1
|
Tambunlertchai S, Geary SM, Salem AK. Topically Applied Resiquimod versus Imiquimod as a Potential Adjuvant in Melanoma Treatment. Pharmaceutics 2022; 14:pharmaceutics14102076. [PMID: 36297510 PMCID: PMC9611754 DOI: 10.3390/pharmaceutics14102076] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 09/17/2022] [Accepted: 09/18/2022] [Indexed: 01/28/2023] Open
Abstract
Melanoma is the most lethal form of skin cancer and surgery remains the preferred and most effective treatment. Nevertheless, there are cases where surgery is not a viable method and alternative treatments are therefore adopted. One such treatment that has been tested is topical 5% imiquimod (IMQ) cream, which, although showing promise as a treatment for melanoma, has been found to have undesirable off-target effects. Resiquimod (RSQ) is an immunomodulatory molecule that can activate immune responses by binding to Toll-like receptors (TLR) 7 and 8 and may be more effective than IMQ in the context of melanoma treatment. RSQ can cross the stratum corneum (SC) easily without requiring pretreatment of the skin. In a gel formulation, RSQ has been studied as a monotherapy and adjuvant for melanoma treatment in pre-clinical studies and as an adjuvant in clinical settings. Although side effects of RSQ in gel formulation were also reported, they were never severe enough for the treatment to be suspended. In this review, we discuss the potential use of RSQ as an adjuvant for melanoma treatment.
Collapse
|
2
|
Lai JCY, Cheng WK, Hopkins PD, Komba M, Carlow DA, Dutz JP. Topical Adjuvant Application during Subcutaneous Vaccination Promotes Resident Memory T Cell Generation. THE JOURNAL OF IMMUNOLOGY 2019; 203:2443-2450. [PMID: 31578270 DOI: 10.4049/jimmunol.1900199] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 08/26/2019] [Indexed: 12/18/2022]
Abstract
Skin tissue resident memory T cells (TRM) provide superior protection to a second infection. In this study, we evaluated the use of topical CpG oligodeoxynucleotide (ODN) as adjuvant to generate skin TRM in mice. Topical or s.c. CpG ODN adjuvant administration at the time of a s.c. Ag injection led to an accumulation of CD103- CD8 T cells in the epidermis. However, only mice with CpG ODN administered topically had significant numbers of CD103+ Ag-specific CD8 T cells persisting in the local epidermal skin, enhanced circulating memory cells in the blood, and showed protection from intradermal challenge with melanoma cells. Generation of Ag-specific CD8 T cells was dependent on TLR9 expression on hematopoietic cells and partially dependent on receptor expression on stromal cells. Topical challenge of immunized mice at a distal site led to significant expansion of Ag-specific T cells in the blood and accumulation in the challenged skin. We demonstrate that local and systemic T cell memory can be generated with topical CpG ODN at the time of s.c. immunization, suggesting a new method of enhancing current vaccine formulations to generate tissue TRM.
Collapse
Affiliation(s)
- Jacqueline C Y Lai
- Department of Dermatology and Skin Science, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia V5Z 4E8, Canada; and.,BC Children's Hospital Research Institute, Vancouver, British Columbia V5Z 4H4, Canada
| | - Wing Ki Cheng
- Department of Dermatology and Skin Science, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia V5Z 4E8, Canada; and.,BC Children's Hospital Research Institute, Vancouver, British Columbia V5Z 4H4, Canada
| | - Patrick D Hopkins
- Department of Dermatology and Skin Science, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia V5Z 4E8, Canada; and.,BC Children's Hospital Research Institute, Vancouver, British Columbia V5Z 4H4, Canada
| | - Mitsuhiro Komba
- BC Children's Hospital Research Institute, Vancouver, British Columbia V5Z 4H4, Canada
| | - Douglas A Carlow
- Department of Dermatology and Skin Science, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia V5Z 4E8, Canada; and.,BC Children's Hospital Research Institute, Vancouver, British Columbia V5Z 4H4, Canada
| | - Jan P Dutz
- Department of Dermatology and Skin Science, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia V5Z 4E8, Canada; and .,BC Children's Hospital Research Institute, Vancouver, British Columbia V5Z 4H4, Canada
| |
Collapse
|
3
|
Mie M, Takahashi T. Current condition and issues of animal evaluation models for cancer immunotherapy. Nihon Yakurigaku Zasshi 2016; 148:144-8. [PMID: 27581962 DOI: 10.1254/fpj.148.144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
4
|
Kitaoka M, Wakabayashi R, Kamiya N, Goto M. Solid-in-oil nanodispersions for transdermal drug delivery systems. Biotechnol J 2016; 11:1375-1385. [PMID: 27529824 PMCID: PMC5132072 DOI: 10.1002/biot.201600081] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Revised: 07/10/2016] [Accepted: 07/11/2016] [Indexed: 12/15/2022]
Abstract
Transdermal administration of drugs has advantages over conventional oral administration or administration using injection equipment. The route of administration reduces the opportunity for drug evacuation before systemic circulation, and enables long-lasting drug administration at a modest body concentration. In addition, the skin is an attractive route for vaccination, because there are many immune cells in the skin. Recently, solid-in-oil nanodisperison (S/O) technique has demonstrated to deliver cosmetic and pharmaceutical bioactives efficiently through the skin. S/O nanodispersions are nanosized drug carriers designed to overcome the skin barrier. This review discusses the rationale for preparation of efficient and stable S/O nanodispersions, as well as application examples in cosmetic and pharmaceutical materials including vaccines. Drug administration using a patch is user-friendly, and may improve patient compliance. The technique is a potent transcutaneous immunization method without needles.
Collapse
Affiliation(s)
- Momoko Kitaoka
- Graduate School of Engineering, Kyushu University, Fukuoka, Japan
| | - Rie Wakabayashi
- Graduate School of Engineering, Kyushu University, Fukuoka, Japan.,Center for Transdermal Drug Delivery, Kyushu University, Fukuoka, Japan
| | - Noriho Kamiya
- Graduate School of Engineering, Kyushu University, Fukuoka, Japan.,Center for Transdermal Drug Delivery, Kyushu University, Fukuoka, Japan.,Center for Future Chemistry, Kyushu University, Fukuoka, Japan
| | - Masahiro Goto
- Graduate School of Engineering, Kyushu University, Fukuoka, Japan.,Center for Transdermal Drug Delivery, Kyushu University, Fukuoka, Japan.,Center for Future Chemistry, Kyushu University, Fukuoka, Japan
| |
Collapse
|
5
|
Cheng WK, Plumb AW, Lai JCY, Abraham N, Dutz JP. Topical CpG Oligodeoxynucleotide Adjuvant Enhances the Adaptive Immune Response against Influenza A Infections. Front Immunol 2016; 7:284. [PMID: 27524984 PMCID: PMC4965457 DOI: 10.3389/fimmu.2016.00284] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Accepted: 07/13/2016] [Indexed: 11/26/2022] Open
Abstract
Current influenza vaccines generate humoral immunity, targeting highly variable epitopes and thus fail to achieve long-term protection. T cells recognize and respond to several highly conserved epitopes across influenza serotypes. A strategy of raising strong cytotoxic T cell memory responses to epitopes conserved across serotypes would provide cross serotype protection, eliminating the need for annual vaccination. We explored the adjuvant potential of epicutaneous (ec) and subcutaneous (sc) delivery of CpG oligodeoxynucleotide in conjunction with sc protein immunization to improve protection against influenza A virus (IAV) infections using a mouse model. We found enhanced long-term protection with epicutaneous CpG ODN (ecCpG) compared to subcutaneous CpG ODN (scCpG) as demonstrated by reduced viral titers in the lungs. This correlated with increased antigen-specific CD8 T cells in the airways and the lungs. The memory T cell response after immunization with ecCpG adjuvant was comparable to memory response by priming with IAV infection in the lungs. In addition, ecCpG was more efficient than scCpG in inducing the generation of IFN-γ producing CD4 T cells. The adjuvant effect of ecCpG was accompanied with its ability to modulate tissue-homing molecules on T cells that may direct them to the site of infection. Together, this work provides evidence for using ecCpG to induce strong antibody and memory T cell responses to confer protection against IAV infection.
