vaccines. Adv. Drug Deliv. Rev. 99(Pt A), 85–96 (2016).
    • 48. Wang N, Chen M, Wang T. Liposomes used as a vaccine adjuvant-delivery system: from basics to clinical immunization. J. Control. Rel. 303, 130–150 (2019).
    • 49. Foged C, Arigita C, Sundblad A, Jiskoot W, Storm G, Frokjaer S. Interaction of dendritic cells with antigen-containing liposomes: effect of bilayer composition. Vaccine 22(15–16), 1903–1913 (2004).
    • 50. Werninghaus K, Babiak A, Gross O et al. Adjuvanticity of a synthetic cord factor analogue for subunit Mycobacterium tuberculosis vaccination requires FcRgamma-Syk-Card9-dependent innate immune activation. J. Exp. Med. 206(1), 89–97 (2009).
    • 51. Henriksen-Lacey M, Christensen D, Bramwell VW et al. Liposomal cationic charge and antigen adsorption are important properties for the efficient deposition of antigen at the injection site and ability of the vaccine to induce a CMI response. J. Control. Rel. 145(2), 102–108 (2010).
    • 52. Norris DA, Puri N, Sinko PJ. The effect of physical barriers and properties on the oral absorption of particulates. Adv. Drug Deliv. Rev. 34(2–3), 135–154 (1998).
    • 53. Schmidt ST, Khadke S, Korsholm KS et al. The administration route is decisive for the ability of the vaccine adjuvant CAF09 to induce antigen-specific CD8(+) T-cell responses: the immunological consequences of the biodistribution profile. J. Control. Rel. 239, 107–117 (2016).
    • 54. Wang T, Zhen Y, Ma X, Wei B, Li S, Wang N. Mannosylated and lipid A-incorporating cationic liposomes constituting microneedle arrays as an effective oral mucosal HBV vaccine applicable in the controlled temperature chain. Colloids Surf. B. Biointerfaces 126, 520–530 (2015).
    • 55. Yuba E. Liposome-based immunity-inducing systems for cancer immunotherapy. Mol. Immunol. 98, 8–12 (2018).
    • 56. Heuts J, Varypataki EM, Van Der Maaden K et al. Cationic liposomes: a flexible vaccine delivery system for physicochemically diverse antigenic peptides. Pharm. Res. 35(11), 207 (2018).
    • 57. Vanbever R, Loira-Pastoriza C, Dauguet N et al. Cationic nanoliposomes are efficiently taken up by alveolar macrophages but have little access to dendritic cells and interstitial macrophages in the normal and CpG-stimulated lungs. Mol. Pharm. 16(5), 2048–2059 (2019).
    • 58. Yan W, Chen W, Huang L. Mechanism of adjuvant activity of cationic liposome: phosphorylation of a MAP kinase, ERK and induction of chemokines. Mol. Immunol. 44(15), 3672–3681 (2007). •• Adjuvant properties of cationic liposome.
    • 59. Chen W, Huang L. Induction of cytotoxic T-lymphocytes and antitumor activity by a liposomal lipopeptide vaccine. Mol. Pharm. 5(3), 464–471 (2008).
    • 60. Chen W, Yan W, Huang L. A simple but effective cancer vaccine consisting of an antigen and a cationic lipid. Cancer Immunol. Immunother. 57(4), 517–530 (2008).
    • 61. Yan W, Chen W, Huang L. Reactive oxygen species play a central role in the activity of cationic liposome based cancer vaccine. J. Control. Rel. 130(1), 22–28 (2008). •• Role of reactive oxygen species in the activity of cationic liposome.
    • 62. Yan W, Huang L. The effects of salt on the physicochemical properties and immunogenicity of protein based vaccine formulated in cationic liposome. Int. J. Pharm. 368(1–2), 56–62 (2009).
    • 63. Yun CH, Bae CS, Ahn T. Cargo-free nanoparticles containing cationic lipids induce reactive oxygen species and cell death in HepG2 cells. Biol. Pharm. Bull. 39(8), 1338–1346 (2016).
    • 64. Wang C, Zhuang Y, Zhang Y et al. Toll-like receptor 3 agonist complexed with cationic liposome augments vaccine-elicited antitumor immunity by enhancing TLR3-IRF3 signaling and type I interferons in dendritic cells. Vaccine 30(32), 4790–4799 (2012).
    • 65. Alipour Talesh G, Ebrahimi Z, Badiee A et al. Poly (I:C)-DOTAP cationic nanoliposome containing multi-epitope HER2-derived peptide promotes vaccine-elicited anti-tumor immunity in a murine model. Immunol. Lett. 176, 57–64 (2016).
