US20060269593A1 - Adjuvant for transcutaneous immunization - Google Patents

Adjuvant for transcutaneous immunization Download PDF

Info

Publication number
US20060269593A1
US20060269593A1 US11/334,349 US33434906A US2006269593A1 US 20060269593 A1 US20060269593 A1 US 20060269593A1 US 33434906 A US33434906 A US 33434906A US 2006269593 A1 US2006269593 A1 US 2006269593A1
Authority
US
United States
Prior art keywords
antigen
immunization
cells
skin
liposomes
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US11/334,349
Inventor
Gregory Glenn
Carl Alving
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
US Government
Original Assignee
US Government
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=27669393&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US20060269593(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Priority claimed from US08/749,164 external-priority patent/US5910306A/en
Application filed by US Government filed Critical US Government
Priority to US11/334,349 priority Critical patent/US20060269593A1/en
Publication of US20060269593A1 publication Critical patent/US20060269593A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/06Ointments; Bases therefor; Other semi-solid forms, e.g. creams, sticks, gels
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/39Medicinal preparations containing antigens or antibodies characterised by the immunostimulating additives, e.g. chemical adjuvants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/08Solutions
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/10Dispersions; Emulsions
    • A61K9/107Emulsions ; Emulsion preconcentrates; Micelles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/10Dispersions; Emulsions
    • A61K9/127Liposomes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/70Web, sheet or filament bases ; Films; Fibres of the matrix type containing drug
    • A61K9/7023Transdermal patches and similar drug-containing composite devices, e.g. cataplasms
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B82NANOTECHNOLOGY
    • B82YSPECIFIC USES OR APPLICATIONS OF NANOSTRUCTURES; MEASUREMENT OR ANALYSIS OF NANOSTRUCTURES; MANUFACTURE OR TREATMENT OF NANOSTRUCTURES
    • B82Y5/00Nanobiotechnology or nanomedicine, e.g. protein engineering or drug delivery
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/53DNA (RNA) vaccination
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/54Medicinal preparations containing antigens or antibodies characterised by the route of administration
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55511Organic adjuvants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55511Organic adjuvants
    • A61K2039/55522Cytokines; Lymphokines; Interferons
    • A61K2039/55527Interleukins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55511Organic adjuvants
    • A61K2039/55544Bacterial toxins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55511Organic adjuvants
    • A61K2039/55555Liposomes; Vesicles, e.g. nanoparticles; Spheres, e.g. nanospheres; Polymers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55511Organic adjuvants
    • A61K2039/55566Emulsions, e.g. Freund's adjuvant, MF59