Collapse
Affiliation(s)
- Wing Ki Cheng
- Department of Dermatology and Skin Science, Faculty of Medicine, Child and Family Research Institute, The University of British Columbia , Vancouver, BC , Canada
| | - Adam William Plumb
- Department of Microbiology and Immunology, Faculty of Science, Life Sciences Institute, The University of British Columbia , Vancouver, BC , Canada
| | - Jacqueline Cheuk-Yan Lai
- Department of Dermatology and Skin Science, Faculty of Medicine, Child and Family Research Institute, The University of British Columbia , Vancouver, BC , Canada
| | - Ninan Abraham
- Department of Microbiology and Immunology, Faculty of Science, Life Sciences Institute, The University of British Columbia, Vancouver, BC, Canada; Department of Zoology, Faculty of Science, Life Sciences Institute, The University of British Columbia, Vancouver, BC, Canada
| | - Jan Peter Dutz
- Department of Dermatology and Skin Science, Faculty of Medicine, Child and Family Research Institute, The University of British Columbia , Vancouver, BC , Canada
| |
Collapse
|
6
|
Hartnett JN, Boisen ML, Oottamasathien D, Jones AB, Millett MM, Nelson DS, Muncy IJ, Goba A, Momoh M, Fullah M, Mire CE, Geisbert JB, Geisbert TW, Holton DL, Rouelle JA, Kannadka CB, Reyna AA, Moses LM, Khan SH, Gevao SM, Grant DS, Robinson JE, Happi C, Pitts KR, Garry RF, Branco LM. Current and emerging strategies for the diagnosis, prevention and treatment of Lassa fever. Future Virol 2015. [DOI: 10.2217/fvl.15.41] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
ABSTRACT Lassa fever (LF) is a potentially fatal disease that affects an estimated 300,000–500,000 people in endemic areas of west Africa each year. Though past studies have identified fatality rates of 5–20% in patients suspected to have contracted Lassa virus (LASV), new studies using more precise clinical diagnoses and modern diagnostic assays show fatalities rates above 60% in acutely ill patients from endemic regions. Currently, there are no approved vaccines or therapeutics, and only one Comformité Européenne (CE) marked rapid immunodiagnostic for acute LASV infection. Therefore, preventing LASV transmission is the primary goal in endemic regions. Development of rapid immunodiagnostics and research into the efficacy of current treatment options continues toward saving lives in west Africa as well as creating a line of defense against the nefarious use of LASV in bioterrorism settings.
Collapse
Affiliation(s)
- Jessica N Hartnett
- Department of Microbiology, Tulane University School of Medicine, 1430 Tulane Avenue, SL-38, New Orleans, LA 70112, USA
| | - Matthew L Boisen
- Department of Microbiology, Tulane University School of Medicine, 1430 Tulane Avenue, SL-38, New Orleans, LA 70112, USA
- Corgenix Medical Corporation, Broomfield, CO 80020, USA
| | | | | | | | | | - Ivana J Muncy
- Corgenix Medical Corporation, Broomfield, CO 80020, USA
| | | | - Mambu Momoh
- Kenema Government Hospital, Kenema, Sierra Leone
- Eastern Polytechnic College, Kenema, Sierra Leone
| | | | - Chad E Mire
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Joan B Geisbert
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Thomas W Geisbert
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Debra L Holton
- Department of Microbiology, Tulane University School of Medicine, 1430 Tulane Avenue, SL-38, New Orleans, LA 70112, USA
| | - Julie A Rouelle
- Department of Microbiology, Tulane University School of Medicine, 1430 Tulane Avenue, SL-38, New Orleans, LA 70112, USA
| | - Chandrika B Kannadka
- Department of Microbiology, Tulane University School of Medicine, 1430 Tulane Avenue, SL-38, New Orleans, LA 70112, USA
| | - Ashley A Reyna
- Department of Microbiology, Tulane University School of Medicine, 1430 Tulane Avenue, SL-38, New Orleans, LA 70112, USA
| | - Lina M Moses
- Department of Microbiology, Tulane University School of Medicine, 1430 Tulane Avenue, SL-38, New Orleans, LA 70112, USA
| | | | - Sahr M Gevao
- Ministry of Health and Sanitation, Freetown, Sierra Leone
- University of Sierra Leone, Freetown, Sierra Leone
| | - Donald S Grant
- Kenema Government Hospital, Kenema, Sierra Leone
- Ministry of Health and Sanitation, Freetown, Sierra Leone
| | - James E Robinson
- Department of Pediatric Infectious Diseases, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | | | - Kelly R Pitts
- Corgenix Medical Corporation, Broomfield, CO 80020, USA
| | - Robert F Garry
- Department of Microbiology, Tulane University School of Medicine, 1430 Tulane Avenue, SL-38, New Orleans, LA 70112, USA
- Zalgen Labs, LLC, Germantown, MD 20876, USA
| | | | | |
Collapse
|
7
|
Topical CpG adjuvantation of a protein-based vaccine induces protective immunity to Listeria monocytogenes. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2014; 21:329-39. [PMID: 24391136 DOI: 10.1128/cvi.00734-13] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Robust CD8(+) T cell responses are essential for immune protection against intracellular pathogens. Using parenteral administration of ovalbumin (OVA) protein as a model antigen, the effect of the Toll-like receptor 9 (TLR9) agonist, CpG oligodeoxynucleotide (ODN) 1826, as an adjuvant delivered either topically, subcutaneously, or intramuscularly on antigen-specific CD8(+) T cell responses in a mouse model was evaluated. Topical CpG adjuvant increased the frequency of OVA-specific CD8(+) T cells in the peripheral blood and in the spleen. The more effective strategy to administer topical CpG adjuvant to enhance CD8(+) T cell responses was single-dose administration at the time of antigen injection with a prime-boost regimen. Topical CpG adjuvant conferred both rapid and long-lasting protection against systemic challenge with recombinant Listeria monocytogenes expressing the cytotoxic T lymphocyte (CTL) epitope of OVA(257-264) (strain Lm-OVA) in a TLR9-dependent manner. Topical CpG adjuvant induced a higher proportion of CD8(+) effector memory T cells than parenteral administration of the adjuvant. Although traditional vaccination strategies involve coformulation of antigen and adjuvant, split administration using topical adjuvant is effective and has advantages of safety and flexibility. Split administration of topical CpG ODN 1826 with parenteral protein antigen is superior to other administration strategies in enhancing both acute and memory protective CD8(+) T cell immune responses to subcutaneous protein vaccines. This vaccination strategy induces rapid and persistent protective immune responses against the intracellular organism L. monocytogenes.
Collapse
|
8
|
|
9
|
Karande P, Mitragotri S. Transcutaneous immunization: an overview of advantages, disease targets, vaccines, and delivery technologies. Annu Rev Chem Biomol Eng 2012; 1:175-201. [PMID: 22432578 DOI: 10.1146/annurev-chembioeng-073009-100948] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Skin is an immunologically active tissue composed of specialized cells and agents that capture and process antigens to confer immune protection. Transcutaneous immunization takes advantage of the skin immune network by inducing a protective immune response against topically applied antigens. This mode of vaccination presents a novel and attractive approach for needle-free immunization that is safe, noninvasive, and overcomes many of the limitations associated with needle-based administrations. In this review we will discuss the developments in the field of transcutaneous immunization in the past decade with special emphasis on disease targets and vaccine delivery technologies. We will also briefly discuss the challenges that need to be overcome to translate early laboratory successes in transcutaneous immunization into the development of effective clinical prophylactics.
Collapse
Affiliation(s)
- Pankaj Karande
- Department of Chemical and Biological Engineering, Rensselaer Polytechnic Institute, Troy, NY 12180, USA.
| | | |
Collapse
|
10
|
Poly D,L-lactide-co-glycolic acid-liposome encapsulated ODN on innate immunity in Epinephelus bruneus against Vibrio alginolyticus. Vet Immunol Immunopathol 2012; 147:77-85. [PMID: 22551979 DOI: 10.1016/j.vetimm.2012.04.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2012] [Revised: 03/26/2012] [Accepted: 04/04/2012] [Indexed: 11/23/2022]
Abstract
The efficacy of poly D,L-lactide-co-glycolic acid (PLGA)-liposome (L) encapsulated oligodeoxynucleotides with unmethylated deoxycytidyl-deoxyguanosine motifs (CpG-ODNs) on innate and adaptive immune response and disease resistance in kelp grouper (Epinephelus bruneus) against Vibrio alginolyticus at weeks 1, 2, and 4 is reported. The superoxide dismutase (SOD), respiratory burst, and lysozyme activities significantly increased in E. bruneus when immunized with ODN, PLGA+ODN, L+ODN, and PLGA+L+ODN on weeks 2 and 4. The serum complement activity was significantly enhanced with L+ODN and PLGA+L+ODN on week 1 while it increased with PLGA+ODN, L+ODN, and PLGA+L+ODN on weeks 2 and 4. The antibody titre consistently was increased with PLGA or L encapsulated with ODN (PLGA+ODN, L+ODN, and PLGA+L+ODN) from weeks 1 to 4. The cumulative mortality was 20% each in PLGA+ODN administered groups and 15% each in ODN, L+ODN, and PLGA+L+ODN groups during a period of 30 days. The present study suggests that PLGA-liposome encapsulated ODN has the potential to modulate the immune system and can serve as a useful tool for further design of immunoprophylatic nano drug formulations against bacterial diseases.
