456
Views
13
CrossRef citations to date
0
Altmetric
Review

Enhancing tumor specific immune responses by transcutaneous vaccination

, , , , , & show all
Pages 1079-1094 | Received 16 Jun 2017, Accepted 18 Sep 2017, Published online: 17 Oct 2017

References

  • Dunn GP, Bruce AT, Ikeda H, et al. Cancer immunoediting: from immunosurveillance to tumor escape. Nat Immunol. 2002;3:991–998.
  • Teng MWL, Galon J, Fridman W-H, et al. From mice to humans: developments in cancer immunoediting. J Clin Invest. 2015;125:3338.
  • Wherry EJ. T cell exhaustion. Nat Immunol. 2011;12:492–499.
  • Combadiere B, Liard C. Transcutaneous and intradermal vaccination. Hum Vaccin. 2011;7:811–827.
  • Kupper TS, Fuhlbrigge RC. Immune surveillance in the skin: mechanisms and clinical consequences. Nat Rev Immunol. 2004;4:211–222.
  • Wosicka H, Cal K. Targeting to the hair follicles: current status and potential. J Dermatol Sci. 2010;57:83–89.
  • Nishibu A, Ward BR, Jester JV, et al. Behavioral responses of epidermal Langerhans cells in situ to local pathological stimuli. J Invest Dermatol. 2006;126:787–796.
  • Henri S, Poulin LF, Tamoutounour S, et al. CD207+ CD103+ dermal dendritic cells cross-present keratinocyte-derived antigens irrespective of the presence of Langerhans cells. J Exp Med. 2010;207:189–206.
  • Yoshida K, Kubo A, Fujita H, et al. Distinct behavior of human Langerhans cells and inflammatory dendritic epidermal cells at tight junctions in patients with atopic dermatitis. J Allergy Clin Immunol. 2014;134:856–864.
  • Ouchi T, Kubo A, Yokouchi M, et al. Langerhans cell antigen capture through tight junctions confers preemptive immunity in experimental staphylococcal scalded skin syndrome. J Exp Med. 2011;208:2607–2613.
  • Hemmi H, Yoshino M, Yamazaki H, et al. Skin antigens in the steady state are trafficked to regional lymph nodes by transforming growth factor-beta1-dependent cells. Int Immunol. 2001;13:695–704.
  • Flacher V, Tripp CH, Stoitzner P, et al. Epidermal Langerhans cells rapidly capture and present antigens from C-type lectin-targeting antibodies deposited in the dermis. J Invest Dermatol. 2010;130:755–762.
  • Flacher V, Tripp CH, Haid B, et al. Skin langerin+ dendritic cells transport intradermally injected anti-DEC-205 antibodies but are not essential for subsequent cytotoxic CD8+ T cell responses. J Immunol Baltim Md. 1950;2012(188):2146–2155.
  • Kissenpfennig A, Henri S, Dubois B, et al. Dynamics and function of langerhans cells in vivo. Immunity. 2005;22:643–654.
  • Ritter U, Meißner A, Scheidig C, et al. CD8α- and Langerin-negative dendritic cells, but not Langerhans cells, act as principal antigen-presenting cells in leishmaniasis. Eur J Immunol. 2004;34:1542–1550.
  • Lee C-H, Chen J-S, Chiu H-C, et al. Differential activation behavior of dermal dendritic cells underlies the strain-specific Th1 responses to single epicutaneous immunization. J Dermatol Sci. 2016;84:248–257.
  • Ebner S, Ehammer Z, Holzmann S, et al. Expression of C-type lectin receptors by subsets of dendritic cells in human skin. Int Immunol. 2004;16:877–887.
  • East L, Isacke CM. The mannose receptor family. Biochim Biophys Acta. 2002;1572:364–386.
  • Kaplan DH, Kissenpfennig A, Clausen BE. Insights into Langerhans cell function from Langerhans cell ablation models. Eur J Immunol. 2008;38:2369–2376.
  • Kautz-Neu K, Noordegraaf M, Dinges S, et al. Langerhans cells are negative regulators of the anti- Leishmania response. J Exp Med. 2011;208:885–891.
  • Cao T, Ueno H, Glaser C, et al. Both Langerhans cells and interstitial DC cross-present melanoma antigens and efficiently activate antigen-specific CTL. Eur J Immunol. 2007;37:2657–2667.
  • Stoitzner P, Tripp CH, Eberhart A, et al. Langerhans cells cross-present antigen derived from skin. Proc Natl Acad Sci USA. 2006;103:7783–7788.