Definitions

  • the invention relates to transcutaneous immunization, and adjuvants useful therein, to induce an antigen-specific immune response.
  • Transcutaneous immunization requires both passage of an antigen through the outer barriers of the skin, which are normally impervious to such passage, and an immune response to the antigen.
  • an antigen elicits a strong antibody response that is highly reproducible; the antigen could be applied in a saline solution to the skin, with or without liposomes.
  • adjuvants such as, for example, bacterial exotoxins, their subunits, and related toxins.
  • transferosomes are used as a carrier for proteins (bovine serum albumin and gap junction proteins) against which the complement-mediated lysis of antigen-sensitized liposomes is directed.
  • proteins bovine serum albumin and gap junction proteins
  • An immune response was not induced when solution containing the protein was placed on the skin; only transferosomes were able to transport antigen across the skin and achieve immunization.
  • transferosomes are not liposomes.
  • FIG. 1 of Paul et al. (1995) showed that only a formulation of antigen and transferosomes induced an immune response, assayed by lysis of antigen-sensitized liposomes.
  • Formulations of antigen in solution, antigen and mixed micelles, and antigen and liposomes (i.e., smectic mesophases) applied to the skin did not induce an immune response equivalent to that induced by subcutaneous injection. Therefore, there was a positive control (i.e., antigen and transfersomes) to validate their negative conclusion that a formulation of antigen and liposomes did not cause transdermal immunization.
  • OVA ovalbumin
  • mice were more extensively patched with OVA in solution for four days every two weeks. This was repeated five times, i.e., the mice wore patches for a total of 20 days. Again, the high dose of OVA did not produce significant IgG2a antibodies. Significant levels of IgE antibodies were produced.
  • the inventors have found that application to the skin of antigen and adjuvant provides a transcutaneous delivery system for antigen that can induce an antigen-specific immune response of IgG or IgA.
  • the adjuvant is preferably an ADP-ribosylating exotoxin.
  • hydration, penetration enhancers, or occlusive dressings may be used in the transcutaneous delivery system.
  • An object of the invention is to provide a system for transcutaneous immunization that induces an immune response (e.g., humoral and/or cellular effectors) in an animal or human.
  • the system provides simple application to intact skin of an organism of a formulation comprised of antigen and adjuvant to induce a specific immune response against the antigen.
  • the adjuvant may activate antigen presenting cells of the immune system (e.g., Langerhans cells in the epidermis, dermal dendritic cells, dendritic cells, macrophages, B lymphocytes) and/or induce the antigen presenting cells to phagocytose the antigen.
  • the antigen presenting cells then present the antigen to T and B cells.
  • the antigen presenting cells then may migrate from the skin to the lymph nodes and present antigen to lymphocytes (e.g., B and/or T cells), thereby inducing an antigen-specific immune response.
  • another object of the invention is to positively and/or negatively regulate components of the immune system by using the transcutaneous immunization system to affect antigen-specific helper (Th1 and/or Th2) or delayed-type hypersensitivity (T DTH ) T-cell subsets.
  • Th1 and/or Th2 antigen-specific helper
  • T DTH delayed-type hypersensitivity
  • a formulation containing antigen and adjuvant is applied to intact skin of an organism, the antigen is presented to immune cells, and an antigen-specific immune response is induced without perforating the skin.
  • the formulation may include additional antigens such that transcutaneous application of the formulation induces an immune response to multiple antigens.
  • the antigens may or may not be derived from the same source, but the antigens will have different chemical structures so as to induce immune responses specific for the different antigens.
  • Antigen-specific lymphocytes may participate in the immune response and, in the case-of participation by B lymphocytes, antigen-specific antibodies may be part of the immune response.
  • the above method is used to treat an organism.
  • the treatment vaccinates the organism against infection by the pathogen or against its pathogenic effects such as those caused by toxin secretion.
  • a formulation that includes a tumor antigen may provide a cancer treatment; a formulation that includes an autoantigen may provide a treatment for a disease caused by the organism's own immune system (e.g., autoimmune disease).
  • a patch for use in the above methods comprises a dressing, and effective amounts of antigen and adjuvant.
  • the dressing may be occlusive or non-occlusive.
  • the patch may include additional antigens such that application of the patch induces an immune response to multiple antigens.
  • the antigens may or may not be derived from the same source, but the antigens will have different chemical structures so as to induce an immune response specific for the different antigens.
  • multiple patches may be applied at frequent intervals or constantly over a period of time.
  • the formulation is applied to intact skin overlying more than one draining lymph node field using either single or multiple applications.
  • the formulation may include additional antigens such that application to intact skin induces an immune response to multiple antigens.
  • the antigens may or may not be derived from the same source, but the antigens will have different chemical structures so as to induce an immune response specific for the different antigens.
  • the products and methods may be used therapeutically to treat existing disease, protectively to prevent disease, or to reduce the severity and/or duration of disease.
  • the formulation may comprise a hydrating agent (e.g., liposomes), a penetration enhancer, or both.
  • a hydrating agent e.g., liposomes
  • the formulation may comprise AQUAPHOR (an emulsion of petrolatum, mineral oil, mineral wax, wool wax, panthenol, bisabol, and glycerin), emulsions (e.g., aqueous creams), oil-in-water emulsions (e.g., oily creams), anhydrous lipids and oil-in-water emulsions, anhydrous lipids and water-in-oil emulsions, fats, waxes, oil, silicones, and humectants (e.g., glycerol).
  • AQUAPHOR an emulsion of petrolatum, mineral oil, mineral wax, wool wax, panthenol, bisabol, and glycerin
  • emulsions e
  • the antigen may be derived from a pathogen that-can infect the organism (e.g., bacterium, virus, fungus, or parasite), or a cell (e.g., tumor cell or normal cell).
  • the antigen may be a tumor antigen or an autoantigen.
  • the antigen may be a carbohydrate, glycolipid, glycoprotein, lipid, lipoprotein, phospholipid, polypeptide, or chemical or recombinant conjugate of the above.
  • the molecular weight of the antigen may be greater than 500 daltons, preferably greater than 800 daltons, and more preferably greater than 1000 daltons.
  • Antigen may be obtained by recombinant means, chemical synthesis, or purification from a natural source. Preferred are proteinaceous antigen or conjugates with polysaccharide. Antigen may be at least partially purified in cell-free form. Alternatively, antigen may be provided in the form of a live virus, an attenuated live virus, or an inactivated virus.
  • an adjuvant may allow potentiation or modulation of the immune response.
  • selection of a suitable antigen or adjuvant may allow preferential induction of a humoral or cellular immune response, specific antibody isotypes (e.g., IgM, IgD, IgA1, IgA2, IgE, IgG1, IgG2, IgG3, and/or IgG4), and/or specific T-cell subsets (e.g., CTL, Th1, Th2 and/or T DTH ).
  • specific antibody isotypes e.g., IgM, IgD, IgA1, IgA2, IgE, IgG1, IgG2, IgG3, and/or IgG4
  • specific T-cell subsets e.g., CTL, Th1, Th2 and/or T DTH .
  • the adjuvant is an ADP-ribosylating exotoxin or a subunit thereof.
  • an activator of Langerhans cells may be used.
  • antigen, adjuvant, or both may be provided in the formulation by means of a nucleic acid (e.g., DNA, RNA, cDNA, cRNA) encoding the antigen or adjuvant as appropriate.
  • a nucleic acid e.g., DNA, RNA, cDNA, cRNA
  • This technique is called genetic immunization.
  • an antigen as used in the invention, is meant to describe a substance that induces a specific immune response when presented to immune cells of an organism.
  • An antigen may comprise a single immunogenic epitope, or a multiplicity of immunogenic epitopes recognized by a B-cell receptor (i.e., antibody on the membrane of the B cell) or a T-cell receptor.
  • a molecule may be both an antigen and an adjuvant (e.g., cholera toxin) and, thus, the formulation may contain only one component.
  • adjuvant as used in the invention, is meant to describe a substance added to the formulation to assist in inducing an immune response to the antigen.
  • an effective amount is meant to describe that amount of antigen which induces an antigen-specific immune response.
  • Such induction of an immune response may provide a treatment such as, for example, immunoprotection, desensitization, immunosuppression, modulation of autoimmune disease, potentiation of cancer immunosurveillance, or therapeutic vaccination against an established infectious disease.
  • draining lymph node field means an anatomic area over which the lymph collected is filtered through a set of defined set of lymph nodes (e.g., cervical, axillary, inguinal, epitrochelear, popliteal, those of the abdomen and thorax).
  • FIG. 1 shows cholera toxin (CT) induces enhanced major histocompatibility complex (MHC) class II expression on Langerhans cells (LC), changes in LC morphology, and loss of LCs (presumably through migration).
  • CT cholera toxin
  • MHC major histocompatibility complex
  • Panels indicate (A) saline alone as a negative control, (B) transcutaneous immunization with CT in saline, (C) transcutaneous immunization with CTB in saline, and (D) intradermal injection with tumor necrosis factor- ⁇ (10 ⁇ g) as a positive control.
  • a transcutaneous immunization system delivers agents to specialized cells (e.g., antigen presentation cell, lymphocyte) that produce an immune response (Bos, 1997). These agents as a class are called antigens.
  • Antigen may be composed of chemicals such as, for example, carbohydrate, glycolipid, glycoprotein, lipid, lipoprotein, phospholipid, polypeptide, conjugates thereof, or any other material known to induce an immune response.
  • Antigen may be provided as a whole organism such as, for example, a bacterium or virion; antigen may be obtained from an extract or lysate, either from whole cells or membrane alone; or antigen may be chemically synthesized or produced by recombinant means.
  • compositions suitable for administration to a human or animal may be applied in the form of an cream, emulsion, gel, lotion, ointment, paste, solution, suspension, or other forms known in the art. In particular, formulations that enhance skin hydration, penetration, or both are preferred. There may also be incorporated other pharmaceutically acceptable additives including, for example, diluents, binders, stabilizers, preservatives, and colorings.
  • penetration enhancer does not include substances such as, for example: water, physiological buffers, saline solutions, and alcohols which would not perforate the skin.
  • An object of the present invention is to provide a novel means for immunization through intact skin without the need for perforating the skin.
  • the transcutaneous immunization system provides a method whereby antigens and adjuvant can be delivered to the immune system, especially specialized antigen presentation cells underlying the skin such as, for example, Langerhans cells.
  • the transcutaneous immunization delivery system carries antigen to cells of the immune system where an immune response is induced.
  • the antigen may pass through the normal protective outer layers of the skin (i.e., stratum corneum) and induce the immune response directly, or through an antigen presenting cell (e.g., macrophage, tissue macrophage, Langerhans cell, dendritic cell, dermal dendritic cell, B lymphocyte, or Kupffer cell) that presents processed antigen to a T lymphocyte.
  • an antigen presenting cell e.g., macrophage, tissue macrophage, Langerhans cell, dendritic cell, dermal dendritic cell, B lymphocyte, or Kupffer cell
  • the antigen may pass through the stratum corneum via a hair follicle or a skin organelle (e.g., sweat gland, oil gland).
  • Transcutaneous immunization with bacterial ADP-ribosylating exotoxins may target the epidermal Langerhans cell, known to be among the most efficient of the antigen presenting cells (APCs) (Udey, 1997).
  • APCs antigen presenting cells
  • bAREs activate Langerhans cells when applied epicutaneously to the skin in saline solution.
  • the Langerhans cells direct specific immune responses through phagocytosis of the antigens, and migration to the lymph nodes where they act as APCs to present the antigen to lymphocytes (Udey, 1997), and thereby induce a potent antibody response.
  • the skin is generally considered a barrier to invading organisms
  • the imperfection of this barrier is attested to by the numerous Langerhans cells distributed throughout the epidermis that are designed to orchestrate the immune response against organisms invading via the skin (Udey, 1997).
  • a transcutaneous immune response using the skin immune system would require delivery of vaccine antigen only to Langerhans cells in the stratum corneum (the outermost layer of the skin consisting of cornified cells and lipids) via passive diffusion and subsequent activation of the Langerhans cells to take up antigen, migrate to B-cell follicles and/or T-cell dependent regions, and present the antigen to B and/or T cells (Stingl et al., 1989).
  • antigens other that bAREs for example BSA
  • BSA antigens other that bAREs
  • these antigens could also be taken to the lymph node for presentation to T-cells and subsequently induce an immune response specific for that antigen (e.g., BSA).
  • a feature of transcutaneous immunization is the activation of the Langerhans cell, presumably by a bacterial ADP-ribosylating exotoxin, ADP-ribosylating exotoxin binding subunits (e.g., cholera toxin B subunit), or other Langerhans cell activating substance.
  • the mechanism of transcutaneous immunization via Langerhans cells activation, migration and antigen presentation is clearly shown by the upregulation of MHC class II expression in the epidermal Langerhans cells from epidermal sheets transcutaneously immunized with CT or CTB.
  • the magnitude of the antibody response induced by transcutaneous immunization and isotype switching to predominantly IgG is generally achieved with T-cell help (Janeway and Travers, 1996), and activation of both Th1 and Th2 pathways is suggested by the production of IgG1 and IgG2a (Paul and Seder, 1994; Seder and Paul, 1994).
  • a large antibody response may be induced by a thymus-independent antigen type 1 (TI-1) which directly activates the B cell (Janeway and Travers, 1996).
  • contact dermatitis a pathogenic manifestation of LC activation
  • Langerhans cells which phagocytose antigen, migrate to lymph nodes, present antigen, and sensitize T cells for the intense destructive cellular response that occurs at the affected skin site (Dahl, 1996; Leung, 1997).
  • Atopic dermatitis may utilize the Langerhans cell in a similar fashion, but is identified with Th2 cells and is generally associated with high levels of IgE antibody (Dahl, 1996; Leung, 1997).
  • Transcutaneous immunization with cholera toxin-and related bAREs is a novel immune response with an absence of superficial and microscopic post-immunization skin findings (i.e., non-inflamed skin) shown by the absence of lymphocyte infiltration 24, 48 and 120 hours after immunization.
  • the uniqueness of the transcutaneous immune response here is also indicated by the both high levels of antigen-specific IgG antibody, and the type of antibody produced (e.g., IgM, IgG1, IgG2a, IgG2b, IgG3 and IgA) and the absence of anti-CT IgE antibody.
  • bacterial-derived toxins applied to the surface of the skin can activate Langerhans cells and induce a potent immune response manifested as high levels of antigen-specific circulating IgG antibodies.
  • adjuvants may be used in transcutaneous immunization to enhance the IgG antibody response to proteins not otherwise immunogenic by themselves when placed on the skin.
  • Transcutaneous targeting of Langerhans cells may also be used to deactivate their antigen presenting function, thereby preventing immunization or sensitization.
  • Techniques to deactivate Langerhans cells include, for example, the use of interleukin-10 (Peguet-Navarro et al., 1995), monoclonal antibody to interleukin-1 ⁇ (Enk et al., 1993), or depletion via superantigens such as through staphylococcal enterotoxin-A (SEA) induced epidermal Langerhans cell depletion (Shankar et al., 1996).
  • SEA staphylococcal enterotoxin-A
  • Transcutaneous immunization may be induced via the ganglioside GM1 binding activity of CT, LT or subunits such as CTB (Craig and Cuatrecasas, 1975).
  • Ganglioside GM1 is a ubiquitous cell membrane glycolipid found in all mammalian cells (Plotkin and Mortimer, 1994).
  • a hydrophilic pore is formed which allows the A subunit to penetrate across the lipid bilayer (Ribi et al., 1988).
  • transcutaneous immunization by CT or CTB may require ganglioside GM1 binding activity.
  • CTA contains the ADP-ribosylating exotoxin activity but only CT and CTB containing the binding activity were able to induce an immune response indicating that the B subunit was necessary and sufficient to immunize through the skin.
  • the Langerhans cell may be activated by CTB binding to its cell surface.
  • Antigen of the invention may be expressed by recombinant means, preferably as a fusion with an affinity or epitope tag (Summers and Smith, 1987; Goeddel, 1990; Ausubel et al., 1996); chemical synthesis of an oligopeptide, either free or conjugated to carrier proteins, may be used to obtain antigen of the invention (Bodanszky, 1993; Wisdom, 1994). Oligopeptides are considered a type of polypeptide.
  • Oligopeptide lengths of 6 residues to 20 residues are preferred.
  • Polypeptides may also by synthesized as branched structures such as those disclosed in U.S. Pat. Nos. 5,229,490 and 5,390,111.
  • Antigenic polypeptides include, for example, synthetic or recombinant B-cell and T-cell epitopes, universal T-cell epitopes, and mixed T-cell epitopes from one organism or disease and B-cell epitopes from another.
  • Antigen obtained through recombinant means or peptide synthesis, as well as antigen of the invention obtained from natural sources or extracts, may be purified by means of the antigen's physical and chemical characteristics, preferably by fractionation or chromatography (Janson and Ryden, 1989; Deutscher, 1990; Scopes, 1993).
  • a multivalent antigen formulation may be used to induce an immune response to more than one antigen at the same time.
  • Conjugates may be used to induce an immune response to multiple antigens, to boost the immune response, or both.
  • toxins may be boosted by the use of toxoids, or toxoids boosted by the use of toxins.
  • Transcutaneous immunization may be used to boost responses induced initially by other routes of immunization such as by injection, or the oral or intranasal routes.
  • Antigen includes, for example, toxins, toxoids, subunits thereof, or combinations thereof (e.g., cholera toxin, tetanus toxoid).
  • Antigen may be solubilized in a buffer.
  • Suitable buffers include, but are not limited to, phosphate buffered saline Ca ++ /Mg ++ free (PBS), normal saline (150 mM NaCl in water), and Tris buffer.
  • Antigen not soluble in neutral buffer can be solubilized in 10 mM acetic acid and then diluted to the desired volume with a neutral buffer such as PBS.
  • acetate-PBS at acid pH may be used as a diluent after solubilization in dilute acetic acid.
  • Glycerol may be a suitable non-aqueous buffer for use in the present invention.
  • Hydrophobic antigen can be solubilized in a detergent, for example a polypeptide containing a membrane-spanning domain.
  • a detergent solution e.g., a cell membrane extract
  • liposomes then may be formed by removal of the detergent by dilution, dialysis, or column chromatography.
  • Certain antigens such as, for example, those from a virus (e.g., hepatitis A) need not be soluble per se, but can be incorporated directly into a liposome in the form of a virosome (Morein and Simons, 1985).
  • Plotkin and Mortimer (1994) provide antigens which can be used to vaccinate animals or humans to induce an immune response specific for particular pathogens, as well as methods of preparing antigen, determining a suitable dose of antigen, assaying for induction of an immune response, and treating infection by a pathogen (e.g., bacterium, virus, fungus, or parasite).
  • a pathogen e.g., bacterium, virus, fungus, or parasite.
  • Bacteria include, for example: anthrax, campylobacter, cholera, diphtheria, enterotoxigenic E. coli, giardia, gonococcus, Helicobacter pylori (Lee and Chen, 1994), Hemophilus influenza B, Hemophilus influenza non-typable, meningococcus, pertussis, pneumococcus, salmonella, shigella, Streptococcus B, tetanus, Vibrio cholerae, and yersinia.
  • Viruses include, for example: adenovirus, dengue serotypes 1 to 4 (Delenda et al., 1994; Fonseca et al., 1994; Smucny et al., 1995), ebola (setting et al., 1996), enterovirus, hepatitis serotypes A to E (Blum, 1995; Katkov, 1996; Lieberman and Greenberg, 1996; Mast, 1996; Shafara et al., 1995; Smedila et al., 1994; U.S. Pat. Nos.
  • herpes simplex virus 1 or 2 human immunodeficiency virus (Deprez et al., 1996), influenza, measles, Norwalk, papilloma virus, parvovirus B19, polio, rabies, rotavirus, rubella, rubeola, vaccinia, vaccinia constructs containing genes coding for other antigens such as malaria antigens, varicella, and yellow fever.
  • Parasites include, for example: Entamoeba histolytica (Zhang et al., 1995); Plasmodium (Bathurst et al., 1993; Chang et al., 1989, 1992, 1994; Fries et al., 1992a, 1992b; Herrington et al., 1991; Khusmith et al., 1991; Malik et al., 1991; Migliorini et al., 1993; Pessi et al., 1991; Tam, 1988; Vreden et al., 1991; White et al., 1993; Wiesmueller et al., 1991), Leishmania (Frankenburg et al., 1996), and the Helminthes.
  • the formulation also contains an adjuvant, although a single molecule may contain both adjuvant and antigen properties (e.g., cholera toxin) (Elson and Dertzbaugh, 1994).
  • adjuvants are substances that are used to specifically or non-specifically potentiate an antigen-specific immune response.
  • the adjuvant and the formulation are mixed prior to presentation of the antigen but, alternatively, they may be separately presented within a short interval of time.
  • Adjuvants include, for example, an oil emulsion (e.g., complete or incomplete Freund's adjuvant), a chemokine (e.g., defensins 1 or 2, RANTES, MIP1- ⁇ , MIP-2, interleukin-8) or a cytokine (e.g., interleukin-1 ⁇ , -2, -6, -10 or -12; ⁇ -interferon; tumor necrosis factor- ⁇ ; or granulocyte-monocyte-colony stimulating factor) (reviewed in Nohria and Rubin, 1994), a muramyl dipeptide derivative (e.g., murabutide, threonyl-MDP or muramyl tripeptide), a heat shock protein or a derivative, a derivative of Leishmania major LeIF (Skeiky et al., 1995), cholera toxin or cholera toxin B, a lipopolysaccharide (LPS) derivative (e.
  • An adjuvant may be chosen to preferentially induce antibody or cellular effectors, specific antibody isotypes (e.g., IgM, IgD, IgA1, IgA2, secretory IgA, IgE, IgG1, IgG2, IgG3, and/or IgG4), or specific T-cell subsets (e.g., CTL, Th1, Th2 and/or T DTH ) (Glenn et al., 1995).
  • specific antibody isotypes e.g., IgM, IgD, IgA1, IgA2, secretory IgA, IgE, IgG1, IgG2, IgG3, and/or IgG4
  • T-cell subsets e.g., CTL, Th1, Th2 and/or T DTH
  • Cholera toxin is a bacterial exotoxin from the family of ADP-ribsoylating exotoxins (referred to as bAREs). Most bAREs are organized as A:B dimer with a binding B subunit and an A subunit containing the ADP-ribosyltransferase.
  • toxins include diphtheria, Pseudomonas exotoxin A, cholera toxin (CT), E. coli heat-labile enterotoxin (LT), pertussis toxin, C. botulinum toxin C2, C. botulinum toxin C3, C. limosum exoenzyme, B. cereus exoenzyme, Pseudomonas exotoxin S, Staphylococcus aureus EDIN, and B. sphaericus toxin.
  • CT cholera toxin
  • LT heat-labile enterotoxin
  • Cholera toxin is an example of a bARE that is organized with A and B subunits.
  • the B subunit is the binding subunit and consists of a B-subunit pentamer which is non-covalently bound to the A subunit.
  • the B-subunit pentamer is arranged in a symmetrical doughnut-shaped structure that binds to GM 1 -ganglioside on the target cell.
  • the A subunit serves to ADP ribosylate the alpha subunit of a subset of the hetero trimeric GTP proteins (G proteins) including the Gs protein which results in the elevated intracellular levels of cyclic AMP. This stimulates release of ions and fluid from intestinal cells in the case of cholera.
  • G proteins hetero trimeric GTP proteins
  • CT Cholera toxin
  • CTB B subunit
  • bARE Pseudomonas exotoxin A (ETA) binds to the ⁇ 2 -macroglobulin receptor-low density lipoprotein receptor-related protein (Kounnas et al., 1992). bAREs are reviewed by Krueger and Barbieri (1995).
  • CT cholera toxin
  • CTB cholera toxin
  • LT E. coli heat-labile enterotoxin
  • pertussis toxin are potent adjuvants for transcutaneous immunization, inducing high levels of IgG antibodies but not IgE antibodies.
  • CTB without CT can also induce high levels of IgG antibodies.
  • both bAREs and a derivative thereof can effectively immunize when epicutaneouly applied to the skin in a simple solution.
  • bAREs can act as adjuvants for non-immunogenic proteins in an transcutaneous immunization system.
  • DPT tetanus
  • toxins can be chemically inactivated to form toxoids which are less toxic but remain immunogenic.
  • the transcutaneous immunization system using toxin-based immunogens and adjuvants can achieve anti-toxin levels adequate for protection against these diseases.
  • the anti-toxin antibodies may be induced through immunization with the toxins, or genetically-detoxified toxoids themselves, or with toxoids and adjuvants such as CT.
  • Genetically toxoided toxins which have altered ADP-ribosylating exotoxin activity, but not binding activity, are envisioned to be especially useful as non-toxic activators of antigen presenting cells used in transcutaneous immunization.
  • CT can also act as an adjuvant to induce antigen-specific CTLs through transcutaneous immunization (see Bowen et al., 1994; Porgador et al., 1997 for the use of CT as an adjuvant in oral immunization).
  • the bARE adjuvant may be chemically conjugated to other antigens including, for example, carbohydrates, polypeptides, glycolipids, and glycoprotein antigens. Chemical conjugation with toxins, their subunits, or toxoids with these antigens would be expected to enhance the immune response to these antigens when applied epicutaneously.
  • diphtheria toxin is known to be so toxic that one molecule can kill a cell
  • several workers have taken a recombinant approach to producing genetically produced toxoids. This is based on inactivating the catalytic activity of the ADP-ribosyl transferase by genetic deletion. These toxins retain the binding capabilities, but lack the toxicity, of the natural toxins. This approach is described by Burnette et al. (1994), Rappuoli et al. (1995), and Rappuoli et al. (1996).
  • Such genetically toxoided exotoxins could be useful for transcutaneous immunization system in that they would not create a safety concern as the toxoids would not be considered toxic. Additionally, several techniques exist to chemically toxoid toxins which can address the same problem (Schneerson et al., 1996). These techniques could be important for certain applications, especially pediatric applications, in which ingested toxins (e.g., diphtheria toxin) might possibly create adverse reactions. optionally, an activator of Langerhans cells may be used as an adjuvant.
  • activators include: inducers of heat shock protein; contact sensitizers (e.g., trinitrochlorobenzene, dinitrofluorobenzene, nitrogen mustard, pentadecylcatechol); toxins (e.g, Shiga toxin, Staph enterotoxin B); lipopolysaccharides, lipid A, or derivatives thereof; bacterial DNA (Stacey et al., 1996); cytokines (e.g., tumor necrosis factor- ⁇ , interleukin-1 ⁇ , -10, -12); and chemokines (e.g., defensins 1 or 2, RANTES, MIP-1 ⁇ , MIP-2, interleukin-8).
  • contact sensitizers e.g., trinitrochlorobenzene, dinitrofluorobenzene, nitrogen mustard, pentadecylcatechol
  • toxins e.g, Shiga toxin, Staph enterotoxin B
  • an immunizing antigen has sufficient Langerhans cell activating capabilities then a separate adjuvant may not be required, as in the case of CT which is both antigen and adjuvant. It is envisioned that whole cell preparations, live viruses, attenuated viruses, DNA plasmids, and bacterial DNA could be sufficient to immunize transcutaneously. It may be possible to use low concentrations of contact sensitizers or other activators of Langerhans cells to induce an immune response without inducing skin lesions.
  • Liposomes are closed vesicles surrounding an internal aqueous space.
  • the internal compartment is separated from the external medium by a lipid bilayer composed of discrete lipid molecules.
  • antigen may be delivered through intact skin to specialized cells of the immune system, whereby an antigen-specific immune response is induced.
  • Transcutaneous immunization may be achieved by using liposomes; however, as shown in the examples, liposomes are not required to elicit an antigen-specific immune response.
  • Liposomes may be prepared using a variety of techniques and membrane lipids (reviewed in Gregoriadis, 1993). Liposomes may be pre-formed and then mixed with antigen. The antigen may be dissolved or suspended, and then added to (a) the pre-formed liposomes in a lyophilized state, (b) dried lipids as a swelling solution or suspension, or (c) the solution of lipids used to form liposomes. They may also be formed from lipids extracted from the stratum corneum including, for example, ceramide and cholesterol derivatives (Wertz, 1992).
  • Chloroform is a preferred solvent for lipids, but it may deteriorate upon storage. Therefore, at one- to three-month intervals, chloroform is redistilled prior to its use as the solvent in forming liposomes. After distillation, 0.7% ethanol can be added as a preservative. Ethanol and methanol are other suitable solvents.
  • the lipid solution used to form liposomes is placed in a round-bottomed flask.
  • Pear-shaped boiling flasks are preferred, particularly those flasks sold by Lurex Scientific (Vineland, N.J., cat. no. JM-5490).
  • the volume of the flask should be more than ten times greater than the volume of the anticipated aqueous suspension of liposomes to allow for proper agitation during liposome formation.
  • solvent is removed at 37° C. under negative pressure for 10 minutes with a filter aspirator attached to a water faucet.
  • the flask is further dried under low vacuum (i.e., less than 50 mm Hg) for 1 hour in a dessicator.
  • an aqueous solution containing antigen may be added to lyophilized liposome lipids in a volume that results in a concentration of approximately 200 mM with respect to liposome lipid, and shaken or vortexed until all the dried liposome lipids are wet.
  • the liposome-antigen mixture may then be incubated for 18 hours to 72 hours at 4° C.
  • the liposome-antigen formulation may be used immediately or stored for several years. It is preferred to employ such a formulation directly in the transcutaneous immunization system without removing unencapsulated antigen. Techniques such as bath sonication may be employed to decrease the size of liposomes, which may augment transcutaneous immunization.
  • Liposomes may be formed as described above but without addition of antigen to the aqueous solution. Antigen may then be added to the pre-formed liposomes and, therefore, antigen would be in solution and/or associated with, but not encapsulated by, the liposomes. This process of making a liposome-containing formulation is preferred because of its simplicity. Techniques such as bath sonication may be employed to alter the size and/or lamellarity of the liposomes to enhance immunization.
  • liposomes have been used as carriers with adjuvants to enhance the immune response to antigens mixed with, encapsulated in, attached to, or associated with liposomes.
  • Efficient immunization can be achieved with the present invention because transcutaneous delivery of antigen may target the Langerhans cell. These cells are found in abundance in the skin and are efficient antigen presenting cells leading to T-cell memory and potent immune responses (Udey, 1997). Because of the presence of large numbers of Langerhans cells in the skin, the efficiency of transcutaneous delivery may be related to the surface area exposed to antigen and adjuvant. In fact, the reason that transcutaneous immunization is so efficient may be that it targets a larger number of these efficient antigen presenting cells than intramuscular immunization.
  • transcutaneous immunization does not involve penetration of the skin and the complications and difficulties thereof, the requirements of trained personnel, sterile technique, and sterile equipment are reduced. Furthermore, the barriers to immunization at multiple sites or to multiple immunizations are diminished. Immunization by a single application of the formulation is also envisioned.
  • Immunization may be achieved using epicutaneous application of a simple solution of antigen and adjuvant impregnated in gauze under an occlusive patch, or by using other patch technologies; creams, immersion, ointments and sprays are other possible methods of application.
  • the immunization could be given by untrained personnel, and is amenable to self-application. Large-scale field immunization could occur given the easy accessibility to immunization. Additionally, a simple immunization procedure would improve access to immunization by pediatric patients and the elderly, and populations in Third World countries.
  • the transcutaneous delivery system of the invention is also not concerned with penetration of intact skin by sound or electrical energy.
  • Such a system that uses an electrical field to induce dielectric breakdown of the stratum corneum is disclosed in U.S. Pat. No. 5,464,386.
  • transcutaneous immunization may be superior to immunization using needles as more immune cells would be targeted by the use of several locations targeting large surface areas of skin.
  • a therapeutically effective amount of antigen sufficient to induce an immune response may be delivered transcutaneously either at a single cutaneous location, or over an area of intact skin covering multiple draining lymph node fields (e.g., cervical, axillary, inguinal, epitrochelear, popliteal, those of the abdomen and thorax).
  • lymph node fields e.g., cervical, axillary, inguinal, epitrochelear, popliteal, those of the abdomen and thorax.
  • Antigen passing through or into the skin may encounter antigen presenting cells which process the antigen in a way that induces an immune response. Multiple immunization sites may recruit a greater number of antigen presenting cells and the larger population of antigen presenting cells that were recruited would result in greater induction of the immune response. It is conceivable that absorption through the skin may deliver antigen to phagocytic cells of the skin such as, for example, dermal dendritic cells, macrophages, and other skin antigen presenting cells; antigen may also be delivered to phagocytic cells of the liver, spleen, and bone marrow that are known to serve as the antigen presenting cells through the blood stream or lymphatic system. The result would be widespread distribution of antigen to antigen presenting cells to a degree that is rarely, if ever achieved, by current immunization practices.
  • the transcutaneous immunization system may be applied directly to the skin and allowed to air dry; rubbed into the skin or scalp; held in place with a dressing, patch, or absorbent material; otherwise held by a device such as a stocking, slipper, glove, or shirt; or sprayed onto the skin to maximize contact with the skin.
  • the formulation may be applied in an absorbant dressing or gauze.
  • the formulation may be covered with an occlusive dressing such as, for example, AQUAPHOR (an emulsion of petrolatum, mineral oil, mineral wax, wool wax, panthenol, bisabol, and glycerin from Beiersdorf, Inc.), plastic film, COMFEEL (Coloplast) or vaseline; or a non-occlusive dressing such as, for example, DUODERM (3M) or OPSITE (Smith & Napheu).
  • An occlusive dressing completely excludes the passage of water.
  • the formulation may be applied to single or multiple sites, to single or multiple limbs, or to large surface areas of the skin by complete immersion.
  • the formulation may be applied directly to the skin.
  • the nucleic acid(s) contained in the formulation may encode the antigen, the adjuvant, or both.