Collapse
|
11
|
Cholera toxin activates nonconventional adjuvant pathways that induce protective CD8 T-cell responses after epicutaneous vaccination. Proc Natl Acad Sci U S A 2012; 109:2072-7. [PMID: 22308317 DOI: 10.1073/pnas.1105771109] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The ability to induce humoral and cellular immunity via antigen delivery through the unbroken skin (epicutaneous immunization, EPI) has immediate relevance for vaccine development. However, it is unclear which adjuvants induce protective memory CD8 T-cell responses by this route, and the molecular and cellular requirements for priming through intact skin are not defined. We report that cholera toxin (CT) is superior to other adjuvants in its ability to prime memory CD8 T cells that control bacterial and viral challenges. Epicutaneous immunization with CT does not require engagement of classic toll-like receptor (TLR) and inflammasome pathways and, surprisingly, is independent of skin langerin-expressing cells (including Langerhans cells). However, CT adjuvanticity required type-I IFN sensitivity, participation of a Batf3-dependent dendritic cell (DC) population and engagement of CT with suitable gangliosides. Chemoenzymatic generation of CT-antigen fusion proteins led to efficient priming of the CD8 T-cell responses, paving the way for development of this immunization strategy as a therapeutic option.
Collapse
|
12
|
Kigasawa K, Kajimoto K, Nakamura T, Hama S, Kanamura K, Harashima H, Kogure K. Noninvasive and efficient transdermal delivery of CpG-oligodeoxynucleotide for cancer immunotherapy. J Control Release 2011; 150:256-65. [PMID: 21256903 DOI: 10.1016/j.jconrel.2011.01.018] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2010] [Revised: 11/30/2010] [Accepted: 01/17/2011] [Indexed: 12/13/2022]
Abstract
Oligodeoxynucleotides containing unmethylated cytosine-phosphate-guanosine motifs (CpG-ODN) possess immunostimulatory effects and potential antitumor activity. Since the skin is an easily available site of administration of CpG-ODN due to its accessibility and the presence of abundant antigen presenting cells, it is expected that the application of CpG-ODN to the skin would induce systemic immune response and antitumor activity. However, it is difficult to deliver hydrophilic macromolecules including CpG-ODN through the skin. We have previously demonstrated that small interfering RNA (siRNA) was efficiently delivered into rat epidermis by iontophoresis. In this report, we investigate the effect of transdermal iontophoretic delivery of CpG-ODN on the induction of immune responses and antitumor activity against B16F1 melanoma in mice. Iontophoresis promoted CpG-ODN delivery into the epidermis and dermis. Furthermore, iontophoretic delivery of CpG-ODN to the skin induced the expression of proinflammatory and Th1-type cytokines in the skin and draining lymph node. Finally, transdermal iontophoretic delivery of CpG-ODN led to antitumor activity against B16F1 melanoma. Interestingly, the CpG-ODN administration site is not restricted to the tumor area. In conclusion, CpG-ODN delivered transdermally induced potent antitumor activity, and our system is expected to serve as a simple and noninvasive approach for cancer immunotherapy.
Collapse
Affiliation(s)
- Kaoru Kigasawa
- Graduate School of Life Science, Hokkaido University, Kita 12 Nishi 6, Kita-ku, Sapporo-City, Hokkaido 060-0812 Japan
| | | | | | | | | | | | | |
Collapse
|
13
|
Abstract
The skin has been investigated as a site for vaccine delivery only since the late 1990s. However, much has been discovered about the cell populations that reside in the skin, their active role in immune responses, and the fate of trans- cutaneously applied antigens. Transcutaneous immunization (TCI) is a safe, effective means of inducing immune responses against a number of pathogens. One of the most notable benefits of TCI is the induction of immune responses in both systemic and mucosal compartments. This chapter focuses on the transport of antigen into and beyond intact skin, the cutaneous sentinel cell populations that play a role in TCI, and the types of mucosal immune responses that have been generated. A number of in vivo studies in murine models have provided information about the broad responses induced by TCI. Cellular and humoral responses and protection against challenge have been noted in the gastrointestinal, reproductive, and respiratory tracts. Clinical trials have demonstrated the benefits of this vaccine delivery route in humans. As with other routes of immunization, the type of vaccine formulation and choice of adjuvant may be critical for achieving appropriate responses and can be tailored to activate specific immune-responsive cells in the skin to increase the efficacy of TCI against mucosal pathogens.
Collapse
|
14
|
Su X, Kim BS, Kim SR, Hammond PT, Irvine DJ. Layer-by-layer-assembled multilayer films for transcutaneous drug and vaccine delivery. ACS NANO 2009; 3:3719-29. [PMID: 19824655 PMCID: PMC3057049 DOI: 10.1021/nn900928u] [Citation(s) in RCA: 111] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
We describe protein- and oligonucleotide-loaded layer-by-layer (LbL)-assembled multilayer films incorporating a hydrolytically degradable polymer for transcutaneous drug or vaccine delivery. Films were constructed based on electrostatic interactions between a cationic poly(beta-amino ester) (denoted Poly-1) with a model protein antigen, ovalbumin (ova), and/or immunostimulatory CpG (cytosine-phosphate diester-guanine-rich) DNA oligonucleotide adjuvant molecules. Linear growth of nanoscale Poly-1/ova bilayers was observed. Dried ova protein-loaded films rapidly deconstructed when rehydrated in saline solutions, releasing ova as nonaggregated/nondegraded protein, suggesting that the structure of biomolecules integrated into these multilayer films is preserved during release. Using confocal fluorescence microscopy and an in vivo murine ear skin model, we demonstrated delivery of ova from LbL films into barrier-disrupted skin, uptake of the protein by skin-resident antigen-presenting cells (Langerhans cells), and transport of the antigen to the skin-draining lymph nodes. Dual incorporation of ova and CpG oligonucleotides into the nanolayers of LbL films enabled dual release of the antigen and adjuvant with distinct kinetics for each component; ova was rapidly released, while CpG was released in a relatively sustained manner. Applied as skin patches, these films delivered ova and CpG to Langerhans cells in the skin. To our knowledge, this is the first demonstration of LbL films applied for the delivery of biomolecules into skin. This approach provides a new route for storage of vaccines and other immunotherapeutics in a solid-state thin film for subsequent delivery into the immunologically rich milieu of the skin.
Collapse
Affiliation(s)
- Xingfang Su
- Department of Material Science and Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139
| | - Byeong-Su Kim
- Department of Chemical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139
| | - Sara R. Kim
- Department of Chemical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139
| | - Paula T. Hammond
- Department of Chemical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139
| | - Darrell J. Irvine
- Department of Material Science and Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139
- Department of Biological Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139
- Ragon Institute of MGH, MIT, and Harvard, Boston, MA 02114
| |
Collapse
|
15
|
Senti G, Graf N, Haug S, Rüedi N, von Moos S, Sonderegger T, Johansen P, Kündig TM. Epicutaneous allergen administration as a novel method of allergen-specific immunotherapy. J Allergy Clin Immunol 2009; 124:997-1002. [PMID: 19733905 DOI: 10.1016/j.jaci.2009.07.019] [Citation(s) in RCA: 142] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2008] [Revised: 06/24/2009] [Accepted: 07/10/2009] [Indexed: 01/30/2023]
Abstract
BACKGROUND Subcutaneous allergen-specific immunotherapy is an effective treatment of IgE-mediated allergies, but it requires repeated allergen injections with a risk of systemic allergic reactions. Transcutaneous immunotherapy may improve patient compliance and safety. OBJECTIVE To assess the safety and efficacy of epicutaneous allergen immunotherapy. METHODS This monocentric, placebo-controlled, double-blind trial was conducted from March 2006 to December 2007 at the University Hospital Zurich. Thirty-seven adult patients with positive skin prick and nasal provocation tests to grass pollen were randomized to receive patches containing either allergen (n = 21) or placebo (n = 16). Treatment took place before and during the pollen season 2006, and follow-up visits took place before (n = 26) and after the pollen season 2007 (n = 30). The primary outcome measures were nasal provocation tests. RESULTS Allergen-treated patients showed significantly decreased scores in nasal provocation tests in the first (P < .001) and second year (P = .003) after treatment. In contrast, placebo-treated patients had decreased scores in the first treatment year, 2006 (P = .03), but the effect diminished in the second year (P = .53). Although improvement of nasal provocation test scores was not significantly better in the verum versus placebo group, the overall treatment success was rated significantly higher by the allergen-treated group than by the placebo group (2006, P = .02; 2007, P = .005). No severe adverse events were observed. Occurrence of eczema after allergen patch applications proved stimulation of specific T-cell responses, but was noted as an adverse effect of the treatment. CONCLUSION Epicutaneous allergen immunotherapy is a promising strategy to treat allergies and merits further investigation.