  • Idoyaga J, Cheong C, Suda K, et al. Cutting edge: langerin/CD207 receptor on dendritic cells mediates efficient antigen presentation on MHC I and II products in vivo. J Immunol Baltim Md. 1950;2008(180):3647–3650.
  • Bobr A, Olvera-Gomez I, Igyarto BZ, et al. Acute ablation of Langerhans cells enhances skin immune responses. J Immunol Baltim Md 1950. 2010;185:4724.
  • Bedoui S, Whitney PG, Waithman J, et al. Cross-presentation of viral and self antigens by skin-derived CD103+ dendritic cells. Nat Immunol. 2009;10:488–495.
  • Flacher V, Tripp CH, Mairhofer DG, et al. Murine Langerin+ dermal dendritic cells prime CD8+ T cells while Langerhans cells induce cross-tolerance. EMBO Mol Med. 2014;6:1191–1204.
  • Dudziak D, Kamphorst AO, Heidkamp GF, et al. Differential antigen processing by dendritic cell subsets in vivo. Science. 2007;315:107–111.
  • Bos JD, Meinardi MM. The 500 Dalton rule for the skin penetration of chemical compounds and drugs. Exp Dermatol. 2000;9:165–169.
  • Kumar S, Sahdev P, Perumal O, et al. Identification of a novel skin penetration enhancement peptide by phage display peptide library screening. Mol Pharm. 2012;9:1320–1330.
  • Afonin S, Frey A, Bayerl S, et al. The cell-penetrating peptide TAT(48-60) induces a non-lamellar phase in DMPC membranes. Chemphyschem Eur J Chem Phys Phys Chem. 2006;7:2134–2142.
  • Gautam A, Nanda JS, Samuel JS, et al. Topical delivery of protein and peptide using novel cell penetrating peptide IMT-P8. Sci Rep. 2016;6:26278.
  • Benson HAE. Transfersomes for transdermal drug delivery. Expert Opin Drug Deliv. 2006;3:727–737.
  • Lee M-Y, Shin M-C, Yang VC. Transcutaneous antigen delivery system. BMB Rep. 2013;46:17–24.
  • Pham QD, Björklund S, Engblom J, et al. Chemical penetration enhancers in stratum corneum — relation between molecular effects and barrier function. J Controlled Release. 2016;232:175–187.
  • Liu X, Grice JE, Lademann J, et al. Hair follicles contribute significantly to penetration through human skin only at times soon after application as a solvent deposited solid in man. Br J Clin Pharmacol. 2011;72:768–774.
  • Blume-Peytavi U, Vogt A. Human hair follicle: reservoir function and selective targeting. Br J Dermatol. 2011;165(Suppl 2):13–17.
  • Kim Y-C, Prausnitz MR. Enabling skin vaccination using new delivery technologies. Drug Deliv Transl Res. 2011;1:7–12.
  • Frerichs DM, Ellingsworth LR, Frech SA, et al. Controlled, single-step, stratum corneum disruption as a pretreatment for immunization via a patch. Vaccine. 2008;26:2782–2787.
  • Sloat BR, Kiguchi K, Xiao G, et al. Transcutaneous DNA immunization following waxing-based hair depilation. J Control Release Off J Control Release Soc. 2012;157:94–102.
  • Engelke L, Winter G, Hook S, et al. Recent insights into cutaneous immunization: how to vaccinate via the skin. Vaccine. 2015;33:4663–4674.
  • Nasr M, Abdel-Hamid S, Alyoussef AA. A highlight on lipid based nanocarriers for transcutaneous immunization. Curr Pharm Biotechnol. 2015;16:371–379.
  • Esser ES, Romanyuk A, Vassilieva EV, et al. Tetanus vaccination with a dissolving microneedle patch confers protective immune responses in pregnancy. J Control Release Off J Control Release Soc. 2016;236:47–56.
  • van der Maaden K, Varypataki EM, Yu H, et al. Parameter optimization toward optimal microneedle-based dermal vaccination. Eur J Pharm Sci Off J Eur Fed Pharm Sci. 2014;64:18–25.
  • An M, Liu H. Dissolving microneedle arrays for transdermal delivery of amphiphilic vaccines. Small weinh. Bergstr Ger. 2017;13(26). doi: 10.1002/smll.201700164.
  • Lee JW, Gadiraju P, Park J-H, et al. Microsecond thermal ablation of skin for transdermal drug delivery. J Control Release Off J Control Release Soc. 2011;154:58–68.
  • Witting M, Boreham A, Brodwolf R, et al. Interactions of hyaluronic acid with the skin and implications for the dermal delivery of biomacromolecules. Mol Pharm. 2015;12:1391–1401.