  • the nucleic acid may or may not be capable of replication; it may be non-integrating and non-infectious.
  • the nucleic acid may further comprise a regulatory region (e.g., promoter, enhancer, silencer, transcription initiation and termination sites, RNA splice acceptor and donor sites, polyadenylation signal, internal ribosome binding site, translation initiation and termination sites) operably linked to the sequence encoding the antigen or adjuvant.
  • the nucleic acid may be complexed with an agent that promotes transfection such as cationic lipid, calcium phosphate, DEAE-dextran, polybrene-DMSO, or a combination thereof.
  • the nucleic acid may comprise regions derived from viral genomes. Such materials and techniques are described by Kriegler (1990) and Murray (1991).
  • An immune response may comprise humoral (i.e., antigen-specific antibody) and/or cellular (i.e., antigen-specific lymphocytes such as B cells, CD4 + T cells, CD8 + T cells, CTL, Th1 cells, Th2 cells, and/or T DTH cells) effector arms.
  • the immune response may comprise NK cells that mediate antibody-dependent cell-mediated cytotoxicity (ADCC)
  • the immune response induced by the formulation of the invention may include the elicitation of antigen-specific antibodies and/or cytotoxic lymphocytes (CTL, reviewed in Alving and Wassef, 1994).
  • Antibody can be detected by immunoassay techniques, and the detection of various isotypes (e.g., IgM, IgD, IgA1, IgA2, secretory IgA, IgE, IgG1, IgG2, IgG3, or IgG4) may be expected.
  • An immune response can also be detected by a neutralizing assay.
  • Antibodies are protective proteins produced by B lymphocytes. They are highly specific, generally targeting one epitope of an antigen. Often, antibodies play a role in protection against disease by specifically reacting with antigens derived from the pathogens causing the disease. Immunization may induce antibodies specific for the immunizing antigen, such as cholera toxin. These antigen-specific antibodies are induced when antigen is delivered through the skin by liposomes.
  • CTLs are particular protective immune cells produced to protect against infection by a pathogen. They are also highly specific. Immunization may induce CTLs specific for the antigen, such as a synthetic oligopeptide based on a malaria protein, in association with self-major histocompatibility antigen. CTLs induced by immunization with the transcutaneous delivery system may kill pathogen infected cells. Immunization may also produce a memory response as indicated by boosting responses in antibodies and CTLs, lymphocyte proliferation by culture of lymphocytes stimulated with the antigen, and delayed type hypersensitivity responses to intradermal skin challenge of the antigen alone.
  • the antigen such as a synthetic oligopeptide based on a malaria protein
  • serial dilutions of sera are added to host cells which are then observed for infection after challenge with infectious virus.
  • serial dilutions of sera may be incubated with infectious titers of virus prior to innoculation of an animal, and the innoculated animals are then observed for signs of infection.
  • the transcutaneous immunization system of the invention may be evaluated using challenge models in either animals or humans, which evaluate the ability of immunization with the antigen to protect the subject from disease. Such protection would demonstrate an antigen-specific immune response.
  • achieving anti-diphtheria antibody titers of 5 IU/ml or greater is generally assumed to indicate optimum protection and serves as a surrogate marker for protection (Plotkin and Mortimer, 1994).
  • the Plasmodium faciparum challenge model may be used as to induce an antigen-specific immune response in humans.
  • Human volunteers may be immunized using the transcutaneous immunization system containing oligopeptides or proteins (polypeptides) derived from the malaria parasite, and then exposed to malaria experimentally or in the natural setting.
  • the Plasmodium yoelii mouse malaria challenge model may be used to evaluate protection in the mouse against malaria (Wang et al., 1995).
  • CSP circumsporozoite protein
  • merozoite surface proteins of Plasmodium falciparum have been encapsulated in liposomes containing lipid A.
  • All of the malaria antigens that have been encapsulated in liposomes containing lipid A have been shown to induce humoral effectors (i.e., antigen-specific antibodies), and some have been shown to induce cell-mediated responses as well.
  • Generation of an immune response and immunoprotection in an animal vaccinated with a malaria antigen may be assayed by immunofluorescence to whole, fixed malaria sporozoites or CTLs killing of target cells transfected with CSP.
  • mice may be transcutaneously immunized with cholera toxin, and then challenged intranasally with an LD 70 (40 ⁇ g) dose of cholera toxin and observed for protection.
  • LD 70 40 ⁇ g
  • Mallet et al. personal communication
  • C57BL/6 mice develop a fatal hemorrhagic pneumonia in response to intranasal challenge with CT.
  • the mice may be challenged with an intraperitoneal dose of CT (Dragunsky et al., 1992).
  • Cholera toxin-specific IgG or IgA antibody may provide protection against cholera toxin challenge (Pierce, 1978; Pierce and Reynolds, 1974).
  • Vaccination has also been used as a treatment for cancer and autoimmune disease.
  • a tumor antigen e.g., prostate specific antigen
  • vaccination with a tumor antigen may induce an immune response in the form of antibodies, CTLs and lymphocyte proliferation which allows the body's immune system to recognize and kill tumor cells.
  • Tumor antigens useful for vaccination have been described for melanoma (U.S. Pat. Nos. 5,102,663, 5,141,742, and 5,262,177), prostate carcinoma (U.S. Pat. No. 5,538,866), and lymphoma (U.S. Pat. Nos. 4,816,249, 5,068,177, and 5,227,159).
  • Vaccination with T-cell receptor oligopeptide may induce an immune response that halts progression of autoimmune disease (U.S. Pat. Nos. 5,612,035 and 5,614,192; Antel et al., 1996; Vandenbark et al., 1996).
  • U.S. Pat. No. 5,552,300 also describes antigens suitable for treating autoimmune disease.
  • mice of 6 to 8 weeks were shaved with a #40 clipper. This shaving could be done without any signs of trauma to the skin. The shaving was done from the mid-thorax to just below the nape of the neck. The mice were then allowed to rest for 24 hours. Prior to this the mice had been ear-tagged for identification, and pre-bled to obtain a sample of pre-immune serum. Mice were also transcutaneously immunized without shaving by applying 50 ⁇ l of immunizing solution to each ear.
  • mice were then immunized in the following way. Mice were anesthetized with 0.03-0.06 ml of a 20 mg/ml solution of xylazine and 0.5 ml of 100 mg/ml ketamine; mice were immobilized by this dose of anesthesia for approximately one hour. The mice were placed ventral side down on a warming blanket.
  • the immunizing solution was then placed on the dorsal shaved skin of a mouse in the following manner: a 1.2 cm ⁇ 1.6 cm stencil made of polystyrene was laid gently on the back and a saline-wetted sterile gauze was used to partially wet the skin (this allowed even application of the immunizing solution), the immunizing solution was then applied with a pipet to the area circumscribed by the stencil to yield a 2 cm 2 patch of immunizing solution. Care was used not to scrape or rub the skin with the pipet tip. The immunizing solution was spread around the area to be covered with the smooth side of the pipet tip.
  • the immunizing solution (between about 100 ⁇ l to about 200 ⁇ l) was left on the back of the mouse for 60 minutes. At the end of 60 minutes, the mouse was held gently by the nape of the neck and the tail under a copious stream of lukewarm tap water, and washed for 10 seconds. The mouse was then gently patted dry with a piece of sterile gauze and a second washing was performed for 10 seconds; the mouse was then patted dry a second time and left in the cage. The mice appeared to exhibit no adverse effects from the anesthesia, immunization, washing procedure, or toxicity from the exotoxins. No skin irritation, swelling or redness was seen after the immunization and the mice appeared to thrive. Immunization using the ear was performed as described above except that fur was not removed prior to immunization.
  • Antibodies specific for CT, LT, ETA, pertussis toxin, diphtheria toxoid, tetanus toxoid, Hemophilus influenza B conjugate, and BSA were determined using ELISA in a technique similar to Glenn et al. (1995). All antigens were dissolved in sterile saline at a concentration of 2 ⁇ g/ml. Fifty microlilters of this solution (0.1 ⁇ g) per well was put on IMMULON-2 polystyrene plates (Dynatech Laboratories, Chantilly, Va.) and incubated at room temperature overnight. The plates were then blocked with a 0.5% casein/0.05% Tween 20 blocking buffer solution for one hour. Sera was diluted with 0.5% casein/0.05% Tween 20 diluent; dilution series were done in columns on the plate. Incubation was for 2 hours at room temperature.
  • the plates were then washed in a PBS-0.05% Tween 20 wash solution four times, and goat anti-mouse IgG(H+L) horseradish peroxidase (HRP)-linked (Bio-Rad Laboratories, Richmond, Calif., Cat #170-6516) secondary antibody was diluted in casein diluent at a dilution of 1/500 and left on the plates for one hour at room temperature.
  • the plates were then washed four times in the PBS-Tween wash solution.
  • IgG( ⁇ ), IgM( ⁇ ) and IgA( ⁇ ) anti-CT antibody levels were determined using ELISA with a technique similar to Glenn et al. (1995).
  • CT was dissolved in sterile saline at a concentration of 2 ⁇ g/ml.
  • Fifty microliters of this solution (0.1 ⁇ g) per well were put on IMMULON-2 polystyrene plates (Dynatech Laboratories, Chantilly, Va.) and incubated at room temperature overnight. The plates were then blocked with a 0.5% casein-Tween 20 blocking buffer solution for one hour.
  • Sera was diluted and casein diluent and serial dilutions were done on the plate. This was incubated for two hours at room temperature.
  • the plates were then washed in a PBS-Tween wash solution four times and goat anti-mouse IgG( ⁇ ) HRP-linked (Bio-Rad Laboratories, Richmond, Calif., Cat #172-1038), goat anti-mouse IgM( ⁇ ) HRP-linked (BioRad Laboratories, Richmond, Calif., Cat #172-1030), or goat anti-mouse IgA HRP-linked (Sigma, St. Louis, Mo., Cat #1158985) secondary antibody was diluted in casein diluent in a dilution of 1/1000 and left on the plates for one hour at room temperature. The plates were then washed four times in a PBS-Tween wash solution.
  • Antigen-specific IgG (IgG1, IgG2a, IgG2b, and IgG3) subclass antibody against CT, LT, ETA, and BSA was performed as described by Glenn et al. (1995).
  • the solid phase ELISA was performed in IMMULON-2 polystyrene plates (Dynatech Laboratories, Chantilly, Va.). Wells were incubated with the respective antigens in saline overnight (0.1 ⁇ g/50 ⁇ l) and blocked with 0.5% casein-Tween 20. Individual mouse sera diluted in 0.5% casein were serially diluted, and incubated at room temperature for four hours.
  • Secondary antibody consisted of horseradish peroxidase-conjugated goat anti-mouse isotype-specific antibody (IgG1, IgG2a, IgG2b, IgG3, The Binding Site, San Diego, Calif.).
  • a standard curve for each subclass was determined using mouse myeloma IgG1, IgG2a, IgG2b, and IgG3 (The Binding Site, San Diego, Calif.).
  • Standard wells were coated with goat anti-mouse IgG(H+L) (Bio-Rad Laboratories, Richmond, CA, Cat #172-1054) to capture the myeloma IgG subclass standards which were added in serial dilutions.
  • the myeloma IgG subclass was also detected using the peroxidase-conjugated goat anti-mouse subclass-specific antibody. Both the test sera and myeloma standards were detected using 2,2′-azino-di(3-ethyl-benzthiazolone) sulphonic acid (Kirkegaard and Perry, Gaithersburg, Md.) as substrate. Absorbances were read at 405 nm. Individual antigen specific subclasses were quantitated using the values from the linear titration curve computed against the myeloma standard curve and reported as ⁇ g/ml.
  • Antigen-specific IgE antibody quantitation was performed using a protocol from Pharmingen Technical Protocols, page 541 of the Research Products Catalog, 1996-1997 (Pharmingen, San Diego, Calif.). Fifty microliters of 2 ⁇ g/ml purified anti-mouse IgE capture mAb (Pharmingen, Cat# 02111D) in 0.1 M NaHCO 3 (pH 8.2) were added to IMMUNO plates(Nunc, Cat #12-565-136). Plates were incubated overnight at room temperature, washed three times with PBS-Tween 20, blocked with 3% BSA in PBS for two hours, and washed three times with PBS-Tween.
  • Sera were diluted in 1% BSA in PBS, added at dilutions of 1/100, and diluted serially down the columns (e.g., 1/100, 1/200, et cetera).
  • Purified mouse IgE standards (Pharmingen, Cat # 0312D) were added with a starting dilution of 0.25 ⁇ g/ml and serially diluted down the columns. Plates were incubated for two hours and washed five times with PBS-Tween.
  • Biotinylated anti-mouse IgE mAB (Pharmingen, Cat #02122D) to 2 ⁇ g/ml in 1% BSA in PBS, incubated for 45 minutes and washed five times with PBS-Tween.
  • Avidin-peroxidase (Sigma A3151, 1:400 of 1 mg/ml solution) was added for 30 min and plates were washed six times with PBS-Tween. Both the test sera and IgE standards were detected using 2,2′-azino-di(3-ethyl-benzthiazolone)sulphonic acid (Kirkegaard and Perry, Gaithersburg, Md.) as substrate.
  • Absorbances were read at 405 nm. Individual antigen specific subclasses were quantitated using the values from the linear titration curve computed against the IgE standard curve and reported as ⁇ g/ml.
  • multilamellar liposomes composed of dimyristoyl phosphatidyl choline, dimyristoyl phosphatidyl glycerol, cholesterol were prepared according to Alving et al. (1993). Dimyristoyl phosphatidylcholine, dimyristoyl phosphatidylglycerol, and cholesterol were purchased from Avanti Polar Lipids Inc. (Alabaster, Ala.). Stock solutions of the lipids in chloroform were removed from ⁇ 20° C. freezer where they were stored.
  • the lipids were mixed in a molar ratio of 0.9:0.1:0.75 dimyristoyl phosphatidyl choline, dimyristoyl phosphatidyl glycerol, and cholesterol in a pear shaped flask. Using a rotary evaporator, the solvent was removed at 37° C. under negative pressure for 10 minutes. The flask was further dried under low vacuum for two hours in a dessicator to remove residual solvent. The liposomes were swollen at 37 mM phospholipid using sterile water, lyophilized and stored at ⁇ 20° C.
  • liposomes were mixed in their lyophilized state with normal saline (pH 7.0) to achieve a designated phospholipid concentration in the saline.
  • the dried lipids were swollen to make liposomes with normal saline (pH 7.0) and were not lyophilized.
  • mice at 6 to 8 weeks of age were immunized transcutaneously as described above for “Immunization Procedure”, in groups of five mice.
  • the mice were immunized using 100 ⁇ l of immunization solution which was prepared as follows: liposomes prepared as described above for “Liposome Preparation” were mixed with saline to form the liposomes.
  • the pre-formed liposomes were then diluted in either saline (liposome alone group) or with CT in saline to yield an immunizing solution containing liposomes at 10-150 mM phospholipid with 100 ⁇ g of CT per 100 ⁇ l of immunizing solution.
  • CT was mixed in saline to make an immunizing solution containing 100 ⁇ g of CT per 100 ⁇ g of solution for the group receiving CT alone. Solutions were vortexed for 10 seconds prior to immunization.
  • mice were immunized transcutaneously at 0 and 3 weeks.
  • Antibody levels were determined using ELISA as described above for “ELISA IgG(H+L)” 3 weeks after the boosting immunization, and compared against pre-immune sera.
  • Table 1 the level of anti-CT antibodies induced by CT without liposomes was not different from the level of anti-CT antibodies generated using liposomes except in the mice where 150 mM liposomes were used.
  • CT in saline alone was able to immunize mice against CT to produce high antibody titers.
  • mice at 6 to 8 weeks of age were immunized transcutaneously as described above for “Immunization Procedure”, in groups of five mice.
  • the mice were immunized at 0 and 3 weeks using 100 ⁇ l of immunization solution prepared as follows: BSA was mixed in saline to make an immunizing solution containing 200 ⁇ g of BSA per 100 ⁇ l of saline for the group receiving BSA alone; BSA and CT were mixed in saline to make an immunizing solution containing 200 ⁇ g of BSA and 100 ⁇ g of CT per 100 ⁇ l of saline for the group receiving BSA and CT.
  • the liposomes were prepared as described above for “Liposome Preparation”, and were first mixed with saline to form the liposomes. They were then diluted in BSA or BSA and CT in saline to yield an immunizing solution containing liposomes at 50 mM phospholipid with 200 ⁇ g of BSA per 100 ⁇ l of immunizing solution, or 200 ⁇ g BSA+100 ⁇ g CT per 100 ⁇ l of immunizing solution. Solutions were vortexed for 10 seconds prior to immunization.
  • mice at 6 to 8 weeks of age were immunized transcutaneously as described above for “Immunization Procedure”, in groups of five mice.
  • the mice were immunized at 0 and 3 weeks using 100 ⁇ l of immunization solution prepared as follows: LT was mixed in saline to make an immunizing solution containing 100 ⁇ g of LT per 100 ⁇ l of saline for the group receiving LT alone.
  • liposomes were used the liposomes prepared as described above for “Liposome Preparation”, and were first mixed with saline to form the liposomes.
  • the pre-formed liposomes were then diluted in LT in saline to yield an immunizing solution containing liposomes at 50 mM phospholipid with 100 ⁇ g of LT per 100 ⁇ l of immunizing solution. Solutions were vortexed for 10 seconds prior to immunization.
  • LT and CT are members of the family of bacterial ADP-ribosylating exotoxins (bAREs). They are organized as A:B proenzymes with the ADP-ribosyltransferase activity contained in the A subunit and the target cell binding a function of the B subunit.
  • LT is 80% homologous with CT at the amino acid level and has a similar non-covalently bound subunit organization, stoichiometry (A:B5), the same binding target, ganglioside GM1, and is similar in size (MW ⁇ 80,000).
  • A:B5 non-covalently bound subunit organization
  • ganglioside GM1 the same binding target
  • MW ⁇ 80,000 the same binding target
  • mice at 6 to 8 weeks of age were immunized transcutaneously as described above for “Immunization Procedure”, in groups of five mice.
  • the mice were immunized once using 100 ⁇ l of immunization solution prepared as follows: LT was mixed in saline to make an immunizing solution containing 100 ⁇ g of LT per 100 ⁇ l of saline. The solution was vortexed for 10 seconds prior to immunization.
  • the anti-LT antibodies were determined using ELISA as described above for “ELISA IgG (H+L)” 3 weeks after the single immunization. The results are shown in Table 4. LT was clearly immunogenic with a single immunization and antibodies were produced by 3 weeks. Rapid enhancement of antibody titers and responses to single immunization would be a useful aspect of the transcutaneous immunization method. It is conceivable that a rapid single immunization would be useful in epidemics, for travelers, and where access to medical care is poor. TABLE 4 Anti-LT antibodies Mouse Number ELISA Units 5141 6,582 5142 198 5143 229 5144 6,115 5145 17,542 Geo Mean 2,000
  • C57BL6 mice at 8 to 12 weeks of age were immunized transcutaneously as described above for “Immunization Procedure”, in groups of five mice.
  • the mice were immunized once using 100 ⁇ l of immunization solution prepared as follows: CT was mixed in saline to make an immunizing solution containing 100 ⁇ g of CT per 100 ⁇ l of saline. The solution was vortexed for 10 seconds prior to immunization.
  • the anti-CT antibodies were determined using ELISA as described above for “ELISA IgG (H+L)” 3 weeks after the single immunization. The results are shown in Table 5.
  • CT was highly immunogenic with a single immunization. Rapid enhancement of antibody titers and responses to single immunication may be a useful aspect of the transcutaeous immunization method. It is conceivable that a rapid single immunization would be useful in epidemics, for travelers, and where access to medical care is poor.
  • TABLE 5 Anti-CT antibodies Mouse Number ELISA Units 2932 18,310 2933 30,878 2934 48,691 2935 7,824 Geo Mean 21,543
  • mice at 6 to 8 weeks of age were immunized transcutaneously as described above for “Immunization Procedure”, in groups of five mice.
  • the mice were immunized at 0 and 3 weeks using 100 ⁇ l of immunization solution prepared as follows: ETA was mixed in saline to make an immunizing solution containing 100 ⁇ g of ETA per 100 ⁇ l of saline for the group receiving ETA alone.
  • the liposomes were prepared as described above for “Liposome Preparation”, and were first mixed with saline to form the liposomes.
  • the pre-formed liposomes were then diluted with ETA in saline to yield an immunizing solution containing liposomes at 50 mM phospholipid with 100 ⁇ g of ETA per 100 ⁇ l of immunizing solution. Solutions were vortexed for 10 seconds prior to immunization.
  • ETA was clearly immunogenic both with and without liposomes, and no significant difference between the groups could be detected.
  • ETA differs from CT and LT in that ETA is a single 613 amino acid peptide with A and B domains on the same peptide and binds to an entirely different receptor, the ⁇ 2-macroglobulin receptor/low density lipoprotein receptor-related protein (Kounnas et al., 1992).
  • ETA also induced a transcutaneous antibody response.
  • mice at 6 to 8 weeks of age were immunized transcutaneously as described above for “Immunization Procedure”, in groups of five mice.
  • the mice were immunized using 100 ⁇ l of immunization solution which was prepared as follows: CT was mixed in saline to make 100 ⁇ g of CT per 100 ⁇ l of immunizing solution, LT was mixed in saline to make 100 ⁇ g of LT per 100 ⁇ l of immunizing solution, ETA was mixed in saline to make 100 ⁇ g of ETA per 100 ⁇ l of immunizing solution, and CT and BSA were mixed in saline to make 100 ⁇ g of CT per 100 ⁇ l of immunizing solution and 200 ⁇ g of BSA per 100 ⁇ l of immunizing solution. Solutions were vortexed for 10 seconds prior to immunization.
  • mice were immunized transcutaneously at 0 and 3 weeks and the antibody levels were determined using ELISA as described above for “ELISA IgG Subclass”, three weeks after the boosting immunization and compared against the pre-immune sera.
  • the IgG subclass response to CT, BSA and LT had similar levels of IgG1 and IgG2a reflecting activation of T help from both Th1 and Th2 lymphocytes (Seder and Paul, 1994), whereas the IgG subclass response to ETA consisted of almost exclusively IgG1 and IgG3, consistent with a Th2-like response (Table 7). Thus, it appears that all IgG subclasses can be produced using transcutaneous immunization.
  • mice at 6 to 8 weeks of age were immunized transcutaneously as described above for “Immunization Procedure”, in groups of five mice.
  • the mice were immunized using 100 ⁇ l of immunization solution which was prepared as follows: LT was mixed in saline to make an immunizing solution containing 100 ⁇ g of LT per 100 ⁇ l of saline for the group receiving LT alone, CT was mixed in saline to make an immunizing solution containing 100 ⁇ g of CT per 100 ⁇ l of saline for the group receiving CT alone, ETA was mixed in saline to make an immunizing solution containing 100 ⁇ g of ETA per 100 ⁇ l of saline for the group receiving ETA alone, and BSA and CT were mixed in saline to make an immunizing solution containing 100 ⁇ g of BSA and 100 ⁇ g of CT per 100 ⁇ l of saline for the group receiving BSA and CT.
  • mice were immunized transcutaneously at 0 and 3 weeks and the antibody levels were determined using ELISA as described above for “ELISA IgE”, one week after the boosting immunization and compared against the pre-immune sera. As shown in Table 8, no IgE antibodies were found although the sensitivity of detection was 0.003 ⁇ g/ml.
  • IgG antibodies were determined in the same mice using “ELISA IgG(H+L)” on sera 3 weeks after the second immunization. The IgG antibody response to LT, ETA, CT and BSA are shown to indicate that the animals were successfully immunized and responded with high titers of antibodies to the respective antigens.
  • mice at 6 to 8 weeks of age immunized transcutaneously as described above for “Immunization Procedure”, in groups of five mice.
  • the mice were immunized at 0 and 3 weeks using 100 ml of immunization solution which was prepared as follows: CT was mixed in saline to make an immunizing solution containing 100 mg of CT per 100 ml of immunizing solution. The immunization solution was vortexed for 10 seconds prior to immunization.
  • mice were immunized transcutaneously at 0 and 3 weeks and the antibody levels were determined using ELISA as described above for “ELISA IgG(H+L)” and “ELISA IgG( ⁇ )”. Determinations were done at 1 and 4 weeks after the initial immunization, and compared against the pre-immune sera. As shown in Table 9, high levels of anti-CT IgG( ⁇ ) antibodies were induced by CT in saline. Small amounts of IgM could be detected by using IgM( ⁇ ) specific secondary antibody. By 4 weeks, the antibody response was primarily IgG. Data are reported in ELISA units. TABLE 9 IgG( ⁇ ) and IgM( ⁇ ) Imm. Group Week IgG( ⁇ ) IgM( ⁇ ) CT 1 72 168 CT 4 21,336 38 L( ) + CT 1 33 38 L( ) + CT 4 22,239 70
  • mice at 6 to 8 weeks of age were immunized transcutaneously as described above for “Immunization Procedure”, in groups of five mice.
  • the mice were immunized once using 100 ⁇ l of immunization solution prepared as follows: CT was mixed in saline to make an immunizing solution containing 100 ⁇ g of CT per 100 ⁇ l of saline. The solution was vortexed for 10 seconds prior to immunization.
  • the mice were immunized transcutaneously at 0 and 3 weeks.
  • Antibody levels were determined using ELISA as described above for “ELISA IgG (H+L)” 5 weeks after the boosting immunization, and compared against pre-immune sera. As shown in Table 10, serum anti-CT IgA antibodies were detected.
  • TABLE 10 Anti-CT IgA antibodies Mouse Number IgA (ng/ml) 1501 232 1502 22 1503 41 1504 16 1505 17
  • mice at 6 to 8 weeks of age were immunized transcutaneously as described above for “Immunization Procedure”, in groups of five mice.
  • the mice were immunized using 100 ⁇ l of immunization solution which was prepared as follows: CT was mixed in saline to make an immunizing solution containing 100 ⁇ g of CT per 100 ⁇ l of immunizing solution. The immunization solution was vortexed for 10 seconds prior to immunization.
  • mice were immunized with 100 ⁇ l of immunizing solution transcutaneously at 0 and 3 weeks and the antibody levels were determined using ELISA as described above for “ELISA IgG(H+L)” and “ELISA IgG( ⁇ )”. Antibody determinations were done at 8 weeks after the initial immunization and compared against the pre-immune sera. As shown in Table 11, high levels of serun anti-CT antibodies were induced by CT in saline. Lung wash IgG could be detected by ELISA using IgG(H+L) or IgG( ⁇ ) specific antibody. The antibody found on the lung mucosal surface is diluted by the lavage method used to collect mucosal antibody and, thus, the exact amounts of antibody detected are not as significant as the mere presence of detectable antibody.
  • Lung washes were obtained after sacrificing the mouse.
  • the trachea and lungs were exposed by gentle dissection and trachea was transected above the bifurcation.
  • a 22 gauge polypropylene tube was inserted and tied off on the trachea to form a tight seal at the edges.
  • Half a milliliter of PBS was infused using a 1 ml syringe attached to the tubing and the lungs were gently inflated with the fluid. The fluid was withdrawn and reinfused for a total of 3 rounds of lavage.
  • the lung wash was then frozen at ⁇ 20° C.
  • Table 11 shows the IgG(H+L) and IgG( ⁇ ) antibody response to cholera toxin in the sera and lung washes at 8 weeks. Data are expressed in ELISA units. Antibodies were clearly detectable for all mice in the lung washes. The presence of antibodies in the mucosa may be important for protection against mucosally active diseases. TABLE 11 Mucosal Antibody to CT Animal# Imm.
  • mice were immunized transcutaneously at 0 and 3 weeks as described above for “Immunization Procedure”, in groups of four mice.
  • Liposomes were prepared as described above for “Liposome Preparation”, and were first mixed with saline to form the liposomes. The pre-formed liposomes were then diluted with either CT, CTA or CTB in saline to yield an immunizing solution containing liposomes at 50 mM phospholipid with 50 ⁇ g of antigen (CT, CTA or CTB) per 100 ⁇ l of immunizing solution. Solutions were vortexed for 10 seconds prior to immunization.
  • mice were immunized transcutaneously as described above for “Immunization Procedure”, in groups of five mice. Mice were immunized at 0 and 3 weeks with 100 ⁇ g of diphtheria toxoid and 10 ⁇ g of pertussis toxin per 100 ⁇ l of saline solution. Solutions were vortexed for 10 seconds prior to immunization.
  • the antibodies were quantitated using ELISA as described for “ELISA IgG(H+L)”.
  • Anti-diphtheria toxoid antibodies were detected only in animals immunized with both pertussis toxin and diphtheria toxoid. The highest responder had anti-diphtheria toxoid antibody ELISA units of 1,038.
  • a small amount of pertussis toxin acts as an adjuvant for diphtheria toxoid antigen.
  • the toxoid alone did not induce an immune response suggesting that the toxoiding process has affected the portion of the molecule responsible for the adjuvant effects found in the ADP-ribosylating exotexin.
  • mice were immunized transcutaneously as described above for “Immunization Procedure”, in groups of five mice. Mice were immunized once at 0 weeks with 50 ⁇ g of pertussis toxin per 100 ⁇ l of saline solution. The solution was vortexed for 10 seconds prior to immunization.
  • mice were immunized transcutaneously as described above for “Immunization Procedure”, in groups of five mice. Mice were immunized once at 0 weeks with 50 ⁇ g of tetanus toxoid and 100 ⁇ g of cholera toxin per 100 ⁇ l of saline solution. The solution was vortexed for 10 seconds prior to immunization.
  • the antibodies were quantitated using ELISA as described for “ELISA IgG(H+L)”. Anti-tetanus toxoid antibodies were detected at 8 weeks in animal 5173 at 443 ELISA units.
  • mice were anesthetized, transcutaneously immunized as described above for “Immunization Procedure” with 100 ⁇ g of 125 I-labeled CT (150,000 cpm/ ⁇ g CT). Control mice remained anesthetized for 6 hours to exclude grooming, and experimental mice were anesthetized for one hour and then allowed to groom after washing. Mice were sacrificed at 6 hours and organs weighed and counted for 125I on a Packard gamma counter.
  • a total of 2-3 ⁇ g of CT was detected on the shaved skin at the site of immunization (14,600 cpm/ ⁇ g tissue) while a maximum of 0.5 ⁇ g of CT was detected in the stomach (661 cpm/ ⁇ g tissue) and intestine (9 cpm/ ⁇ g tissue).
  • Induction of an immune response to orally fed CT requires the addition of NaHCO 3 to the immunizing solution (Piece, 1978; Lycke and Holmgren, 1986).
  • oral immunization does not significantly contribute to the antibodies detected when CT is applied epicutaneously to the skin.
  • BALB/c mouse ears were coated on the dorsal side with either 100 ⁇ g of CT in saline, 100 ⁇ g of CTB in saline, saline alone, or an intradermal injection of the positive controls 100 ⁇ g LPS or 10 ⁇ g TNF ⁇ , for one hour while the mouse was anesthetized. The ears were then throughly washed and, after 24 hours, the ears were removed and epidermal sheets were harvested and stained for MHC class II expression as described by Caughman et al. (1986).
  • Epidermal sheets were stained with MKD6 (anti-I-A d ) or negative control Y3P (anti-I-A k ), and goat anti-mouse FITC F(ab) 2 was used as a second step reagent. Mice transcutaneously immunized on the ear (as described above without shaving) had previously been found to have anti-CT antibodies of 7,000 ELISA units three weeks after a single immunization.
  • Langerhans cells represent the epidermal contingent of a family of potent accessory cells termed ‘dendritic cells’. Langerhans cells (and perhaps related cells in the dermis) are thought to be required for immune responses directed against foreign antigens that are encountered in skin. The ‘life cycle’ of the Langerhans cell is characterized by at least two distinct stages. Langerhans cells in epidermis (the ‘sentinels’) can ingest particulates and process antigens efficiently, but are weak stimulators of unprimed T cells.
  • Langerhans cells that have been induced to migrate to lymph nodes after contact with antigen in epidermis are poorly phagocytic and have limited antigen-processing capabilities, but are potent stimulators of naive T cells. If Langerhans cells are to fulfill both their ‘sentinel‘ and ‘messenger’ roles, they must be able to persist in epidermis, and also be able to exit epidermis in a controlled fashion after exposure to antigen. Thus, regulation of Langerhans cell-keratinocyte adhesion represents a key control point in Langerhans cell trafficking and function.
  • Langerhans cells express E-cadherin (Blauvelt et al., 1995), a homophilic adhesion molecule that is prominently represented in epithelia. Keratinocytes also express this adhesion molecule, and E-cadherin clearly mediates adhesion of murine Langerhans cells to keratinocytes in vitro. It is known that E-cadherin is involved in the localization of Langerhans cells in epidermis. See Stingl et al. (1989) for a review of the characterization and properties of Langerhans cells and keratinocytes.
  • LC epidermal Langerhans cells
  • LC epidermal Langerhans cells
  • LC epidermal Langerhans cells
  • LC epidermal Langerhans cells
  • phenotypic changes required for their movement from the skin and acquisition of the capacity for antigen presentation.
  • adhesion molecules that regulate interactions with the surrounding tissue matrix and with T lymphocytes.
  • the migration of the Langerhan cell is known to be associated with a marked reduction in the expression of E-cadherin (Schwarzenberger and Udey, 1996, and a parallel upregulation of ICAM-1 (Udey, 1997).
  • Transcutaneous immunization with bacterial ADP ribosylating exotoxins target the Langerhans cells in the epidermis.
  • the bAREs activate the Langerhans cell, transforming it from its sentinel role to its messenger role. Ingested antigen is then taken to the lymph node where it is presented to B and T cells (Streilein and Grammer, 1989; Kripke et al., 1990; Tew et al., 1997).
  • the epidermal Langerhans cell matures into an antigen-presenting dendritic cell in the lymph node (Schuler and Steinman, 1985); lymphocytes entering a lymph node segregate into B-cell follicles and T-cell regions.
  • the activation of the Langerhans cell to become a migratory Langerhans cell is known to be associated with not only a marked increase in MHC class II molecules, but also marked reduction in the expression of E-cadherin, and upregulation of ICAM-1.
  • CT or CTB acts as an adjuvant by freeing the sentinel Langerhans cell to present antigens such as BSA or diphtheria toxoid phagocytosed by the Langerhans cell at the same location and time as the encounter with the CT or CTB when they are acting as adjuvant.
  • the activation of a Langerhans cells to upregulate the expression of ICAM-1 and dowregulate the expression of E-cadherin may be mediated by cytokine release including TNF ⁇ and IL-1 ⁇ from the epidermal cells or the Langerhans cells themselves.
  • This method of adjuvancy for transcutaneous immunization is envisioned to work for any compound that activates the Langerhans cell. Activation could occur in such manner as to downregulate the E-cadherin and upregulate ICAM-1. Langerhans cells would then carry antigens made of mixtures of such Langerhans cell-activating compounds and antigens (such as diphtheria toxoid or BSA) to the lymph nodes where the antigens are presented to T cells and evoke an immune response.
  • antigens made of mixtures of such Langerhans cell-activating compounds and antigens (such as diphtheria toxoid or BSA) to the lymph nodes where the antigens are presented to T cells and evoke an immune response.
  • the activating substance such as a bARE can be used as an adjuvant for an other wise transcutaneously non-immunogenic antigen such as Diphtheria toxoid by activating the Langerhans cell to phagocytose the antigen such as diphtheria toxoid, migrate to the lymph node, mature into a dendritic cell, and present the antigen to T cells.
  • an other wise transcutaneously non-immunogenic antigen such as Diphtheria toxoid by activating the Langerhans cell to phagocytose the antigen such as diphtheria toxoid, migrate to the lymph node, mature into a dendritic cell, and present the antigen to T cells.
  • the T-cell helper response to antigens used in transcutaneous immunization may be influenced by the application of cytokines and/or chemokines.
  • cytokines and/or chemokines For example, interleukin-10 (IL-10) may skew the antibody response towards a Th2 IgG1/IgE response whereas anti-IL-10 may enhance the production of IgG2a (Bellinghausen et al., 1996).