Collapse
Affiliation(s)
- Gabriela Senti
- Unit for Experimental Immunotherapy, University Hospital of Zurich, Zurich, Switzerland
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Chang BA, Cross JL, Najar HM, Dutz JP. Topical resiquimod promotes priming of CTL to parenteral antigens. Vaccine 2009; 27:5791-9. [PMID: 19660592 DOI: 10.1016/j.vaccine.2009.07.062] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2008] [Revised: 07/14/2009] [Accepted: 07/19/2009] [Indexed: 01/12/2023]
Abstract
We explored the topical use of resiquimod (R-848), a Toll-like receptor (TLR) 7/8 agonist, in gel formulation, to enhance cross-priming to subcutaneously administered protein antigen in a murine model. Resiquimod application at the time of subcutaneous administration of ovalbumin generated robust antigen-specific CTL as detected by tetramers, IFN-gamma ELISPOT assays and standard cytotoxicity assays. Induced CTL were capable of mediating antigen-specific killing in vivo as measured by in vivo cytotoxicity assays and an ability to protect against B16-OVA tumor challenge. Multiple serial applications of topical resiquimod increased the frequency of antigen-specific CTL when compared to single application. This enhanced frequency was noted despite a marked inhibition of adjuvant mediated pro-inflammatory cytokine release following repeated administration. Topical resiquimod is a potent adjuvant for locally administered subcutaneous vaccines, inducing clinically relevant CTL responses following single application at the time of subcutaneous vaccination.
Collapse
Affiliation(s)
- Brent A Chang
- Child and Family Research Institute, Department of Dermatology & Skin Science, University of British Columbia, 835 West Tenth Ave, Vancouver, BC, Canada V5Z 4E8
| | | | | | | |
Collapse
|
17
|
Flacher V, Sparber F, Tripp CH, Romani N, Stoitzner P. Targeting of epidermal Langerhans cells with antigenic proteins: attempts to harness their properties for immunotherapy. Cancer Immunol Immunother 2009; 58:1137-47. [PMID: 18677477 PMCID: PMC11030799 DOI: 10.1007/s00262-008-0563-9] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2008] [Accepted: 07/12/2008] [Indexed: 12/16/2022]
Abstract
Langerhans cells, a subset of skin dendritic cells in the epidermis, survey peripheral tissue for invading pathogens. In recent functional studies it was proven that Langerhans cells can present exogenous antigen not merely on major histocompatibility complexes (MHC)-class II molecules to CD4+ T cells, but also on MHC-class I molecules to CD8+ T cells. Immune responses against topically applied antigen could be measured in skin-draining lymph nodes. Skin barrier disruption or co-application of adjuvants was required for maximal induction of T cell responses. Cytotoxic T cells induced by topically applied antigen inhibited tumor growth in vivo, thus underlining the potential of Langerhans cells for immunotherapy. Here we review recent work and report novel observations relating to the potential use of Langerhans cells for immunotherapy. We investigated the potential of epicutaneous immunization strategies in which resident skin dendritic cells are loaded with tumor antigen in situ. This contrasts with current clinical approaches, where dendritic cells generated from progenitors in blood are loaded with tumor antigen ex vivo before injection into cancer patients. In the current study, we applied either fluorescently labeled protein antigen or targeting antibodies against DEC-205/CD205 and langerin/CD207 topically onto barrier-disrupted skin and examined antigen capture and transport by Langerhans cells. Protein antigen could be detected in Langerhans cells in situ, and they were the main skin dendritic cell subset transporting antigen during emigration from skin explants. Potent in vivo proliferative responses of CD4+ and CD8+ T cells were measured after epicutaneous immunization with low amounts of protein antigen. Targeting antibodies were mainly transported by langerin+ migratory dendritic cells of which the majority represented migratory Langerhans cells and a smaller subset the new langerin+ dermal dendritic cell population located in the upper dermis. The preferential capture of topically applied antigen by Langerhans cells and their ability to induce potent CD4+ and CD8+ T cell responses emphasizes their potential for epicutaneous immunization strategies.
Collapse
Affiliation(s)
- Vincent Flacher
- Department of Dermatology and Venereology, Innsbruck Medical University, Anichstrasse 35, 6020 Innsbruck, Austria
| | - Florian Sparber
- Department of Dermatology and Venereology, Innsbruck Medical University, Anichstrasse 35, 6020 Innsbruck, Austria
| | - Christoph H. Tripp
- Department of Dermatology and Venereology, Innsbruck Medical University, Anichstrasse 35, 6020 Innsbruck, Austria
| | - Nikolaus Romani
- Department of Dermatology and Venereology, Innsbruck Medical University, Anichstrasse 35, 6020 Innsbruck, Austria
- Kompetenzzentrum Medizin Tirol (CEMIT), Innsbruck, Austria
| | - Patrizia Stoitzner
- Department of Dermatology and Venereology, Innsbruck Medical University, Anichstrasse 35, 6020 Innsbruck, Austria
| |
Collapse
|
18
|
Ghoreishi M, Bach P, Obst J, Komba M, Fleet JC, Dutz JP. Expansion of antigen-specific regulatory T cells with the topical vitamin d analog calcipotriol. THE JOURNAL OF IMMUNOLOGY 2009; 182:6071-8. [PMID: 19414758 DOI: 10.4049/jimmunol.0804064] [Citation(s) in RCA: 113] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
1,25-Dihydroxyvitamin D(3) is immunosuppressive both in vivo and in vitro. Topical vitamin D analogs such as calcipotriol alter keratinocyte function, but their effects on cutaneous immune responses are less well understood. We demonstrate that exposure of the skin to calcipotriol before transcutaneous immunization with OVA protein and CpG adjuvant prevents Ag-specific CD8(+) T cell priming coincident with Langerhans cell depletion in the skin. Immunization through calcipotriol-treated skin induces CD4(+)CD25(+) regulatory T cells (Treg) that prevent subsequent Ag-specific CD8(+) T cell proliferation and IFN-gamma production. Treg induced by calcipotriol are able to inhibit the induction and the elicitation of protein contact hypersensitivity. Topical calcipotriol treatment also induces RANKL (receptor activator of NF-kappaB ligand) expression by keratinocytes, a TNF family member involved in modulation of skin dendritic cells. UV light B induces Ag-specific tolerance when it is applied before transcutaneous immunization. We suggest that UV light B-induced tolerance is induced via a vitamin D receptor-dependent mechanism as vitamin D receptor (VDR) knockout mice fail to increase FoxP3(+) Treg in their peripheral draining lymph node following irradiation. Additionally, keratinocytes of VDR(-/-) mice fail to induce RANKL upon UV irradiation or calcipotriol treatment. The in vivo expansion of Ag-specific Treg with the topical application of the vitamin D analog calcipotriol followed by transcutaneous immunization is a simple method to augment functional Ag-specific CD4(+)CD25(+)Foxp3(+) Treg populations and mimics Ag-specific UV-induced tolerance.
Collapse
Affiliation(s)
- Mehran Ghoreishi
- Department of Dermatology & Skin Science and Child and Family Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | | | | | | | | | | |
Collapse
|
19
|
Skountzou I, Kang SM. Transcutaneous Immunization with Influenza Vaccines. Curr Top Microbiol Immunol 2009; 333:347-68. [DOI: 10.1007/978-3-540-92165-3_17] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
20
|
Abstract
Advances in the engineering of peptides, adjuvants and delivery systems have renewed the enthusiasm for peptide-based vaccination regimens in the setting of cancer, and there are a variety of clinical trials being conducted by pharmaceutical companies based on the use of peptides. The challenges to successful cancer immunotherapy are common to all immunotherapeutic strategies and not unique to peptide-based vaccination regimens. This review will describe the advances in the identification, design and delivery of peptides, the challenges to successful immunotherapy and will discuss potential options for the future.
Collapse
Affiliation(s)
- Shreya Kanodia
- Department of Molecular Microbiology and Immunology, Norris Comprehensive Cancer Center, NRT 7517, University of Southern California, 1450 Biggy Street, Los Angeles, CA 90033, USA.
| | | |
Collapse
|
21
|
Wang L, Bursch LS, Kissenpfennig A, Malissen B, Jameson SC, Hogquist KA. Langerin Expressing Cells Promote Skin Immune Responses under Defined Conditions. THE JOURNAL OF IMMUNOLOGY 2008; 180:4722-7. [DOI: 10.4049/jimmunol.180.7.4722] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
22
|
Abstract
This chapter provides an overview of the application of electroporation to areas other than gene delivery. These areas include the delivery of drugs and vaccines to tissues and tumors as well as into and through the skin. Achievements and limitations of electroporation in these areas are presented. Alternative physical methods for gene and drug delivery besides electroporation are described. The advantages and drawbacks of electroporation, compared with these methods, are also discussed.