  • Glenn GM, Villar CP, Flyer DC, et al. Safety and immunogenicity of an enterotoxigenic Escherichia coli vaccine patch containing heat-labile toxin: use of skin pretreatment to disrupt the stratum corneum. Infect Immun. 2007;75:2163–2170.
  • Vogt A, Hadam S, Deckert I, et al. Hair follicle targeting, penetration enhancement and Langerhans cell activation make cyanoacrylate skin surface stripping a promising delivery technique for transcutaneous immunization with large molecules and particle-based vaccines. Exp Dermatol. 2015;24:73–75.
  • Ali AA, McCrudden CM, McCaffrey J, et al. DNA vaccination for cervical cancer; a novel technology platform of RALA mediated gene delivery via polymeric microneedles. Nanomed Nanotechnol Biol Med. 2017;13:921–932.
  • Tawde SA, Chablani L, Akalkotkar A, et al. Evaluation of microparticulate ovarian cancer vaccine via transdermal route of delivery. J Controlled Release. 2016;235:147–154.
  • Khong H, Overwijk WW. Adjuvants for peptide-based cancer vaccines. J Immunother Cancer. 2016;4:56.
  • Kohli AK, Alpar HO. Potential use of nanoparticles for transcutaneous vaccine delivery: effect of particle size and charge. Int J Pharm. 2004;275:13–17.
  • Larese Filon F, Mauro M, Adami G, et al. Nanoparticles skin absorption: new aspects for a safety profile evaluation. Regul Toxicol Pharmacol. 2015;72:310–322.
  • Ascenso A, Raposo S, Batista C, et al. Development, characterization, and skin delivery studies of related ultradeformable vesicles: transfersomes, ethosomes, and transethosomes. Int J Nanomedicine. 2015;10:5837–5851.
  • Patzelt A, Richter H, Knorr F, et al. Selective follicular targeting by modification of the particle sizes. J Controlled Release. 2011;150:45–48.
  • Xiang SD, Scholzen A, Minigo G, et al. Pathogen recognition and development of particulate vaccines: does size matter? Methods San Diego Calif. 2006;40:1–9.
  • Foged C, Brodin B, Frokjaer S, et al. Particle size and surface charge affect particle uptake by human dendritic cells in an in vitro model. Int J Pharm. 2005;298:315–322.
  • Vogt A, Combadiere B, Hadam S, et al. 40 nm, but not 750 or 1,500 nm, nanoparticles enter epidermal CD1a+ cells after transcutaneous application on human skin. J Invest Dermatol. 2006;126:1316–1322.
  • Jeanbart L, Ballester M, De Titta A, et al. Enhancing efficacy of anticancer vaccines by targeted delivery to tumor-draining lymph nodes. Cancer Immunol Res. 2014;2:436–447.
  • Fan Y, Moon JJ. Nanoparticle drug delivery systems designed to improve cancer vaccines and immunotherapy. Vaccines. 2015;3:662–685.
  • Reddy ST, Rehor A, Schmoekel HG, et al. In vivo targeting of dendritic cells in lymph nodes with poly(propylene sulfide) nanoparticles. J Controlled Release. 2006;112:26–34.
  • Reddy ST, van der Vlies AJ, Simeoni E, et al. Exploiting lymphatic transport and complement activation in nanoparticle vaccines. Nat Biotechnol. 2007;25:1159–1164.
  • Fifis T, Gamvrellis A, Crimeen-Irwin B, et al. Size-dependent immunogenicity: therapeutic and protective properties of nano-vaccines against tumors. J Immunol Baltim Md. 1950;2004(173):3148–3154.
  • Mottram PL, Leong D, Crimeen-Irwin B, et al. Type 1 and 2 immunity following vaccination is influenced by nanoparticle size: formulation of a model vaccine for respiratory syncytial virus. Mol Pharm. 2007;4:73–84.
  • Dragicevic-Curic N, Gräfe S, Gitter B, et al. Surface charged temoporfin-loaded flexible vesicles: in vitro skin penetration studies and stability. Int J Pharm. 2010;384:100–108.
  • Wischke C, Borchert -H-H, Zimmermann J, et al. Stable cationic microparticles for enhanced model antigen delivery to dendritic cells. J Control Release Off J Control Release Soc. 2006;114:359–368.
  • Nakanishi T, Kunisawa J, Hayashi A, et al. Positively charged liposome functions as an efficient immunoadjuvant in inducing cell-mediated immune response to soluble proteins. J Control Release Off J Control Release Soc. 1999;61:233–240.