Abstract

A transcutaneous immunization system delivers antigen to immune cells without perforation of the skin, and induces an immune response in an animal or human. The system uses an adjuvant, preferably an ADP-ribosylating exotoxin, to induce an antigen-specific immune response (e.g., humoral and/or cellular effectors) after transcutaneous application of a formulation containing antigen and adjuvant to intact skin of the animal or human. The efficiency of immunization may be enhanced by adding hydrating agents (e.g., liposomes), penetration enhancers, or occlusive dressings to the transcutaneous delivery system. This system may allow activation of Langerhans cells in the skin, migration of the Langerhans cells to lymph nodes, and antigen presentation.

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • This is a continuation in-part of U.S. application Ser. No. 08/749,164 filed Nov. 14, 1996.
  • GOVERNMENT RIGHTS
  • The U.S. government may retain certain rights in this invention.
  • BACKGROUND
  • The invention relates to transcutaneous immunization, and adjuvants useful therein, to induce an antigen-specific immune response.
  • Transcutaneous immunization requires both passage of an antigen through the outer barriers of the skin, which are normally impervious to such passage, and an immune response to the antigen. We showed in U.S. application Ser. No. 08/749,164 that using cholera toxin as an antigen elicits a strong antibody response that is highly reproducible; the antigen could be applied in a saline solution to the skin, with or without liposomes. In the present application, we show transcutaneous immunization using adjuvants such as, for example, bacterial exotoxins, their subunits, and related toxins.
  • There is a report of transdermal immunization with transferosomes by Paul et al. (1995). In this publication, the transferosomes are used as a carrier for proteins (bovine serum albumin and gap junction proteins) against which the complement-mediated lysis of antigen-sensitized liposomes is directed. An immune response was not induced when solution containing the protein was placed on the skin; only transferosomes were able to transport antigen across the skin and achieve immunization. As discussed in U.S. application Ser. No. 08/749,164, transferosomes are not liposomes.
  • FIG. 1 of Paul et al. (1995) showed that only a formulation of antigen and transferosomes induced an immune response, assayed by lysis of antigen-sensitized liposomes. Formulations of antigen in solution, antigen and mixed micelles, and antigen and liposomes (i.e., smectic mesophases) applied to the skin did not induce an immune response equivalent to that induced by subcutaneous injection. Therefore, there was a positive control (i.e., antigen and transfersomes) to validate their negative conclusion that a formulation of antigen and liposomes did not cause transdermal immunization.
  • Paul et al. (1995) stated on page 3521 that the skin is an effective protective barrier that is “impenetrable to substances with a molecular mass at most 750 DA”, precluding non-invasive immunization with large immunogen through intact skin. Therefore, the reference would teach away from using a molecule like cholera toxin (which is 85,000 daltons) because such molecules would not be expected to penetrate the skin and, therefore, would not be expected to achieve immunization. Thus, skin represents a barrier that would make penetration by an adjuvant or antigen like cholera toxin unexpected without the disclosure of the present invention.
  • Paul and Cevc (1995) stated on page 145, “Large molecules normally do not get across the intact mammalian skin. It is thus impossible to immunize epicutaneously with simple peptide or protein solutions.” They concluded, “The dermally applied liposomal or mixed micellar immunogens are biologically as inactive as simple protein solutions, whether or not they are combined with the immunoadjuvant lipid A.”
  • Wang et al. (1996) placed a solution of ovalbumin (OVA) in water on the skin of shaved mice to induce an allergic type response as a model for atopic dermatitis. Mice were anesthetized and covered with an occlusive patch containing up to 10 mg of OVA, which was placed on the skin continuously for four days. This procedure was repeated after two weeks.
  • In FIG. 2 of Wang et al. (1996), an ELISA assay done to determine the IgG2a antibody response showed no IgG2a antibody response to OVA. However, IgE antibodies that are associated with allergic responses could be detected. In a further experiment, the mice were more extensively patched with OVA in solution for four days every two weeks. This was repeated five times, i.e., the mice wore patches for a total of 20 days. Again, the high dose of OVA did not produce significant IgG2a antibodies. Significant levels of IgE antibodies were produced.
  • The authors stated on page 4079 that “we established an animal model to show that epicutaneous exposure to protein Ag, in the absence of adjuvant, can sensitize animals and induce a dominant Th2-like response with high levels of IgE”. Extensive epicutaneous exposure to high doses of protein antigen could not produce significant IgG antibodies but could induce IgE antibodies, the hallmark of an allergic type reaction. Thus, Wang et al. (1996) teaches that OVA exposure as described is a model for atopic dermatitis and not a mode of immunization. Therefore, following the teaching of the reference, one would have expected that transcutaneous immunization with antigen would induce high levels of IgE antibodies if it were to pass through the skin and induce an immune response. Instead, we have unexpectedly found that antigen placed on the skin in a saline solution with adjuvant induces high levels of IgG and some IgA, but not IgE.
  • In contrast to the cited references, the inventors have found that application to the skin of antigen and adjuvant provides a transcutaneous delivery system for antigen that can induce an antigen-specific immune response of IgG or IgA. The adjuvant is preferably an ADP-ribosylating exotoxin. Optionally, hydration, penetration enhancers, or occlusive dressings may be used in the transcutaneous delivery system.
  • SUMMARY OF THE INVENTION
  • An object of the invention is to provide a system for transcutaneous immunization that induces an immune response (e.g., humoral and/or cellular effectors) in an animal or human. The system provides simple application to intact skin of an organism of a formulation comprised of antigen and adjuvant to induce a specific immune response against the antigen. In particular, the adjuvant may activate antigen presenting cells of the immune system (e.g., Langerhans cells in the epidermis, dermal dendritic cells, dendritic cells, macrophages, B lymphocytes) and/or induce the antigen presenting cells to phagocytose the antigen. The antigen presenting cells then present the antigen to T and B cells. In the instance of Langerhans cells, the antigen presenting cells then may migrate from the skin to the lymph nodes and present antigen to lymphocytes (e.g., B and/or T cells), thereby inducing an antigen-specific immune response.
  • In addition to eliciting immune reactions leading to generation of an antigen-specific B lymphocyte and/or T lymphocyte, including a cytotoxic T lymphocyte (CTL), another object of the invention is to positively and/or negatively regulate components of the immune system by using the transcutaneous immunization system to affect antigen-specific helper (Th1 and/or Th2) or delayed-type hypersensitivity (TDTH) T-cell subsets.
  • In a first embodiment of the invention, a formulation containing antigen and adjuvant is applied to intact skin of an organism, the antigen is presented to immune cells, and an antigen-specific immune response is induced without perforating the skin. The formulation may include additional antigens such that transcutaneous application of the formulation induces an immune response to multiple antigens. In such a case, the antigens may or may not be derived from the same source, but the antigens will have different chemical structures so as to induce immune responses specific for the different antigens. Antigen-specific lymphocytes may participate in the immune response and, in the case-of participation by B lymphocytes, antigen-specific antibodies may be part of the immune response.
  • In a second embodiment of the invention, the above method is used to treat an organism. If the antigen is derived from a pathogen, the treatment vaccinates the organism against infection by the pathogen or against its pathogenic effects such as those caused by toxin secretion. A formulation that includes a tumor antigen may provide a cancer treatment; a formulation that includes an autoantigen may provide a treatment for a disease caused by the organism's own immune system (e.g., autoimmune disease).
  • In a third embodiment of the invention, a patch for use in the above methods is provided. The patch comprises a dressing, and effective amounts of antigen and adjuvant. The dressing may be occlusive or non-occlusive. The patch may include additional antigens such that application of the patch induces an immune response to multiple antigens. In such a case, the antigens may or may not be derived from the same source, but the antigens will have different chemical structures so as to induce an immune response specific for the different antigens. For effective treatment, multiple patches may be applied at frequent intervals or constantly over a period of time.
  • Moreover, in a fourth embodiment of the invention, the formulation is applied to intact skin overlying more than one draining lymph node field using either single or multiple applications. The formulation may include additional antigens such that application to intact skin induces an immune response to multiple antigens. In such a case, the antigens may or may not be derived from the same source, but the antigens will have different chemical structures so as to induce an immune response specific for the different antigens.
  • The products and methods may be used therapeutically to treat existing disease, protectively to prevent disease, or to reduce the severity and/or duration of disease.
  • In addition to antigen and adjuvant, the formulation may comprise a hydrating agent (e.g., liposomes), a penetration enhancer, or both. For example, the formulation may comprise AQUAPHOR (an emulsion of petrolatum, mineral oil, mineral wax, wool wax, panthenol, bisabol, and glycerin), emulsions (e.g., aqueous creams), oil-in-water emulsions (e.g., oily creams), anhydrous lipids and oil-in-water emulsions, anhydrous lipids and water-in-oil emulsions, fats, waxes, oil, silicones, and humectants (e.g., glycerol).
  • The antigen may be derived from a pathogen that-can infect the organism (e.g., bacterium, virus, fungus, or parasite), or a cell (e.g., tumor cell or normal cell). The antigen may be a tumor antigen or an autoantigen. Chemically, the antigen may be a carbohydrate, glycolipid, glycoprotein, lipid, lipoprotein, phospholipid, polypeptide, or chemical or recombinant conjugate of the above. The molecular weight of the antigen may be greater than 500 daltons, preferably greater than 800 daltons, and more preferably greater than 1000 daltons.
  • Antigen may be obtained by recombinant means, chemical synthesis, or purification from a natural source. Preferred are proteinaceous antigen or conjugates with polysaccharide. Antigen may be at least partially purified in cell-free form. Alternatively, antigen may be provided in the form of a live virus, an attenuated live virus, or an inactivated virus.
  • Inclusion of an adjuvant may allow potentiation or modulation of the immune response. Moreover, selection of a suitable antigen or adjuvant may allow preferential induction of a humoral or cellular immune response, specific antibody isotypes (e.g., IgM, IgD, IgA1, IgA2, IgE, IgG1, IgG2, IgG3, and/or IgG4), and/or specific T-cell subsets (e.g., CTL, Th1, Th2 and/or TDTH).
  • Preferably, the adjuvant is an ADP-ribosylating exotoxin or a subunit thereof. Optionally, an activator of Langerhans cells may be used.
  • Optionally, antigen, adjuvant, or both may be provided in the formulation by means of a nucleic acid (e.g., DNA, RNA, cDNA, cRNA) encoding the antigen or adjuvant as appropriate. This technique is called genetic immunization.
  • The term “antigen” as used in the invention, is meant to describe a substance that induces a specific immune response when presented to immune cells of an organism. An antigen may comprise a single immunogenic epitope, or a multiplicity of immunogenic epitopes recognized by a B-cell receptor (i.e., antibody on the membrane of the B cell) or a T-cell receptor. A molecule may be both an antigen and an adjuvant (e.g., cholera toxin) and, thus, the formulation may contain only one component.
  • The term “adjuvant” as used in the invention, is meant to describe a substance added to the formulation to assist in inducing an immune response to the antigen.
  • The term “effective amount” as used in the invention, is meant to describe that amount of antigen which induces an antigen-specific immune response. Such induction of an immune response may provide a treatment such as, for example, immunoprotection, desensitization, immunosuppression, modulation of autoimmune disease, potentiation of cancer immunosurveillance, or therapeutic vaccination against an established infectious disease.
  • The term “draining lymph node field” as used in the invention means an anatomic area over which the lymph collected is filtered through a set of defined set of lymph nodes (e.g., cervical, axillary, inguinal, epitrochelear, popliteal, those of the abdomen and thorax).
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1 shows cholera toxin (CT) induces enhanced major histocompatibility complex (MHC) class II expression on Langerhans cells (LC), changes in LC morphology, and loss of LCs (presumably through migration). BALB/c mice (H-2d) were transcutaneously immunized with cholera CT or its B subunit (CTB) in saline solution on the ear (7,000 anti-CT ELISA units after a single immunization). Previous experiments had established that mice were readily immunized when using the skin of the ear. After 16 hours, epidermal sheets were prepared and stained for MHC class II molecules (scale bar is 50 μm). Panels indicate (A) saline alone as a negative control, (B) transcutaneous immunization with CT in saline, (C) transcutaneous immunization with CTB in saline, and (D) intradermal injection with tumor necrosis factor-α (10 μg) as a positive control.
  • DETAILED DESCRIPTION OF THE INVENTION
  • A transcutaneous immunization system delivers agents to specialized cells (e.g., antigen presentation cell, lymphocyte) that produce an immune response (Bos, 1997). These agents as a class are called antigens. Antigen may be composed of chemicals such as, for example, carbohydrate, glycolipid, glycoprotein, lipid, lipoprotein, phospholipid, polypeptide, conjugates thereof, or any other material known to induce an immune response. Antigen may be provided as a whole organism such as, for example, a bacterium or virion; antigen may be obtained from an extract or lysate, either from whole cells or membrane alone; or antigen may be chemically synthesized or produced by recombinant means.
  • Processes for preparing a pharmaceutical formulation are well-known in the art, whereby the antigen and adjuvant is combined with a pharmaceutically acceptable carrier vehicle. Suitable vehicles and their preparation are described, for example, in Remington's Pharmaceutical Sciences by E. W. Martin. Such formulations will contain an effective amount of the antigen and adjuvant together with a suitable amount of vehicle in order to prepare pharmaceutically acceptable compositions suitable for administration to a human or animal. The formulation may be applied in the form of an cream, emulsion, gel, lotion, ointment, paste, solution, suspension, or other forms known in the art. In particular, formulations that enhance skin hydration, penetration, or both are preferred. There may also be incorporated other pharmaceutically acceptable additives including, for example, diluents, binders, stabilizers, preservatives, and colorings.
  • Increasing hydration of the stratum corneum will increase the rate of percutaneous absorbtion of a given solute (Roberts and Walker, 1993). As used in the present invention, “penetration enhancer” does not include substances such as, for example: water, physiological buffers, saline solutions, and alcohols which would not perforate the skin.
  • An object of the present invention is to provide a novel means for immunization through intact skin without the need for perforating the skin. The transcutaneous immunization system provides a method whereby antigens and adjuvant can be delivered to the immune system, especially specialized antigen presentation cells underlying the skin such as, for example, Langerhans cells.
  • Without being bound to any particular theory but only to provide an explanation for our observations, it is presumed that the transcutaneous immunization delivery system carries antigen to cells of the immune system where an immune response is induced. The antigen may pass through the normal protective outer layers of the skin (i.e., stratum corneum) and induce the immune response directly, or through an antigen presenting cell (e.g., macrophage, tissue macrophage, Langerhans cell, dendritic cell, dermal dendritic cell, B lymphocyte, or Kupffer cell) that presents processed antigen to a T lymphocyte. Optionally, the antigen may pass through the stratum corneum via a hair follicle or a skin organelle (e.g., sweat gland, oil gland).
  • Transcutaneous immunization with bacterial ADP-ribosylating exotoxins (bAREs) may target the epidermal Langerhans cell, known to be among the most efficient of the antigen presenting cells (APCs) (Udey, 1997). We have found that bAREs activate Langerhans cells when applied epicutaneously to the skin in saline solution. The Langerhans cells direct specific immune responses through phagocytosis of the antigens, and migration to the lymph nodes where they act as APCs to present the antigen to lymphocytes (Udey, 1997), and thereby induce a potent antibody response. Although the skin is generally considered a barrier to invading organisms, the imperfection of this barrier is attested to by the numerous Langerhans cells distributed throughout the epidermis that are designed to orchestrate the immune response against organisms invading via the skin (Udey, 1997).
  • According to Udey (1997):
      • “Langerhans cells are bone-marrow derived cells that are present in all mammalian stratified squamous epithelia. They comprise all of the accessory cell activity that is present in uninflammed epidermis, an in the current paradigm are essential for the initiation and propagation of immune responses directed against epicutaneously applied antigens. Langerhans cells are members of a family of potent accessory cells (‘dendritic cells’) that are widely distributed, but infrequently represented, in epithelia and solid organs as well as in lymphoid tissue . . .
      • “It is now recognized that Langerhans cells (and presumably other dendritic cells) have a life cycle with at least two distinct stages. Langerhans cells that are located in epidermis constitute a regular network of antigen-trapping ‘sentinel’ cells. Epidermal Langerhans cells can ingest particulates, including microorganisms, and are efficient processors of complex antigens. However, they express only low levels of MHC class I and II antigens and costimulatory molecules (ICAM-1, B7-1 and B7-2) and are poor stimulators of unprimed T cells. After contact with antigen, some Langerhans cells become activated, exit the epidermis and migrate to T-cell-dependent regions of regional lymph nodes where they local as mature dendritic cells. In the course of exiting the epidermis and migrating to lymph nodes, antigen-bearing epidermal Langerhans cells (now the ‘messengers’) exhibit dramatic changes in morphology, surface phenotype and function. In contrast to epidermal Langerhans cells, lymphoid dendritic cells are essentially non-phagocytic and process protein antigens inefficiently, but express high levels of MHC class I and class II antigens and various costimulatory molecules and are the most potent stimulators of naive T cells that have been identified.”
  • We envision that the potent antigen presenting capability of the epidermal Langerhans cells can be exploited for transcutaneously delivered vaccines. A transcutaneous immune response using the skin immune system would require delivery of vaccine antigen only to Langerhans cells in the stratum corneum (the outermost layer of the skin consisting of cornified cells and lipids) via passive diffusion and subsequent activation of the Langerhans cells to take up antigen, migrate to B-cell follicles and/or T-cell dependent regions, and present the antigen to B and/or T cells (Stingl et al., 1989). If antigens other that bAREs (for example BSA) were to be phagocytosed by the Langerhans cells, then these antigens could also be taken to the lymph node for presentation to T-cells and subsequently induce an immune response specific for that antigen (e.g., BSA). Thus, a feature of transcutaneous immunization is the activation of the Langerhans cell, presumably by a bacterial ADP-ribosylating exotoxin, ADP-ribosylating exotoxin binding subunits (e.g., cholera toxin B subunit), or other Langerhans cell activating substance.
  • The mechanism of transcutaneous immunization via Langerhans cells activation, migration and antigen presentation is clearly shown by the upregulation of MHC class II expression in the epidermal Langerhans cells from epidermal sheets transcutaneously immunized with CT or CTB. In addition, the magnitude of the antibody response induced by transcutaneous immunization and isotype switching to predominantly IgG is generally achieved with T-cell help (Janeway and Travers, 1996), and activation of both Th1 and Th2 pathways is suggested by the production of IgG1 and IgG2a (Paul and Seder, 1994; Seder and Paul, 1994). Alternatively, a large antibody response may be induced by a thymus-independent antigen type 1 (TI-1) which directly activates the B cell (Janeway and Travers, 1996).
  • The spectrum of more commonly known skin immune responses is represented by contact dermatitis and atopy. Contact dermatitis, a pathogenic manifestation of LC activation, is directed by Langerhans cells which phagocytose antigen, migrate to lymph nodes, present antigen, and sensitize T cells for the intense destructive cellular response that occurs at the affected skin site (Dahl, 1996; Leung, 1997). Atopic dermatitis may utilize the Langerhans cell in a similar fashion, but is identified with Th2 cells and is generally associated with high levels of IgE antibody (Dahl, 1996; Leung, 1997).
  • Transcutaneous immunization with cholera toxin-and related bAREs on the other hand is a novel immune response with an absence of superficial and microscopic post-immunization skin findings (i.e., non-inflamed skin) shown by the absence of lymphocyte infiltration 24, 48 and 120 hours after immunization. This indicates that Langerhans cells “comprise all of the accessory cell activity that is present in uninflammed epidermis, and in the current paradigm are essential for the initiation and propagation of immune responses directed against epicutaneously applied antigens” (Udey, 1997). The uniqueness of the transcutaneous immune response here is also indicated by the both high levels of antigen-specific IgG antibody, and the type of antibody produced (e.g., IgM, IgG1, IgG2a, IgG2b, IgG3 and IgA) and the absence of anti-CT IgE antibody.
  • Thus, we have found that bacterial-derived toxins applied to the surface of the skin can activate Langerhans cells and induce a potent immune response manifested as high levels of antigen-specific circulating IgG antibodies. Such adjuvants may be used in transcutaneous immunization to enhance the IgG antibody response to proteins not otherwise immunogenic by themselves when placed on the skin.
  • Transcutaneous targeting of Langerhans cells may also be used to deactivate their antigen presenting function, thereby preventing immunization or sensitization. Techniques to deactivate Langerhans cells include, for example, the use of interleukin-10 (Peguet-Navarro et al., 1995), monoclonal antibody to interleukin-1β (Enk et al., 1993), or depletion via superantigens such as through staphylococcal enterotoxin-A (SEA) induced epidermal Langerhans cell depletion (Shankar et al., 1996).
  • Transcutaneous immunization may be induced via the ganglioside GM1 binding activity of CT, LT or subunits such as CTB (Craig and Cuatrecasas, 1975). Ganglioside GM1 is a ubiquitous cell membrane glycolipid found in all mammalian cells (Plotkin and Mortimer, 1994). When the pentameric CT B subunit binds to the cell surface a hydrophilic pore is formed which allows the A subunit to penetrate across the lipid bilayer (Ribi et al., 1988).
  • We have shown that transcutaneous immunization by CT or CTB may require ganglioside GM1 binding activity. When mice were transcutaneously immunized with CT, CTA and CTB, only CT and CTB resulted in an immune response. CTA contains the ADP-ribosylating exotoxin activity but only CT and CTB containing the binding activity were able to induce an immune response indicating that the B subunit was necessary and sufficient to immunize through the skin. We conclude that the Langerhans cell may be activated by CTB binding to its cell surface.
  • Antigen
  • Antigen of the invention may be expressed by recombinant means, preferably as a fusion with an affinity or epitope tag (Summers and Smith, 1987; Goeddel, 1990; Ausubel et al., 1996); chemical synthesis of an oligopeptide, either free or conjugated to carrier proteins, may be used to obtain antigen of the invention (Bodanszky, 1993; Wisdom, 1994). Oligopeptides are considered a type of polypeptide.
  • Oligopeptide lengths of 6 residues to 20 residues are preferred. Polypeptides may also by synthesized as branched structures such as those disclosed in U.S. Pat. Nos. 5,229,490 and 5,390,111. Antigenic polypeptides include, for example, synthetic or recombinant B-cell and T-cell epitopes, universal T-cell epitopes, and mixed T-cell epitopes from one organism or disease and B-cell epitopes from another.
  • Antigen obtained through recombinant means or peptide synthesis, as well as antigen of the invention obtained from natural sources or extracts, may be purified by means of the antigen's physical and chemical characteristics, preferably by fractionation or chromatography (Janson and Ryden, 1989; Deutscher, 1990; Scopes, 1993).
  • A multivalent antigen formulation may be used to induce an immune response to more than one antigen at the same time. Conjugates may be used to induce an immune response to multiple antigens, to boost the immune response, or both. Additionally, toxins may be boosted by the use of toxoids, or toxoids boosted by the use of toxins. Transcutaneous immunization may be used to boost responses induced initially by other routes of immunization such as by injection, or the oral or intranasal routes.
  • Antigen includes, for example, toxins, toxoids, subunits thereof, or combinations thereof (e.g., cholera toxin, tetanus toxoid).
  • Antigen may be solubilized in a buffer. Suitable buffers include, but are not limited to, phosphate buffered saline Ca++/Mg++ free (PBS), normal saline (150 mM NaCl in water), and Tris buffer. Antigen not soluble in neutral buffer can be solubilized in 10 mM acetic acid and then diluted to the desired volume with a neutral buffer such as PBS. In the case of antigen soluble only at acid pH, acetate-PBS at acid pH may be used as a diluent after solubilization in dilute acetic acid. Glycerol may be a suitable non-aqueous buffer for use in the present invention.
  • Hydrophobic antigen can be solubilized in a detergent, for example a polypeptide containing a membrane-spanning domain. Furthermore, for formulations containing liposomes, an antigen in a detergent solution (e.g., a cell membrane extract) may be mixed with lipids, and liposomes then may be formed by removal of the detergent by dilution, dialysis, or column chromatography. Certain antigens such as, for example, those from a virus (e.g., hepatitis A) need not be soluble per se, but can be incorporated directly into a liposome in the form of a virosome (Morein and Simons, 1985).
  • Plotkin and Mortimer (1994) provide antigens which can be used to vaccinate animals or humans to induce an immune response specific for particular pathogens, as well as methods of preparing antigen, determining a suitable dose of antigen, assaying for induction of an immune response, and treating infection by a pathogen (e.g., bacterium, virus, fungus, or parasite).
  • Bacteria include, for example: anthrax, campylobacter, cholera, diphtheria, enterotoxigenic E. coli, giardia, gonococcus, Helicobacter pylori (Lee and Chen, 1994), Hemophilus influenza B, Hemophilus influenza non-typable, meningococcus, pertussis, pneumococcus, salmonella, shigella, Streptococcus B, tetanus, Vibrio cholerae, and yersinia.
  • Viruses include, for example: adenovirus, dengue serotypes 1 to 4 (Delenda et al., 1994; Fonseca et al., 1994; Smucny et al., 1995), ebola (Jahrling et al., 1996), enterovirus, hepatitis serotypes A to E (Blum, 1995; Katkov, 1996; Lieberman and Greenberg, 1996; Mast, 1996; Shafara et al., 1995; Smedila et al., 1994; U.S. Pat. Nos. 5,314-,808 and 5,436,126), herpes simplex virus 1 or 2, human immunodeficiency virus (Deprez et al., 1996), influenza, measles, Norwalk, papilloma virus, parvovirus B19, polio, rabies, rotavirus, rubella, rubeola, vaccinia, vaccinia constructs containing genes coding for other antigens such as malaria antigens, varicella, and yellow fever.
  • Parasites include, for example: Entamoeba histolytica (Zhang et al., 1995); Plasmodium (Bathurst et al., 1993; Chang et al., 1989, 1992, 1994; Fries et al., 1992a, 1992b; Herrington et al., 1991; Khusmith et al., 1991; Malik et al., 1991; Migliorini et al., 1993; Pessi et al., 1991; Tam, 1988; Vreden et al., 1991; White et al., 1993; Wiesmueller et al., 1991), Leishmania (Frankenburg et al., 1996), and the Helminthes.
  • Adjuvant
  • The formulation also contains an adjuvant, although a single molecule may contain both adjuvant and antigen properties (e.g., cholera toxin) (Elson and Dertzbaugh, 1994). Adjuvants are substances that are used to specifically or non-specifically potentiate an antigen-specific immune response. Usually, the adjuvant and the formulation are mixed prior to presentation of the antigen but, alternatively, they may be separately presented within a short interval of time.
  • Adjuvants include, for example, an oil emulsion (e.g., complete or incomplete Freund's adjuvant), a chemokine (e.g., defensins 1 or 2, RANTES, MIP1-α, MIP-2, interleukin-8) or a cytokine (e.g., interleukin-1β, -2, -6, -10 or -12; γ-interferon; tumor necrosis factor-α; or granulocyte-monocyte-colony stimulating factor) (reviewed in Nohria and Rubin, 1994), a muramyl dipeptide derivative (e.g., murabutide, threonyl-MDP or muramyl tripeptide), a heat shock protein or a derivative, a derivative of Leishmania major LeIF (Skeiky et al., 1995), cholera toxin or cholera toxin B, a lipopolysaccharide (LPS) derivative (e.g., lipid A or monophosphoryl lipid A), or superantigen (Saloga et al., 1996). Also, see Richards et al. (1995) for adjuvants useful in immunization.
  • An adjuvant may be chosen to preferentially induce antibody or cellular effectors, specific antibody isotypes (e.g., IgM, IgD, IgA1, IgA2, secretory IgA, IgE, IgG1, IgG2, IgG3, and/or IgG4), or specific T-cell subsets (e.g., CTL, Th1, Th2 and/or TDTH) (Glenn et al., 1995).
  • Cholera toxin is a bacterial exotoxin from the family of ADP-ribsoylating exotoxins (referred to as bAREs). Most bAREs are organized as A:B dimer with a binding B subunit and an A subunit containing the ADP-ribosyltransferase. Such toxins include diphtheria, Pseudomonas exotoxin A, cholera toxin (CT), E. coli heat-labile enterotoxin (LT), pertussis toxin, C. botulinum toxin C2, C. botulinum toxin C3, C. limosum exoenzyme, B. cereus exoenzyme, Pseudomonas exotoxin S, Staphylococcus aureus EDIN, and B. sphaericus toxin.
  • Cholera toxin is an example of a bARE that is organized with A and B subunits. The B subunit is the binding subunit and consists of a B-subunit pentamer which is non-covalently bound to the A subunit. The B-subunit pentamer is arranged in a symmetrical doughnut-shaped structure that binds to GM1-ganglioside on the target cell. The A subunit serves to ADP ribosylate the alpha subunit of a subset of the hetero trimeric GTP proteins (G proteins) including the Gs protein which results in the elevated intracellular levels of cyclic AMP. This stimulates release of ions and fluid from intestinal cells in the case of cholera.
  • Cholera toxin (CT) and its B subunit (CTB) have adjuvant properties when used as either an intramuscular or oral immunogen (Elson and Dertzbaugh, 1994; Trach et al., 1997). Another antigen, heat-labile enterotoxin from E. coli (LT) is 80% homologous at the amino acid level with CT and possesses similar binding properties; it also appears to bind the GM1-ganglioside receptor in the gut and has similar ADP-ribosylating exotoxin activities. Another bARE, Pseudomonas exotoxin A (ETA), binds to the α2-macroglobulin receptor-low density lipoprotein receptor-related protein (Kounnas et al., 1992). bAREs are reviewed by Krueger and Barbieri (1995).
  • The examples below show that cholera toxin (CT),-its B subunit (CTB), E. coli heat-labile enterotoxin (LT), and pertussis toxin are potent adjuvants for transcutaneous immunization, inducing high levels of IgG antibodies but not IgE antibodies. Also shown is that CTB without CT can also induce high levels of IgG antibodies. Thus, both bAREs and a derivative thereof can effectively immunize when epicutaneouly applied to the skin in a simple solution.
  • When an adjuvant such as CT is mixed with BSA, a protein not usually immunogenic when applied to the skin, anti-BSA antibodies are induced. An immune response to diphtheria toxoid was induced using pertussis toxin as adjuvant, but not with diphtheria toxoid alone. Thus, bAREs can act as adjuvants for non-immunogenic proteins in an transcutaneous immunization system.
  • Protection against the life-threatening infections diphtheria, pertussis, and tetanus (DPT) can be achieved by inducing high levels of circulating anti-toxin antibodies. Pertussis may be an exception in that some investigators feel that antibodies directed to other portions of the invading organism are necessary for protection, although this is controversial (see Schneerson et al., 1996) and most new generation acellular pertussis vaccines have PT as a component of the vaccine (Krueger and Barbieri, 1995). The pathologies in the diseases caused by DPT are directly related to the effects of their toxins and anti-toxin antibodies most certainly play a role in protection (Schneerson et al., 1996).
  • In general, toxins can be chemically inactivated to form toxoids which are less toxic but remain immunogenic. We envision that the transcutaneous immunization system using toxin-based immunogens and adjuvants can achieve anti-toxin levels adequate for protection against these diseases. The anti-toxin antibodies may be induced through immunization with the toxins, or genetically-detoxified toxoids themselves, or with toxoids and adjuvants such as CT. Genetically toxoided toxins which have altered ADP-ribosylating exotoxin activity, but not binding activity, are envisioned to be especially useful as non-toxic activators of antigen presenting cells used in transcutaneous immunization.
  • We envision that CT can also act as an adjuvant to induce antigen-specific CTLs through transcutaneous immunization (see Bowen et al., 1994; Porgador et al., 1997 for the use of CT as an adjuvant in oral immunization).
  • The bARE adjuvant may be chemically conjugated to other antigens including, for example, carbohydrates, polypeptides, glycolipids, and glycoprotein antigens. Chemical conjugation with toxins, their subunits, or toxoids with these antigens would be expected to enhance the immune response to these antigens when applied epicutaneously.
  • To overcome the problem of the toxicity of the toxins, (e.g., diphtheria toxin is known to be so toxic that one molecule can kill a cell) and to overcome the difficulty of working with such potent toxins as tetanus, several workers have taken a recombinant approach to producing genetically produced toxoids. This is based on inactivating the catalytic activity of the ADP-ribosyl transferase by genetic deletion. These toxins retain the binding capabilities, but lack the toxicity, of the natural toxins. This approach is described by Burnette et al. (1994), Rappuoli et al. (1995), and Rappuoli et al. (1996). Such genetically toxoided exotoxins could be useful for transcutaneous immunization system in that they would not create a safety concern as the toxoids would not be considered toxic. Additionally, several techniques exist to chemically toxoid toxins which can address the same problem (Schneerson et al., 1996). These techniques could be important for certain applications, especially pediatric applications, in which ingested toxins (e.g., diphtheria toxin) might possibly create adverse reactions. optionally, an activator of Langerhans cells may be used as an adjuvant. Examples of such activators include: inducers of heat shock protein; contact sensitizers (e.g., trinitrochlorobenzene, dinitrofluorobenzene, nitrogen mustard, pentadecylcatechol); toxins (e.g, Shiga toxin, Staph enterotoxin B); lipopolysaccharides, lipid A, or derivatives thereof; bacterial DNA (Stacey et al., 1996); cytokines (e.g., tumor necrosis factor-α, interleukin-1β, -10, -12); and chemokines (e.g., defensins 1 or 2, RANTES, MIP-1α, MIP-2, interleukin-8).
  • If an immunizing antigen has sufficient Langerhans cell activating capabilities then a separate adjuvant may not be required, as in the case of CT which is both antigen and adjuvant. It is envisioned that whole cell preparations, live viruses, attenuated viruses, DNA plasmids, and bacterial DNA could be sufficient to immunize transcutaneously. It may be possible to use low concentrations of contact sensitizers or other activators of Langerhans cells to induce an immune response without inducing skin lesions.
  • Liposomes and Their Preparation
  • Liposomes are closed vesicles surrounding an internal aqueous space. The internal compartment is separated from the external medium by a lipid bilayer composed of discrete lipid molecules. In the present invention, antigen may be delivered through intact skin to specialized cells of the immune system, whereby an antigen-specific immune response is induced. Transcutaneous immunization may be achieved by using liposomes; however, as shown in the examples, liposomes are not required to elicit an antigen-specific immune response.
  • Liposomes may be prepared using a variety of techniques and membrane lipids (reviewed in Gregoriadis, 1993). Liposomes may be pre-formed and then mixed with antigen. The antigen may be dissolved or suspended, and then added to (a) the pre-formed liposomes in a lyophilized state, (b) dried lipids as a swelling solution or suspension, or (c) the solution of lipids used to form liposomes. They may also be formed from lipids extracted from the stratum corneum including, for example, ceramide and cholesterol derivatives (Wertz, 1992).
  • Chloroform is a preferred solvent for lipids, but it may deteriorate upon storage. Therefore, at one- to three-month intervals, chloroform is redistilled prior to its use as the solvent in forming liposomes. After distillation, 0.7% ethanol can be added as a preservative. Ethanol and methanol are other suitable solvents.
  • The lipid solution used to form liposomes is placed in a round-bottomed flask. Pear-shaped boiling flasks are preferred, particularly those flasks sold by Lurex Scientific (Vineland, N.J., cat. no. JM-5490). The volume of the flask should be more than ten times greater than the volume of the anticipated aqueous suspension of liposomes to allow for proper agitation during liposome formation.
  • Using a rotary evaporator, solvent is removed at 37° C. under negative pressure for 10 minutes with a filter aspirator attached to a water faucet. The flask is further dried under low vacuum (i.e., less than 50 mm Hg) for 1 hour in a dessicator.
  • To encapsulate antigen into liposomes, an aqueous solution containing antigen may be added to lyophilized liposome lipids in a volume that results in a concentration of approximately 200 mM with respect to liposome lipid, and shaken or vortexed until all the dried liposome lipids are wet. The liposome-antigen mixture may then be incubated for 18 hours to 72 hours at 4° C. The liposome-antigen formulation may be used immediately or stored for several years. It is preferred to employ such a formulation directly in the transcutaneous immunization system without removing unencapsulated antigen. Techniques such as bath sonication may be employed to decrease the size of liposomes, which may augment transcutaneous immunization.
  • Liposomes may be formed as described above but without addition of antigen to the aqueous solution. Antigen may then be added to the pre-formed liposomes and, therefore, antigen would be in solution and/or associated with, but not encapsulated by, the liposomes. This process of making a liposome-containing formulation is preferred because of its simplicity. Techniques such as bath sonication may be employed to alter the size and/or lamellarity of the liposomes to enhance immunization.
  • Although not required to practice the present invention, hydration and/or penetration of the stratum corneum may be enhanced by adding liposomes to the formulation. Liposomes have been used as carriers with adjuvants to enhance the immune response to antigens mixed with, encapsulated in, attached to, or associated with liposomes.
  • Transcutaneous Delivery of Antigen
  • Efficient immunization can be achieved with the present invention because transcutaneous delivery of antigen may target the Langerhans cell. These cells are found in abundance in the skin and are efficient antigen presenting cells leading to T-cell memory and potent immune responses (Udey, 1997). Because of the presence of large numbers of Langerhans cells in the skin, the efficiency of transcutaneous delivery may be related to the surface area exposed to antigen and adjuvant. In fact, the reason that transcutaneous immunization is so efficient may be that it targets a larger number of these efficient antigen presenting cells than intramuscular immunization.
  • We envision the present invention will enhance access to immunization, while inducing a potent immune response. Because transcutaneous immunization does not involve penetration of the skin and the complications and difficulties thereof, the requirements of trained personnel, sterile technique, and sterile equipment are reduced. Furthermore, the barriers to immunization at multiple sites or to multiple immunizations are diminished. Immunization by a single application of the formulation is also envisioned.
  • Immunization may be achieved using epicutaneous application of a simple solution of antigen and adjuvant impregnated in gauze under an occlusive patch, or by using other patch technologies; creams, immersion, ointments and sprays are other possible methods of application. The immunization could be given by untrained personnel, and is amenable to self-application. Large-scale field immunization could occur given the easy accessibility to immunization. Additionally, a simple immunization procedure would improve access to immunization by pediatric patients and the elderly, and populations in Third World countries.
  • For previous vaccines, their formulations were injected through the skin with needles. Injection of vaccines using needles carries certain drawbacks including the need for sterile needles and syringes, trained medical personnel to administer the vaccine, discomfort from the injection, and potential complications brought about by puncturing the skin with the needle. Immunization through the skin without the use of needles (i.e., transcutaneous immunization) represents a major advance for vaccine delivery by avoiding the aforementioned drawbacks.
  • The transcutaneous delivery system of the invention is also not concerned with penetration of intact skin by sound or electrical energy. Such a system that uses an electrical field to induce dielectric breakdown of the stratum corneum is disclosed in U.S. Pat. No. 5,464,386.
  • Moreover, transcutaneous immunization may be superior to immunization using needles as more immune cells would be targeted by the use of several locations targeting large surface areas of skin. A therapeutically effective amount of antigen sufficient to induce an immune response may be delivered transcutaneously either at a single cutaneous location, or over an area of intact skin covering multiple draining lymph node fields (e.g., cervical, axillary, inguinal, epitrochelear, popliteal, those of the abdomen and thorax). Such locations close to numerous different lymphatic nodes at locations all over the body will provide a more widespread stimulus to the immune system than when a small amount of antigen is injected at a single location by intradermal subcutaneous or intramuscular injection.
  • Antigen passing through or into the skin may encounter antigen presenting cells which process the antigen in a way that induces an immune response. Multiple immunization sites may recruit a greater number of antigen presenting cells and the larger population of antigen presenting cells that were recruited would result in greater induction of the immune response. It is conceivable that absorption through the skin may deliver antigen to phagocytic cells of the skin such as, for example, dermal dendritic cells, macrophages, and other skin antigen presenting cells; antigen may also be delivered to phagocytic cells of the liver, spleen, and bone marrow that are known to serve as the antigen presenting cells through the blood stream or lymphatic system. The result would be widespread distribution of antigen to antigen presenting cells to a degree that is rarely, if ever achieved, by current immunization practices.
  • The transcutaneous immunization system may be applied directly to the skin and allowed to air dry; rubbed into the skin or scalp; held in place with a dressing, patch, or absorbent material; otherwise held by a device such as a stocking, slipper, glove, or shirt; or sprayed onto the skin to maximize contact with the skin. The formulation may be applied in an absorbant dressing or gauze. The formulation may be covered with an occlusive dressing such as, for example, AQUAPHOR (an emulsion of petrolatum, mineral oil, mineral wax, wool wax, panthenol, bisabol, and glycerin from Beiersdorf, Inc.), plastic film, COMFEEL (Coloplast) or vaseline; or a non-occlusive dressing such as, for example, DUODERM (3M) or OPSITE (Smith & Napheu). An occlusive dressing completely excludes the passage of water.
  • The formulation may be applied to single or multiple sites, to single or multiple limbs, or to large surface areas of the skin by complete immersion. The formulation may be applied directly to the skin.
  • Genetic immunization has been described in U.S. Pat. Nos. 5,589,466 and 5,593,972. The nucleic acid(s) contained in the formulation may encode the antigen, the adjuvant, or both. The nucleic acid may or may not be capable of replication; it may be non-integrating and non-infectious. The nucleic acid may further comprise a regulatory region (e.g., promoter, enhancer, silencer, transcription initiation and termination sites, RNA splice acceptor and donor sites, polyadenylation signal, internal ribosome binding site, translation initiation and termination sites) operably linked to the sequence encoding the antigen or adjuvant. The nucleic acid may be complexed with an agent that promotes transfection such as cationic lipid, calcium phosphate, DEAE-dextran, polybrene-DMSO, or a combination thereof. The nucleic acid may comprise regions derived from viral genomes. Such materials and techniques are described by Kriegler (1990) and Murray (1991).
  • An immune response may comprise humoral (i.e., antigen-specific antibody) and/or cellular (i.e., antigen-specific lymphocytes such as B cells, CD4+ T cells, CD8+ T cells, CTL, Th1 cells, Th2 cells, and/or TDTH cells) effector arms. Moreover, the immune response may comprise NK cells that mediate antibody-dependent cell-mediated cytotoxicity (ADCC) The immune response induced by the formulation of the invention may include the elicitation of antigen-specific antibodies and/or cytotoxic lymphocytes (CTL, reviewed in Alving and Wassef, 1994). Antibody can be detected by immunoassay techniques, and the detection of various isotypes (e.g., IgM, IgD, IgA1, IgA2, secretory IgA, IgE, IgG1, IgG2, IgG3, or IgG4) may be expected. An immune response can also be detected by a neutralizing assay.
  • Antibodies are protective proteins produced by B lymphocytes. They are highly specific, generally targeting one epitope of an antigen. Often, antibodies play a role in protection against disease by specifically reacting with antigens derived from the pathogens causing the disease. Immunization may induce antibodies specific for the immunizing antigen, such as cholera toxin. These antigen-specific antibodies are induced when antigen is delivered through the skin by liposomes.
  • CTLs are particular protective immune cells produced to protect against infection by a pathogen. They are also highly specific. Immunization may induce CTLs specific for the antigen, such as a synthetic oligopeptide based on a malaria protein, in association with self-major histocompatibility antigen. CTLs induced by immunization with the transcutaneous delivery system may kill pathogen infected cells. Immunization may also produce a memory response as indicated by boosting responses in antibodies and CTLs, lymphocyte proliferation by culture of lymphocytes stimulated with the antigen, and delayed type hypersensitivity responses to intradermal skin challenge of the antigen alone.
  • In a viral neutralization assay, serial dilutions of sera are added to host cells which are then observed for infection after challenge with infectious virus. Alternatively, serial dilutions of sera may be incubated with infectious titers of virus prior to innoculation of an animal, and the innoculated animals are then observed for signs of infection.
  • The transcutaneous immunization system of the invention may be evaluated using challenge models in either animals or humans, which evaluate the ability of immunization with the antigen to protect the subject from disease. Such protection would demonstrate an antigen-specific immune response. In lieu of challenge, achieving anti-diphtheria antibody titers of 5 IU/ml or greater is generally assumed to indicate optimum protection and serves as a surrogate marker for protection (Plotkin and Mortimer, 1994).
  • Furthermore, the Plasmodium faciparum challenge model may be used as to induce an antigen-specific immune response in humans. Human volunteers may be immunized using the transcutaneous immunization system containing oligopeptides or proteins (polypeptides) derived from the malaria parasite, and then exposed to malaria experimentally or in the natural setting. The Plasmodium yoelii mouse malaria challenge model may be used to evaluate protection in the mouse against malaria (Wang et al., 1995).
  • Alving et al. (1986) injected liposomes comprising lipid A as an adjuvant for inducing an immune response to cholera toxin (CT) in rabbits and to a synthetic protein consisting of a malaria oligopeptide containing four tetra-peptides (Asn-Ala-Asn-Pro) conjugated to BSA. The authors found that the immune response to cholera toxin or to the synthetic malaria protein was markedly enhanced by encapsulating the antigen within the liposomes containing lipid A, compared to similar liposomes lacking lipid A. Several antigens derived either from the circumsporozoite protein (CSP) or from merozoite surface proteins of Plasmodium falciparum have been encapsulated in liposomes containing lipid A. All of the malaria antigens that have been encapsulated in liposomes containing lipid A have been shown to induce humoral effectors (i.e., antigen-specific antibodies), and some have been shown to induce cell-mediated responses as well. Generation of an immune response and immunoprotection in an animal vaccinated with a malaria antigen may be assayed by immunofluorescence to whole, fixed malaria sporozoites or CTLs killing of target cells transfected with CSP.
  • Mice may be transcutaneously immunized with cholera toxin, and then challenged intranasally with an LD70 (40 μg) dose of cholera toxin and observed for protection. Mallet et al. (personal communication) have found that C57BL/6 mice develop a fatal hemorrhagic pneumonia in response to intranasal challenge with CT. Alternatively, the mice may be challenged with an intraperitoneal dose of CT (Dragunsky et al., 1992). Cholera toxin-specific IgG or IgA antibody may provide protection against cholera toxin challenge (Pierce, 1978; Pierce and Reynolds, 1974).
  • Vaccination has also been used as a treatment for cancer and autoimmune disease. For example, vaccination with a tumor antigen (e.g., prostate specific antigen) may induce an immune response in the form of antibodies, CTLs and lymphocyte proliferation which allows the body's immune system to recognize and kill tumor cells. Tumor antigens useful for vaccination have been described for melanoma (U.S. Pat. Nos. 5,102,663, 5,141,742, and 5,262,177), prostate carcinoma (U.S. Pat. No. 5,538,866), and lymphoma (U.S. Pat. Nos. 4,816,249, 5,068,177, and 5,227,159). Vaccination with T-cell receptor oligopeptide may induce an immune response that halts progression of autoimmune disease (U.S. Pat. Nos. 5,612,035 and 5,614,192; Antel et al., 1996; Vandenbark et al., 1996). U.S. Pat. No. 5,552,300 also describes antigens suitable for treating autoimmune disease.
  • The following is meant to be illustrative of the present invention; however, the practice of the invention is not limited or restricted in any way by the examples.
  • EXAMPLES
  • Immunization Procedure
  • BALB/c mice of 6 to 8 weeks were shaved with a #40 clipper. This shaving could be done without any signs of trauma to the skin. The shaving was done from the mid-thorax to just below the nape of the neck. The mice were then allowed to rest for 24 hours. Prior to this the mice had been ear-tagged for identification, and pre-bled to obtain a sample of pre-immune serum. Mice were also transcutaneously immunized without shaving by applying 50 μl of immunizing solution to each ear.
  • The mice were then immunized in the following way. Mice were anesthetized with 0.03-0.06 ml of a 20 mg/ml solution of xylazine and 0.5 ml of 100 mg/ml ketamine; mice were immobilized by this dose of anesthesia for approximately one hour. The mice were placed ventral side down on a warming blanket.
  • The immunizing solution was then placed on the dorsal shaved skin of a mouse in the following manner: a 1.2 cm×1.6 cm stencil made of polystyrene was laid gently on the back and a saline-wetted sterile gauze was used to partially wet the skin (this allowed even application of the immunizing solution), the immunizing solution was then applied with a pipet to the area circumscribed by the stencil to yield a 2 cm2 patch of immunizing solution. Care was used not to scrape or rub the skin with the pipet tip. The immunizing solution was spread around the area to be covered with the smooth side of the pipet tip.
  • The immunizing solution (between about 100 μl to about 200 μl) was left on the back of the mouse for 60 minutes. At the end of 60 minutes, the mouse was held gently by the nape of the neck and the tail under a copious stream of lukewarm tap water, and washed for 10 seconds. The mouse was then gently patted dry with a piece of sterile gauze and a second washing was performed for 10 seconds; the mouse was then patted dry a second time and left in the cage. The mice appeared to exhibit no adverse effects from the anesthesia, immunization, washing procedure, or toxicity from the exotoxins. No skin irritation, swelling or redness was seen after the immunization and the mice appeared to thrive. Immunization using the ear was performed as described above except that fur was not removed prior to immunization.
  • Antigen
  • The following antigens were used for immunization and ELISA, and were mixed using sterile PBS or normal saline. Cholera toxin or CT (List Biologicals, Cat #101B, lot #10149CB), CT B subunit (List Biologicals, Cat #BT01, lot #CVXG-14E), CT A subunit (List Biologicals, Cat #102A, lot #CVXA-17B), CT A subunit (Calbiochem, Cat #608562); pertussis toxin, salt-free (List Biologicals, lot #181120a); tetanus toxoid (List Biologicals, lots #1913a and #1915a); Pseudomonas exotoxin A (List Biologicals, lot #ETA25a); diphtheria toxoid (List Biologicals, lot #15151); heat-labile enterotoxin from E. coli (Sigma, lot #9640625); bovine serum albumin or BSA (Sigma, Cat #3A-4503, lot #31F-0116); and Hemophilus influenza B conjugate (Connaught, lot#6J81401).
  • ELISA—IgG(H+L)
  • Antibodies specific for CT, LT, ETA, pertussis toxin, diphtheria toxoid, tetanus toxoid, Hemophilus influenza B conjugate, and BSA were determined using ELISA in a technique similar to Glenn et al. (1995). All antigens were dissolved in sterile saline at a concentration of 2 μg/ml. Fifty microlilters of this solution (0.1 μg) per well was put on IMMULON-2 polystyrene plates (Dynatech Laboratories, Chantilly, Va.) and incubated at room temperature overnight. The plates were then blocked with a 0.5% casein/0.05% Tween 20 blocking buffer solution for one hour. Sera was diluted with 0.5% casein/0.05% Tween 20 diluent; dilution series were done in columns on the plate. Incubation was for 2 hours at room temperature.
  • The plates were then washed in a PBS-0.05% Tween 20 wash solution four times, and goat anti-mouse IgG(H+L) horseradish peroxidase (HRP)-linked (Bio-Rad Laboratories, Richmond, Calif., Cat #170-6516) secondary antibody was diluted in casein diluent at a dilution of 1/500 and left on the plates for one hour at room temperature. The plates were then washed four times in the PBS-Tween wash solution. One hundred microliters of 2,2′-azino-di(3-ethyl-benzthiazolone)sulphonic acid substrate (Kirkegaard and Perry) were added to each well and the plates were read at 405 nm after 20-40 minutes of development. Results are reported as the geometric mean of individual sera and standard error of the mean of ELISA units (the serum dilution at which the absorbance in equal to 1.0) or as individual antibody responses in ELISA units.
  • ELISA—IgG(γ), IgM(μ) and IgA(α)
  • IgG(γ), IgM(μ) and IgA(α) anti-CT antibody levels were determined using ELISA with a technique similar to Glenn et al. (1995). CT was dissolved in sterile saline at a concentration of 2 μg/ml. Fifty microliters of this solution (0.1 μg) per well were put on IMMULON-2 polystyrene plates (Dynatech Laboratories, Chantilly, Va.) and incubated at room temperature overnight. The plates were then blocked with a 0.5% casein-Tween 20 blocking buffer solution for one hour. Sera was diluted and casein diluent and serial dilutions were done on the plate. This was incubated for two hours at room temperature.
  • The plates were then washed in a PBS-Tween wash solution four times and goat anti-mouse IgG(γ) HRP-linked (Bio-Rad Laboratories, Richmond, Calif., Cat #172-1038), goat anti-mouse IgM(μ) HRP-linked (BioRad Laboratories, Richmond, Calif., Cat #172-1030), or goat anti-mouse IgA HRP-linked (Sigma, St. Louis, Mo., Cat #1158985) secondary antibody was diluted in casein diluent in a dilution of 1/1000 and left on the plates for one hour at room temperature. The plates were then washed four times in a PBS-Tween wash solution. One hundred microliters of 2,2′-azino-di(3-ethyl benzthiazolone) sulphonic acid substrate from (Kirkegaard and Perry, Gaithersburg, Md.) were added to the wells and the plates were read at 405 nm. Results are reported as the geometric mean of individual sera and standard error of the mean of ELISA units (the serum dilution at which the absorbance in equal to 1.0).
  • ELISA—IgG Subclass
  • Antigen-specific IgG (IgG1, IgG2a, IgG2b, and IgG3) subclass antibody against CT, LT, ETA, and BSA was performed as described by Glenn et al. (1995). The solid phase ELISA was performed in IMMULON-2 polystyrene plates (Dynatech Laboratories, Chantilly, Va.). Wells were incubated with the respective antigens in saline overnight (0.1 μg/50 μl) and blocked with 0.5% casein-Tween 20. Individual mouse sera diluted in 0.5% casein were serially diluted, and incubated at room temperature for four hours. Secondary antibody consisted of horseradish peroxidase-conjugated goat anti-mouse isotype-specific antibody (IgG1, IgG2a, IgG2b, IgG3, The Binding Site, San Diego, Calif.). A standard curve for each subclass was determined using mouse myeloma IgG1, IgG2a, IgG2b, and IgG3 (The Binding Site, San Diego, Calif.). Standard wells were coated with goat anti-mouse IgG(H+L) (Bio-Rad Laboratories, Richmond, CA, Cat #172-1054) to capture the myeloma IgG subclass standards which were added in serial dilutions. The myeloma IgG subclass was also detected using the peroxidase-conjugated goat anti-mouse subclass-specific antibody. Both the test sera and myeloma standards were detected using 2,2′-azino-di(3-ethyl-benzthiazolone) sulphonic acid (Kirkegaard and Perry, Gaithersburg, Md.) as substrate. Absorbances were read at 405 nm. Individual antigen specific subclasses were quantitated using the values from the linear titration curve computed against the myeloma standard curve and reported as μg/ml.
  • ELISA—IgE
  • Antigen-specific IgE antibody quantitation was performed using a protocol from Pharmingen Technical Protocols, page 541 of the Research Products Catalog, 1996-1997 (Pharmingen, San Diego, Calif.). Fifty microliters of 2 μg/ml purified anti-mouse IgE capture mAb (Pharmingen, Cat# 02111D) in 0.1 M NaHCO3 (pH 8.2) were added to IMMUNO plates(Nunc, Cat #12-565-136). Plates were incubated overnight at room temperature, washed three times with PBS-Tween 20, blocked with 3% BSA in PBS for two hours, and washed three times with PBS-Tween. Sera were diluted in 1% BSA in PBS, added at dilutions of 1/100, and diluted serially down the columns (e.g., 1/100, 1/200, et cetera). Purified mouse IgE standards (Pharmingen, Cat # 0312D) were added with a starting dilution of 0.25 μg/ml and serially diluted down the columns. Plates were incubated for two hours and washed five times with PBS-Tween.
  • Biotinylated anti-mouse IgE mAB (Pharmingen, Cat #02122D) to 2 μg/ml in 1% BSA in PBS, incubated for 45 minutes and washed five times with PBS-Tween. Avidin-peroxidase (Sigma A3151, 1:400 of 1 mg/ml solution) was added for 30 min and plates were washed six times with PBS-Tween. Both the test sera and IgE standards were detected using 2,2′-azino-di(3-ethyl-benzthiazolone)sulphonic acid (Kirkegaard and Perry, Gaithersburg, Md.) as substrate. Absorbances were read at 405 nm. Individual antigen specific subclasses were quantitated using the values from the linear titration curve computed against the IgE standard curve and reported as μg/ml.
  • Liposome Preparation
  • Where liposomes were included in the formulation for transcutaneous immunization, multilamellar liposomes composed of dimyristoyl phosphatidyl choline, dimyristoyl phosphatidyl glycerol, cholesterol were prepared according to Alving et al. (1993). Dimyristoyl phosphatidylcholine, dimyristoyl phosphatidylglycerol, and cholesterol were purchased from Avanti Polar Lipids Inc. (Alabaster, Ala.). Stock solutions of the lipids in chloroform were removed from −20° C. freezer where they were stored.
  • The lipids were mixed in a molar ratio of 0.9:0.1:0.75 dimyristoyl phosphatidyl choline, dimyristoyl phosphatidyl glycerol, and cholesterol in a pear shaped flask. Using a rotary evaporator, the solvent was removed at 37° C. under negative pressure for 10 minutes. The flask was further dried under low vacuum for two hours in a dessicator to remove residual solvent. The liposomes were swollen at 37 mM phospholipid using sterile water, lyophilized and stored at −20° C. These liposomes were mixed in their lyophilized state with normal saline (pH 7.0) to achieve a designated phospholipid concentration in the saline. Alternatively, the dried lipids were swollen to make liposomes with normal saline (pH 7.0) and were not lyophilized.
  • Example 1
  • BALB/c mice at 6 to 8 weeks of age were immunized transcutaneously as described above for “Immunization Procedure”, in groups of five mice. The mice were immunized using 100 μl of immunization solution which was prepared as follows: liposomes prepared as described above for “Liposome Preparation” were mixed with saline to form the liposomes. The pre-formed liposomes were then diluted in either saline (liposome alone group) or with CT in saline to yield an immunizing solution containing liposomes at 10-150 mM phospholipid with 100 μg of CT per 100 μl of immunizing solution. CT was mixed in saline to make an immunizing solution containing 100 μg of CT per 100 μg of solution for the group receiving CT alone. Solutions were vortexed for 10 seconds prior to immunization.
  • The mice were immunized transcutaneously at 0 and 3 weeks. Antibody levels were determined using ELISA as described above for “ELISA IgG(H+L)” 3 weeks after the boosting immunization, and compared against pre-immune sera. As shown in Table 1, the level of anti-CT antibodies induced by CT without liposomes was not different from the level of anti-CT antibodies generated using liposomes except in the mice where 150 mM liposomes were used. CT in saline alone was able to immunize mice against CT to produce high antibody titers.
    TABLE 1
    Anti-CT antibodies
    Group ELISA Units SEM
    CT alone 27,482 (16,635-48,051)
    CT + 150 mM Liposomes 4,064 *(2,845-5,072) 
    CT + 100 mM Liposomes 35,055 (25,932-44,269)
    CT + 50 mM Liposomes 9,168  (4,283-12,395)
    CT + 25 mM Liposomes 18,855 (12,294-40,374)
    CT + 10 mM Liposomes 28,660 (18,208-31,498)
    50 mM Liposomes 0