Collapse
|
23
|
Glenn GM, Flyer DC, Ellingsworth LR, Frech SA, Frerichs DM, Seid RC, Yu J. Transcutaneous immunization with heat-labile enterotoxin: development of a needle-free vaccine patch. Expert Rev Vaccines 2007; 6:809-19. [PMID: 17931160 DOI: 10.1586/14760584.6.5.809] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The skin is an attractive target for vaccine delivery. Adjuvants and antigens delivered into the skin can result in potent immune responses and an unmatched safety profile. The heat-labile enterotoxin (LT) from Escherichia coli, which acts both as antigen and adjuvant, has been shown to be delivered to human skin efficiently when used in a patch, resulting in strong immune responses. Iomai scientists have capitalized on these observations to develop late-stage products based on LT. This has encouraged commercial-level product development of a delivery system that is efficient, user-friendly and designed to address important medical needs. Over the past 2 years, extensive clinical testing and optimization has allowed the patch to evolve to a late-stage product. As a strategy for approval of a revolutionary vaccine-delivery system, the singular focus on optimization of LT delivery has enabled technical progress to extend patch-vaccine product development beyond LT. The field efficacy of the LT-based travelers' diarrhea vaccine has validated this approach. The discussion of transcutaneous immunization is unique, in that any consideration of the adjuvant must also include delivery, and the significant advances in a commercial patch application system are described. In this review, we integrate these concepts, update the clinical data and look to the future.
Collapse
Affiliation(s)
- Gregory M Glenn
- Iomai Corporation, 20 Firstfield Road, Suite 250, Gaithersburg, MD 20878, USA.
| | | | | | | | | | | | | |
Collapse
|
24
|
Najar HM, Dutz JP. Topical TLR9 agonists induce more efficient cross-presentation of injected protein antigen than parenteral TLR9 agonists do. Eur J Immunol 2007; 37:2242-56. [PMID: 17634951 DOI: 10.1002/eji.200636212] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Topical application of adjuvant to the skin promotes the generation of immune responses to co-administered peptide or protein antigen. We demonstrate that topical administration of CpG adjuvant (a TLR9 agonist) induces the cross-presentation of, and antigen-specific CTL induction to, locally injected soluble protein antigen. C57BL/6 mice were immunized by subcutaneous or intramuscular injection with ovalbumin (OVA) protein as model antigen. Application of CpG to the local skin induced more efficient cross-presentation of the injected antigen than co-injected adjuvant. Robust antigen-specific CTL responses were generated, as determined by antigen-specific CTL enumeration using tetramers, IFN-gamma ELISPOT analysis and cytotoxicity assays. Long-term memory CTL responses were induced. Topical administration of adjuvant induced Langerhans cell migration, local type 1 IFN-dependent myxovirus-resistance protein A expression and bystander dendritic cell (DC) activation. Soluble antigen-bearing DC within the skin draining lymph nodes were mainly CD11chiCD11bhilangerinloDEC205lo. Topical administration did not result in the splenomegaly or systemic cytokine induction (including TNF-alpha, IL-12, IFN-gamma and MCP-1) noted with parenteral administration. Topical TLR9 family agonists may be used to modulate the immune response to soluble protein vaccines administered by standard percutaneous route. Topical adjuvant administration increases efficacy of CTL induction and reduces toxicity when compared to parenteral adjuvant administration.
Collapse
Affiliation(s)
- Hossain M Najar
- Department of Dermatology, and Child and Family Research Institute, University British Colombia, Vancouver, British Colombia, Canada
| | | |
Collapse
|
25
|
de Jong S, Chikh G, Sekirov L, Raney S, Semple S, Klimuk S, Yuan N, Hope M, Cullis P, Tam Y. Encapsulation in liposomal nanoparticles enhances the immunostimulatory, adjuvant and anti-tumor activity of subcutaneously administered CpG ODN. Cancer Immunol Immunother 2007; 56:1251-64. [PMID: 17242927 PMCID: PMC11030982 DOI: 10.1007/s00262-006-0276-x] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2006] [Accepted: 12/15/2006] [Indexed: 12/19/2022]
Abstract
Immunostimulatory oligodeoxynucleotides (ODN) containing cytosine-guanine (CpG) motifs are powerful stimulators of innate as well as adaptive immune responses, exerting their activity through triggering of the Toll-like receptor 9. We have previously shown that encapsulation in liposomal nanoparticles (LN) enhances the immunostimulatory activity of CpG ODN (LN-CpG ODN) (Mui et al. in J Pharmacol Exp Ther 298:1185, 2001). In this work we investigate the effect of encapsulation on the immunopotency of subcutaneously (s.c.) administered CpG ODN with regard to activation of innate immune cells as well as its ability to act as a vaccine adjuvant with tumor-associated antigens (TAAs) to induce antigen (Ag)-specific, adaptive responses and anti-tumor activity in murine models. It is shown that encapsulation specifically targets CpG ODN for uptake by immune cells. This may provide the basis, at least in part, for the significantly enhanced immunostimulatory activity of LN-CpG ODN, inducing potent innate (as judged by immune cell activation and plasma cytokine/chemokine levels) and adaptive, Ag-specific (as judged by MHC tetramer positive T lymphocytes, IFN-gamma secretion and cytotoxicity) immune responses. Finally, in efficacy studies, it is shown that liposomal encapsulation enhances the ability of CpG ODN to adjuvanate adaptive immune responses against co-administered TAAs after s.c. immunization, inducing effective anti-tumor activity against both model and syngeneic tumor Ags in murine tumor models of thymoma and melanoma.
Collapse
MESH Headings
- Adjuvants, Immunologic/administration & dosage
- Adjuvants, Immunologic/pharmacokinetics
- Adjuvants, Immunologic/pharmacology
- Animals
- Antineoplastic Agents/administration & dosage
- Antineoplastic Agents/pharmacokinetics
- Antineoplastic Agents/pharmacology
- CD8-Positive T-Lymphocytes/drug effects
- CD8-Positive T-Lymphocytes/immunology
- Cancer Vaccines/therapeutic use
- Chemokine CCL2/blood
- Drug Compounding
- Drug Screening Assays, Antitumor
- Female
- Fluorescent Dyes/analysis
- Injections, Subcutaneous
- Interferon-gamma/blood
- Interleukin-10/blood
- Interleukin-6/blood
- Intramolecular Oxidoreductases/immunology
- Liposomes/administration & dosage
- Liposomes/pharmacokinetics
- Lung Neoplasms/secondary
- Lung Neoplasms/therapy
- Lymphocyte Activation/drug effects
- Melanoma, Experimental/immunology
- Melanoma, Experimental/secondary
- Melanoma, Experimental/therapy
- Mice
- Mice, Inbred C57BL
- Mice, Inbred ICR
- Nanoparticles/administration & dosage
- Oligodeoxyribonucleotides/administration & dosage
- Oligodeoxyribonucleotides/pharmacokinetics
- Oligodeoxyribonucleotides/pharmacology
- Ovalbumin/immunology
- Thymoma/immunology
- Thymoma/secondary
- Thymoma/therapy
Collapse
Affiliation(s)
- Susan de Jong
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC Canada
| | - Ghania Chikh
- Inex Pharmaceuticals Corporation, Burnaby, BC Canada
| | - Laura Sekirov
- Inex Pharmaceuticals Corporation, Burnaby, BC Canada
| | - Sam Raney
- Inex Pharmaceuticals Corporation, Burnaby, BC Canada
| | - Sean Semple
- Inex Pharmaceuticals Corporation, Burnaby, BC Canada
| | - Sandra Klimuk
- Inex Pharmaceuticals Corporation, Burnaby, BC Canada
| | - Ning Yuan
- Inex Pharmaceuticals Corporation, Burnaby, BC Canada
| | - Micheal Hope
- Inex Pharmaceuticals Corporation, Burnaby, BC Canada
| | - Pieter Cullis
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC Canada
| | - Ying Tam
- Inex Pharmaceuticals Corporation, Burnaby, BC Canada
| |
Collapse
|
26
|
Wilson KD, Raney SG, Sekirov L, Chikh G, deJong SD, Cullis PR, Tam YK. Effects of intravenous and subcutaneous administration on the pharmacokinetics, biodistribution, cellular uptake and immunostimulatory activity of CpG ODN encapsulated in liposomal nanoparticles. Int Immunopharmacol 2007; 7:1064-75. [PMID: 17570323 DOI: 10.1016/j.intimp.2007.04.002] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2007] [Revised: 04/02/2007] [Accepted: 04/04/2007] [Indexed: 11/23/2022]
Abstract
We have previously demonstrated that the immune response to an unmethylated cytidine-guanosine (CpG)-containing oligonucleotide (ODN) is greatly enhanced when encapsulated in a lipid nanoparticle (LN-CpG ODN). In this study, the pharmacokinetics, biodistribution and cellular uptake of LN-CpG ODN following intravenous (i.v.) and subcutaneous (s.c.) administration was characterized and correlated with immunostimulatory activity. It is shown that, despite dramatic differences in tissue distribution profiles and considerable differences in uptake by CD11c-positive, CD11b-positive, Mac-3-positive and CD45R/B220-positive cells following i.v. and s.c. administration, the resultant immune response is very similar with respect to levels of cellular activation (DX5, Mac-3, CD11b, CD45/B220, CD4, CD8 and CD11c) and cytolytic activity of immune cells [natural killer (NK) cells and monocytes/macrophages] in the spleen and blood compartments. Some differences in response kinetics and antibody-dependent cellular cytotoxicity (ADCC) activity were noted in the peripheral blood NK cell population. Analyses of particle biodistribution and cell types involved in uptake leads to the conclusion that the inherent ability of antigen-presenting cells (APCs) to sequester LN-CpG ODN results in efficient uptake of the particle, even when present at very low concentrations, leading to similar responses following i.v. and s.c. administration. These results contrast with the behavior of free CpG ODN, for which distinctly different immune responses are observed following i.v. or s.c. administration.