  • Cui Z, Mumper RJ. Chitosan-based nanoparticles for topical genetic immunization. J Control Release Off J Control Release Soc. 2001;75:409–419.
  • Patzelt A, Mak WC, Jung S, et al. Do nanoparticles have a future in dermal drug delivery? J Controlled Release. 2017;246:174–182.
  • Ansaldi F, Orsi A, de Florentiis D, et al. Head-to-head comparison of an intradermal and a virosome influenza vaccine in patients over the age of 60: evaluation of immunogenicity, cross-protection, safety and tolerability. Hum Vaccines Immunother. 2013;9:591–598.
  • Levin Y, Kochba E, Shukarev G, et al. A phase 1, open-label, randomized study to compare the immunogenicity and safety of different administration routes and doses of virosomal influenza vaccine in elderly. Vaccine. 2016;34:5262–5272.
  • Ambrosch F, Wiedermann G, Jonas S, et al. Immunogenicity and protectivity of a new liposomal hepatitis A vaccine. Vaccine. 1997;15:1209–1213.
  • Jain S, Patel N, Shah MK, et al. Recent advances in lipid-based vesicles and particulate carriers for topical and transdermal application. J Pharm Sci. 2017;106:423–445.
  • Fan Y, Zhang Q. Development of liposomal formulations: from concept to clinical investigations. Asian J Pharm Sci. 2013;8:81–87.
  • Schwendener RA. Liposomes as vaccine delivery systems: a review of the recent advances. Ther Adv Vaccines. 2014;2:159.
  • Mezei M, Gulasekharam V. Liposomes–a selective drug delivery system for the topical route of administration. Lotion dosage form. Life Sci. 1980;26:1473–1477.
  • Elsayed MMA, Abdallah OY, Naggar VF, et al. Lipid vesicles for skin delivery of drugs: reviewing three decades of research. Int J Pharm. 2007;332:1–16.
  • Cevc G, Blume G. Lipid vesicles penetrate into intact skin owing to the transdermal osmotic gradients and hydration force. Biochim Biophys Acta. 1992;1104:226–232.
  • Matsuo K, Hirobe S, Okada N, et al. Frontiers of transcutaneous vaccination systems: novel technologies and devices for vaccine delivery. Vaccine. 2013;31:2403–2415.
  • Wang J, Hu J, Li F, et al. Strong cellular and humoral immune responses induced by transcutaneous immunization with HBsAg DNA–cationic deformable liposome complex. Exp Dermatol. 2007;16:724–729.
  • Gupta V, Dhote V, Paul BN, et al. Development of novel topical drug delivery system containing cisplatin and imiquimod for dual therapy in cutaneous epithelial malignancy. J Liposome Res. 2014;24:150–162.
  • Tyagi RK, Garg NK, Dalai SK, et al. Transdermal immunization of P. falciparum surface antigen (MSP-119) via elastic liposomes confers robust immunogenicity. Hum Vaccines Immunother. 2016;12:990–992.
  • Rattanapak T, Young K, Rades T, et al. Comparative study of liposomes, transfersomes, ethosomes and cubosomes for transcutaneous immunisation: characterisation and in vitro skin penetration. J Pharm Pharmacol. 2012;64:1560–1569.
  • Song CK, Balakrishnan P, Shim C-K, et al. A novel vesicular carrier, transethosome, for enhanced skin delivery of voriconazole: characterization and in vitro/in vivo evaluation. Colloids Surf B Biointerfaces. 2012;92:299–304.
  • Espuelas S, Thumann C, Heurtault B, et al. Influence of ligand valency on the targeting of immature human dendritic cells by mannosylated liposomes. Bioconjug Chem. 2008;19:2385–2393.
  • Thomann J-S, Heurtault B, Weidner S, et al. Antitumor activity of liposomal ErbB2/HER2 epitope peptide-based vaccine constructs incorporating TLR agonists and mannose receptor targeting. Biomaterials. 2011;32:4574–4583.
  • Press Announcements - FDA Approves a Cellular Immunotherapy for Men with Advanced Prostate Cancer [Internet]. [cited 2017 Jan 22]. Available from: http://www.fda.gov/NewsEvents/Newsroom/PressAnnouncements/ucm210174.htm.
  • Press Announcements - FDA approves first-of-its-kind product for the treatment of melanoma [Internet]. [cited 2017 Jan 22]. Available from: http://www.fda.gov/NewsEvents/Newsroom/PressAnnouncements/ucm469571.htm.
  • Karkada M, Berinstein NL, Mansour M. Therapeutic vaccines and cancer: focus on DPX-0907. Biol Targets Ther. 2014;8:27.