    • 66. Zhuang Y, Ma Y, Wang C et al. PEGylated cationic liposomes robustly augment vaccine-induced immune responses: role of lymphatic trafficking and biodistribution. J. Control. Rel. 159(1), 135–142 (2012). •• Functionalized cationic lipsome and enhanced immune response.
    • 67. Zhang J, Tian H, Li C et al. Antitumor effects obtained by autologous Lewis lung cancer cell vaccine engineered to secrete mouse interleukin 27 by means of cationic liposome. Mol. Immunol. 55(3–4), 264–274 (2013).
    • 68. Mansourian M, Badiee A, Jalali SA et al. Effective induction of anti-tumor immunity using p5 HER-2/neu derived peptide encapsulated in fusogenic DOTAP cationic liposomes co-administrated with CpG-ODN. Immunol. Lett. 162(1 Pt A), 87–93 (2014).
    • 69. Yazdani M, Hatamipour M, Alani B et al. Liposomal gp100 vaccine combined with CpG ODN sensitizes established B16F10 melanoma tumors to anti PD-1 therapy Iran. J. Basic Med. Sci. 23(8), 1065–1077 (2020).
    • 70. Varypataki EM, Van Der Maaden K, Bouwstra J, Ossendorp F, Jiskoot W. Cationic liposomes loaded with a synthetic long peptide and poly(I:C): a defined adjuvanted vaccine for induction of antigen-specific T cell cytotoxicity. Aaps. J. 17(1), 216–226 (2015).
    • 71. Sayour EJ, De Leon G, Pham C et al. Systemic activation of antigen-presenting cells via RNA-loaded nanoparticles. Oncoimmunology 6(1), e1256527 (2017).
    • 72. Shen KY, Liu HY, Li HJ et al. A novel liposomal recombinant lipoimmunogen enhances anti-tumor immunity. J. Control. Rel. 233, 57–63 (2016).
    • 73. Barati N, Nikpoor AR, Razazan A et al. Nanoliposomes carrying HER2/neu-derived peptide AE36 with CpG-ODN exhibit therapeutic and prophylactic activities in a mice TUBO model of breast cancer. Immunol. Lett. 190, 108–117 (2017).
    • 74. Liu C, Chu X, Sun P et al. Synergy effects of Polyinosinic-polycytidylic acid, CpG oligodeoxynucleotide, and cationic peptides to adjuvant HPV E7 epitope vaccine through preventive and therapeutic immunization in a TC-1 grafted mouse model. Hum. Vaccin. Immunother. 14(4), 931–940 (2018).
    • 75. Siegel DP, Epand RM. The mechanism of lamellar-to-inverted hexagonal phase transitions in phosphatidylethanolamine: implications for membrane fusion mechanisms. Biophys. J. 73(6), 3089–3111 (1997).
    • 76. Kunisawa J, Mayumi T. [Application of novel drug delivery system, fusogenic liposome, for cancer therapy. Gan. To. Kagaku. Ryoho. 28(5), 577–583 (2001). • Fusogenic liposome in cancer therapy.
    • 77. Kunisawa J, Nakagawa S, Mayumi T. Pharmacotherapy by intracellular delivery of drugs using fusogenic liposomes: application to vaccine development. Adv. Drug Deliv. Rev. 52(3), 177–186 (2001).
    • 78. Okazaki S, Iwasaki T, Yuba E, Watarai S. Evaluation of pH-sensitive fusogenic polymer-modified liposomes co-loaded with antigen and α-galactosylceramide as an anti-tumor vaccine. J. Vet. Med. Sci. 80(2), 197–204 (2018).
    • 79. Razazan A, Behravan J, Arab A et al. Conjugated nanoliposome with the HER2/neu-derived peptide GP2 as an effective vaccine against breast cancer in mice xenograft model. PLoS ONE 12(10), e0185099 (2017).
    • 80. Farzad N, Barati N, Momtazi-Borojeni AA et al. P435 HER2/neu-derived peptide conjugated to liposomes containing DOPE as an effective prophylactic vaccine formulation for breast cancer. Artif. Cells Nanomed. Biotechnol. 47(1), 665–673 (2019).
    • 81. Persano S, Guevara ML, Li Z et al. Lipopolyplex potentiates anti-tumor immunity of mRNA-based vaccination. Biomaterials 125, 81–89 (2017).