Collapse
Affiliation(s)
- Kaley D Wilson
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia, Canada
| | | | | | | | | | | | | |
Collapse
|
27
|
Pivarcsi A. Toll-like receptor 9-independent suppression of skin inflammation by oligonucleotides. J Invest Dermatol 2007; 127:746-8. [PMID: 17363956 DOI: 10.1038/sj.jid.5700679] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
It has been well established that cytidine-phosphate-guanosine (CpG) oligodeoxynucleotides (ODNs) activate innate and adaptive immune responses in keratinocytes by stimulating Toll-like receptor 9 (TLR9)-dependent signaling pathways. However, as Dorn et al. report, keratinocytes possess another, yet uncharacterized, TLR9-independent mechanism for the recognition of ODNs. Surprisingly, the activation of the pathway leads to suppressed chemokine production in vitro and decreased skin inflammation in vivo.
Collapse
Affiliation(s)
- Andor Pivarcsi
- Clinical Allergy Research Unit, Department of Medicine, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
28
|
Warger T, Schild H, Rechtsteiner G. Initiation of adaptive immune responses by transcutaneous immunization. Immunol Lett 2007; 109:13-20. [PMID: 17320194 DOI: 10.1016/j.imlet.2007.01.007] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2007] [Accepted: 01/20/2007] [Indexed: 10/23/2022]
Abstract
The development of new, effective, easy-to-use and lower-cost vaccination approaches for the combat against malignant and infectious diseases is a pre-eminent need: cancer is a leading cause of morbidity in the Western World; there are numerous pathogenic diseases for which we still have no protective or therapeutic cure; and the financial limitations of developing countries to fight these diseases. In this mini-review we focus on transcutaneous immunization (TCI), a relatively new route for antigen delivery. TCI protocols appear to be particularly promising by gaining access to skin resident APC, which are highly efficient for the initiation of humoral and/or cellular immune responses. Consisting of an adjuvant as a stimulus in combination with an antigen which defines the target, TCI offers a most attractive immunization strategy to mount highly specific full-blown adaptive immune responses. As a topically applicable cell-free adjuvant/antigen mixture, TCI might be suitable to improve patient compliance, as well as feasible economically for the use in Third World countries. In addition, this non-invasive procedure might increase the safety of vaccinations by eliminating the risk of infections related to the recycling and improper disposal of needles. The dissection of antigen and adjuvant is important because it allows "free" combinations in contrast to classical immunizations which are based on application of the pathogen of interest. The most relevant ways and means to find new, effective pathogenic target antigens are "reverse vaccinology" and the direct peptide-epitope identification from MHC molecules with mass-spectrometry. Due to these efficient approaches the variety of antigenic epitopes for potential protective/therapeutic use is perpetually expanding. The most studied adjuvants in TCI approaches are cholera toxin (CT) and its less toxic relative, the heat-labile enterotoxin (LT). Both CT and LT can serve as antigen as well. In contrast to these large proteins, which can only penetrate "pre-treated" skin barrier, the immune response modifier, TLR7 agonist R-837 (Imiquimod) is a small compound adjuvant that easily passages non-disrupted epidermis. It remains currently elusive which cells of the complex-structured "skin-associated lymphoid tissue" (SALT) respond to the adjuvant and which APC carries the antigen to the draining lymphnodes for subsequent initiation of adaptive immune responses.
Collapse
Affiliation(s)
- Tobias Warger
- Institute for Immunology, Johannes Gutenberg-University, Obere Zahlbacher Str. 67, D-55131 Mainz, Germany
| | | | | |
Collapse
|
29
|
Wang LF, Hsu CJ, Miaw SC, Chiu HC, Liu CY, Yu HS. Cross-priming with an epicutaneously introduced soluble protein antigen generates Tc1 cells. Eur J Immunol 2007; 36:2904-11. [PMID: 17048268 DOI: 10.1002/eji.200535770] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Epicutaneous sensitization with a protein antigen was demonstrated to induce a predominant type 2 CD4 T cell response with high IgE production in mice. On the other hand, its CD8 T cell responses have not been addressed probably partly because of the generally accepted concept that cross-priming of soluble protein is an inefficient process. Here, we used an established patch-applied murine model to demonstrate that cross-priming with an epicutaneously introduced soluble protein antigen, though inefficient, generated mainly Tc1 cells, but not Tc2 cells. In the presence of an irritant or hapten, the efficiency of this cross-priming process could be enhanced and more Tc1 cells were generated. CpG oligonucleotides also promote the generation of Tc1 cells. In contrast, lipopolysaccharide and poly (inosinic-cytidylic) acid [poly (I:C)] have no effect. Together, these results provide supportive evidence of the epicutaneous sensitization of human cutaneous lymphocyte-associated antigen-positive CD8 T cells found in the peripheral blood or tissues of patients. The surprising observation of the type 1 character of the generated CD8 T cells will also help us to better understand the complicated pathogenesis of atopic and cutaneous inflammatory diseases.
Collapse
Affiliation(s)
- Li-Fang Wang
- Department of Dermatology, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | | | | | | | | | | |
Collapse
|
30
|
Inoue J, Aramaki Y. Cyclooxygenase-2 inhibition promotes enhancement of antitumor responses by transcutaneous vaccination with cytosine-phosphate-guanosine-oligodeoxynucleotides and model tumor antigen. J Invest Dermatol 2006; 127:614-21. [PMID: 17159912 DOI: 10.1038/sj.jid.5700656] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
One of the principal goals in tumor immune prophylaxis and tumor therapy is the induction of antitumor responses by generating sufficient numbers of tumor antigen-specific helper T (Th)1 cells and cytotoxic T lymphocytes (CTLs). We have demonstrated that the administration of cytosine-phosphate-guanosine-oligodeoxynucleotide (CpG-ODN) through tape-stripped skin induced a Th1-type immune response and suggested that the skin is a potential site for vaccination. CpG-ODN induces the expression of cyclooxygenase (COX)-2, and its product prostaglandin (PG) E2 underlies an immunosuppressive network, therefore it is a simple strategy to use a COX-2 inhibitor for tumor vaccination with CpG-ODN. In this study, we examined whether a COX-2 inhibitor enhances the antitumor immune response induced by CpG-ODN with model tumor antigen, ovalbumin (OVA), applied to tape-stripped skin in mice. The COX-2 inhibitor remarkably enhanced antigen-specific Th1-type immune responses and generation of CTLs induced by transcutaneous vaccination with CpG-ODN and OVA. PGE2 and IL-10 levels in the skin were significantly decreased and production of IL-12 was enhanced. This vaccination also induces an effective antitumor immunity in tumor-challenged mice. These results suggested that transcutaneous vaccination with a COX-2 inhibitor, CpG-ODN, and tumor antigen is a very simple and cost-effective strategy for tumor vaccine and may be readily achievable.
Collapse
Affiliation(s)
- Joe Inoue
- School of Pharmacy, Tokyo University of Pharmacy and Life Science, Hachioji, Tokyo, Japan
| | | |
Collapse
|
31
|
Graham SP, Saya R, Awino E, Ngugi D, Nyanjui JK, Hecker R, Taracha ELN, Nene V. Immunostimulatory CpG oligodeoxynucleotides enhance the induction of bovine CD4+ cytotoxic T-lymphocyte responses against the polymorphic immunodominant molecule of the protozoan parasite Theileria parva. Vet Immunol Immunopathol 2006; 115:383-9. [PMID: 17197038 DOI: 10.1016/j.vetimm.2006.11.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2006] [Revised: 10/18/2006] [Accepted: 11/15/2006] [Indexed: 11/21/2022]
Abstract
Enhancement of the induction of cytotoxic T-cell responses by immunostimulatory CpG oligodeoxynucleotides has been described in humans and mouse models. The present study attempted to address whether CpG has a similar effect in cattle. Immunisation of cattle with a recombinant form of the polymorphic immunodominant molecule from Theileria parva emulsified with immunostimulatory CpG oligodeoxynucleotides in adjuvant had no effect on the induction of antibody responses including the isotype profile, but significantly enhanced the induction of cytolytic responses that were mediated by CD4+CD3+ T cells utilizing the perforin-granzyme pathway.