    *Significantly different from the Group CT alone (P < 0.05)
  • Example 2
  • BALB/c mice at 6 to 8 weeks of age were immunized transcutaneously as described above for “Immunization Procedure”, in groups of five mice. The mice were immunized at 0 and 3 weeks using 100 μl of immunization solution prepared as follows: BSA was mixed in saline to make an immunizing solution containing 200 μg of BSA per 100 μl of saline for the group receiving BSA alone; BSA and CT were mixed in saline to make an immunizing solution containing 200 μg of BSA and 100 μg of CT per 100 μl of saline for the group receiving BSA and CT. Where liposomes were used, the liposomes were prepared as described above for “Liposome Preparation”, and were first mixed with saline to form the liposomes. They were then diluted in BSA or BSA and CT in saline to yield an immunizing solution containing liposomes at 50 mM phospholipid with 200 μg of BSA per 100 μl of immunizing solution, or 200 μg BSA+100 μg CT per 100 μl of immunizing solution. Solutions were vortexed for 10 seconds prior to immunization.
  • The antibodies were determined using ELISA as described above for “ELISA IgG(H+L)” on sera 3 weeks after the second immunization. The results are shown in Table 2. BSA alone, with or without liposomes, was not able to elicit an antibody response. However, the addition of CT stimulated an immune response to BSA. CT acted as a adjuvant for the immune response to BSA, and anti-BSA antibodies of high titer were produced.
    TABLE 2
    Anti-BSA antibodies
    Group ELISA Units SEM
    BSA in saline 0
    BSA + 50 mM Liposomes 0
    CT + BSA in saline 8,198 (5,533-11,932)
    CT + BSA + 50 mM 3,244  (128-3,242)
  • Example 3
  • BALB/c mice at 6 to 8 weeks of age were immunized transcutaneously as described above for “Immunization Procedure”, in groups of five mice. The mice were immunized at 0 and 3 weeks using 100 μl of immunization solution prepared as follows: LT was mixed in saline to make an immunizing solution containing 100 μg of LT per 100 μl of saline for the group receiving LT alone. Where liposomes were used the liposomes prepared as described above for “Liposome Preparation”, and were first mixed with saline to form the liposomes. The pre-formed liposomes were then diluted in LT in saline to yield an immunizing solution containing liposomes at 50 mM phospholipid with 100 μg of LT per 100 μl of immunizing solution. Solutions were vortexed for 10 seconds prior to immunization.
  • The anti-LT antibodies were determined using ELISA as described above for “ELISA IgG(H+L)” 3 weeks after the second immunization. The results are shown in Table 3. LT was clearly immunogenic both with and without liposomes, and no significant difference between the groups could be detected. LT and CT are members of the family of bacterial ADP-ribosylating exotoxins (bAREs). They are organized as A:B proenzymes with the ADP-ribosyltransferase activity contained in the A subunit and the target cell binding a function of the B subunit. LT is 80% homologous with CT at the amino acid level and has a similar non-covalently bound subunit organization, stoichiometry (A:B5), the same binding target, ganglioside GM1, and is similar in size (MW ˜80,000). The similarities of LT and CT appear to influence their immunogenicity by the transcutaneous route as reflected by the similar magnitude of the antibody response to both CT and LT (Tables 1 and 3).
    TABLE 3
    Anti-LT antibodies
    Group ELISA Units SEM
    LT in saline 23,461 (20,262-27,167)
    LT + 50 mM Liposomes 27,247 (19,430-38,211)
  • Example 4
  • C57BL/6 mice at 6 to 8 weeks of age were immunized transcutaneously as described above for “Immunization Procedure”, in groups of five mice. The mice were immunized once using 100 μl of immunization solution prepared as follows: LT was mixed in saline to make an immunizing solution containing 100 μg of LT per 100 μl of saline. The solution was vortexed for 10 seconds prior to immunization.
  • The anti-LT antibodies were determined using ELISA as described above for “ELISA IgG (H+L)” 3 weeks after the single immunization. The results are shown in Table 4. LT was clearly immunogenic with a single immunization and antibodies were produced by 3 weeks. Rapid enhancement of antibody titers and responses to single immunization would be a useful aspect of the transcutaneous immunization method. It is conceivable that a rapid single immunization would be useful in epidemics, for travelers, and where access to medical care is poor.
    TABLE 4
    Anti-LT antibodies
    Mouse Number ELISA Units
    5141 6,582
    5142 198
    5143 229
    5144 6,115
    5145 17,542
    Geo Mean 2,000
  • Example 5
  • C57BL6 mice at 8 to 12 weeks of age were immunized transcutaneously as described above for “Immunization Procedure”, in groups of five mice. The mice were immunized once using 100 μl of immunization solution prepared as follows: CT was mixed in saline to make an immunizing solution containing 100 μg of CT per 100 μl of saline. The solution was vortexed for 10 seconds prior to immunization.
  • The anti-CT antibodies were determined using ELISA as described above for “ELISA IgG (H+L)” 3 weeks after the single immunization. The results are shown in Table 5. CT was highly immunogenic with a single immunization. Rapid enhancement of antibody titers and responses to single immunication may be a useful aspect of the transcutaeous immunization method. It is conceivable that a rapid single immunization would be useful in epidemics, for travelers, and where access to medical care is poor.
    TABLE 5
    Anti-CT antibodies
    Mouse Number ELISA Units
    2932 18,310
    2933 30,878
    2934 48,691
    2935 7,824
    Geo Mean 21,543
  • Example 6
  • BALB/c mice at 6 to 8 weeks of age were immunized transcutaneously as described above for “Immunization Procedure”, in groups of five mice. The mice were immunized at 0 and 3 weeks using 100 μl of immunization solution prepared as follows: ETA was mixed in saline to make an immunizing solution containing 100 μg of ETA per 100 μl of saline for the group receiving ETA alone. Where liposomes were used, the liposomes were prepared as described above for “Liposome Preparation”, and were first mixed with saline to form the liposomes. The pre-formed liposomes were then diluted with ETA in saline to yield an immunizing solution containing liposomes at 50 mM phospholipid with 100 μg of ETA per 100 μl of immunizing solution. Solutions were vortexed for 10 seconds prior to immunization.
  • The antibodies were determined using ELISA as described above for “ELISA IgG(H+L)” on sera 3 weeks after the second immunization. The results are shown in Table 6. ETA was clearly immunogenic both with and without liposomes, and no significant difference between the groups could be detected. ETA differs from CT and LT in that ETA is a single 613 amino acid peptide with A and B domains on the same peptide and binds to an entirely different receptor, the α2-macroglobulin receptor/low density lipoprotein receptor-related protein (Kounnas et al., 1992). Despite the dissimilarities between ETA and CT in size, structure, and binding target, ETA also induced a transcutaneous antibody response.
    TABLE 6
    Anti-ETA antibodies
    Group ELISA Units SEM
    ETA in saline 3,756 (1,926-7,326)
    ETA + 50 mM Liposomes 857   (588-1,251)
  • Example 7
  • BALB/c mice at 6 to 8 weeks of age were immunized transcutaneously as described above for “Immunization Procedure”, in groups of five mice. The mice were immunized using 100 μl of immunization solution which was prepared as follows: CT was mixed in saline to make 100 μg of CT per 100 μl of immunizing solution, LT was mixed in saline to make 100 μg of LT per 100 μl of immunizing solution, ETA was mixed in saline to make 100 μg of ETA per 100 μl of immunizing solution, and CT and BSA were mixed in saline to make 100 μg of CT per 100 μl of immunizing solution and 200 μg of BSA per 100 μl of immunizing solution. Solutions were vortexed for 10 seconds prior to immunization.
  • The mice were immunized transcutaneously at 0 and 3 weeks and the antibody levels were determined using ELISA as described above for “ELISA IgG Subclass”, three weeks after the boosting immunization and compared against the pre-immune sera. The IgG subclass response to CT, BSA and LT had similar levels of IgG1 and IgG2a reflecting activation of T help from both Th1 and Th2 lymphocytes (Seder and Paul, 1994), whereas the IgG subclass response to ETA consisted of almost exclusively IgG1 and IgG3, consistent with a Th2-like response (Table 7). Thus, it appears that all IgG subclasses can be produced using transcutaneous immunization.
    TABLE 7
    IgG subclasses of induced antibodies
    Imm. Antibody IgG1 IgG2a IgG2b IgG3
    Antigen Specificity (μg/μl) (μg/μl) (μg/μl) (μg/μl)
    CT CT 134 25 27 0
    CT + BSA BSA 108 17 12 5
    LT LT 155 28 10 8
    ETA ETA 50 0 1 10
  • Example 8
  • BALB/c mice at 6 to 8 weeks of age were immunized transcutaneously as described above for “Immunization Procedure”, in groups of five mice. The mice were immunized using 100 μl of immunization solution which was prepared as follows: LT was mixed in saline to make an immunizing solution containing 100 μg of LT per 100 μl of saline for the group receiving LT alone, CT was mixed in saline to make an immunizing solution containing 100 μg of CT per 100 μl of saline for the group receiving CT alone, ETA was mixed in saline to make an immunizing solution containing 100 μg of ETA per 100 μl of saline for the group receiving ETA alone, and BSA and CT were mixed in saline to make an immunizing solution containing 100 μg of BSA and 100 μg of CT per 100 μl of saline for the group receiving BSA and CT.
  • The mice were immunized transcutaneously at 0 and 3 weeks and the antibody levels were determined using ELISA as described above for “ELISA IgE”, one week after the boosting immunization and compared against the pre-immune sera. As shown in Table 8, no IgE antibodies were found although the sensitivity of detection was 0.003 μg/ml. IgG antibodies were determined in the same mice using “ELISA IgG(H+L)” on sera 3 weeks after the second immunization. The IgG antibody response to LT, ETA, CT and BSA are shown to indicate that the animals were successfully immunized and responded with high titers of antibodies to the respective antigens.
    TABLE 8
    IgE antibodies to LT, ETA, CT and BSA
    Antibody
    Group Specificity IgE (μg/ml) IgG (ELISA Units)
    LT Anti-LT 0 23,461
    ETA Anti-ETA 0 3,756
    CT Anti-CT 0 39,828
    CT + BSA Anti-BSA 0 8,198
  • Example 9
  • BALB/c mice at 6 to 8 weeks of age immunized transcutaneously as described above for “Immunization Procedure”, in groups of five mice. The mice were immunized at 0 and 3 weeks using 100 ml of immunization solution which was prepared as follows: CT was mixed in saline to make an immunizing solution containing 100 mg of CT per 100 ml of immunizing solution. The immunization solution was vortexed for 10 seconds prior to immunization.
  • The mice were immunized transcutaneously at 0 and 3 weeks and the antibody levels were determined using ELISA as described above for “ELISA IgG(H+L)” and “ELISA IgG(γ)”. Determinations were done at 1 and 4 weeks after the initial immunization, and compared against the pre-immune sera. As shown in Table 9, high levels of anti-CT IgG(γ) antibodies were induced by CT in saline. Small amounts of IgM could be detected by using IgM(μ) specific secondary antibody. By 4 weeks, the antibody response was primarily IgG. Data are reported in ELISA units.
    TABLE 9
    IgG(γ) and IgM(μ)
    Imm. Group Week IgG(γ) IgM(μ)
    CT 1 72 168
    CT 4 21,336 38
    L( ) + CT 1 33 38
    L( ) + CT 4 22,239 70
  • Example 10
  • BALB/c mice at 6 to 8 weeks of age were immunized transcutaneously as described above for “Immunization Procedure”, in groups of five mice. The mice were immunized once using 100 μl of immunization solution prepared as follows: CT was mixed in saline to make an immunizing solution containing 100 μg of CT per 100 μl of saline. The solution was vortexed for 10 seconds prior to immunization. The mice were immunized transcutaneously at 0 and 3 weeks. Antibody levels were determined using ELISA as described above for “ELISA IgG (H+L)” 5 weeks after the boosting immunization, and compared against pre-immune sera. As shown in Table 10, serum anti-CT IgA antibodies were detected.
    TABLE 10
    Anti-CT IgA antibodies
    Mouse Number IgA (ng/ml)
    1501 232
    1502 22
    1503 41
    1504 16
    1505 17
  • Example 11
  • BALB/c mice at 6 to 8 weeks of age were immunized transcutaneously as described above for “Immunization Procedure”, in groups of five mice. The mice were immunized using 100 μl of immunization solution which was prepared as follows: CT was mixed in saline to make an immunizing solution containing 100 μg of CT per 100 μl of immunizing solution. The immunization solution was vortexed for 10 seconds prior to immunization.
  • The mice were immunized with 100 μl of immunizing solution transcutaneously at 0 and 3 weeks and the antibody levels were determined using ELISA as described above for “ELISA IgG(H+L)” and “ELISA IgG(γ)”. Antibody determinations were done at 8 weeks after the initial immunization and compared against the pre-immune sera. As shown in Table 11, high levels of serun anti-CT antibodies were induced by CT in saline. Lung wash IgG could be detected by ELISA using IgG(H+L) or IgG(γ) specific antibody. The antibody found on the lung mucosal surface is diluted by the lavage method used to collect mucosal antibody and, thus, the exact amounts of antibody detected are not as significant as the mere presence of detectable antibody.
  • Lung washes were obtained after sacrificing the mouse. The trachea and lungs were exposed by gentle dissection and trachea was transected above the bifurcation. A 22 gauge polypropylene tube was inserted and tied off on the trachea to form a tight seal at the edges. Half a milliliter of PBS was infused using a 1 ml syringe attached to the tubing and the lungs were gently inflated with the fluid. The fluid was withdrawn and reinfused for a total of 3 rounds of lavage. The lung wash was then frozen at −20° C.
  • Table 11 shows the IgG(H+L) and IgG(γ) antibody response to cholera toxin in the sera and lung washes at 8 weeks. Data are expressed in ELISA units. Antibodies were clearly detectable for all mice in the lung washes. The presence of antibodies in the mucosa may be important for protection against mucosally active diseases.
    TABLE 11
    Mucosal Antibody to CT
    Animal# Imm. Group IgG(H + L) IgG(γ) Source
    1501 CT 133 34 Lungs
    1502 CT 75 12 Lungs
    1503 CT 162 28 Lungs
    1504 CT 144 18 Lungs
    1505 CT 392 56 Lungs
    Geo Mean 156 26
    1501 CT 34,131 13,760 Sera
    1502 CT 11,131 2,928 Sera
    1503 CT 21,898 10,301 Sera
    1504 CT 22,025 8,876 Sera
    1505 CT 34,284 10,966 Sera
    Geo Mean 23,128 8,270
  • Example 12
  • BALB/c mice were immunized transcutaneously at 0 and 3 weeks as described above for “Immunization Procedure”, in groups of four mice. Liposomes were prepared as described above for “Liposome Preparation”, and were first mixed with saline to form the liposomes. The pre-formed liposomes were then diluted with either CT, CTA or CTB in saline to yield an immunizing solution containing liposomes at 50 mM phospholipid with 50 μg of antigen (CT, CTA or CTB) per 100 μl of immunizing solution. Solutions were vortexed for 10 seconds prior to immunization.
  • The antibodies were determined using ELISA as described above for “ELISA IgG(H+L)”, one week after the boosting immunization and compared against the pre-immune sera. The results are shown in Table 12. CT and CTB were clearly immunogenic whereas CTA was not. Thus, the B subunit of CT is necessary and sufficient to induce a strong antibody response.
    TABLE 12
    Antibodies to CT, CTA and CTB
    Group Anti-CT Anti-CTA Anti-CTB
    CT + 50 mM Liposomes 12,636 136 7,480
    CTB + 50 mM Liposomes 757 20 1,986
    CTA + 50 mM Liposomes 0 0 0
  • Example 13
  • BALB/c mice were immunized transcutaneously as described above for “Immunization Procedure”, in groups of five mice. Mice were immunized at 0 and 3 weeks with 100 μg of diphtheria toxoid and 10 μg of pertussis toxin per 100 μl of saline solution. Solutions were vortexed for 10 seconds prior to immunization.
  • The antibodies were quantitated using ELISA as described for “ELISA IgG(H+L)”. Anti-diphtheria toxoid antibodies were detected only in animals immunized with both pertussis toxin and diphtheria toxoid. The highest responder had anti-diphtheria toxoid antibody ELISA units of 1,038. Thus, a small amount of pertussis toxin acts as an adjuvant for diphtheria toxoid antigen. The toxoid alone did not induce an immune response suggesting that the toxoiding process has affected the portion of the molecule responsible for the adjuvant effects found in the ADP-ribosylating exotexin.
    TABLE 13
    Antibody to Diphtheria
    Mouse Number Immunizing Antigen IgG ELISA Units
    4731 DT + PT 1,039
    4732 DT + PT 1
    4733 DT + PT 28
    4734 DT + PT 15
    4735 DT + PT 20
    4621 DT 0
    4622 DT 0
    4623 DT 0
    4624 DT 0
    4625 DT 0
  • Example 14
  • BALB/c mice were immunized transcutaneously as described above for “Immunization Procedure”, in groups of five mice. Mice were immunized once at 0 weeks with 50 μg of pertussis toxin per 100 μl of saline solution. The solution was vortexed for 10 seconds prior to immunization.
  • The antibodies were quantitated using ELISA as described for “ELISA IgG(H+L)”. Anti-pertussis toxin antibodies were detected at 8 weeks in animals immunized with pertussis. The highest responder had anti-petussis toxin antibody ELISA units of 73. Thus, pertussis toxin acts as an adjuvant for itself and immunizes after a single immunization.
    TABLE 14
    Antibody to Pertussis
    Mouse Number Immunizing Antigen IgG ELISA Units
    4731 PT 56
    4732 PT 60
    4733 PT 3
    4734 PT 13
    4735 PT 73
  • Example 15
  • BALB/c mice were immunized transcutaneously as described above for “Immunization Procedure”, in groups of five mice. Mice were immunized once at 0 weeks with 50 μg of tetanus toxoid and 100 μg of cholera toxin per 100 μl of saline solution. The solution was vortexed for 10 seconds prior to immunization.
  • The antibodies were quantitated using ELISA as described for “ELISA IgG(H+L)”. Anti-tetanus toxoid antibodies were detected at 8 weeks in animal 5173 at 443 ELISA units.
  • Example 16
  • The possibility that oral immunization occurred through grooming after epicutaneous application and subsequent washing of the site of application was evaluated using 125I-labeled CT to track the fate of the antigen/adjuvant. Mice were anesthetized, transcutaneously immunized as described above for “Immunization Procedure” with 100 μg of 125I-labeled CT (150,000 cpm/μg CT). Control mice remained anesthetized for 6 hours to exclude grooming, and experimental mice were anesthetized for one hour and then allowed to groom after washing. Mice were sacrificed at 6 hours and organs weighed and counted for 125I on a Packard gamma counter. A total of 2-3 μg of CT was detected on the shaved skin at the site of immunization (14,600 cpm/μg tissue) while a maximum of 0.5 μg of CT was detected in the stomach (661 cpm/μg tissue) and intestine (9 cpm/μg tissue).
  • Oral immunization (n=5) with 10 μg if CT in saline at 0 and 3 weeks (without NaHCO3) induced a 6 week mean IgG antibody response of <1,000 ELISA units whereas transcutaneous immunization with 100 μg of CT, shown above to result in less than 5 μg of CT retained in the skin after washing, resulted in an anti-CT response of 42,178 ELISA units at 6 weeks. Induction of an immune response to orally fed CT requires the addition of NaHCO3 to the immunizing solution (Piece, 1978; Lycke and Holmgren, 1986). Thus, oral immunization does not significantly contribute to the antibodies detected when CT is applied epicutaneously to the skin.
  • Example 17
  • In vivo evidence of Langerhans cell activation was obtained using cholera toxin (CT) in saline applied epicutaneously to the skin, specifically the ears of the mouse, where large populations of Langerhans cells can be readily visualized (Enk et al., 1993; Bacci et al., 1997), and staining for major histocompatibility complex (MHC) class II molecules which is upregulated in activated Langerhans cells (Shimada et al., 1987).
  • BALB/c mouse ears were coated on the dorsal side with either 100 μg of CT in saline, 100 μg of CTB in saline, saline alone, or an intradermal injection of the positive controls 100 μg LPS or 10 μg TNFα, for one hour while the mouse was anesthetized. The ears were then throughly washed and, after 24 hours, the ears were removed and epidermal sheets were harvested and stained for MHC class II expression as described by Caughman et al. (1986). Epidermal sheets were stained with MKD6 (anti-I-Ad) or negative control Y3P (anti-I-Ak), and goat anti-mouse FITC F(ab)2 was used as a second step reagent. Mice transcutaneously immunized on the ear (as described above without shaving) had previously been found to have anti-CT antibodies of 7,000 ELISA units three weeks after a single immunization.
  • Enhanced expression of MHC class II molecules as detected by staining intensity, the reduced number of Langerhans cells (especially with cholera toxin), and changes in Langerhans cell morphology were found in the epidermal sheets of the mice immunized with CT and CTB comparable to controls (FIG. 1), suggesting that the Langerhans cells were activated by the epicutaneously applied cholera toxin (Aiba and Katz, 1990; Enk et al., 1993).
  • Example 18
  • Langerhans cells represent the epidermal contingent of a family of potent accessory cells termed ‘dendritic cells’. Langerhans cells (and perhaps related cells in the dermis) are thought to be required for immune responses directed against foreign antigens that are encountered in skin. The ‘life cycle’ of the Langerhans cell is characterized by at least two distinct stages. Langerhans cells in epidermis (the ‘sentinels’) can ingest particulates and process antigens efficiently, but are weak stimulators of unprimed T cells. In contrast, Langerhans cells that have been induced to migrate to lymph nodes after contact with antigen in epidermis (the ‘messengers’) are poorly phagocytic and have limited antigen-processing capabilities, but are potent stimulators of naive T cells. If Langerhans cells are to fulfill both their ‘sentinel‘ and ‘messenger’ roles, they must be able to persist in epidermis, and also be able to exit epidermis in a controlled fashion after exposure to antigen. Thus, regulation of Langerhans cell-keratinocyte adhesion represents a key control point in Langerhans cell trafficking and function. Langerhans cells express E-cadherin (Blauvelt et al., 1995), a homophilic adhesion molecule that is prominently represented in epithelia. Keratinocytes also express this adhesion molecule, and E-cadherin clearly mediates adhesion of murine Langerhans cells to keratinocytes in vitro. It is known that E-cadherin is involved in the localization of Langerhans cells in epidermis. See Stingl et al. (1989) for a review of the characterization and properties of Langerhans cells and keratinocytes.
  • The migration of epidermal Langerhans cells (LC) and their transport of antigen from the skin to draining lymph nodes are known to be important in the induction of cutaneous immune responses, such as contact sensitization. While in transit to the lymph nodes, Langerhans cells are subject to a number of phenotypic changes required for their movement from the skin and acquisition of the capacity for antigen presentation. In addition to the upregulation of MHC class II molecules, are alterations in the expression of adhesion molecules that regulate interactions with the surrounding tissue matrix and with T lymphocytes. The migration of the Langerhan cell is known to be associated with a marked reduction in the expression of E-cadherin (Schwarzenberger and Udey, 1996, and a parallel upregulation of ICAM-1 (Udey, 1997).
  • Transcutaneous immunization with bacterial ADP ribosylating exotoxins (bARE's) target the Langerhans cells in the epidermis. The bAREs activate the Langerhans cell, transforming it from its sentinel role to its messenger role. Ingested antigen is then taken to the lymph node where it is presented to B and T cells (Streilein and Grammer, 1989; Kripke et al., 1990; Tew et al., 1997). In the process, the epidermal Langerhans cell matures into an antigen-presenting dendritic cell in the lymph node (Schuler and Steinman, 1985); lymphocytes entering a lymph node segregate into B-cell follicles and T-cell regions. The activation of the Langerhans cell to become a migratory Langerhans cell is known to be associated with not only a marked increase in MHC class II molecules, but also marked reduction in the expression of E-cadherin, and upregulation of ICAM-1.
  • We envision that cholera toxin (CT) and its B subunit (CTB) upregulate the expression of ICAM-1 and downregulate the expression of E-cadherin on Langerhans cells as well as upregulate the expression of MHC class II molecules on the Langerhans cell. CT or CTB acts as an adjuvant by freeing the sentinel Langerhans cell to present antigens such as BSA or diphtheria toxoid phagocytosed by the Langerhans cell at the same location and time as the encounter with the CT or CTB when they are acting as adjuvant. The activation of a Langerhans cells to upregulate the expression of ICAM-1 and dowregulate the expression of E-cadherin may be mediated by cytokine release including TNFα and IL-1β from the epidermal cells or the Langerhans cells themselves.
  • This method of adjuvancy for transcutaneous immunization is envisioned to work for any compound that activates the Langerhans cell. Activation could occur in such manner as to downregulate the E-cadherin and upregulate ICAM-1. Langerhans cells would then carry antigens made of mixtures of such Langerhans cell-activating compounds and antigens (such as diphtheria toxoid or BSA) to the lymph nodes where the antigens are presented to T cells and evoke an immune response. Thus, the activating substance such as a bARE can be used as an adjuvant for an other wise transcutaneously non-immunogenic antigen such as Diphtheria toxoid by activating the Langerhans cell to phagocytose the antigen such as diphtheria toxoid, migrate to the lymph node, mature into a dendritic cell, and present the antigen to T cells.
  • The T-cell helper response to antigens used in transcutaneous immunization may be influenced by the application of cytokines and/or chemokines. For example, interleukin-10 (IL-10) may skew the antibody response towards a Th2 IgG1/IgE response whereas anti-IL-10 may enhance the production of IgG2a (Bellinghausen et al., 1996).
  • The disclosures of all patents, as well as all other printed documents, cited in this specification are incorporated herein by reference in their entirety.
  • While the present invention has been described in connection with what is presently considered to be practical and preferred embodiments, it is understood that the present invention is not to be limited or restricted to the disclosed embodiments but, on the contrary, is intended to cover various modifications and equivalent arrangements included within the spirit and scope of the appended claims.
  • Thus, it is to be understood that variations in the described invention will be obvious to those skilled in the art without departing from the novel aspects of the present invention and such variations are intended to come within the scope of the claims below.
  • REFERENCES
    • Aiba, S., and Katz, S. I. (1990) Phenotypic and functional characteristics of in vivo-activated Langerhans cells. J. Immunol., 145:2791-2796
    • Alving, C. R., et al. (1986) Effectiveness of liposomes as potential carriers of vaccines: Applications to cholera toxin and human malaria sporozoite antigen. Vaccine, 4:166-172.
    • Alving, C. R., and Wassef, N. M. (1994) Cytotoxic T lymphocytes induced by liposomal antigens: Mechanisms of immunological presentation. AIDS Res. Hum. Retro., 10(sup. 2):S91-S94.
    • Alving, C. R., et al. (1993) The preparation and use of liposomes in immunological studies. In: Liposome Technology, vol. 3, (Ed., Gregoriadis, G.), CRC Press, Boca Raton, pp. 317-343.
    • Antel, J. P., et al. (1996) Immunotherapy for multiple sclerosis: From theory to practice. Nature Medicine, 2:1074-1075.
    • Ausubel, F. M., et al. (1996) Current Protocols in Molecular Biology, Wiley, New York.
    • Bacci, S., et al. (1997) High and low doses of haptens dictate whether dermal or epidermal antigen-presenting cells promote contact hypersensitivity. Eur. J. Immunol., 27:442-448.
    • Bathurst, I. C., et al. (1993) An experimental vaccine cocktail for Plasmodium falciparum malaria. Vaccine, 11:449-456.
    • Bellinghausen, I. et al. (1996) Epidermal cells enhance interleukin 4 and immunoglobulin E production after stimulation with protein allergen. J. Invest. Dermatol., 107:582-588.
    • Blauvelt, A., et al. (1995) Human Langerhans cells express E-cadherin. J. Invest. Dermatol., 104:293-296.
    • Blum, H. E. (1995) Variants of hepatitis B, C and D viruses: Molecular biology and clinical significance. Digestion, 56:85-95.
    • Bodanszky, M. (1993) Peptide Chemistry, Springer-Verlag, New York.
    • Bos, J. D. (1997) The skin as an organ of immunity. Clin. Exp. Immunol., 107 (suppl. 1):3-5.
    • Bowen, J. C., et al. (1994) Cholera toxin acts as a potent adjuvant for the induction of cytotoxic T-lymphocyte responses with non-replicating antigens. Immunology, 81:338-342.
    • Burnette, W. N., et al. (1994) Recombinant microbial ADP-ribosylating toxins of Bordetella pertussis, Vibrio cholerae, and enterotoxigenic Escherichia coli: Structure, function, and toxoid vaccine Development. In: Bioprocess Technology, (Eds. Burnette, W. N., et al.), pp. 185-203.
    • Caughman, S. W., et al. (1986) Ia+ murine epidermal Langerhans cells are deficient in surface expression of the class I major histocompatibility complex. Proc. Natl. Acad. Sci. USA, 83:7438-7442.
    • Chang, S. P., et al. (1989) Generalized immunological recognition of the major merozoite surface antigen (gp 195) of Plasmodium falciparum. Proc. Natl. Acad. Sci. USA, 86:6343-6347.
    • Chang, S. P., et al. (1992) A carboxyl-terminal fragment of Plasmodium falciparum gp195 expressed by a recombinant baculovirus induces antibodies that completely inhibit parasite growth. J. Immunol., 139:548-555.
    • Chang, S. P., et al. (1994) Regulation of antibody specificity to Plasmodium falciparum merozoite surface protein-1 by adjuvant and MHC haplotype. J. Immunol., 152:3483-3490.
    • Craig, S. W., and Cuatrecasas, P. (1975) Mobility of the cholera toxin receptors on rat lymphocyte membranes. Proc. Natl. Acad. Sci. USA, 72:3384-2288.
    • Dahl, M. V. (1996) Atopic dermatitis. In: Clinical Inmunodermatology, 3rd Ed. Mosby, St. Louis, pp. 345-352.
    • Delenda, C., et al. (1994) Analysis of C-terminally truncated dengue 2 and dengue 3 virus envelope glycoproteins: Processing in insect cells and immunogenic properties in mice. J. Gen. Virol., 75:1569-1578.
    • Deprez, B., et al. (1996) Comparative efficiencies of simple lipopeptide constructs for in vivo induction of virus-specific CTL. Vaccine, 14:375-382.
    • Deutscher, M. P. (1990) Guide to Protein Purification, Academic Press, San Diego.
    • Dragunsky, E. M., et al. (1992) Experimental evaluation of antitoxic protective effect of new cholera vaccines in mice. Vaccine, 10:735-736.
    • Elson, C. O., and Dertzbaugh, M. T. (1994) Mucosal adjuvants. In: Handbook of Mucosal Immunology (Eds. Ogra, P. L., et al.) Academic Press, San Diego, p. 391.
    • Enk, A. H., et al. (1993) An essential role for Langerhans cell-derived IL-1 beta in the initiation of primary immune responses in skin. J. Immunol., 150:3698-3704.
    • Fonseca, B. A., et al. (1994) Recombinant vaccinia viruses co-expressing dengue-1 glycoproteins prM and E induce neutralizing antibodies in mice. Vaccine, 12:279-285.
    • Frankenburg, S., et al. (1996) Effective immunization of mice against cutaneous leishmaniasis using an intrinsically adjuvanted synthetic lipopeptide vaccine. Vaccine, 14:923-929.
    • Fries, L. F., et al. (1992a) Liposomal malaria vaccine in humans: A safe and potent adjuvant strategy. Proc. Natl. Acad. Sci. USA, 89:358-362.
    • Fries, L. F., et al. (1992b) Safety, immunogenicity, and efficacy of a Plasmodium falciparum vaccine comprising a circumsporozoite protein repeat region peptide conjugated to Pseudomonas aeruginosa toxin A. Infect. Immun., 60:1834-1839.
    • Glenn, G. M., et al. (1995) Murine IgG subclass antibodies to antigens incorporated in liposomes containing lipid A. Immunol. Lett., 47:73-78.
    • Goeddel, D. V. (1990) Gene Expression Technology, Academic Press, San Diego.
    • Gregoriadis, G. (1993) Liposome Preparation and Related Techniques, 2nd Ed., CRC Press, Boca Raton.
    • Herrington, D. A., et al. (1991) Safety and immunogenicity of a recombinant sporozoite malaria vaccine against Plasmodium vivax. Am. J. Trop. Med. Hyg., 45:695-701.
    • Jahrling, P. B., et al. (1996) Passive immunization of Ebola virus-infected cynomolgus monkeys with immunoglobulin from hyperimmune horses. Arch. Virol. Suppl., 11:135-140.
    • Janeway, C. A., and Travers, P. (1996). Immunobiology, Churchill Livingstone, New York.
    • Janson, J.-C., and Ryden, L. (1989) Protein Purification, VCH, New York.
    • Katkov, W. N. (1996) Hepatitis vaccines. Med. Clin. North Am., 80:189-200.
    • Khusmith, S., et al. (1991) Protection against malaria by vaccination with sporozoite surface protein 2 plus CS protein. Science, 252:715-718.
    • Kounnas, M. Z., et al. (1992) The α2-macroglobulin receptor/low density lipoprotein receptor-related protein binds and internalizes Pseudomonas exotoxin A. J. Biol. Chem., 267:12420-12423.
    • Kriegler, M. (1990) Gene Transfer and Expression, Stockton Press, New York.
    • Kripke, M. L., et al. (1990) Evidence that cutaneous antigen-presenting cells migrate to regional lymph nodes during contact sensitization. J. Immunol., 145:2833-2838.
    • Krueger, K. M., and Barbieri, J. T. (1995) The family of bacterial ADP-ribosylating exotoxins. Clin. Microbiol. Rev., 8:34-47.
    • Lee, A., and Chen, M. (1994) Successful immunization against gastric infection with Helicobacter species: Use of a cholera toxin B-subunit-whole-cell vaccine. Infect. Immun., 62:3594-3597.
    • Leung, D. Y. (1997) Atopic dermatitis: Immunobiology and treatment with immune modulators. Clin. Exp. Immunol., 107 (Suppl. 1):25-30.
    • Lycke, N., and Holmgren, J. (1986) Strong adjuvant properties of cholera toxin on gut mucosal immune responses to orally presented antigens. Immunology, 59:301-308.
    • Lieberman, J. M., and Greenberg, D. P. (1996) Hepatitis A and B vaccines in children. Adv. Pediatr. Infect. Dis., 11:333-363.
    • Malik, A., et al. (1991) Human cytotoxic T lymphocytes against the Plasmodium falciparum circumsporozoite protein. Proc. Natl. Acad. Sci. USA, 88:3300-3304.
    • Mast, E. E., and Krawczynski, K. (1996) Hepatitis E: An overview. Annu. Rev. Med., 47:257-266.
    • Migliorini, P., et al. (1993) Malaria vaccine: Immunization of mice with a synthetic T cell helper epitope alone leads to protective immunity. Eur. J. Immunol., 23:582-585.
    • Morein, B., and Simons, K. (1985) Subunit vaccines against enveloped viruses: Virosomes, micelles and other protein complexes. Vaccine, 3:83-93.
    • Murray, E. J. (1991) Gene Transfer and Expression Protocols. Humana Press, Clifton, N.J.
    • Nohria, A., and Rubin, R. H. (1994) Cytokines as potential vaccine adjuvants. Biotherapy, 7:261-269.
    • Paul, A., and Cevc, G. (1995) Noninvasive administration of protein antigens: Transdermal immunization with bovine serum albumin in transfersomes. Vaccine Res., 3:145-164.
    • Paul, A., et al. (1995) Transdermal immunization with large proteins by means of ultradeformable drug carriers. Eur. J. Immunol., 25:3521-3524, 1995.
    • Paul, W. E., and Seder, R. A. (1994) Lymphocyte responses and cytokines. Cell, 76:241-251.
    • Peguet-Navarro, J., et al. (1995) Inhibitory effect on human Langerhans cell antigen presenting function. Adv. Exp. Med. Biol., 378:359-361.
    • Pessi, A., et al. (1991) Lack of H-2 restriction of the Plasmodium falciparum (NANP) sequence as multiple antigen peptide, Eur. J. Immunol., 24:2273-2276.
    • Pierce, N. F. (1978) The role of antigen form and function in the primary and secondary intestinal immune responses to cholera toxin and toxoid in rats. J. Exp. Med., 148:195-206.
    • Pierce, N. F., and Reynolds, H. Y. (1974) Immunity to experimental cholera. I. Protective effect of humoral IgG antitoxin demonstrated by passive immunization. J. Immunol., 113:1017-1023.
    • Plotkin, S. A., and Mortimer Jr., E. A. (1994) Vaccines, 2nd Ed., W. B. Saunders, Philadelphia.
    • Porgador, A., et al. (1997) Intranasal immunization with CTL epitope peptides from HIV-1 or ovalbumin and the mucosal adjuvant cholera toxin induces peptide-specific CTLs and protection against tumor development in vivo. J. Immunol., 158:834-841.
    • Rappuoli, R., et al. (1995) Genetic detoxification of bacterial toxins: A new approach to vaccine development. Int. Archiv. Allergy Immunol., 108:327-333.
    • Rappuoli, R., et al. (1996) New vaccines against bacterial toxins. Adv. Exp. Med. Biol., 397:55-60.
    • Ribi, H. O., et al. (1988). Three-dimensional structure of cholera toxin penetrating a lipid membrane. Science, 239:1272-1276.
    • Richards, R. L., et al. (1995) A compendium of vaccine adjuvants and excipients. In: Vaccine Design (Eds., Powell, M. F., and Newman, M. J.), Plenum, New York.
    • Roberts, M. S., and Walker, M. (1993) Water, the most natural penetration enhancer. In: Pharmaceutical Skin Penetration Enhancement (Eds., Walters, K. A., and Hadgraft, J.), Marcel Dekker, New York.
    • Saloga, J., et al. (1996) Superantigens. Exp. Dermatol., 5:65-71.
    • Schneerson, R. E., et al. (1996) A toxoid vaccine for pertussis as well as diphtheria? Lessons to be relearned. Lancet 348:1289-1292.
    • Schuler, G., and Steinman, R. M. (1985) Murine epidermal Langerhans cells mature into potent immunostimulatory dendritic cells in vitro. J. Exp. Med., 161:526-546.
    • Schwarzenberger, K., and Udey, M. C. (1996) Contact allergens and epidermal proinflammatory cytokines modulate Langerhans cell E-cadherin expression in situ. J. Invest. Dermatol., 106:553-558.
    • Scopes, R. K. (1993) Protein Purification, Springer-Verlag, New York.
    • Seder, R. A., and Paul, W. E. (1994) Acquisition of lymphokine-producing phenotype by CD4+ T cells. Annu. Rev. Immunol., 12:635-673.
    • Shafara, A., et al. (1995) Hepatitis C. Ann. Intern. Med., 125:658-668.
    • Shankar, G., et al. (1996) Superantigen-induced Langerhans cell depletion is mediated by epidermal cell-derived IL-1α and TNFα. Cell. Immunol., 171:240-245.
    • Skeiky, Y. A. W., et al. (1995) A recombinant Leishmania antigen that stimulates human peripheral blood mononuclear cells to express a Th1-type cytokine profile and to produce interleukin 12. J. Exp. Med., 181:1527-1537.
    • Smedile, A., et al. (1994) Advances in hepatitis D virus biology and disease. Prog. Liver Dis., 12:157-175.
    • Smucny, J. J., et al. (1995) Murine immunoglobulin G subclass responses following immunization with live dengue virus or a recombinant dengue envelope protein. Am. J. Trop. Med. Hyg., 53:432-437.
    • Stacey, K. J., et al. (1996) Macrophages ingest and are activated by bacterial DNA. J. Immunol., 157:2116-2122.
    • Stingl, G., et al. (1989) The immune functions of epidermal cells. Immunol. Ser., 46:3-42.
    • Streilein, J. W., and Grammer, S. F. (1989) In vitro evidence that Langerhans cells can adopt two functionally distinct forms capable of antigen presentation to T lymphocytes. J. Immunol., 143:3925-3933.
    • Summers, M. D., and Smith, G. E. (1987) A manual of methods for baculovirus vectors and insect cell culture procedure. Texas Agricultural-Experiment Station Bulletin, No. 1555.
    • Tam, J. P. (1988) Synthetic peptide vaccine design: Synthesis and properties of a high-density multiple antigenic peptide system. Proc. Natl. Acad. Sci. USA, 85:5409-5413.
    • Tew, J. G., et al. (1997) Follicular dendritic cells and presentation of antigen and costimulatory signals to B cells. Immunol. Rev., 156:39-52.
    • Trach, D. D., et al. (1997) Field trial of a locally produced, killed, oral cholera vaccine in Vietnam. Lancet, 349:231-235.
    • Udey, M. C. (1997) Cadherins and Langerhans cell immunobiology. Clin. Exp. Immunol., 107 (Suppl. 1):6-8.
    • Vandenbark, A. A., et al. (1996) Treatment of multiple sclerosis with T-cell receptor peptides: Results of a double-blind pilot trial. Nature Medicine, 2:1109-1115.
    • Vreden, S. G. S., et al. (1991) Phase I clinical trial of a recombinant malaria vaccine consisting of the circumsporozoite repeat region of Plasmodium falciparum coupled to hepatitus B surface antigen, Am. J. Trop. Med. Hyg., 45:533-538.
    • Wang, R., et al. (1995) Induction of protective polyclonal antibodies by immunization with a Plasmodium yoelii circumsporozoite protein multiple antigen peptide vaccine. J. Immunol., 154:2784-2793.
    • Wertz, P. W. (1992) Liposome dramatics: Chemicals aspects of the skin lipid approach. In: Liposome Dramatics (Eds., Braun-Falco, O. et al.), Springer-Verlag, New York, pp. 38-43.
    • White, K., et al. (1993) Induction of cytolytic and antibody responses using Plasmodium falciparum repeatless circumsporozoite protein encapsulated in liposomes. Vaccine, 11:1341-1346.
    • Wiesmueller, K.-H., et al. (1991) The antibody response in BALB/c mice to the Plasmodium falciparum circumsporozoite repetitive epitope covalently coupled to synthetic lipopeptide adjuvant. Immunology, 72:109-113.
    • Wisdom, G. B. (1994) Peptide Antigens, IRL Press, Oxford.
    • Zhang, T., et al. (1995) Oral immunization with the dodecapeptide repeat of the serine-rich Entamoeba histolytica protein (SREHP) fused to the cholera toxin B subunit induces a mucosal and systemic anti-SREHP antibody response. Infect. Immun., 63:1349-1355.