  • Berinstein NL, Karkada M, Morse MA, et al. First-in-man application of a novel therapeutic cancer vaccine formulation with the capacity to induce multi-functional T cell responses in ovarian, breast and prostate cancer patients. J Transl Med. 2012;10:156.
  • Wurz GT, Kao C-J, Wolf M, et al. Tecemotide: an antigen-specific cancer immunotherapy. Hum Vaccines Immunother. 2014;10:3383–3393.
  • Davis ID, Chen W, Jackson H, et al. Recombinant NY-ESO-1 protein with ISCOMATRIX adjuvant induces broad integrated antibody and CD4+ and CD8+ T cell responses in humans. Proc Natl Acad Sci USA. 2004;101:10697.
  • Mitchell P, Thatcher N, Socinski MA, et al. Tecemotide in unresectable stage III non-small-cell lung cancer in the phase III START study: updated overall survival and biomarker analyses. Ann Oncol. 2015;26:1134–1142.
  • Nicholaou T, Chen W, Davis ID, et al. Immunoediting and persistence of antigen-specific immunity in patients who have previously been vaccinated with NY-ESO-1 protein formulated in ISCOMATRIXTM. Cancer Immunol Immunother. 2011;60:1625.
  • Nicholaou T, Ebert LM, Davis ID, et al. Regulatory T-cell–mediated attenuation of T-cell responses to the NY-ESO-1 ISCOMATRIX vaccine in patients with advanced malignant melanoma. Clin Cancer Res. 2009;15:2166–2173.
  • Chen J-L, Dawoodji A, Tarlton A, et al. NY-ESO-1 specific antibody and cellular responses in melanoma patients primed with NY-ESO-1 protein in ISCOMATRIX and boosted with recombinant NY-ESO-1 fowlpox virus. Int J Cancer. 2015;136:E590–E601.
  • Nemunaitis J, Jahan T, Ross H, et al. Phase 1/2 trial of autologous tumor mixed with an allogeneic GVAX vaccine in advanced-stage non-small-cell lung cancer. Cancer Gene Ther. 2006;13:555–562.
  • Sabado RL, Pavlick A, Gnjatic S, et al. Resiquimod as an immunologic adjuvant for NY-ESO-1 protein vaccination in patients with high-risk melanoma. Cancer Immunol Res. 2015;3:278–287.
  • Adams S, O’Neill DW, Nonaka D, et al. Immunization of malignant melanoma patients with full-length NY-ESO-1 protein using TLR7 agonist imiquimod as vaccine adjuvant. J Immunol Baltim Md. 1950;2008(181):776–784.
  • Burkhardt UE, Hainz U, Stevenson K, et al. Autologous CLL cell vaccination early after transplant induces leukemia-specific T cells. J Clin Invest. 2013;123:3756–3765.
  • Dhodapkar MV, Sznol M, Zhao B, et al. Induction of antigen-specific immunity with a vaccine targeting NY-ESO-1 to the dendritic cell receptor DEC-205. Sci Transl Med. 2014;6:232ra51.
  • Fujiyama T, Oze I, Yagi H, et al. Induction of cytotoxic T cells as a novel independent survival factor in malignant melanoma with percutaneous peptide immunization. J Dermatol Sci. 2014;75:43–48.
  • Iwama T, Uchida T, Sawada Y, et al. Vaccination with liposome-coupled glypican-3-derived epitope peptide stimulates cytotoxic T lymphocytes and inhibits GPC3-expressing tumor growth in mice. Biochem Biophys Res Commun. 2016;469:138–143.
  • 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. 2014;355:54–60.
  • Kakhi Z, Frisch B, Heurtault B, et al. Liposomal constructs for antitumoral vaccination by the nasal route. Biochimie. 2016;130:14–22.
  • Kakhi Z, Frisch B, Bourel-Bonnet L, et al. Airway administration of a highly versatile peptide-based liposomal construct for local and distant antitumoral vaccination. Int J Pharm. 2015;496:1047–1056.
  • Karkada M, Weir GM, Quinton T, et al. A liposome-based platform, VacciMax®, and its modified water-free platform DepoVaxTM enhance efficacy of in vivo nucleic acid delivery. Vaccine. 2010;28:6176–6182.
  • Butts C, Socinski MA, Mitchell PL, et al. Tecemotide (L-BLP25) versus placebo after chemoradiotherapy for stage III non-small-cell lung cancer (START): a randomised, double-blind, phase 3 trial. Lancet Oncol. 2014;15:59–68.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.