Collapse
Affiliation(s)
- Simon P Graham
- International Livestock Research Institute, PO Box 30709, Nairobi 00100, Kenya.
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Inoue J, Aramaki Y. Toll-like receptor-9 expression induced by tape-stripping triggers on effective immune response with CpG-oligodeoxynucleotides. Vaccine 2006; 25:1007-13. [PMID: 17049678 DOI: 10.1016/j.vaccine.2006.09.075] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2006] [Revised: 09/05/2006] [Accepted: 09/26/2006] [Indexed: 12/22/2022]
Abstract
Recently, there has been a lot of interest in the potential of non-invasive routes, such as via the skin, for vaccine delivery. CpG-oligodeoxynucleotides (ODN) are an effective adjuvant for the induction of cellular and humoral immunities when administered with an antigen. We demonstrated here that tape-stripping induces the expression of toll-like receptor (TLR)-9 in the skin, and enhances the Th1-type immune response triggered by CpG-ODN administered through the tape-stripped skin. Tape-stripping induces expression of TLR-9 and tumor necrosis factor (TNF)-alpha in the skin, and CpG-ODN treatment through the tape-stripped skin enhances the migration of antigen presenting cells (APCs) to the draining lymph nodes. On the other hand, TLR-9 mRNA and TNF-alpha mRNA were not observed in the skin when CpG-ODN was injected intradermally in a volume of 10 microL, or in a Th1-type immune response. The transdermal application of CpG-ODN with an antigen through the tape-stripped skin is an effective way to induce a Th1-type immune response, and is also a simple, cost-effective and needle-free vaccination system.
Collapse
Affiliation(s)
- Joe Inoue
- School of Pharmacy, Tokyo University of Pharmacy and Life Science, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan
| | | |
Collapse
|
33
|
Epidermal Langerhans cells--changing views on their function in vivo. Immunol Lett 2006; 106:119-25. [PMID: 16828878 DOI: 10.1016/j.imlet.2006.05.010] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2006] [Revised: 05/17/2006] [Accepted: 05/18/2006] [Indexed: 12/01/2022]
Abstract
New experimental models and methods have rendered the field of Langerhans cells very lively. An interesting and productive scientific debate as to the functions of Langerhans cells in vivo is currently going on. We have not yet reached the point where the "pros" would weigh out the "cons", or vice versa. There is good evidence for a lack of Langerhans cell function and for down-regulatory Langerhans cell function in some models. On the other hand, there is also evidence for an active immunogenic and tolerogenic role of Langerhans cells. These recent developments will be discussed.
Collapse
|
34
|
Stoitzner P, Tripp CH, Eberhart A, Price KM, Jung JY, Bursch L, Ronchese F, Romani N. Langerhans cells cross-present antigen derived from skin. Proc Natl Acad Sci U S A 2006; 103:7783-8. [PMID: 16672373 PMCID: PMC1472522 DOI: 10.1073/pnas.0509307103] [Citation(s) in RCA: 158] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Dendritic cells (DC) efficiently cross-present exogenous antigen on MHC class I molecules to CD8+ T cells. However, little is known about cross-presentation by Langerhans cells (LC), the DCs of the epidermis. Therefore, we investigated this issue in detail. Isolated murine LCs were able to cross-present soluble ovalbumin protein on MHC-class I molecules to antigen-specific CD8+ T cells, albeit less potently than the CD8+ DC subsets from spleen. Furthermore, LCs cross-presented cell-associated ovalbumin peptide and protein expressed by neighboring keratinocytes. Use of transporter associated with antigen processing (TAP-1)-deficient mice suggested a TAP-dependent pathway. Similar observations were made with migratory LC. Antigen expressed in the epidermis was ingested by LCs during migration from the epidermis and presented to antigen-specific T cells in vitro. Cross-presentation of ovalbumin protein by LCs induced IFN-gamma production and cytotoxicity in antigen-specific CD8+ T cells. Additionally, epicutaneous application of ovalbumin protein induced in vivo proliferation of OT-I T cells in the draining lymph nodes; this was markedly enhanced when antigen was applied to inflamed, barrier-disrupted skin. Thus, LCs cross-present exogenous antigen to CD8+ T cells and induce effector functions, like cytokine production and cytotoxicity, and may thereby critically contribute in epicutaneous vaccination approaches.
Collapse
Affiliation(s)
- Patrizia Stoitzner
- Departments of *Dermatology and
- Malaghan Institute of Medical Research, Wellington 6005, New Zealand; and
| | | | - Andreas Eberhart
- Biochemical Pharmacology, Innsbruck Medical University, 6020 Innsbruck, Austria
| | - Kylie M. Price
- Malaghan Institute of Medical Research, Wellington 6005, New Zealand; and
| | - Jae Y. Jung
- Malaghan Institute of Medical Research, Wellington 6005, New Zealand; and
| | - Laura Bursch
- Center for Immunology, Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455
| | - Franca Ronchese
- Malaghan Institute of Medical Research, Wellington 6005, New Zealand; and
| | - Nikolaus Romani
- Departments of *Dermatology and
- To whom correspondence should be addressed. E-mail:
| |
Collapse
|
35
|
Ghoreishi M, Dutz JP. Tolerance induction by transcutaneous immunization through ultraviolet-irradiated skin is transferable through CD4+CD25+ T regulatory cells and is dependent on host-derived IL-10. THE JOURNAL OF IMMUNOLOGY 2006; 176:2635-44. [PMID: 16456026 DOI: 10.4049/jimmunol.176.4.2635] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
UV radiation of the skin impairs immune responses to haptens and to tumor Ags. Transcutaneous immunization (TCI) is an effective method of inducing immune responses to protein and peptide Ag. We explore the effect of UV irradiation on TCI. The generation of Ag-specific CTL to OVA protein, but not class I MHC-restricted OVA peptide, is inhibited by TCI through UV-irradiated skin. Consequently, the induction of protein contact hypersensitivity and in vivo Ag-specific CTL activity following OVA protein immunization is prevented. Application of haptens to UV-exposed skin induces hapten-specific tolerance. We demonstrate that application of protein or class II MHC-restricted OVA peptide to UV-irradiated skin induces transferable Ag-specific tolerance. This tolerance is mediated by CD4(+)CD25(+) T regulatory (T(reg)) cells. These Ag-specific T(reg) cells inhibit the priming of CTL following protein immunization in the presence of CpG adjuvant. IL-10 deficiency is known to prevent hapten-specific tolerance induction. In this study, we demonstrate, using IL-10-deficient mice and adoptive T cell transfer, that IL-10 is required for the direct inhibition of CTL priming following immunization through UV-irradiated skin. However, IL-10 is not required for the induction of T(reg) cells through UV-irradiated skin as IL-10-deficient T(reg) cells are able to mediate tolerance. Rather, host-derived IL-10 is required for the function of UV-generated T(reg) cells. These experiments indicate that protein and peptide TCI through UV-irradiated skin may be used to induce robust Ag-specific tolerance to neo-Ags and that UV-induced T(reg) cells mediate their effects in part through the modulation of IL-10.
Collapse
Affiliation(s)
- Mehran Ghoreishi
- Department of Medicine and British Colombia Research Institute of Children and Women's Health, University of British Colombia, Vancouver, Canada
| | | |
Collapse
|
36
|
Dell K, Koesters R, Gissmann L. Transcutaneous immunization in mice: Induction of T-helper and cytotoxic T lymphocyte responses and protection against human papillomavirus-induced tumors. Int J Cancer 2006; 118:364-72. [PMID: 16052529 DOI: 10.1002/ijc.21360] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Previous reports have shown that transcutaneous immunization (TCI) with proteins or peptides in combination with adjuvants efficiently induces specific cellular and humoral immune responses. However, depending on the kind of skin pretreatment, induction of cellular immune responses was restricted to generation of either specific cytotoxic T lymphocytes (CTLs) or T-helper (Th) cells. In this study, we induced antigen-specific CTL responses together with the appropriate Th responses by TCI of C57BL/6 mice. We applied ovalbumin protein or an ovalbumin-derived fusion peptide containing a CTL and Th epitope together with a combination of cholera toxin (CT) and CpG oligodeoxynucleotide (CpG) onto cold wax-depilated and hydrated bare skin. TCI with the ovalbumin fusion peptide induced more robust CTL and Th responses than that with ovalbumin protein. The fusion peptide in combination with the nontoxic CT derivative CTA1-D2D1 and CpG induced an antigen-specific CTL response, albeit less efficiently than in combination with complete CT. Further, we compared the potency of HPV-16 E7 oncoprotein-derived peptides containing single (CTL) or multiple (CTL + Th + B cell) epitopes to induce effective CTL responses. Strong E7-specific CTL responses were detected only after TCI with the E7 multiepitope peptide. This peptide was also shown to protect mice against tumor growth after challenge with HPV-16 E7-positive tumor cells. TCI with E7 protein and CT/CpG led to formation of an E7-specific humoral immune response.