Claims (11)

1-49. (canceled)
50: A method of inducing an immune response to at least one antigen comprising applying a formulation to hydrated skin of an organism, wherein the formulation comprises an antigen which is derived from a pathogen, and wherein an effective amount of the antigen induces the immune response to the at least one antigen in the organism.
51: The method of claim 50, wherein the pathogen is selected from the group consisting of bacterium, virus, fungus and parasite.
52: The method of claim 50, wherein the antigen is selected from the group consisting of carbohydrate, glycolipid, glycoprotein, lipid, protein, lipoprotein, phospholipid, and polypeptide.
53: The method of claim 50, wherein the pathogen is a live or an attenuated live virus and the antigen is expressed by the live or attenuated live virus.
54: The method of claim 51, wherein the bacterium is anthrax.
55: The method of claim 51, wherein the virus is rabies.
56: The method of claim 50, wherein the formulation is a cream or gel or emulsion or ointment or lotion or paste or solution or suspension.
57: The method of claim 50, wherein the formulation is applied with a patch.
58: The method of claim 50, wherein the formulation further comprises a dressing.
59: The method of claim 58, wherein the dressing is occlusive or non-occlusive.
US11/334,349 1996-11-14 2006-01-19 Adjuvant for transcutaneous immunization Abandoned US20060269593A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US11/334,349 US20060269593A1 (en) 1996-11-14 2006-01-19 Adjuvant for transcutaneous immunization

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US08/749,164 US5910306A (en) 1996-11-14 1996-11-14 Transdermal delivery system for antigen
US08/896,085 US5980898A (en) 1996-11-14 1997-07-17 Adjuvant for transcutaneous immunization
US09/266,803 US7037499B1 (en) 1996-11-14 1999-03-12 Adjuvant for transcutaneous immunization
US11/334,349 US20060269593A1 (en) 1996-11-14 2006-01-19 Adjuvant for transcutaneous immunization

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US09/266,803 Division US7037499B1 (en) 1996-11-14 1999-03-12 Adjuvant for transcutaneous immunization

Publications (1)

Publication Number Publication Date
US20060269593A1 true US20060269593A1 (en) 2006-11-30

Family

ID=27669393

Family Applications (3)

Application Number Title Priority Date Filing Date
US08/896,085 Expired - Lifetime US5980898A (en) 1996-11-14 1997-07-17 Adjuvant for transcutaneous immunization
US09/266,803 Expired - Fee Related US7037499B1 (en) 1996-11-14 1999-03-12 Adjuvant for transcutaneous immunization
US11/334,349 Abandoned US20060269593A1 (en) 1996-11-14 2006-01-19 Adjuvant for transcutaneous immunization

Family Applications Before (2)

Application Number Title Priority Date Filing Date
US08/896,085 Expired - Lifetime US5980898A (en) 1996-11-14 1997-07-17 Adjuvant for transcutaneous immunization
US09/266,803 Expired - Fee Related US7037499B1 (en) 1996-11-14 1999-03-12 Adjuvant for transcutaneous immunization

Country Status (14)

Country Link
US (3) US5980898A (en)
EP (4) EP2272526A3 (en)
JP (1) JP4584361B2 (en)
KR (1) KR100517028B1 (en)
CN (1) CN1279976C (en)
AP (1) AP9901540A0 (en)
AT (2) ATE274805T1 (en)
AU (1) AU744537B2 (en)
BR (1) BR9712952A (en)
CA (1) CA2272417C (en)
DE (2) DE69730534T2 (en)
IL (1) IL129919A (en)
NZ (1) NZ335749A (en)
WO (1) WO1998020734A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7848801B2 (en) 2005-12-30 2010-12-07 Tti Ellebeau, Inc. Iontophoretic systems, devices, and methods of delivery of active agents to biological interface