    • 82. Arab A, Behravan J, Razazan A et al. A nano-liposome vaccine carrying E75, a HER-2/neu-derived peptide, exhibits significant antitumour activity in mice. J. Drug Target. 26(4), 365–372 (2018). •• Significant anticancer activity of nanoliposome loaded with HER-2/neu-derived peptide.
    • 83. Shariat S, Badiee A, Jalali SA et al. P5 HER2/neu-derived peptide conjugated to liposomes containing MPL adjuvant as an effective prophylactic vaccine formulation for breast cancer. Cancer Lett. 355(1), 54–60 (2014).
    • 84. Zamani P, Navashenaq JG, Nikpoor AR et al. MPL nano-liposomal vaccine containing P5 HER2/neu-derived peptide pulsed PADRE as an effective vaccine in a mice TUBO model of breast cancer. J. Control. Rel. 303, 223–236 (2019).
    • 85. Vitor MT, Bergami-Santos PC, Zômpero RHF et al. Cationic liposomes produced via ethanol injection method for dendritic cell therapy. J. Liposome Res. 27(4), 249–263 (2017).
    • 86. Lechanteur A, Furst T, Evrard B, Delvenne P, Hubert P, Piel G. Development of anti-E6 pegylated lipoplexes for mucosal application in the context of cervical preneoplastic lesions. Int. J. Pharm. 483(1–2), 268–277 (2015).
    • 87. Simberg D, Weisman S, Talmon Y, Barenholz Y. DOTAP (and other cationic lipids): chemistry, biophysics, and transfection. Crit. Rev. Ther. Drug Carrier Syst. 21(4), 257–317 (2004).
    • 88. Porteous DJ, Dorin JR, Mclachlan G et al. Evidence for safety and efficacy of DOTAP cationic liposome mediated CFTR gene transfer to the nasal epithelium of patients with cystic fibrosis. Gene Ther. 4(3), 210–218 (1997).
    • 89. Fasol U, Frost A, Büchert M et al. Vascular and pharmacokinetic effects of EndoTAG-1 in patients with advanced cancer and liver metastasis. Ann. Oncol. 23(4), 1030–1036 (2012).
    • 90. Strieth S, Dunau C, Michaelis U et al. Phase I/II clinical study on safety and antivascular effects of paclitaxel encapsulated in cationic liposomes for targeted therapy in advanced head and neck cancer. Head Neck 36(7), 976–984 (2014).
    • 91. Ignatiadis M, Zardavas D, Lemort M et al. Feasibility study of EndoTAG-1, a tumor endothelial targeting agent, in combination with paclitaxel followed by FEC as induction therapy in HER2-negative breast cancer. PLoS ONE 11(7), e0154009 (2016).
    • 92. Awada A, Bondarenko IN, Bonneterre J et al. A randomized controlled Phase II trial of a novel composition of paclitaxel embedded into neutral and cationic lipids targeting tumor endothelial cells in advanced triple-negative breast cancer (TNBC). Ann. Oncol. 25(4), 824–831 (2014).
    • 93. ClinicalTrials.gov, National Library of Medicine (U.S.). EndoTAG-1 plus gemcitabine versus gemcitabine alone in patients with measurable locally advanced and/or metastatic adenocarcinoma of the pancreas failed on FOLFIRINOX treatment NCT03126435. (2017). https://clinicaltrials.gov/ct2/show/NCT03126435
    • 94. Cho EA, Moloney FJ, Cai H et al. Safety and tolerability of an intratumorally injected DNAzyme, Dz13, in patients with nodular basal-cell carcinoma: a Phase I first-in-human trial (DISCOVER). Lancet 381(9880), 1835–1843 (2013).
    • 95. Lu C, Stewart DJ, Lee JJ et al. Phase I clinical trial of systemically administered TUSC2(FUS1)-nanoparticles mediating functional gene transfer in humans. PLoS ONE 7(4), e34833 (2012).
    • 96. ClinicalTrials.gov, National Library of Medicine (U.S.). TUSC2-nanoparticles and erlotinib in stage IV lung cancer NCT01455389. (2011). https://clinicaltrials.gov/ct2/show/NCT01455389
    • 97. Zhi D, Zhang S, Wang B, Zhao Y, Yang B, Yu S. Transfection efficiency of cationic lipids with different hydrophobic domains in gene delivery. Bioconjug. Chem. 21(4), 563–577 (2010).
    • 98. Sayour EJ, Mendez-Gomez HR, Mitchell DA. Cancer vaccine immunotherapy with RNA-loaded liposomes. Int. J. Mol. Sci. 19(10), 2890 (2018).