Collapse
Affiliation(s)
- Kerstin Dell
- Deutsches Krebsforschungszentrum, Heidelberg, Germany.
| | | | | |
Collapse
|
37
|
Inoue J, Yotsumoto S, Sakamoto T, Tsuchiya S, Aramaki Y. Changes in immune responses to antigen applied to tape-stripped skin with CpG-oligodeoxynucleotide in mice. J Control Release 2005; 108:294-305. [PMID: 16209897 DOI: 10.1016/j.jconrel.2005.08.014] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2005] [Revised: 07/22/2005] [Accepted: 08/08/2005] [Indexed: 12/11/2022]
Abstract
CpG-oligodeoxynucleotide (CpG-ODN) plays a critical role in immunity via the augmentation of Th1 and suppression of Th2 responses. We examined here the effect of CpG-ODN on the immune response to an antigen applied to tape-stripped mouse skin by evaluating the production of cytokines and Ig isotypes. Confocal laser scanning microscopy revealed that the model antigen, OVA, and CpG-ODN easily penetrated the tape-stripped skin. Co-administration of CpG-ODN and OVA to the disrupted skin elicited an antigen-specific Th1-predominant immune response and enhanced the production of Th1-type cytokines, IL-12 and IFN-gamma. On the other hand, the production of a Th2-type cytokine, IL-4, was drastically suppressed. Cytokine production was supported by the expression of mRNA in the draining lymph node. In terms of antigen-specific antibody production, the level of IgG2a which is regulated by IFN-gamma was increased by CpG-ODN, but IgE production regulated by IL-4 was suppressed. Furthermore, administration of CpG-ODN via the skin drastically attenuated the production of IgE in mice undergoing IgE-type immune response. Administration of CpG-ODN through the skin may shift the immune response from Th2 to Th1-like response. These results suggested that administration of CpG-ODN via skin is a simple strategy for patients with diseases like AD, which is characterized by Th2-dominated inflammation.
Collapse
Affiliation(s)
- Joe Inoue
- School of Pharmacy, Tokyo University of Pharmacy and Life Science 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan
| | | | | | | | | |
Collapse
|
38
|
Inoue J, Yotsumoto S, Sakamoto T, Tsuchiya S, Aramaki Y. Changes in immune responses to antigen applied to tape-stripped skin with CpG-oligodeoxynucleotide in NC/Nga mice. Pharm Res 2005; 22:1627-33. [PMID: 16180119 DOI: 10.1007/s11095-005-6834-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2005] [Accepted: 06/10/2005] [Indexed: 12/11/2022]
Abstract
PURPOSE CpG-oligodeoxynucleotide (CpG-ODN) plays a critical role in immunity via the augmentation of Th1 and the suppression of Th2 responses. We examined here the effect of CpG-ODN on the immune response to an antigen applied to a tape-stripped skin of NC/Nga mouse, a human atopic dermatitis (AD) model, by evaluating the production of cytokines and immunoglobulin isotypes. METHODS Model antigen, ovalbumin (OVA), and CpG-ODN were applied on to the shaved skin. The penetration of OVA and CpG-ODN was evaluated using confocal laser scanning microscopy (CLSM). Secretion of cytokine from splenocytes and changes in immunoglobulin isotype levels were determined by enzyme-linked immunosorbent assay (ELISA). RESULTS Through CLSM it was revealed that the model antigen, OVA, and CpG-ODN easily penetrated the tape-stripped skin. Coadministration of CpG-ODN and OVA to the skin elicited an antigen-specific, Th1-predominant immune response and enhanced the production of IFN-gamma. On the other hand, the production of a Th2-type cytokine, IL-4, was drastically suppressed. In terms of antigen-specific antibody production, the level of IgG2a regulated by IFN-gamma was increased by CpG-ODN, but IgE production regulated by IL-4 was suppressed. CONCLUSIONS Administration of CpG-ODN with antigen through the skin may shift the immune response from a Th2- to Th1-like response. These results suggested that administration of CpG-ODN via skin is a simple strategy for patients with diseases such as AD, which is characterized by Th2-dominated inflammation.
Collapse
Affiliation(s)
- Joe Inoue
- School of Pharmacy, Tokyo University of Pharmacy and Life Science, 1432-1 Horinouchi, Hachioji, Tokyo, 192-0392, Japan,
| | | | | | | | | |
Collapse
|
39
|
Rechtsteiner G, Warger T, Osterloh P, Schild H, Radsak MP. Cutting Edge: Priming of CTL by Transcutaneous Peptide Immunization with Imiquimod. THE JOURNAL OF IMMUNOLOGY 2005; 174:2476-80. [PMID: 15728450 DOI: 10.4049/jimmunol.174.5.2476] [Citation(s) in RCA: 133] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CTL are important in combating cancer and viruses. Therefore, triggering the complete potential of CTL effector functions by new vaccination strategies will not only improve prophylaxis of tumor or virus-related diseases, but also open opportunities for effective therapeutic immunizations. Using transcutaneous immunization, we show that epicutaneous (e.c.)(4) application of an ointment containing a CTL epitope and the TLR7 ligand imiquimod is highly effective in activating T cells in mice using TCR-transgenic CTL or in wild-type mice. Transcutaneous immunization-activated CTL mount a full-blown immune response against the target epitope characterized by proliferation, cytolytic activity, and the production of IFN-gamma that is completely restricted to the epitope used for vaccination. Our results obtained by simple e.c. application of an ointment, without further skin irritating procedures, provide the basis for the development of new, easy to use vaccines against cancer or virus-associated diseases.
Collapse
MESH Headings
- Administration, Cutaneous
- Adoptive Transfer
- Aminoquinolines/administration & dosage
- Aminoquinolines/immunology
- Animals
- Cells, Cultured
- Cytotoxicity, Immunologic/genetics
- Epitopes, T-Lymphocyte/administration & dosage
- Epitopes, T-Lymphocyte/immunology
- Imiquimod
- Lymph Nodes/immunology
- Lymph Nodes/pathology
- Lymphocyte Activation/genetics
- Lymphocyte Activation/immunology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Protein Transport/immunology
- Receptors, Antigen, T-Cell/genetics
- Resting Phase, Cell Cycle/genetics
- Resting Phase, Cell Cycle/immunology
- Spleen/cytology
- Spleen/immunology
- Spleen/transplantation
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Cytotoxic/metabolism
- Vaccines, Subunit/administration & dosage
- Vaccines, Subunit/immunology
Collapse
Affiliation(s)
- Gerd Rechtsteiner
- Institute for Immunology, Johannes Gutenberg-University, Mainz, Germany
| | | | | | | | | |
Collapse
|
40
|
Partidos CD, Moreau E, Chaloin O, Tunis M, Briand JP, Desgranges C, Muller S. A synthetic HIV-1 Tat protein breaches the skin barrier and elicits Tat-neutralizing antibodies and cellular immunity. Eur J Immunol 2004; 34:3723-31. [PMID: 15549730 DOI: 10.1002/eji.200425313] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
The HIV-1 Tat protein plays a critical role in the pathogenesis of HIV and has been considered as a candidate vaccine antigen. In an effort to design a non-invasive vaccination strategy against HIV-1 that stimulates the induction of systemic and mucosal immune responses, we studied the transcutaneous delivery of a synthetic Tat protein using cholera toxin as an adjuvant. Following immunization of BALB/c mice with various doses of Tat, IgG and IgA antibody responses were measured in the serum and vaginal washes, respectively. Serum antibodies predominantly recognized the N-terminal and basic functional domains of the protein and exhibited neutralizing capacity against Tat-driven transactivation. Transcutaneous immunization also elicited potent cellular immune responses against Tat and the secretion of high levels of IL-2, IFN-gamma and IL-6. These findings demonstrate for the first time that by using a simple and safe immunization procedure, a synthetic Tat protein can elicit potentially protective immune responses. Transcutaneous immunization may be advantageous for the non-invasive delivery of other HIV candidate vaccine antigens.
Collapse
|