Families Citing this family (278)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20020032316A1 (en) * 1995-03-31 2002-03-14 Cantab Pharmaceuticals Research Limited Hapten-carrier conjugates for use in drug-abuse therapy and methods for preparation of same
US6500437B1 (en) 1995-09-22 2002-12-31 Corixa Corporation Leishmania antigens for use in the therapy and diagnosis of leishmaniasis
US6375955B1 (en) * 1995-09-22 2002-04-23 Corixa Corporation Leishmania antigens for use in the therapy and diagnosis of leishmaniasis
US6607731B1 (en) 1995-09-22 2003-08-19 Corixa Corporation Leishmania antigens for use in the therapy and diagnosis of leishmaniasis
US6365165B1 (en) 1995-09-22 2002-04-02 Corixa Corporation Leishmania antigens for use in the therapy and diagnosis of Leishmaniasis
US6613337B1 (en) 1997-02-12 2003-09-02 Corixa Corporation Leishmania antigens for use in the therapy and diagnosis of leishmaniasis
US6638517B2 (en) 1995-09-22 2003-10-28 Corixa Corporation Leishmania antigens for use in the therapy and diagnosis of leishmaniasis
DK0909323T3 (en) 1996-01-04 2007-05-21 Novartis Vaccines & Diagnostic Helicobacter pylori bacterioferritin
US5980898A (en) 1996-11-14 1999-11-09 The United States Of America As Represented By The U.S. Army Medical Research & Material Command Adjuvant for transcutaneous immunization
US6797276B1 (en) * 1996-11-14 2004-09-28 The United States Of America As Represented By The Secretary Of The Army Use of penetration enhancers and barrier disruption agents to enhance the transcutaneous immune response
US20060002949A1 (en) 1996-11-14 2006-01-05 Army Govt. Of The Usa, As Rep. By Secretary Of The Office Of The Command Judge Advocate, Hq Usamrmc. Transcutaneous immunization without heterologous adjuvant
ATE516034T1 (en) * 1997-01-02 2011-07-15 Univ Jefferson METHOD FOR MODULATING THE IMMUNE RESPONSE IN AN INFECTED MAMMAL BY TRANSMUCOSAL ADMINISTRATION OF A MODULATING AGENT
US6818222B1 (en) * 1997-03-21 2004-11-16 Chiron Corporation Detoxified mutants of bacterial ADP-ribosylating toxins as parenteral adjuvants
EP1486215A3 (en) * 1997-03-21 2006-04-12 Chiron Corporation Detoxified mutants of bacterial ADP-ribosylating toxins as parenteral adjuvants
FR2766193B1 (en) * 1997-07-18 2001-09-14 Inst Curie CHEMICAL POLYPEPTIDE COMPRISING FRAGMENT B OF TOXIN SHIGA AND PEPTIDES OF THERAPEUTIC INTEREST
US20030125278A1 (en) * 1997-08-13 2003-07-03 Tang De-Chu C. Immunization of animals by topical applications of a salmonella-based vector
US5993852A (en) * 1997-08-29 1999-11-30 Pharmaderm Laboratories Ltd. Biphasic lipid vesicle composition for transdermal administration of an immunogen
CN1263854C (en) 1997-11-06 2006-07-12 启龙股份公司 Neisserial antigens
US6914131B1 (en) 1998-10-09 2005-07-05 Chiron S.R.L. Neisserial antigens
GB9800487D0 (en) * 1998-01-09 1998-03-04 Oratol Limited Therapies
SG152917A1 (en) 1998-01-14 2009-06-29 Chiron Srl Neisseria meningitidis antigens
CA2321596C (en) * 1998-02-25 2012-05-29 Gregory M. Glenn Use of skin penetration enhancers and barrier disruption agents to enhance the transcutaneous immune response induced by adp-ribosylating exotoxin
EP1849477A1 (en) * 1998-02-25 2007-10-31 THE GOVERNMENT OF THE UNITED STATES, as represented by THE SECRETARY OF THE ARMY Use of skin penetration enhancers and barrier disruption agents to enhance the transcutaneous immune response induced by ADP-ribosylating exotoxin
EP1356821B1 (en) * 1998-02-25 2007-06-20 THE GOVERNMENT OF THE UNITED STATES, as represented by THE SECRETARY OF THE ARMY Use of skin penetration enhancers and barrier disruption agents to enhance transcutaneous immune response
JP4820000B2 (en) * 1998-05-15 2011-11-24 アラン エム グリーン Verotoxin B subunit for immunization
CA2335487A1 (en) * 1998-06-19 1999-12-23 Merieux Oravax Lt and ct in parenteral immunization methods against helicobacter infection
US6210672B1 (en) 1998-10-20 2001-04-03 Torrey Pines Institute For Molecular Studies Topical immunostimulation to induce Langerhans cell migration
US7229621B2 (en) * 1998-10-20 2007-06-12 Torrey Pines Institute For Molecular Studies Method to enhance the immunogenicity of an antigen
CN1191357C (en) * 1998-12-22 2005-03-02 博伊斯·汤普生植物研究所 Orally immunogenic bacterial enterotoxins expressed in transgenic plants
EP1148902A1 (en) * 1999-01-22 2001-10-31 PowderJect Research Limited Method of enhancing needleless transdermal powdered drug delivery
DE69901377T2 (en) * 1999-01-27 2003-01-02 Idea Ag Non-invasive skin vaccination
EP2368575B1 (en) 1999-04-08 2014-10-01 Intercell USA, Inc. Dry formulation for transcutaneous immunization
EP2322210A1 (en) 1999-04-19 2011-05-18 GlaxoSmithKline Biologicals S.A. Adjuvant composition comprising saponin and an immunostimulatory oligonucleotide
US6558670B1 (en) 1999-04-19 2003-05-06 Smithkline Beechman Biologicals S.A. Vaccine adjuvants
WO2000066741A2 (en) 1999-04-30 2000-11-09 Chiron S.P.A. Conserved neisserial antigens
US20040009936A1 (en) 1999-05-03 2004-01-15 Tang De-Chu C. Vaccine and drug delivery by topical application of vectors and vector extracts
KR100863367B1 (en) 1999-05-13 2008-10-13 와이어쓰 홀딩스 코포레이션 Adjuvant combination formulations
GB9911683D0 (en) 1999-05-19 1999-07-21 Chiron Spa Antigenic peptides
AU5315000A (en) * 1999-06-03 2000-12-28 Gregory M. Glenn Indicators for monitoring the technique of transcutaneous immunization
US20020151515A1 (en) * 1999-06-18 2002-10-17 Roberts Bruce L. Preparation and use of superior vaccines
GB9916529D0 (en) 1999-07-14 1999-09-15 Chiron Spa Antigenic peptides
US6835184B1 (en) 1999-09-24 2004-12-28 Becton, Dickinson And Company Method and device for abrading skin
GB9923060D0 (en) * 1999-09-29 1999-12-01 Chiron Spa Vaccine
GB9924351D0 (en) 1999-10-14 1999-12-15 Brennan Frank Immunomodulation methods and compositions
BR0015137A (en) 1999-10-29 2003-03-25 Chiron Spa Neisserial antigenic peptides
US7041296B1 (en) * 1999-11-12 2006-05-09 The United States Of America As Represented By The Department Of Health And Human Services Methods of treating inflammatory bowel disease using cholera toxin B subunit
GB9928196D0 (en) 1999-11-29 2000-01-26 Chiron Spa Combinations of B, C and other antigens
RU2279889C2 (en) 2000-01-17 2006-07-20 Чирон С.Р.Л. OUTER MEMBRANE VESICLE VACCINE (OMV) CONTAINING PROTEINS OF SEROGROUP B N.Meningitis OUTER MEMBRANE
CA2689666C (en) 2000-02-28 2015-02-24 Novartis Vaccines And Diagnostics S.R.L. Heterologous expression of neisserial proteins
WO2001066700A1 (en) * 2000-03-09 2001-09-13 Emory University Transcutaneous immunization for large particulate antigens
US6595947B1 (en) 2000-05-22 2003-07-22 Becton, Dickinson And Company Topical delivery of vaccines
GB0013810D0 (en) 2000-06-06 2000-07-26 Celltech Chiroscience Ltd Biological products
GB0017999D0 (en) 2000-07-21 2000-09-13 Smithkline Beecham Biolog Novel device
GB0022742D0 (en) 2000-09-15 2000-11-01 Smithkline Beecham Biolog Vaccine
PL211151B1 (en) 2000-10-18 2012-04-30 Glaxosmithkline Biolog Sa Vaccines
NZ594877A (en) 2000-10-27 2012-07-27 Novartis Vaccines & Diagnostic Nucleic acids and proteins from streptococcus groups A & B
ATE317267T1 (en) * 2000-11-07 2006-02-15 Immunovaccine Technologies Inc VACCINES WITH INCREASED IMMUNE RESPONSE AND METHOD FOR THE PRODUCTION THEREOF
PE20020530A1 (en) 2000-11-10 2002-06-18 Wyeth Corp ADJUVANT COMBINATION FORMULATIONS
BR0115646A (en) 2000-11-27 2005-12-13 Powderject Vaccines Inc Composition, particle release device, use of a composition, and method for enhancing an immune response against an antigen of interest in a patient.
US20030162733A1 (en) * 2000-11-27 2003-08-28 Haynes Joel R. Nucleic acid adjuvants
CA2437899C (en) * 2001-02-13 2012-05-15 Gregory M. Glenn Vaccine for transcutaneous immunization against etec-caused traveler's diarrhea
DE60238471D1 (en) * 2001-03-09 2011-01-13 Id Biomedical Corp Quebec Proteosome liposaccharide-VACCINE ADJUVANT
US20040109869A1 (en) * 2001-03-19 2004-06-10 Iomai Corporation Transcutaneous immunostimulation
JP2004529906A (en) * 2001-03-19 2004-09-30 イオマイ コーポレイシヨン Percutaneous immunostimulation
EP1381381A1 (en) * 2001-03-19 2004-01-21 Gregory M. Glenn Patch for transcutaneous immunization
GB0107658D0 (en) 2001-03-27 2001-05-16 Chiron Spa Streptococcus pneumoniae
GB0107661D0 (en) 2001-03-27 2001-05-16 Chiron Spa Staphylococcus aureus
WO2002093998A2 (en) * 2001-05-23 2002-11-28 Duotol Ab Suppression of allergic reactions by transdermal administration of allergens in conjunction with or conjugated to toxin subunits or fragments thereof
AU2002312380A1 (en) * 2001-06-08 2002-12-23 Becton, Dickinson And Company Device for manipulating a needle or abrader array
GB0115176D0 (en) 2001-06-20 2001-08-15 Chiron Spa Capular polysaccharide solubilisation and combination vaccines
GB0118249D0 (en) 2001-07-26 2001-09-19 Chiron Spa Histidine vaccines
ATE471990T1 (en) 2001-08-13 2010-07-15 Univ Rochester TRANSCUTANE IMMUNIZATION AGAINST PAPILLOMAVIRUS WITH VIRUS-LIKE PAPILLOMAVIRUS PARTICLES
ES2629395T3 (en) 2001-10-04 2017-08-09 Genetics Institute, Llc Methods and compositions to modulate the activity of interleukin-21
GB0124317D0 (en) 2001-10-10 2001-11-28 Celltech R&D Ltd Biological products
ATE420676T1 (en) * 2001-10-29 2009-01-15 Becton Dickinson Co DEVICE FOR DELIVERING A SUBSTANCE
US20040120964A1 (en) * 2001-10-29 2004-06-24 Mikszta John A. Needleless vaccination using chimeric yellow fever vaccine-vectored vaccines against heterologous flaviviruses
DE60228758D1 (en) 2001-12-12 2008-10-16 Novartis Vaccines & Diagnostic IMMUNIZATION AGAINST CHLAMYDIA TRACHEOMATIS
US20040137004A1 (en) * 2002-03-19 2004-07-15 Glenn Gregory M Patch for transcutaneous immunization
MY139721A (en) * 2002-04-19 2009-10-30 Cpex Pharmaceuticals Inc Pharmaceutical composition
GB0210121D0 (en) 2002-05-02 2002-06-12 Celltech R&D Ltd Biological products
EP2371392B1 (en) 2002-05-02 2015-07-08 Wyeth Holdings LLC Calicheamicin derivative-carrier conjugates
EP1534753B1 (en) 2002-05-28 2011-08-03 UCB Pharma, S.A. Peg positional isomer of an anti-tnfalpha antibody (cdp870)
GB0218921D0 (en) * 2002-08-14 2002-09-25 Glaxosmithkline Biolog Sa Novel vaccine
WO2004046177A2 (en) 2002-11-15 2004-06-03 Chiron Srl Unexpected surface proteins in neisseria meningitidis
EP1447080A1 (en) * 2003-02-13 2004-08-18 Bestewil Holding B.V. Method for producing virosome-like particles
GB0303337D0 (en) 2003-02-13 2003-03-19 Celltech R&D Ltd Biological products
CA2515779A1 (en) 2003-02-14 2004-09-02 The Curators Of The University Of Missouri Contraceptive method and compositions related to proteasomal interference
GB2398783A (en) 2003-02-26 2004-09-01 Antonio Lanzavecchia A method for producing immortalised human B memory lymphocytes
GB0307989D0 (en) * 2003-04-07 2003-05-14 Mcewen Lab Ltd Therapeutic composition
GB0308198D0 (en) 2003-04-09 2003-05-14 Chiron Srl ADP-ribosylating bacterial toxin
EP1629008A1 (en) 2003-05-21 2006-03-01 Ares Trading S.A. Tnf-like secreted protein
US20060035242A1 (en) 2004-08-13 2006-02-16 Michelitsch Melissa D Prion-specific peptide reagents
AU2004268616B2 (en) 2003-08-25 2010-10-07 3M Innovative Properties Company Delivery of immune response modifier compounds
US20050048090A1 (en) * 2003-09-03 2005-03-03 Rau Allen H. Temperature changing skin care product
GB0323103D0 (en) 2003-10-02 2003-11-05 Chiron Srl De-acetylated saccharides
IL159273A0 (en) * 2003-12-09 2004-06-01 Transpharma Medical Ltd Transdermal delivery system for sustained release of polypeptides
WO2005079841A2 (en) * 2003-12-09 2005-09-01 Iomai Corporation Gm1 binding deficient exotoxins for use as immunoadjuvants
DE602005004014T2 (en) * 2004-03-12 2008-12-11 Intercell Ag PROCESS FOR SOLUBILIZING PEPTIDE MIXTURES
US20090017056A1 (en) * 2004-06-15 2009-01-15 Iomai Corporation Skin immunization using lt-sta fusion proteins
TW200613554A (en) 2004-06-17 2006-05-01 Wyeth Corp Plasmid having three complete transcriptional units and immunogenic compositions for inducing an immune response to HIV
EP1768742A4 (en) * 2004-07-06 2007-10-17 Transpharma Medical Ltd Delivery system for transdermal immunization
CA2580137A1 (en) 2004-09-22 2006-03-30 Glaxosmithkline Biologicals S.A. Immunogenic composition
EP1676602A1 (en) 2005-01-04 2006-07-05 Institut National De La Sante Et De La Recherche Medicale (Inserm) Continuous administration of epitopes derived from protein present in atherosclerotic plaque for the treatment of atherosclerosis
ATE463515T1 (en) 2005-03-08 2010-04-15 Sigma Tau Ind Farmaceuti CHIMERIC RECOMBINANT ANTIGENS OF TOXOPLASMA GONDII
EP1865981A2 (en) 2005-04-07 2007-12-19 Chiron Corporation Cacna1e in cancer diagnosis, detection and treatment
WO2006110585A2 (en) 2005-04-07 2006-10-19 Novartis Vaccines And Diagnostics Inc. Cancer-related genes (prlr)
JP2006296511A (en) * 2005-04-15 2006-11-02 Transcutaneous Technologies Inc External preparation, method for applying external preparation, iontophoresis device, and transdermal patch
CA2610757A1 (en) * 2005-06-10 2006-12-14 Transpharma Medical, Ltd. Patch for transdermal drug delivery
FR2887457B1 (en) 2005-06-23 2007-10-05 Fond Bettencourt Schueller TRANSCUTANE TARGETING VACCINATION
US20070009542A1 (en) * 2005-07-05 2007-01-11 Galit Levin Method and device for transdermal immunization
EP2258441A3 (en) 2005-09-02 2011-09-21 Intercell USA, Inc. Devices for transcutaneous delivery of vaccines and transdermal delivery of drugs
JP2009509677A (en) * 2005-09-30 2009-03-12 Tti・エルビュー株式会社 Iontophoretic delivery of vesicle encapsulated active substances
KR20080066712A (en) * 2005-09-30 2008-07-16 티티아이 엘뷰 가부시키가이샤 Functionalized microneedles transdermal drug delivery systems, devices, and methods
WO2007041322A2 (en) * 2005-09-30 2007-04-12 Tti Ellebeau, Inc. Iontophoretic delivery of active agents conjugated to nanoparticles
CA2523032A1 (en) * 2005-10-07 2007-04-07 Immunovaccine Technologies Inc. Vaccines for cancer therapy
JP5186678B2 (en) * 2005-10-13 2013-04-17 財団法人ヒューマンサイエンス振興財団 Antigen kit for transdermal antigen administration
US20090169602A1 (en) * 2005-11-23 2009-07-02 Universität Zürich Allergy Treatment by Epicutaneous Allergen Administration
UA96141C2 (en) 2005-12-09 2011-10-10 Юсиби Фарма, С.А. Neutralising antibody having specificity for human il-6
GB0607088D0 (en) 2006-04-07 2006-05-17 Glaxosmithkline Biolog Sa Vaccine
AR058592A1 (en) 2005-12-22 2008-02-13 Glaxosmithkline Biolog Sa VACCINE
WO2007079466A2 (en) * 2006-01-04 2007-07-12 Nano Mist International, Llc Air driven delivery system for sprayable media
KR101541383B1 (en) 2006-03-30 2015-08-03 글락소스미스클라인 바이오로지칼즈 에스.에이. immunogenic composition
ATE522541T1 (en) 2006-06-09 2011-09-15 Novartis Ag BACTERIAL ADHESIN CONFORMERS
EP2041270B1 (en) 2006-07-13 2013-11-20 Wyeth LLC Production of glycoproteins
GB0614780D0 (en) 2006-07-25 2006-09-06 Ucb Sa Biological products
EP2086582B1 (en) 2006-10-12 2012-11-14 GlaxoSmithKline Biologicals s.a. Vaccine comprising an oil in water emulsion adjuvant
ES2397714T3 (en) 2006-10-12 2013-03-08 Glaxosmithkline Biologicals S.A. Vaccine comprising an oil-in-water emulsion adjuvant
GB0620729D0 (en) 2006-10-18 2006-11-29 Ucb Sa Biological products
US20080193514A1 (en) * 2006-11-02 2008-08-14 Transcu Ltd. Compostions and methods for iontophoresis delivery of active ingredients through hair follicles
WO2008055260A2 (en) 2006-11-03 2008-05-08 Wyeth Glycolysis-inhibiting substances in cell culture
US20080214987A1 (en) 2006-12-22 2008-09-04 Nanomed Devices, Inc. Microdevice And Method For Transdermal Delivery And Sampling Of Active Substances
JPWO2008087803A1 (en) * 2007-01-16 2010-05-06 国立大学法人北海道大学 Liposome preparation for iontophoresis encapsulating antioxidant components
DK2115126T3 (en) 2007-03-02 2015-05-04 Wyeth Llc Use of copper and glutamate in cell culture for the preparation of polypeptides
US20080286349A1 (en) * 2007-05-18 2008-11-20 Youhei Nomoto Systems, devices, and methods for passive transdermal delivery of active agents to a biological interface
JP2010187707A (en) * 2007-06-12 2010-09-02 Hokkaido Univ Liposome preparation for iontophoresis comprising insulin encapsulated therein
WO2009000825A2 (en) 2007-06-26 2008-12-31 Glaxosmithkline Biologicals S.A. Vaccine comprising streptococcus pneumoniae capsular polysaccharide conjugates
GB0713880D0 (en) 2007-07-17 2007-08-29 Novartis Ag Conjugate purification
US9498493B2 (en) 2007-09-27 2016-11-22 Immunovaccine Technologies Inc. Use of liposomes in a carrier comprising a continuous hydrophobic phase for delivery of polynucleotides in vivo
FR2924349B1 (en) 2007-12-03 2010-01-01 Dbv Tech ALLERGEN DISENSIBILITY METHOD
EP2612680B1 (en) 2008-04-16 2018-05-23 GlaxoSmithKline Biologicals SA Vaccine
GB0807413D0 (en) 2008-04-23 2008-05-28 Ucb Pharma Sa Biological products
EP2296696B1 (en) 2008-06-05 2014-08-27 ImmunoVaccine Technologies Inc. Compositions comprising liposomes, an antigen, a polynucleotide and a carrier comprising a continuous phase of a hydrophobic substance
BRPI0913972B1 (en) 2008-07-03 2018-06-26 Infectious Disease Research Institute FUSION POLYPEPTIDE UNDERSTANDING LEISHMANIA ANTIGENS, POLINUCLEOTIDE, PHARMACEUTICAL COMPOSITION, AND METHODS FOR INFECTION DETECTION AND IDENTIFICATION FOR LEISHMANIOSIS AND DIAGNOSTIC KIT FOR DETECTION AND IDENTIFICATION KIT
US20100069877A1 (en) * 2008-09-10 2010-03-18 Smith Gregory A Apparatus and method to dispense hpc-based viscous liquids into porous substrates, e.g., continuous web-based process
JP5722782B2 (en) 2008-09-26 2015-05-27 ナノバイオ コーポレーション Nanoemulsion therapeutic composition and method of use thereof
EP2376089B1 (en) 2008-11-17 2018-03-14 The Regents of the University of Michigan Cancer vaccine compositions and methods of using the same
HUE042114T2 (en) 2009-02-17 2019-06-28 Ucb Biopharma Sprl Antibody molecules having specificity for human ox40
US8606366B2 (en) 2009-02-18 2013-12-10 Syneron Medical Ltd. Skin treatment apparatus for personal use and method for using same
GB0904214D0 (en) 2009-03-11 2009-04-22 Ucb Pharma Sa Biological products
CA2765511C (en) 2009-06-16 2015-05-12 The Regents Of The University Of Michigan Nanoemulsion vaccines
GB0913681D0 (en) 2009-08-05 2009-09-16 Glaxosmithkline Biolog Sa Immunogenic composition
US20120283415A1 (en) 2009-09-10 2012-11-08 Ucb Pharma S.A. Multivalent Antibodies
GB0922434D0 (en) 2009-12-22 2010-02-03 Ucb Pharma Sa antibodies and fragments thereof
CA2778673A1 (en) 2009-10-27 2011-05-05 Karen Margrete Miller Function modifying nav 1.7 antibodies
GB0922435D0 (en) 2009-12-22 2010-02-03 Ucb Pharma Sa Method
US9234037B2 (en) 2009-10-27 2016-01-12 Ucb Biopharma Sprl Method to generate antibodies to ion channels
GB0920127D0 (en) 2009-11-17 2009-12-30 Ucb Pharma Sa Antibodies
GB0920324D0 (en) 2009-11-19 2010-01-06 Ucb Pharma Sa Antibodies
GB201000467D0 (en) 2010-01-12 2010-02-24 Ucb Pharma Sa Antibodies
GB201003922D0 (en) 2010-03-09 2010-04-21 Glaxosmithkline Biolog Sa Conjugation process
GB201003920D0 (en) 2010-03-09 2010-04-21 Glaxosmithkline Biolog Sa Method of treatment
TW201134488A (en) 2010-03-11 2011-10-16 Ucb Pharma Sa PD-1 antibodies
JP5920929B2 (en) 2010-03-11 2016-05-18 ユセベ ファルマ ソシエテ アノニム PD-1 antibody
GB201005064D0 (en) 2010-03-25 2010-05-12 Ucb Pharma Sa Biological products
TR201903279T4 (en) 2010-03-25 2019-03-21 Ucb Biopharma Sprl Disulfide stabilized DVD-IG molecules.
US8658603B2 (en) 2010-06-16 2014-02-25 The Regents Of The University Of Michigan Compositions and methods for inducing an immune response
GB201014033D0 (en) 2010-08-20 2010-10-06 Ucb Pharma Sa Biological products
SG10201510762YA (en) 2011-01-14 2016-01-28 Ucb Pharma Sa Antibody molecules which bind il-17a and il-17f
GB201103836D0 (en) 2011-03-07 2011-04-20 Glaxosmithkline Biolog Sa Conjugation process
US10183069B2 (en) 2011-03-21 2019-01-22 Altimmune Inc. Rapid and prolonged immunologic-therapeutic
CA2829916C (en) 2011-03-21 2019-08-20 Vaxin Inc. Intranasal administration of an adenovirus vector to induce a protective immune response to an inhalation pathogen
WO2012136653A1 (en) 2011-04-08 2012-10-11 Novvac Aps Proteins and nucleic acids useful in vaccines targeting staphylococcus aureus
MX339058B (en) 2011-05-17 2016-05-09 Glaxosmithkline Biolog Sa Vaccine against streptococcus pneumoniae.
DK2748300T3 (en) 2011-08-24 2019-01-14 Dupont Nutrition Biosci Aps ENZYM-PRODUCING BACILLUS STUMS
PT2758432T (en) 2011-09-16 2019-06-14 Ucb Biopharma Sprl Neutralising antibodies to the major exotoxins tcda and tcdb of clostridium difficile
WO2013049941A1 (en) 2011-10-06 2013-04-11 Immunovaccine Technologies Inc. Liposome compositions comprising an adjuvant that activates or increases the activity of tlr2 and uses thereof
EP2766388A1 (en) 2011-10-12 2014-08-20 Møller, Niels Iversen Peptides derived from campylobacter jejuni and their use in vaccination
UA112203C2 (en) 2011-11-11 2016-08-10 Юсб Фарма С.А. Fusion protein of a biospecific antibody that binds to human OX40 and serum human albumin
GB201203071D0 (en) 2012-02-22 2012-04-04 Ucb Pharma Sa Biological products
GB201203051D0 (en) 2012-02-22 2012-04-04 Ucb Pharma Sa Biological products
GB201208370D0 (en) 2012-05-14 2012-06-27 Ucb Pharma Sa Antibodies
WO2014044690A1 (en) 2012-09-18 2014-03-27 Valneva Austria Gmbh Improved vaccines
KR101819803B1 (en) 2012-10-24 2018-01-17 플레이틀렛 타게티드 테라퓨틱스, 엘엘씨 Platelet targeted treatment
GB201223276D0 (en) 2012-12-21 2013-02-06 Ucb Pharma Sa Antibodies and methods of producing same
EP2762157A1 (en) * 2013-02-05 2014-08-06 Nitto Denko Corporation Vaccine composition for transdermal or mucosal administration
GB201315487D0 (en) 2013-08-30 2013-10-16 Ucb Pharma Sa Antibodies
US10208102B2 (en) 2013-11-01 2019-02-19 University Of Oslo Albumin variants and uses thereof
GB201320066D0 (en) 2013-11-13 2013-12-25 Ucb Pharma Sa Biological products
CA2929126C (en) 2013-11-13 2020-01-07 University Of Oslo Outer membrane vesicles and uses thereof
DK3069138T3 (en) 2013-11-15 2019-04-08 Univ Oslo Hf CTL PEPTID EPITOPES AND ANTIGEN-SPECIFIC T-CELLS, METHODS OF RECOGNITION THEREOF, AND APPLICATIONS THEREOF
CA2931570A1 (en) 2013-12-03 2015-06-11 Virometix Ag Proline-rich peptides protective against s. pneumoniae
US20160368952A1 (en) 2013-12-03 2016-12-22 Evaxion Biotech Aps Proteins and nucleic acids useful in vaccines targeting staphylococcus aureus
US9067998B1 (en) 2014-07-15 2015-06-30 Kymab Limited Targeting PD-1 variants for treatment of cancer
US8992927B1 (en) 2014-07-15 2015-03-31 Kymab Limited Targeting human NAV1.7 variants for treatment of pain
US8986694B1 (en) 2014-07-15 2015-03-24 Kymab Limited Targeting human nav1.7 variants for treatment of pain
US9914769B2 (en) 2014-07-15 2018-03-13 Kymab Limited Precision medicine for cholesterol treatment
US9045545B1 (en) 2014-07-15 2015-06-02 Kymab Limited Precision medicine by targeting PD-L1 variants for treatment of cancer
GB201403775D0 (en) 2014-03-04 2014-04-16 Kymab Ltd Antibodies, uses & methods
RU2016139006A (en) 2014-03-05 2018-04-05 Юсб Биофарма Спрл MULTI-DIMENSIONAL FC PROTEINS
GB201409558D0 (en) 2014-05-29 2014-07-16 Ucb Biopharma Sprl Method
GB201411320D0 (en) 2014-06-25 2014-08-06 Ucb Biopharma Sprl Antibody construct
US9139648B1 (en) 2014-07-15 2015-09-22 Kymab Limited Precision medicine by targeting human NAV1.9 variants for treatment of pain
GB201412659D0 (en) 2014-07-16 2014-08-27 Ucb Biopharma Sprl Molecules
GB201412658D0 (en) 2014-07-16 2014-08-27 Ucb Biopharma Sprl Molecules
JP6863888B2 (en) 2014-10-01 2021-04-21 メディミューン,エルエルシー How to conjugate a polypeptide
EP3204039B1 (en) 2014-10-10 2022-06-08 The Regents Of The University Of Michigan Nanoemulsion compositions for preventing, suppressing or eliminating allergic and inflammatory disease
AU2015252119A1 (en) 2014-11-07 2016-05-26 Takeda Vaccines, Inc. Hand, foot, and mouth vaccines and methods of manufacture and use thereof
AR102547A1 (en) 2014-11-07 2017-03-08 Takeda Vaccines Inc VACCINES AGAINST DISEASE OF HANDS, FEET AND MOUTH AND MANUFACTURING METHODS AND THEIR USE
US20180169211A1 (en) 2014-11-13 2018-06-21 Evaxion Biotech Aps Peptides derived from acinetobacter baumannii and their use in vaccination
EP3244918A2 (en) 2015-01-12 2017-11-22 Evaxion Biotech ApS Proteins and nucleic acids useful in vaccines targeting klebsiella pneumoniae
US10647964B2 (en) 2015-03-05 2020-05-12 Northwestern University Non-neuroinvasive viruses and uses thereof
GB201506869D0 (en) 2015-04-22 2015-06-03 Ucb Biopharma Sprl Method
GB201506870D0 (en) 2015-04-22 2015-06-03 Ucb Biopharma Sprl Method
SG11201708323SA (en) 2015-04-30 2017-11-29 Harvard College Anti-ap2 antibodies and antigen binding agents to treat metabolic disorders
GB201508180D0 (en) 2015-05-13 2015-06-24 Ucb Biopharma Sprl Antibodies
EP3317295B1 (en) 2015-07-04 2022-05-18 Evaxion Biotech A/S Proteins and nucleic acids useful in vaccines targeting pseudomonas aeruginosa
GB201601075D0 (en) 2016-01-20 2016-03-02 Ucb Biopharma Sprl Antibodies molecules
GB201601073D0 (en) 2016-01-20 2016-03-02 Ucb Biopharma Sprl Antibodies
GB201601077D0 (en) 2016-01-20 2016-03-02 Ucb Biopharma Sprl Antibody molecule
GB201518684D0 (en) 2015-10-21 2015-12-02 Glaxosmithkline Biolog Sa Vaccine
EP3368570A1 (en) 2015-10-27 2018-09-05 UCB Biopharma SPRL Methods of treatment using anti-il-17a/f antibodies
GB201521383D0 (en) 2015-12-03 2016-01-20 Ucb Biopharma Sprl And Ucb Celltech Method
GB201521389D0 (en) 2015-12-03 2016-01-20 Ucb Biopharma Sprl Method
GB201521382D0 (en) 2015-12-03 2016-01-20 Ucb Biopharma Sprl Antibodies
GB201521391D0 (en) 2015-12-03 2016-01-20 Ucb Biopharma Sprl Antibodies
GB201521393D0 (en) 2015-12-03 2016-01-20 Ucb Biopharma Sprl Antibodies
GB201522394D0 (en) 2015-12-18 2016-02-03 Ucb Biopharma Sprl Antibodies
GB201610198D0 (en) 2016-06-10 2016-07-27 Ucb Biopharma Sprl Anti-ige antibodies
WO2017216384A1 (en) 2016-06-17 2017-12-21 Evaxion Biotech Aps Vaccination targeting ichthyophthirius multifiliis
GB201610599D0 (en) 2016-06-17 2016-08-03 Glaxosmithkline Biologicals Sa Immunogenic Composition
WO2017220787A1 (en) 2016-06-24 2017-12-28 Evaxion Biotech Aps Vaccines against aearomonas salmonicida infection
EP3487872A1 (en) 2016-07-22 2019-05-29 Evaxion Biotech ApS Chimeric proteins for inducing immunity towards infection with s. aureus
WO2018038684A1 (en) 2016-08-26 2018-03-01 Agency For Science, Technology And Research Macrophage stimulating protein receptor (or ron - recepteur d' origine nantais) antibodies and uses thereof
GB201616596D0 (en) 2016-09-29 2016-11-16 Nascient Limited Epitope and antibodies
US11779604B2 (en) 2016-11-03 2023-10-10 Kymab Limited Antibodies, combinations comprising antibodies, biomarkers, uses and methods
WO2018096396A1 (en) 2016-11-22 2018-05-31 University Of Oslo Albumin variants and uses thereof
US11718648B2 (en) 2017-01-05 2023-08-08 Evaxion Biotech A/S Vaccines targeting Pseudomonas aeruginosa
WO2018183366A1 (en) 2017-03-28 2018-10-04 Syndax Pharmaceuticals, Inc. Combination therapies of csf-1r or csf-1 antibodies and a t-cell engaging therapy
WO2018213665A1 (en) 2017-05-19 2018-11-22 Syndax Pharmaceuticals, Inc. Combination therapies
CN110621676B (en) 2017-05-22 2024-02-09 托帕杜制药公司 Dual mode of action of soluble guanylate cyclase activators and phosphodiesterase inhibitors and uses thereof
AR111963A1 (en) 2017-05-26 2019-09-04 Univ California METHOD AND MOLECULES
SG11201912601RA (en) 2017-06-23 2020-01-30 Nosocomial Vaccine Corp Immunogenic compositions
WO2019048936A1 (en) 2017-09-07 2019-03-14 University Of Oslo Vaccine molecules
EP3678698A1 (en) 2017-09-07 2020-07-15 University Of Oslo Vaccine molecules
KR20200085302A (en) 2017-11-03 2020-07-14 다케다 백신즈 인코포레이티드 Zika vaccine and immunogenic composition and methods of use thereof
GB201721582D0 (en) 2017-12-21 2018-02-07 Glaxosmithkline Biologicals Sa S aureus antigens and immunogenic compositions
GB201721576D0 (en) 2017-12-21 2018-02-07 Glaxosmithkline Biologicals Sa Hla antigens and glycoconjugates thereof
WO2019145399A1 (en) 2018-01-24 2019-08-01 Evaxion Biotech Aps Vaccines for prophylaxis of s. aureus infections
CN112739698A (en) 2018-08-24 2021-04-30 捷豹治疗有限公司 Tetrahydropyrimidine derivatives as AHR modulators
AU2019331665A1 (en) 2018-08-31 2021-03-25 Jaguahr Therapeutics Pte Ltd Heterocyclic compounds as AHR modulators
SG11202104073XA (en) 2018-10-09 2021-05-28 Aslan Pharmaceuticals Pte Ltd Malonate salt of varlitinib
US20220144941A1 (en) 2018-10-16 2022-05-12 UCB Biopharma SRL Method for the treatment of myasthenia gravis
WO2020083904A1 (en) 2018-10-22 2020-04-30 Evaxion Biotech Aps Vaccines targeting m. catharrhalis
EP3887376A1 (en) 2018-11-28 2021-10-06 Topadur Pharma AG Novel dual mode of action soluble guanylate cyclase activators and phosphodiesterase inhibitors and uses thereof
WO2020144358A1 (en) 2019-01-10 2020-07-16 Expres2Ion Biotechnologies Aps Glyco-engineered immunization antigens
US20220143168A1 (en) 2019-02-27 2022-05-12 Evaxion Biotech A/S Vaccines targeting H. influenzae
EP4028763A1 (en) 2019-09-13 2022-07-20 Evaxion Biotech A/S Method for identifying t-cell epitopes
WO2021048381A1 (en) 2019-09-13 2021-03-18 Evaxion Biotech Aps Method for identifying stable mhc binding peptides using mass spectrometry
EP4087593A1 (en) 2020-01-06 2022-11-16 Evaxion Biotech A/S Vaccines targeting neisseria gonorrhoeae
JP2023515128A (en) 2020-02-26 2023-04-12 ジャガー セラピューティクス ピーティーイーリミテッド Pyridopyrimidine Derivatives Useful for Modulating AHR Signaling
WO2021245192A1 (en) 2020-06-04 2021-12-09 Topadur Pharma Ag Novel dual mode of action soluble guanylate cyclase activators and phosphodiesterase inhibitors and uses thereof
EP4189686A2 (en) 2020-07-30 2023-06-07 Evaxion Biotech A/S Process for preparation of neopepitope-containing vaccine agents
US20230355602A1 (en) 2020-08-31 2023-11-09 Aslan Pharmaceuticals Pte Ltd Treatment and/or prevention of a viral infection
CA3198049A1 (en) 2020-10-15 2022-04-21 UCB Biopharma SRL Binding molecules that multimerise cd45
EP4228640A1 (en) 2020-10-15 2023-08-23 ASLAN Pharmaceuticals Pte Ltd Treatment of autoimmune diseases with a dihydroorotate hehydrogenase (dhodh) inhibitor
KR102393776B1 (en) 2020-12-30 2022-05-04 (주)이노베이션바이오 Humanized antibody specific for CD22 and chimeric antigen receptor using the same
GB202102227D0 (en) 2021-02-17 2021-03-31 UCB Biopharma SRL Antibodies
EP4067381A1 (en) 2021-04-01 2022-10-05 Julius-Maximilians-Universität Würzburg Novel tnfr2 binding molecules
WO2022248531A1 (en) 2021-05-26 2022-12-01 Evaxion Biotech A/S Vaccination targeting intracellular pathogens
AU2022307747A1 (en) 2021-07-05 2024-01-25 Evaxion Biotech A/S Vaccines targeting neisseria gonorrhoeae
US20230233667A1 (en) 2021-09-08 2023-07-27 Affinivax, Inc. Coronavirus vaccine
GB202115122D0 (en) 2021-10-21 2021-12-08 Dualyx Nv Binding molecules targeting IL-2 receptor
WO2023111306A1 (en) 2021-12-17 2023-06-22 Evaxion Biotech A/S Personalized cancer therapy targeting normally non-expressed sequences
WO2023149841A1 (en) 2022-02-03 2023-08-10 Aslan Pharmaceuticals Pte Ltd Dhodh inhibitor polymorph
WO2023204754A1 (en) 2022-04-21 2023-10-26 Aslan Pharmaceuticals Pte Ltd Treatment of autoimmune skin disease
WO2023213393A1 (en) 2022-05-04 2023-11-09 Evaxion Biotech A/S Staphylococcal protein variants and truncates
GB202210680D0 (en) 2022-07-21 2022-09-07 Dualyx Nv Binding molecules targeting il-35r
GB202210679D0 (en) 2022-07-21 2022-09-07 Dualyx Nv Binding molecules targeting il-12rb2

Citations (98)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3948263A (en) * 1974-08-14 1976-04-06 Minnesota Mining And Manufacturing Company Ballistic animal implant
US3964482A (en) * 1971-05-17 1976-06-22 Alza Corporation Drug delivery device
US4196191A (en) * 1975-09-29 1980-04-01 Burroughs Wellcome Co. Biological preparations
US4394448A (en) * 1978-02-24 1983-07-19 Szoka Jr Francis C Method of inserting DNA into living cells
US4455142A (en) * 1980-07-07 1984-06-19 Alza Corporation Method of coadministering an antigen and an immunopotentiator
US4497796A (en) * 1980-03-26 1985-02-05 The Regents Of The University Of California Gene transfer in intact mammals
US4587044A (en) * 1983-09-01 1986-05-06 The Johns Hopkins University Linkage of proteins to nucleic acids
US4725271A (en) * 1983-10-17 1988-02-16 Enquay Pharmaceutical Associates Synthetic resin matrix drug storage and topical drug delivery dressing for veterinary usage
US4732892A (en) * 1985-07-12 1988-03-22 American Home Products Corporation L-α-amino acids as transdermal penetration enhancers
US4743588A (en) * 1984-06-13 1988-05-10 Allergan Pharmaceuticals, Inc. Compositions and methods of enhancing transdermal and transmembrane penetration systemic agents
US4810499A (en) * 1984-10-01 1989-03-07 Biotek, Inc. Transdermal drug delivery system and method
US4834985A (en) * 1986-06-05 1989-05-30 Euroceltique S.A. Controlled release pharmaceutical composition
US4892737A (en) * 1988-09-22 1990-01-09 University Of Florida Composition and method for enhancing permeability of topical drugs
US4904448A (en) * 1987-08-21 1990-02-27 G-C Dental Industrial Corp. Patch test materials for the detection of metal allergies
US4908389A (en) * 1986-08-27 1990-03-13 Warner-Lambert Company Penetration enhancement system
US4917895A (en) * 1987-11-02 1990-04-17 Alza Corporation Transdermal drug delivery device
US4917688A (en) * 1987-01-14 1990-04-17 Nelson Research & Development Co. Bandage for transdermal delivery of systemically-active drug
US4921757A (en) * 1985-04-26 1990-05-01 Massachusetts Institute Of Technology System for delayed and pulsed release of biologically active substances
US4929442A (en) * 1986-09-26 1990-05-29 Exovir, Inc. Compositions suitable for human topical application including a growth factor and/or related materials
US5003987A (en) * 1987-09-11 1991-04-02 Grinwald Paul M Method and apparatus for enhanced drug permeation of skin
US5008050A (en) * 1984-06-20 1991-04-16 The Liposome Company, Inc. Extrusion technique for producing unilamellar vesicles
US5008111A (en) * 1984-10-11 1991-04-16 Schering Corporation Physiological means of enhancing transdermal delivery of drugs
US5023252A (en) * 1985-12-04 1991-06-11 Conrex Pharmaceutical Corporation Transdermal and trans-membrane delivery of drugs
US5028435A (en) * 1989-05-22 1991-07-02 Advanced Polymer Systems, Inc. System and method for transdermal drug delivery
US5030629A (en) * 1987-01-12 1991-07-09 Rajadhyaksha Vithal J Compositions and method comprising heterocyclic compounds containing two heteroatoms as membrane penetration enhancers
US5082866A (en) * 1988-06-01 1992-01-21 Odontex, Inc. Biodegradable absorption enhancers
US5108921A (en) * 1989-04-03 1992-04-28 Purdue Research Foundation Method for enhanced transmembrane transport of exogenous molecules
US5182109A (en) * 1988-04-08 1993-01-26 National Institute Of Health Vaccine preparation comprising a bacterial toxin adjuvant
US5196410A (en) * 1986-10-31 1993-03-23 Pfizer Inc. Transdermal flux enhancing compositions
US5200393A (en) * 1989-02-17 1993-04-06 The Liposome Company, Inc. Lipid excipient for nasal delivery and topical application
US5204339A (en) * 1986-01-31 1993-04-20 Whitby Research, Inc. Penetration enhancers for transdermal delivery of systemic agents
US5215520A (en) * 1991-09-17 1993-06-01 Centre Internationale De Recherches Dermatologiques Galderma (C.I.R.D. Galderma) Method for delivering an active substance topically or percutaneously
US5279544A (en) * 1990-12-13 1994-01-18 Sil Medics Ltd. Transdermal or interdermal drug delivery devices
US5308835A (en) * 1984-07-09 1994-05-03 Praxis Biologics, Inc. Production of the E. coli LT-B enterotoxin subunit
US5399346A (en) * 1989-06-14 1995-03-21 The United States Of America As Represented By The Department Of Health And Human Services Gene therapy
US5411738A (en) * 1989-03-17 1995-05-02 Hind Health Care, Inc. Method for treating nerve injury pain associated with shingles (herpes-zoster and post-herpetic neuralgia) by topical application of lidocaine
US5428132A (en) * 1987-10-11 1995-06-27 United States Of America Conjugate and method for integration of foreign DNA into cells
US5482965A (en) * 1991-03-19 1996-01-09 Rajadhyaksha; Vithal J. Compositions and method comprising aminoalcohol derivatives as membrane penetration enhancers for physiological active agents
US5492698A (en) * 1991-05-15 1996-02-20 Lts Lohmann Therapie-Systeme Gmbh & Co. Kg Lanoline derivatives as penetration enhancing substances
US5505958A (en) * 1994-10-31 1996-04-09 Algos Pharmaceutical Corporation Transdermal drug delivery device and method for its manufacture
US5505956A (en) * 1992-11-30 1996-04-09 Pacific Chemical Co., Ltd. Medicinal adhesive for percutaneous administration
US5518725A (en) * 1989-09-25 1996-05-21 University Of Utah Research Foundation Vaccine compositions and method for induction of mucosal immune response via systemic vaccination
US5593972A (en) * 1993-01-26 1997-01-14 The Wistar Institute Genetic immunization
US5601827A (en) * 1992-06-18 1997-02-11 President And Fellows Of Harvard College Diphtheria toxin vaccines
US5607691A (en) * 1992-06-12 1997-03-04 Affymax Technologies N.V. Compositions and methods for enhanced drug delivery
US5612382A (en) * 1994-07-15 1997-03-18 Frances B. Fike Composition for percutaneous absorption of pharmaceutically active ingredients
US5614212A (en) * 1992-04-08 1997-03-25 International Medical Associates, Inc. Method of transdermally administering high molecular weight drugs with a polymer skin enhancer
US5614503A (en) * 1993-11-12 1997-03-25 Aronex Pharmaceuticals, Inc. Amphipathic nucleic acid transporter
US5620896A (en) * 1992-03-23 1997-04-15 University Of Massachusetts Medical Center DNA vaccines against rotavirus infections
US5626866A (en) * 1994-03-07 1997-05-06 Theratech, Inc. Drug-containing adhesive composite transdermal delivery device
US5705151A (en) * 1995-05-18 1998-01-06 National Jewish Center For Immunology & Respiratory Medicine Gene therapy for T cell regulation
US5718914A (en) * 1992-06-01 1998-02-17 Pharmaderm Laboratories Ltd. Topical patch for liposomal drug delivery system
US5720948A (en) * 1995-11-07 1998-02-24 Helene Curtis Inc. Non-ionic surfactant emulsion vehicles and their use for deposition of drug into and across skin
US5722397A (en) * 1993-11-15 1998-03-03 Altea Technologies, Inc. Enhancement of transdermal monitoring applications with ultrasound and chemical enhancers
US5723114A (en) * 1993-03-19 1998-03-03 Cellegy Pharmaceuticals Inc. Penetration enhancing compositions and methods of their use
US5731303A (en) * 1985-12-04 1998-03-24 Conrex Pharmaceutical Corporation Transdermal and trans-membrane delivery compositions
US5733572A (en) * 1989-12-22 1998-03-31 Imarx Pharmaceutical Corp. Gas and gaseous precursor filled microspheres as topical and subcutaneous delivery vehicles
US5733762A (en) * 1994-04-28 1998-03-31 I.D.M. Immuno-Designed Molecules Complexes of nucleic acid and polymer, their process of preparation and their use for the transfection of cells
US5736392A (en) * 1995-06-07 1998-04-07 Life Technologies, Inc. Peptide-enhanced cationic lipid transfections
US5736154A (en) * 1996-03-11 1998-04-07 Fuisz Technologies Ltd. Transdermal delivery system
US5736524A (en) * 1994-11-14 1998-04-07 Merck & Co.,. Inc. Polynucleotide tuberculosis vaccine
US5739118A (en) * 1994-04-01 1998-04-14 Apollon, Inc. Compositions and methods for delivery of genetic material
US5738647A (en) * 1996-09-27 1998-04-14 Becton Dickinson And Company User activated iontophoretic device and method for activating same
US5741510A (en) * 1994-03-30 1998-04-21 Lectec Corporation Adhesive patch for applying analgesic medication to the skin
US5756117A (en) * 1992-04-08 1998-05-26 International Medical Asscociates, Inc. Multidose transdermal drug delivery system
US5760096A (en) * 1996-10-18 1998-06-02 Thornfeldt; Carl R. Potent penetration enhancers
US5766899A (en) * 1995-02-27 1998-06-16 Board Of Regents , The University Of Texas System Targeted nucleic acid delivery into liver cells
US5770580A (en) * 1992-04-13 1998-06-23 Baylor College Of Medicine Somatic gene therapy to cells associated with fluid spaces
US5773022A (en) * 1994-04-05 1998-06-30 Astra Ab Topical dressing
US5856187A (en) * 1993-03-17 1999-01-05 The United States Of America As Represented By The Department Of Health And Human Services Immunogenic chimeras comprising nucleic acid sequences encoding endoplasmic reticulum signal sequence peptides and at least one other peptide, and their uses in vaccines and disease treatments
US5858784A (en) * 1991-12-17 1999-01-12 The Regents Of The University Of California Expression of cloned genes in the lung by aerosol- and liposome-based delivery
US5866553A (en) * 1994-06-30 1999-02-02 Merck & Co., Inc. Polynucleotide vaccine for papillomavirus
US5877302A (en) * 1994-03-23 1999-03-02 Case Western Reserve University Compacted nucleic acids and their delivery to cells
US5877159A (en) * 1995-05-03 1999-03-02 University Of Maryland At Baltimore Method for introducing and expressing genes in animal cells and live invasive bacterial vectors for use in the same
US5879326A (en) * 1995-05-22 1999-03-09 Godshall; Ned Allen Method and apparatus for disruption of the epidermis
US5885971A (en) * 1995-03-24 1999-03-23 The Regents Of The University Of California Gene therapy by secretory gland expression
US5910306A (en) * 1996-11-14 1999-06-08 The United States Of America As Represented By The Secretary Of The Army Transdermal delivery system for antigen
US5910488A (en) * 1993-06-07 1999-06-08 Vical Incorporated Plasmids suitable for gene therapy
US5914114A (en) * 1995-06-02 1999-06-22 The United States Of America As Represented By The Secretary Of The Army Method of raising antibodies against E. coli of the family CS4-CFA/I
US5916879A (en) * 1996-11-12 1999-06-29 St. Jude Children's Research Hospital DNA transcription unit vaccines that protect against avian influenza viruses and methods of use thereof
US6019982A (en) * 1994-08-26 2000-02-01 The Administrators Of The Tulane Educational Fund Mutant enterotoxin effective as a non-toxic oral adjuvant
US6022316A (en) * 1998-03-06 2000-02-08 Spectrx, Inc. Apparatus and method for electroporation of microporated tissue for enhancing flux rates for monitoring and delivery applications
US6033684A (en) * 1996-08-28 2000-03-07 Jonor, Inc. Compositions and methods for wound management
US6033673A (en) * 1998-03-18 2000-03-07 The Administrators Of Tulane Educational Fund Double mutant enterotoxin for use as an adjuvant
US6039969A (en) * 1996-10-25 2000-03-21 3M Innovative Properties Company Immune response modifier compounds for treatment of TH2 mediated and related diseases
US6063399A (en) * 1996-12-20 2000-05-16 Roehm Gmbh Chemische Fabrik Adhesive binders for dermal or transdermal therapy systems
US6173202B1 (en) * 1998-03-06 2001-01-09 Spectrx, Inc. Method and apparatus for enhancing flux rates of a fluid in a microporated biological tissue
US6180136B1 (en) * 1998-11-10 2001-01-30 Idexx Laboratories, Inc. Phospholipid-coated microcrystals for the sustained release of pharmacologically active compounds and methods of their manufacture and use
US6183434B1 (en) * 1996-07-03 2001-02-06 Spectrx, Inc. Multiple mechanical microporation of skin or mucosa
US6190689B1 (en) * 1994-05-13 2001-02-20 Lts Lohmann Therapie-Systeme Gmbh Hydrophilic pressure sensitive hot-melt adhesives
US6190367B1 (en) * 1999-09-22 2001-02-20 Becton, Dickinson And Company Medical site prep device
US6207184B1 (en) * 1998-06-18 2001-03-27 Ssp Co., Ltd. Hydrophilic adhesive masses
US6210672B1 (en) * 1998-10-20 2001-04-03 Torrey Pines Institute For Molecular Studies Topical immunostimulation to induce Langerhans cell migration
US6248212B1 (en) * 1997-12-30 2001-06-19 Kimberly-Clark Worldwide, Inc. Through-air-dried post bonded creped fibrous web
US6348450B1 (en) * 1997-08-13 2002-02-19 The Uab Research Foundation Noninvasive genetic immunization, expression products therefrom and uses thereof
US6365178B1 (en) * 1996-09-06 2002-04-02 Watson Pharmaceuticals, Inc. Method of making pressure sensitive adhesive matrix patches for transdermal drug delivery using hydrophilic salts of drugs and hydrophobic pressure sensitive adhesive dispersions
US6379324B1 (en) * 1999-06-09 2002-04-30 The Procter & Gamble Company Intracutaneous microneedle array apparatus
US6406705B1 (en) * 1997-03-10 2002-06-18 University Of Iowa Research Foundation Use of nucleic acids containing unmethylated CpG dinucleotide as an adjuvant

Family Cites Families (68)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3837340A (en) 1971-10-20 1974-09-24 Lilly Co Eli Device for administering immunication against virus
US3982536A (en) 1974-11-15 1976-09-28 Minnesota Mining And Manufacturing Company Ballistic inoculation of animals and projectile therefor
US4285931A (en) 1978-01-30 1981-08-25 Merck & Co., Inc. E. coli enterotoxin vaccine for veterinary and human use
US4220584A (en) 1978-01-30 1980-09-02 Merck & Co., Inc. E. coli enterotoxin vaccine for veterinary and human use
US4235871A (en) 1978-02-24 1980-11-25 Papahadjopoulos Demetrios P Method of encapsulating biologically active materials in lipid vesicles
US4761372A (en) 1981-01-12 1988-08-02 New York University Mutant enterotoxin of E. coli
US4411888A (en) 1981-06-22 1983-10-25 The University Of Rochester Composition of a novel immunogen for protection against diarrheal disease caused by enterotoxigenic Escherichia coli
US4484923A (en) * 1982-03-25 1984-11-27 Alza Corporation Method for administering immunopotentiator
US5169637A (en) * 1983-03-24 1992-12-08 The Liposome Company, Inc. Stable plurilamellar vesicles
SE443914B (en) 1984-10-01 1986-03-17 Torkel Ingemar Fischer MEANS OF A SENSITIVITY TEST
US5049386A (en) 1985-01-07 1991-09-17 Syntex (U.S.A.) Inc. N-ω,(ω-1)-dialkyloxy)- and N-(ω,(ω-1)-dialkenyloxy)Alk-1-YL-N,N,N-tetrasubstituted ammonium lipids and uses therefor
US4692462A (en) 1985-03-18 1987-09-08 Menley & James Laboratories, Ltd. Compositions and method of controlling transdermal penetration of topical and systemic agents
US4877612A (en) * 1985-05-20 1989-10-31 Frank M. Berger Immunological adjuvant and process for preparing the same, pharmaceutical compositions, and process
US5059421A (en) * 1985-07-26 1991-10-22 The Liposome Company, Inc. Preparation of targeted liposome systems of a defined size distribution
DE3527893A1 (en) 1985-08-03 1987-02-05 Merck Patent Gmbh EPICUTANE TEST PLASTER
GB2191941B (en) * 1985-08-27 1990-03-14 Glyzinc Pharma Ltd Zinc glycerolate complex and additions for pharmaceutical applications
US4970206A (en) 1985-11-29 1990-11-13 Merck & Co., Inc. Pyroglutamic acid esters used as dermal penetration enhancers for drugs
US4764381A (en) 1985-12-06 1988-08-16 Key Pharmaceuticals, Inc. Percutaneous penetration enhancer of oleic acid and 2-ethyl-1, 3-hexanediol
US5270346A (en) 1986-01-31 1993-12-14 Whitby Research, Inc. Penetration enhancers for transdermal delivery of systemic agents
US4775361A (en) 1986-04-10 1988-10-04 The General Hospital Corporation Controlled removal of human stratum corneum by pulsed laser to enhance percutaneous transport
US5142044A (en) 1986-04-23 1992-08-25 Whitby Research, Inc. Penetration enhancers for transdermal delivery of systemic agents
US5352449A (en) * 1986-05-30 1994-10-04 Cambridge Biotech Corporation Vaccine comprising recombinant feline leukemia antigen and saponin adjuvant
US5041439A (en) 1986-06-13 1991-08-20 The Procter & Gamble Company Penetrating topical pharmaceutical compositions
US5326790A (en) * 1986-11-19 1994-07-05 Dermatologic Research Corporation Administration of skin medications by use of dicarboxylic acids and derivatives
JP3011939B2 (en) * 1987-03-02 2000-02-21 ホワイトヘツド・インスチチユート・フオー・バイオメデイカル・リサーチ Recombinant mycobacterial vaccine
US5049387A (en) 1987-03-09 1991-09-17 Alza Corporation Inducing skin tolerance to a sensitizing drug
US4783450A (en) 1987-04-13 1988-11-08 Warner-Lambert Company Use of commercial lecithin as skin penetration enhancer
US5166320A (en) 1987-04-22 1992-11-24 University Of Connecticut Carrier system and method for the introduction of genes into mammalian cells
IL86170A (en) 1987-05-01 1992-12-01 Elan Transdermal Ltd Preparations and compositions comprising nicotine for percutaneous administration
SE458339B (en) 1987-05-25 1989-03-20 Pharmacia Ab TEST REMEMBER TO PROVIDE CONTACT ALLERGY
US5045317A (en) 1987-07-16 1991-09-03 The Regents Of The University Of California Enhancing the cutaneous penetration of pharmacologically active agents
US5059189A (en) 1987-09-08 1991-10-22 E. R. Squibb & Sons, Inc. Method of preparing adhesive dressings containing a pharmaceutically active ingredient
US5472946A (en) 1988-04-08 1995-12-05 Peck; James V. Transdermal penetration enhancers
US5234959A (en) 1988-04-08 1993-08-10 Whitby Research, Inc. Penetration enhancers for transdermal delivery of systemic agents
US5164406A (en) 1988-06-02 1992-11-17 Bristol-Myers Squibb Co. Method for enhancing transdermal penetration and compositions useful therein
US4960771A (en) 1988-07-12 1990-10-02 Rajadhyaksha Vithal J Oxazolidinone penetration enhancing compounds
US5032402A (en) 1988-10-27 1991-07-16 The University Of Kentucky Research Foundation 3-hydroxy-N-methylpyrrolidone and use as transdermal enhancer
US5238944A (en) 1988-12-15 1993-08-24 Riker Laboratories, Inc. Topical formulations and transdermal delivery systems containing 1-isobutyl-1H-imidazo[4,5-c]quinolin-4-amine
LU87410A1 (en) * 1988-12-20 1990-07-10 Cird COSMETIC OR PHARMACEUTICAL COMPOSITION CONTAINING POLYMERIC OR FATTY BODY MICROSPHERES CHARGED WITH AT LEAST ONE ACTIVE PRODUCT
US5332577A (en) 1988-12-27 1994-07-26 Dermamed Transdermal administration to humans and animals
IE62662B1 (en) 1989-01-06 1995-02-22 Elan Corp Plc Use of nicotine in the treatment of conditions susceptible to said treatment
US5534260A (en) 1989-02-23 1996-07-09 University Of Utah Percutaneous drug delivery system
US5540931A (en) 1989-03-03 1996-07-30 Charles W. Hewitt Methods for inducing site-specific immunosuppression and compositions of site specific immunosuppressants
US5328470A (en) 1989-03-31 1994-07-12 The Regents Of The University Of Michigan Treatment of diseases by site-specific instillation of cells or site-specific transformation of cells and kits therefor
US5162315A (en) 1989-05-08 1992-11-10 Rajadhyaksha Vithal J Penetration enhancers
US4956171A (en) 1989-07-21 1990-09-11 Paco Pharmaceutical Services, Inc. Transdermal drug delivery using a dual permeation enhancer and method of performing the same
DE3924393A1 (en) 1989-07-24 1991-01-31 Roehm Gmbh WATER-SOLUBLE PRESSURE-SENSITIVE SKIN ADHESIVE, THE USE THEREOF AND MEANS THEREFORE
US5240846A (en) 1989-08-22 1993-08-31 The Regents Of The University Of Michigan Gene therapy vector for cystic fibrosis
US5252334A (en) 1989-09-08 1993-10-12 Cygnus Therapeutic Systems Solid matrix system for transdermal drug delivery
EP0429842B1 (en) 1989-10-27 1996-08-28 Korea Research Institute Of Chemical Technology Device for the transdermal administration of protein or peptide drug
IE904098A1 (en) * 1989-11-13 1991-05-22 Nova Pharm Corp Lipospheres for controlled delivery of substances
WO1992003122A1 (en) * 1990-08-24 1992-03-05 Gregor Cevc Preparation for application of active substances in the form of minimum-sized droplets
US5332576A (en) * 1991-02-27 1994-07-26 Noven Pharmaceuticals, Inc. Compositions and methods for topical administration of pharmaceutically active agents
US5256422A (en) * 1991-03-28 1993-10-26 Micro Vesicular Systems, Inc. Lipid vesicle containing water-in-oil emulsions
US5326566A (en) 1991-05-17 1994-07-05 Bristol-Myers Squibb Company Use of dibutyl adipate and isopropyl myristate in topical and transdermal products
GB9114346D0 (en) 1991-07-03 1991-08-21 Dow Corning Sa A method of accelerating drug permeation
NZ244306A (en) 1991-09-30 1995-07-26 Boehringer Ingelheim Int Composition for introducing nucleic acid complexes into eucaryotic cells, complex containing nucleic acid and endosomolytic agent, peptide with endosomolytic domain and nucleic acid binding domain and preparation
US5225182A (en) 1991-10-31 1993-07-06 Sharma Yash P Vectored drug delivery system using a cephaloplastin linking agent and a methed of using the system
JP3456994B2 (en) 1991-11-13 2003-10-14 エラン コーポレーション ピーエルシー Drug administration device
US5260066A (en) * 1992-01-16 1993-11-09 Srchem Incorporated Cryogel bandage containing therapeutic agent
US5464386A (en) * 1992-08-17 1995-11-07 Genetronics, Inc. Transdermal drug delivery by electroincorporation of vesicles
US5462743A (en) 1992-10-30 1995-10-31 Medipro Sciences Limited Substance transfer system for topical application
US5445611A (en) 1993-12-08 1995-08-29 Non-Invasive Monitoring Company (Nimco) Enhancement of transdermal delivery with ultrasound and chemical enhancers
GB9326174D0 (en) * 1993-12-22 1994-02-23 Biocine Sclavo Mucosal adjuvant
US5686100A (en) * 1994-11-22 1997-11-11 E.R. Squibb & Sons, Inc. Prophylactic and therapeutic treatment of skin sensitization and irritation
US5573778A (en) 1995-03-17 1996-11-12 Adhesives Research, Inc. Drug flux enhancer-tolerant pressure sensitive adhesive composition
US5601839A (en) * 1995-04-26 1997-02-11 Theratech, Inc. Triacetin as a penetration enhancer for transdermal delivery of a basic drug
US5980898A (en) 1996-11-14 1999-11-09 The United States Of America As Represented By The U.S. Army Medical Research & Material Command Adjuvant for transcutaneous immunization

Patent Citations (99)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3964482A (en) * 1971-05-17 1976-06-22 Alza Corporation Drug delivery device
US3948263A (en) * 1974-08-14 1976-04-06 Minnesota Mining And Manufacturing Company Ballistic animal implant
US4196191A (en) * 1975-09-29 1980-04-01 Burroughs Wellcome Co. Biological preparations
US4394448A (en) * 1978-02-24 1983-07-19 Szoka Jr Francis C Method of inserting DNA into living cells
US4497796A (en) * 1980-03-26 1985-02-05 The Regents Of The University Of California Gene transfer in intact mammals
US4455142A (en) * 1980-07-07 1984-06-19 Alza Corporation Method of coadministering an antigen and an immunopotentiator
US4587044A (en) * 1983-09-01 1986-05-06 The Johns Hopkins University Linkage of proteins to nucleic acids
US4725271A (en) * 1983-10-17 1988-02-16 Enquay Pharmaceutical Associates Synthetic resin matrix drug storage and topical drug delivery dressing for veterinary usage
US4743588A (en) * 1984-06-13 1988-05-10 Allergan Pharmaceuticals, Inc. Compositions and methods of enhancing transdermal and transmembrane penetration systemic agents
US5008050A (en) * 1984-06-20 1991-04-16 The Liposome Company, Inc. Extrusion technique for producing unilamellar vesicles
US5308835A (en) * 1984-07-09 1994-05-03 Praxis Biologics, Inc. Production of the E. coli LT-B enterotoxin subunit
US4810499A (en) * 1984-10-01 1989-03-07 Biotek, Inc. Transdermal drug delivery system and method
US5008111A (en) * 1984-10-11 1991-04-16 Schering Corporation Physiological means of enhancing transdermal delivery of drugs
US4921757A (en) * 1985-04-26 1990-05-01 Massachusetts Institute Of Technology System for delayed and pulsed release of biologically active substances
US4732892A (en) * 1985-07-12 1988-03-22 American Home Products Corporation L-α-amino acids as transdermal penetration enhancers
US5731303A (en) * 1985-12-04 1998-03-24 Conrex Pharmaceutical Corporation Transdermal and trans-membrane delivery compositions
US5023252A (en) * 1985-12-04 1991-06-11 Conrex Pharmaceutical Corporation Transdermal and trans-membrane delivery of drugs
US5204339A (en) * 1986-01-31 1993-04-20 Whitby Research, Inc. Penetration enhancers for transdermal delivery of systemic agents
US4834985A (en) * 1986-06-05 1989-05-30 Euroceltique S.A. Controlled release pharmaceutical composition
US4908389A (en) * 1986-08-27 1990-03-13 Warner-Lambert Company Penetration enhancement system
US4929442A (en) * 1986-09-26 1990-05-29 Exovir, Inc. Compositions suitable for human topical application including a growth factor and/or related materials
US5196410A (en) * 1986-10-31 1993-03-23 Pfizer Inc. Transdermal flux enhancing compositions
US5030629A (en) * 1987-01-12 1991-07-09 Rajadhyaksha Vithal J Compositions and method comprising heterocyclic compounds containing two heteroatoms as membrane penetration enhancers
US4917688A (en) * 1987-01-14 1990-04-17 Nelson Research & Development Co. Bandage for transdermal delivery of systemically-active drug
US4904448A (en) * 1987-08-21 1990-02-27 G-C Dental Industrial Corp. Patch test materials for the detection of metal allergies
US5003987A (en) * 1987-09-11 1991-04-02 Grinwald Paul M Method and apparatus for enhanced drug permeation of skin
US5428132A (en) * 1987-10-11 1995-06-27 United States Of America Conjugate and method for integration of foreign DNA into cells
US4917895A (en) * 1987-11-02 1990-04-17 Alza Corporation Transdermal drug delivery device
US5182109A (en) * 1988-04-08 1993-01-26 National Institute Of Health Vaccine preparation comprising a bacterial toxin adjuvant
US5182109C1 (en) * 1988-04-08 2001-10-02 Nat Inst Health Vaccine preparation comprising a bacterial toxin adjuvant
US5082866A (en) * 1988-06-01 1992-01-21 Odontex, Inc. Biodegradable absorption enhancers
US4892737A (en) * 1988-09-22 1990-01-09 University Of Florida Composition and method for enhancing permeability of topical drugs
US5200393A (en) * 1989-02-17 1993-04-06 The Liposome Company, Inc. Lipid excipient for nasal delivery and topical application
US5411738A (en) * 1989-03-17 1995-05-02 Hind Health Care, Inc. Method for treating nerve injury pain associated with shingles (herpes-zoster and post-herpetic neuralgia) by topical application of lidocaine
US5108921A (en) * 1989-04-03 1992-04-28 Purdue Research Foundation Method for enhanced transmembrane transport of exogenous molecules
US5028435A (en) * 1989-05-22 1991-07-02 Advanced Polymer Systems, Inc. System and method for transdermal drug delivery
US5399346A (en) * 1989-06-14 1995-03-21 The United States Of America As Represented By The Department Of Health And Human Services Gene therapy
US5518725A (en) * 1989-09-25 1996-05-21 University Of Utah Research Foundation Vaccine compositions and method for induction of mucosal immune response via systemic vaccination
US5733572A (en) * 1989-12-22 1998-03-31 Imarx Pharmaceutical Corp. Gas and gaseous precursor filled microspheres as topical and subcutaneous delivery vehicles
US5279544A (en) * 1990-12-13 1994-01-18 Sil Medics Ltd. Transdermal or interdermal drug delivery devices
US5482965A (en) * 1991-03-19 1996-01-09 Rajadhyaksha; Vithal J. Compositions and method comprising aminoalcohol derivatives as membrane penetration enhancers for physiological active agents
US5492698A (en) * 1991-05-15 1996-02-20 Lts Lohmann Therapie-Systeme Gmbh & Co. Kg Lanoline derivatives as penetration enhancing substances
US5215520A (en) * 1991-09-17 1993-06-01 Centre Internationale De Recherches Dermatologiques Galderma (C.I.R.D. Galderma) Method for delivering an active substance topically or percutaneously
US5858784A (en) * 1991-12-17 1999-01-12 The Regents Of The University Of California Expression of cloned genes in the lung by aerosol- and liposome-based delivery
US5620896A (en) * 1992-03-23 1997-04-15 University Of Massachusetts Medical Center DNA vaccines against rotavirus infections
US5614212A (en) * 1992-04-08 1997-03-25 International Medical Associates, Inc. Method of transdermally administering high molecular weight drugs with a polymer skin enhancer
US5756117A (en) * 1992-04-08 1998-05-26 International Medical Asscociates, Inc. Multidose transdermal drug delivery system
US5770580A (en) * 1992-04-13 1998-06-23 Baylor College Of Medicine Somatic gene therapy to cells associated with fluid spaces
US5718914A (en) * 1992-06-01 1998-02-17 Pharmaderm Laboratories Ltd. Topical patch for liposomal drug delivery system
US5607691A (en) * 1992-06-12 1997-03-04 Affymax Technologies N.V. Compositions and methods for enhanced drug delivery
US5601827A (en) * 1992-06-18 1997-02-11 President And Fellows Of Harvard College Diphtheria toxin vaccines
US5505956A (en) * 1992-11-30 1996-04-09 Pacific Chemical Co., Ltd. Medicinal adhesive for percutaneous administration
US5593972A (en) * 1993-01-26 1997-01-14 The Wistar Institute Genetic immunization
US5856187A (en) * 1993-03-17 1999-01-05 The United States Of America As Represented By The Department Of Health And Human Services Immunogenic chimeras comprising nucleic acid sequences encoding endoplasmic reticulum signal sequence peptides and at least one other peptide, and their uses in vaccines and disease treatments
US5723114A (en) * 1993-03-19 1998-03-03 Cellegy Pharmaceuticals Inc. Penetration enhancing compositions and methods of their use
US5910488A (en) * 1993-06-07 1999-06-08 Vical Incorporated Plasmids suitable for gene therapy
US5614503A (en) * 1993-11-12 1997-03-25 Aronex Pharmaceuticals, Inc. Amphipathic nucleic acid transporter
US5722397A (en) * 1993-11-15 1998-03-03 Altea Technologies, Inc. Enhancement of transdermal monitoring applications with ultrasound and chemical enhancers
US5626866A (en) * 1994-03-07 1997-05-06 Theratech, Inc. Drug-containing adhesive composite transdermal delivery device
US5877302A (en) * 1994-03-23 1999-03-02 Case Western Reserve University Compacted nucleic acids and their delivery to cells
US5741510A (en) * 1994-03-30 1998-04-21 Lectec Corporation Adhesive patch for applying analgesic medication to the skin
US5739118A (en) * 1994-04-01 1998-04-14 Apollon, Inc. Compositions and methods for delivery of genetic material
US5773022A (en) * 1994-04-05 1998-06-30 Astra Ab Topical dressing
US5733762A (en) * 1994-04-28 1998-03-31 I.D.M. Immuno-Designed Molecules Complexes of nucleic acid and polymer, their process of preparation and their use for the transfection of cells
US6190689B1 (en) * 1994-05-13 2001-02-20 Lts Lohmann Therapie-Systeme Gmbh Hydrophilic pressure sensitive hot-melt adhesives
US5866553A (en) * 1994-06-30 1999-02-02 Merck & Co., Inc. Polynucleotide vaccine for papillomavirus
US5612382A (en) * 1994-07-15 1997-03-18 Frances B. Fike Composition for percutaneous absorption of pharmaceutically active ingredients
US6019982A (en) * 1994-08-26 2000-02-01 The Administrators Of The Tulane Educational Fund Mutant enterotoxin effective as a non-toxic oral adjuvant
US5505958A (en) * 1994-10-31 1996-04-09 Algos Pharmaceutical Corporation Transdermal drug delivery device and method for its manufacture
US5736524A (en) * 1994-11-14 1998-04-07 Merck & Co.,. Inc. Polynucleotide tuberculosis vaccine
US5766899A (en) * 1995-02-27 1998-06-16 Board Of Regents , The University Of Texas System Targeted nucleic acid delivery into liver cells
US5885971A (en) * 1995-03-24 1999-03-23 The Regents Of The University Of California Gene therapy by secretory gland expression
US5877159A (en) * 1995-05-03 1999-03-02 University Of Maryland At Baltimore Method for introducing and expressing genes in animal cells and live invasive bacterial vectors for use in the same
US5705151A (en) * 1995-05-18 1998-01-06 National Jewish Center For Immunology & Respiratory Medicine Gene therapy for T cell regulation
US5879326A (en) * 1995-05-22 1999-03-09 Godshall; Ned Allen Method and apparatus for disruption of the epidermis
US5914114A (en) * 1995-06-02 1999-06-22 The United States Of America As Represented By The Secretary Of The Army Method of raising antibodies against E. coli of the family CS4-CFA/I
US5736392A (en) * 1995-06-07 1998-04-07 Life Technologies, Inc. Peptide-enhanced cationic lipid transfections
US5720948A (en) * 1995-11-07 1998-02-24 Helene Curtis Inc. Non-ionic surfactant emulsion vehicles and their use for deposition of drug into and across skin
US5736154A (en) * 1996-03-11 1998-04-07 Fuisz Technologies Ltd. Transdermal delivery system
US6183434B1 (en) * 1996-07-03 2001-02-06 Spectrx, Inc. Multiple mechanical microporation of skin or mucosa
US6033684A (en) * 1996-08-28 2000-03-07 Jonor, Inc. Compositions and methods for wound management
US6365178B1 (en) * 1996-09-06 2002-04-02 Watson Pharmaceuticals, Inc. Method of making pressure sensitive adhesive matrix patches for transdermal drug delivery using hydrophilic salts of drugs and hydrophobic pressure sensitive adhesive dispersions
US5738647A (en) * 1996-09-27 1998-04-14 Becton Dickinson And Company User activated iontophoretic device and method for activating same
US5760096A (en) * 1996-10-18 1998-06-02 Thornfeldt; Carl R. Potent penetration enhancers
US6039969A (en) * 1996-10-25 2000-03-21 3M Innovative Properties Company Immune response modifier compounds for treatment of TH2 mediated and related diseases
US5916879A (en) * 1996-11-12 1999-06-29 St. Jude Children's Research Hospital DNA transcription unit vaccines that protect against avian influenza viruses and methods of use thereof
US5910306A (en) * 1996-11-14 1999-06-08 The United States Of America As Represented By The Secretary Of The Army Transdermal delivery system for antigen
US6063399A (en) * 1996-12-20 2000-05-16 Roehm Gmbh Chemische Fabrik Adhesive binders for dermal or transdermal therapy systems
US6406705B1 (en) * 1997-03-10 2002-06-18 University Of Iowa Research Foundation Use of nucleic acids containing unmethylated CpG dinucleotide as an adjuvant
US6348450B1 (en) * 1997-08-13 2002-02-19 The Uab Research Foundation Noninvasive genetic immunization, expression products therefrom and uses thereof
US6248212B1 (en) * 1997-12-30 2001-06-19 Kimberly-Clark Worldwide, Inc. Through-air-dried post bonded creped fibrous web
US6173202B1 (en) * 1998-03-06 2001-01-09 Spectrx, Inc. Method and apparatus for enhancing flux rates of a fluid in a microporated biological tissue
US6022316A (en) * 1998-03-06 2000-02-08 Spectrx, Inc. Apparatus and method for electroporation of microporated tissue for enhancing flux rates for monitoring and delivery applications
US6033673A (en) * 1998-03-18 2000-03-07 The Administrators Of Tulane Educational Fund Double mutant enterotoxin for use as an adjuvant
US6207184B1 (en) * 1998-06-18 2001-03-27 Ssp Co., Ltd. Hydrophilic adhesive masses
US6210672B1 (en) * 1998-10-20 2001-04-03 Torrey Pines Institute For Molecular Studies Topical immunostimulation to induce Langerhans cell migration
US6180136B1 (en) * 1998-11-10 2001-01-30 Idexx Laboratories, Inc. Phospholipid-coated microcrystals for the sustained release of pharmacologically active compounds and methods of their manufacture and use
US6379324B1 (en) * 1999-06-09 2002-04-30 The Procter & Gamble Company Intracutaneous microneedle array apparatus
US6190367B1 (en) * 1999-09-22 2001-02-20 Becton, Dickinson And Company Medical site prep device

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7848801B2 (en) 2005-12-30 2010-12-07 Tti Ellebeau, Inc. Iontophoretic systems, devices, and methods of delivery of active agents to biological interface

Also Published As

Publication number Publication date
IL129919A (en) 2008-04-13
CA2272417C (en) 2003-07-29
KR100517028B1 (en) 2005-09-26
DE69730534D1 (en) 2004-10-07
EP1014787A1 (en) 2000-07-05
IL129919A0 (en) 2000-02-29
EP1014787A4 (en) 2002-08-07
DE69737218D1 (en) 2007-02-15
WO1998020734A1 (en) 1998-05-22
DE69730534T2 (en) 2005-09-15
BR9712952A (en) 1999-12-07
JP2001511115A (en) 2001-08-07
CN1241906A (en) 2000-01-19
KR20000053306A (en) 2000-08-25
DE69737218T2 (en) 2007-11-08
ATE274805T1 (en) 2004-09-15
AU5265298A (en) 1998-06-03
ATE349890T1 (en) 2007-01-15
US7037499B1 (en) 2006-05-02
CN1279976C (en) 2006-10-18
US5980898A (en) 1999-11-09
AU744537B2 (en) 2002-02-28
EP1557177A1 (en) 2005-07-27
NZ335749A (en) 2001-01-26
EP2272526A2 (en) 2011-01-12
EP1014787B1 (en) 2004-09-01
CA2272417A1 (en) 1998-05-22
EP2272526A3 (en) 2012-02-08
EP1384403A1 (en) 2004-01-28
EP1384403B1 (en) 2007-01-03
JP4584361B2 (en) 2010-11-17
AP9901540A0 (en) 1999-06-30

Similar Documents

Publication Publication Date Title
US5980898A (en) Adjuvant for transcutaneous immunization
US6797276B1 (en) Use of penetration enhancers and barrier disruption agents to enhance the transcutaneous immune response
EP1061951B1 (en) Use of skin penetration enhancers and barrier disruption agents to enhance the transcutaneous immune response induced by adp-ribosylating exotoxin
US8911742B2 (en) Transcutaneous immunization without heterologous adjuvant
US20060002959A1 (en) Skin-sctive adjuvants for transcutaneous immuization
JP2010059201A (en) Adjuvant for transcutaneous immunization
US20040258703A1 (en) Skin-active adjuvants for transcutaneous immunization
EP1356821B1 (en) Use of skin penetration enhancers and barrier disruption agents to enhance transcutaneous immune response
AU774610B2 (en) Adjuvant for transcutaneous immunization
AU2004216682B2 (en) Adjuvant for transcutaneous immunization
EP1849477A1 (en) Use of skin penetration enhancers and barrier disruption agents to enhance the transcutaneous immune response induced by ADP-ribosylating exotoxin
MXPA99004452A (en) Adjuvant for transcutaneous immunization

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION