US20020160011A1 - Adjuvant compositions for vaccines - Google Patents

Adjuvant compositions for vaccines Download PDF

Info

Publication number
US20020160011A1
US20020160011A1 US09/445,903 US44590300A US2002160011A1 US 20020160011 A1 US20020160011 A1 US 20020160011A1 US 44590300 A US44590300 A US 44590300A US 2002160011 A1 US2002160011 A1 US 2002160011A1
Authority
US
United States
Prior art keywords
adjuvant
antigen
mice
cells
peptide
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
US09/445,903
Inventor
Thierry Boon
Silvia Silla
Catherine Uyttenhove
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.)
GlaxoSmithKline Biologicals SA
Ludwig Institute for Cancer Research Ltd
Original Assignee
SmithKline Beecham Biologicals SA
Ludwig Institute for Cancer Research Ltd
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
Application filed by SmithKline Beecham Biologicals SA, Ludwig Institute for Cancer Research Ltd filed Critical SmithKline Beecham Biologicals SA
Assigned to SMITHKLINE BEECHAM BIOLOGICALS S.A., LUDWIG INSTITUTE FOR CANCER RESEARCH reassignment SMITHKLINE BEECHAM BIOLOGICALS S.A. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: BOON, THIERRY, SILLA, SILVIA, UYTTENHOVE, CATHERINE
Publication of US20020160011A1 publication Critical patent/US20020160011A1/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
    • A61K39/39Medicinal preparations containing antigens or antibodies characterised by the immunostimulating additives, e.g. chemical adjuvants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • 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
    • A61K2039/55538IL-12
    • 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
    • A61K2039/55572Lipopolysaccharides; Lipid A; Monophosphoryl lipid A
    • 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
    • A61K2039/55577Saponins; Quil A; QS21; ISCOMS

Definitions

  • the present invention relates to improved adjuvant compositions, for the stimulation of an immune response suitable for immunotherapy applications.
  • the present invention relates to compositions comprising mixture of a saponin adjuvant with monophosphoryl lipid A or derivative thereof and interleukin 12 .
  • the invention relates to compositions comprising 3 de-o-acylated monosphoryl lipid A, QS 21 , and IL12. Such compositions are particularly useful in the immunotherapy of tumours.
  • Cancer is a disease developing from a single cell due to genetic changes. Clinical detection of these tumours occurs mostly in a relatively late stage of disease, when the primary tumour can be removed by surgery, and the existence of micro metastases settled in different organs has often already occurred. Chemotherapy does often not completely eliminate these cells, which then remain as a source for recurrent disease.
  • tumour cells are able to control all different tissues (with the exception of the brain) and, due to their memory function, can also eliminate hidden cells reentering the circulation (metastasis). Therefore, an activated immune response to tumour cells is expected to be of clinical benefit.
  • tumour cells are in many aspects indistinguishable from normal cells, and over-expression of certain proteins or expression of mutated proteins is in most cases not sufficient to activate the immune response. This situation results in failure of immune surveillance.
  • strategies for therapy of disseminated tumours need to specifically activate the immune response to tumour cells and to trigger migratory activity of cytotoxic T cells for example leading to elimination of most and possibly every single tumour cell. Genetic mutation in tumour cells is intense, and strong immune responses are therefore required to prevent further genetic changes of the tumour cells (escape variants) under the pressure of the immune system.
  • tumour antigens which are not cell surface proteins per se can be the targets of immune rejection through their recognition by immune regulatory and cytotoxic T cells.
  • New potential target antigens for immune-mediated tumour rejection are being identified, based on their recognition by immune T cells, rather than by antibodies. Such antigens may or may not induce antibody formation.
  • the expression of tumour antigens by a cell is in itself not sufficient for induction of an immune response to these antigens. Initiation of a tumour rejection response requires a series of immune amplification phenomena dependent on the intervention of antigen presenting cells, which are responsible for delivery of a series of activation signals which ultimately leads to the rejection of the tumour.
  • tumour rejection antigens which are presented on tumour cells and which are recognised by cytotoxic T cells can lead to lysis of the cell.
  • a vaccine composition comprising a tumour rejection antigen needs to be presented in a suitable adjuvant system to enable a suitable immune response to be mounted.
  • activation of the immune systems requires activation signals which are initiated by antigen presenting cells and are not activated by the tumour cells themselves.
  • Vaccination with isolated tumour rejection antigens has been envisaged either by recombinant proteins, by the use of live recombinant vectors or by DNA vectors. Preferably subunit antigens will be used. However, to ensure these are effective, powerful adjuvant systems are required.
  • the present invention provides an adjuvant composition
  • an adjuvant composition comprising a combination of a saponin adjuvant in combination with monophosphoryl lipid A or derivative thereof together with the cytokine Interleukin 12.
  • Immunologically active saponin fractions having adjuvant activity derived from the bark of the South American tree Quillaja Saponaria Molina are known in the art.
  • QS 21 also known as QA21, is an Hplc purified fraction from the Quillaja Saponaria Molina tree and it's method of its production is disclosed (as QA21) in U.S. Pat. No. 5,057,540.
  • Quillaja saponin has also been disclosed as an adjuvant by Scott et al, Int. Archs. Allergy Appl. Immun., 1985, 77, 409.
  • Monosphoryl lipid A and derivatives thereof are known in the art.
  • a preferred derivative is 3 de-o-acylated monophosphoryl lipid A, and is known from British Patent No. 2220211.
  • Interleukin 12 is known. For a review see Trinchieri G. Interleukin-12-A proinflammatory cytokine with immunoregulatory functions that bridge innate resistance and antigen-specific adaptive immunity. Immunology 13: 251.276, 1995. It is a heterodimeric cytokine produced mostly by phagocytic cells in response to bacteria, bacterial products, and intracellular parasites, and to some degree by B lymphocytes. In particular, IL-12 is produced by antigen presenting cells and instrumental in induction of TH- 1 cell responses. IL-12 induces IFN-gamma from NK and T cells, acts as a growth factor for activated NK and T cells, enhances the cytotoxic activity of NK cells, and induces cytotoxic T lymphocyte generation.
  • IL-12 and IL-12-induced IFN-gamma favor Th1 cell differentiation by priming CD4 (+) T cells for high IFN-gamma production.
  • cytokines such as IFN- ⁇ , IL-2, IL-6, IL-7, GM-CSF or MCP were unable to enhance the effect of the QS21/MPL adjuvant.
  • compositions of the invention contain the immunologically active saponin fraction in substantially pure form.
  • compositions of the invention contain QS21 in substantially pure form, that is to say, the QS21 is at least 90% pure, preferably at least 95% pure and most preferably at least 98% pure.
  • Other immunologically active saponin fractions useful in compositions of the invention include QA17/QS17.
  • the composition also comprises a sterol such as cholesterol wherein the sterol is present in an excess ratio to that of the saponin.
  • a sterol such as cholesterol wherein the sterol is present in an excess ratio to that of the saponin.
  • the adjuvant compositions can be utilised for the treatment or prophylaxis of a range of disease, they find particular utility in the field of cancer immunotherapy.
  • the adjuvant formulation finds utility particularly with tumour rejection antigens such as those for prostrate, breast, colorectal, pancreatic, renal or melanoma cancers.
  • exemplary antigens include MAGE 1 and MAGE 3 or other MAGE antigens for the treatment of melanoma, BAGE or GAGE LAGE (NY-eso 1 ) 1)PRAME or Her-2/neu; Robbins and Kawakami (1996), Current Opinions in Immunology 8, pps 628-636; Van den Eynde et al., International Journal of Clinical & Laboratory Research (submitted 1997); Correale et al. (1997), Journal of the National Cancer Institute 89 , p293.
  • antigens are expressed in a wide range of tumour types such as melanoma, lung carcinoma, sarcoma and bladder carcinoma.
  • Other classes of antigens useful in the context of the present invention include tissue specific antigens such as Prostate Specific antigen (PSA); Prostate Specific Membrane antigen (PMSA), Melan A/Mart 1, gp100, tyrosinase TRP1 or TRP2.
  • a vaccine comprising an adjuvant composition according to the invention and a tumour rejection antigen, or tissue specific antigen.
  • antigens or antigenic compositions include for example, polysaccharide antigens, protein antigens or DNA encoding antigens or antigenic compositions derived from HIV-1, (such as gp 120 or gp 160), any of Feline Immunodeficiency virus, human or animal herpes viruses, such as gD or derivatives thereof or Immediate Early protein such as ICP27 from HSV1 of HSV2, cytomegalovirus (especially human) (such as gB or derivatives thereof), Varicella Zoster Virus (such as gpl, II or III), or from a hepatitis virus such as hepatitis B virus for example Hepatitis B Surface antigen or a derivative thereof, hepatitis A virus, hepatitis C virus and hepatitis E virus, of from other viral pathogens, such as Respiratory Syncytial virus (for example RSV F and G proteins or immunogenic fragments thereof disclosed in U.S.
  • Respiratory Syncytial virus for
  • meningitis strains such as meningitis A, B and C, Streptococcus Pneumonia, human papillom
  • the P815 tumour is a mastocytoma, induced in a D BA/2 mouse with methylcholanthrene and cultured as both an in vitro tumour and cell line. This represents an excellent model system for the human.
  • the model system described in this application is a murine system whereby a a murine tumour antigen, P1A, expressed in the mouse mastocytoma P815, is being tested for its ability to stimulate CTL in the mouse with and without adjuvant.
  • P1A is a true tumour rejection antigen in that its gene is the same in both normal and tumour cells but the gene is silent in normal cells and only expressed in tumour cells. This is in comparison to other P815 antigens that were previously found and which are created by mutation of normal alleles. These are called tum- variants or tum- antigens. Mutations in the tumantigens create new antigenic peptides which can then be recognised by CTL.
  • tumour- antigens are likely to be tumour-specific whereas true tumour antigens will be shared between different tumours and patients and therefore the latter will be better candidates for vaccine formulations.
  • Human tumour rejection antigens analogous to P1A include the MAGE, BAGE, GAGE etc. families as described earlier. These genes are found in both normal and tumour tissues but the corresponding proteins are expressed only in tumours and in normal testis. As the testis is an immune privileged site it is unlikely to be affected by any vaccine.
  • P1A is a true murine TRAs. Therefore, one can test a large number of adjuvants with a variety of different assays in mice giving a good indication as to which formulations should be used with human tumour rejection antigens in human clinical trials.
  • compositions of the invention are those forming a liposome structure.
  • Compositions where the sterol/immunologically active saponin fraction forms an ISCOM structure also form an aspect of the invention.
  • the ratio of QS21 : sterol will typically be in the order of 1:100 to 1:1 weight to weight. Preferably excess sterol is present, the ratio of QS21: sterol being at least 1:2 w/w.
  • QS21 and sterol will be present in a vaccine in the range of about 1 ng to about 100 ⁇ g, preferably about 10 ⁇ g to about 50 ⁇ g per dose.
  • the liposomes preferably contain a neutral lipid, for example phosphatidylcholine, which is preferably non-crystalline at room temperature, for example eggyolk phosphatidylcholine, dioleoyl phosphatidylcholine or dilauryl phosphatidylcholine.
  • the liposomes may also contain a charged lipid which increases the stability of the lipsome-QS21 structure for liposomes composed of saturated lipids. In these cases the amount of charged lipid is preferably 1-20% w/w, most preferably 5-10%.
  • the ratio of sterol to phospholipid is 1-50% (mol/mol), most preferably 20-25%.
  • compositions of the invention also contain a 3 deacylated monophosphoryl lipid A derivative (3-de- 0 -acylated monophosphoryl lipid A, also known as 3D-MPL) and is manufactured by Ribi Immunochem, Montana.
  • 3D-MPL 3 deacylated monophosphoryl lipid A derivative
  • compositions of the invention are those wherein liposomes are initially prepared without 3D-MPL, and 3D-MPL is then added, preferably as 100nm particles.
  • the 3D-MPL is therefore not contained within the vesicle membrane (known as 3D-MPL out).
  • Compositions where the 3D-MPL is contained within the vesicle membrane also form an aspect of the invention.
  • the antigen can be contained within the vesicle membrane or contained outside the vesicle membrane or encapsulated.
  • Preferably soluble antigens are outside and hydrophobic or lipidated antigens are either contained inside or outside the membrane.
  • the 3D-MPL will be present in the range of about 1Ug to 100Ug and preferably about 10 to 50 ⁇ g per dose of human vaccine.
  • the vaccines of the invention will not require any specific carrier and formulated in an aqueous or other pharmaceutically acceptable buffer. In some cases it may be advantageous that the vaccines of the present invention will further contain alum.
  • An adjuvant composition comprising QS21, lipids (DQ) and 3 de-o-acylated monophosphoryl lipid A (3D-MPL) was prepared.
  • lipid such as phosphatidyicholine either from egg-yolk or synthetic
  • cholesterol in organic solvent
  • An aqueous solution such as phosphate buffered saline
  • This suspension is then microfluidised until the liposome size is reduced to 100 nm, and then sterile filtered through a 0.2 Um filter. Extrusion or sonication could replace this step.
  • the cholesterol phosphatidylcholine ratio is 1:4 (w/w), and the aqueous solution is added to give a final cholesterol concentration of 5 to 50 ng/ml.
  • the liposomes have a defined size of 100 nm and are referred to as SUV (for small unilamellar vesicles). If this solution is repeatedly frozen and thawed the vesicles fuse to form large multilamellar structures (MLV) of size ranging from 500nm to 15 ⁇ m.
  • MLV multilamellar structures
  • FIG. 1 Immunisation of DBA/2 mice with peptide p198 ⁇ DQS21/3D-MPL +IL12
  • mice On d1 and 2, we injected locally an additional dose of IL12 100ng (1000U) or PBS.
  • the mice were bled and stimulation of blood lymphocytes was performed by mixing 3 ⁇ 10 6 Ficoll purified lymphocytes with 10 5 irradiated stimulating cells (100 Gy) and 2 ⁇ 10 6 irradiated normal syngeneic spleen cells (30 Gy) as feeder cells.
  • the stimulating cells were P1983 cells, an azaguanine-resistant variant derived from the P198 TUM-clone.
  • the cells were incubated in 48-well plates in a final volume of 0,8ml MLTC medium described in Warnier et al. (Int. J. Cancer, 1996, 67, 303-310).
  • CTL activity was measured in a standard chromium-release assay using 1,000 5 1 Cr-labelled targets. Two targets were used: the P1983 cells or the P511 cells (azaguanine-resistant variant derived from the P815 TUM+cells) not expressing the P198 antigen. To eliminate non-specific lysis, 10 5 cold P511 cells were added as competitors. On day 26, the mice received a second injection of peptide, adjuvant and IL12 or PBS, followed by two local injections of 100ng (1000U) IL12 or PBS. A second bleeding of the mice was performed on day 41 to estimate CTL activity after two injections. Data are expressed in lytic units (LU)/10 6 lymphocytes as described in Brichard et al.
  • LU lytic units
  • FIG. 2 CTL activity in mice injected with peptide P198 ⁇ DQS21/3D-MPL ⁇ IL12
  • FIG. 3 Immunisation of DBA/2 mice with peptide P198 ⁇ DQS21/3D-MPL ⁇ IL12 in footpads or flanks.
  • FIG. 4 CTL activity in mice injected with peptide P198 ⁇ DQS21/3D-MPL ⁇ IL12 in the footpads or the flanks.
  • mice receiving the peptide, the adjuvant and IL12 combination in the footpads exhibited high CTL activity. All the mice injected with the peptide and the IL12 without adjuvant also showed a specific CTL activity, but much weaker. The same situation is observed for the mice injected in the flank with the peptide, the adjuvant and the IL12 while in the absence of IL12 no response is observed after injection in the flank. After the fourth injection, all the mice that received peptide, adjuvant and IL12 in the footpads had a CTL activity located in the high values.
  • mice injected without adjuvant or receiving the peptide, the adjuvant and the IL12 in the flank We also observed an increase in the average of CTL activity for the mice injected without adjuvant or receiving the peptide, the adjuvant and the IL12 in the flank. Even after 4 injections we did not observe any response in the mice injected in the flank without IL12.
  • FIG. 5 IL12 dose curve
  • mice were injected with the P198 peptide mixed with the DQS21/3D-MPL adjuvant.
  • Different doses of murine IL12 3ng (30U), 10ng (100U), 30ng (300U), 100ng (1000U) were mixed with the peptide and the adjuvant and also repeated locally the two following days.
  • the control group received the peptide and the adjuvant but no IL12.
  • Mice were bled after each of the two immunisations to monitor the appearance and level of CTL activity.
  • FIG. 6 CTL activity in mice receiving peptide P198 ⁇ DQS21/3D-MPL ⁇ various doses of IL12
  • FIG. 7 Immunisation of DBA/2 mice with peptide P1A ⁇ DQS21/3D-MPL ⁇ IL12
  • the P815A antigen represents a good mouse model for the human MAGE, BAGE and GAGE antigens.
  • mice were injected s.c. in the two footpads with 50 ⁇ g of P1A peptide (LPYLGWLVF described in Lethé et al. Eur. J. Immunol. 1992, 22: 2283-2288) mixed with the adjuvant DQS21/3D-MPL.
  • 100ng (1000U) of IL-12 was added to the peptide and the adjuvant.
  • These mice received additional doses of 100ng (1000U) IL-12 injected locally the two following days. This injection scheme was repeated four times and the mice were bled after the second and the fourth injection.
  • the lymphocytes were restimulated in vitro for 7 days and the CTL activity was measured in a conventional 51 Cr assay. We used L1210.
  • P1A cells as stimulating cells.
  • the syngeneic L1210 P1A transfectant cells expressing the antigen P815AB were generated as described in Uyttenhove et al. Int. J. Cancer (1997) 70: 349-356.
  • target cells we used P511 cells expressing all the P815 antigens and P1-204 cells, an antigen-loss variant not expressing the P815 AB antigen described in Uyttenhove et al (J. Exp. Med., 157, 1040-1052, 1983).
  • cold P1-204 were added as competitors.
  • FIG. 8 CTL activity in mice injected with the P1A peptide ⁇ DQS21/13D-MPL ⁇ IL12
  • mice out of ten showed significant CTL activity specific for the P815A antigen when IL12 was added to the peptide and adjuvant. Half of those mice exhibited high CTL activity levels. In the group injected without IL12, a positive response was detected only in one mouse and this activity was rather low.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Public Health (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Immunology (AREA)
  • Microbiology (AREA)
  • Mycology (AREA)
  • Epidemiology (AREA)
  • Communicable Diseases (AREA)
  • Oncology (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)

Abstract

The present invention provides improved adjuvant compositions comprising QS21/3DMPL and Interleukin 12. These find utility in a range of prophylatic and therapeutic vaccines, including cancer vaccines. 09/445903

Description

  • The present invention relates to improved adjuvant compositions, for the stimulation of an immune response suitable for immunotherapy applications. In particular the present invention relates to compositions comprising mixture of a saponin adjuvant with monophosphoryl lipid A or derivative thereof and [0001] interleukin 12. In particular, the invention relates to compositions comprising 3 de-o-acylated monosphoryl lipid A, QS21, and IL12. Such compositions are particularly useful in the immunotherapy of tumours.
  • Cancer is a disease developing from a single cell due to genetic changes. Clinical detection of these tumours occurs mostly in a relatively late stage of disease, when the primary tumour can be removed by surgery, and the existence of micro metastases settled in different organs has often already occurred. Chemotherapy does often not completely eliminate these cells, which then remain as a source for recurrent disease. [0002]
  • Immune cells are able to control all different tissues (with the exception of the brain) and, due to their memory function, can also eliminate hidden cells reentering the circulation (metastasis). Therefore, an activated immune response to tumour cells is expected to be of clinical benefit. Despite their undifferentiated growth, tumour cells are in many aspects indistinguishable from normal cells, and over-expression of certain proteins or expression of mutated proteins is in most cases not sufficient to activate the immune response. This situation results in failure of immune surveillance. Thus, strategies for therapy of disseminated tumours need to specifically activate the immune response to tumour cells and to trigger migratory activity of cytotoxic T cells for example leading to elimination of most and possibly every single tumour cell. Genetic mutation in tumour cells is intense, and strong immune responses are therefore required to prevent further genetic changes of the tumour cells (escape variants) under the pressure of the immune system. [0003]
  • It is now well established that cellular antigens which are not cell surface proteins per se can be the targets of immune rejection through their recognition by immune regulatory and cytotoxic T cells. New potential target antigens for immune-mediated tumour rejection are being identified, based on their recognition by immune T cells, rather than by antibodies. Such antigens may or may not induce antibody formation. It is now recognized that the expression of tumour antigens by a cell is in itself not sufficient for induction of an immune response to these antigens. Initiation of a tumour rejection response requires a series of immune amplification phenomena dependent on the intervention of antigen presenting cells, which are responsible for delivery of a series of activation signals which ultimately leads to the rejection of the tumour. [0004]
  • Tumour rejection antigens which are presented on tumour cells and which are recognised by cytotoxic T cells can lead to lysis of the cell. To achieve this, in a clinical setting a vaccine composition comprising a tumour rejection antigen needs to be presented in a suitable adjuvant system to enable a suitable immune response to be mounted. However, activation of the immune systems requires activation signals which are initiated by antigen presenting cells and are not activated by the tumour cells themselves. [0005]
  • Vaccination with isolated tumour rejection antigens has been envisaged either by recombinant proteins, by the use of live recombinant vectors or by DNA vectors. Preferably subunit antigens will be used. However, to ensure these are effective, powerful adjuvant systems are required. [0006]
  • Accordingly, the present invention provides an adjuvant composition comprising a combination of a saponin adjuvant in combination with monophosphoryl lipid A or derivative thereof together with the cytokine Interleukin 12. [0007]
  • Immunologically active saponin fractions having adjuvant activity derived from the bark of the South American tree Quillaja Saponaria Molina are known in the art. For example QS[0008] 21, also known as QA21, is an Hplc purified fraction from the Quillaja Saponaria Molina tree and it's method of its production is disclosed (as QA21) in U.S. Pat. No. 5,057,540. Quillaja saponin has also been disclosed as an adjuvant by Scott et al, Int. Archs. Allergy Appl. Immun., 1985, 77, 409.
  • Monosphoryl lipid A and derivatives thereof are known in the art. A preferred derivative is 3 de-o-acylated monophosphoryl lipid A, and is known from British Patent No. 2220211. [0009]
  • Interleukin [0010] 12 (IL-12) is known. For a review see Trinchieri G. Interleukin-12-A proinflammatory cytokine with immunoregulatory functions that bridge innate resistance and antigen-specific adaptive immunity. Immunology 13: 251.276, 1995. It is a heterodimeric cytokine produced mostly by phagocytic cells in response to bacteria, bacterial products, and intracellular parasites, and to some degree by B lymphocytes. In particular, IL-12 is produced by antigen presenting cells and instrumental in induction of TH-1 cell responses. IL-12 induces IFN-gamma from NK and T cells, acts as a growth factor for activated NK and T cells, enhances the cytotoxic activity of NK cells, and induces cytotoxic T lymphocyte generation.
  • IL-12 and IL-12-induced IFN-gamma favor Th1 cell differentiation by priming CD4 (+) T cells for high IFN-gamma production. However, we surprisingly found that other cytokines, such as IFN-γ, IL-2, IL-6, IL-7, GM-CSF or MCP were unable to enhance the effect of the QS21/MPL adjuvant. [0011]
  • Preferably the compositions of the invention contain the immunologically active saponin fraction in substantially pure form. Preferably the compositions of the invention contain QS21 in substantially pure form, that is to say, the QS21 is at least 90% pure, preferably at least 95% pure and most preferably at least 98% pure. Other immunologically active saponin fractions useful in compositions of the invention include QA17/QS17. [0012]
  • In a preferred embodiment the composition also comprises a sterol such as cholesterol wherein the sterol is present in an excess ratio to that of the saponin. These show decreased reactogenicity when compared to compositions in which the cholesterol is absent, while the adjuvant effect is maintained. In addition it is known that QS21 degrades under basic conditions where the pH is about 7 or greater. Thus a further advantage is that the stability of QS[0013] 21 to basemediated hydrolysis is enhanced in formulations containing cholesterol.
  • Although the adjuvant compositions can be utilised for the treatment or prophylaxis of a range of disease, they find particular utility in the field of cancer immunotherapy. [0014]
  • In particular, the adjuvant formulation finds utility particularly with tumour rejection antigens such as those for prostrate, breast, colorectal, pancreatic, renal or melanoma cancers. Exemplary antigens include [0015] MAGE 1 and MAGE 3 or other MAGE antigens for the treatment of melanoma, BAGE or GAGE LAGE (NY-eso1) 1)PRAME or Her-2/neu; Robbins and Kawakami (1996), Current Opinions in Immunology 8, pps 628-636; Van den Eynde et al., International Journal of Clinical & Laboratory Research (submitted 1997); Correale et al. (1997), Journal of the National Cancer Institute 89, p293. Indeed these antigens are expressed in a wide range of tumour types such as melanoma, lung carcinoma, sarcoma and bladder carcinoma. Other classes of antigens useful in the context of the present invention include tissue specific antigens such as Prostate Specific antigen (PSA); Prostate Specific Membrane antigen (PMSA), Melan A/Mart 1, gp100, tyrosinase TRP1 or TRP2.
  • Accordingly in one aspect of the present invention there is provided a vaccine comprising an adjuvant composition according to the invention and a tumour rejection antigen, or tissue specific antigen. [0016]
  • Other antigens or antigenic compositions include for example, polysaccharide antigens, protein antigens or DNA encoding antigens or antigenic compositions derived from HIV-1, (such as gp 120 or gp 160), any of Feline Immunodeficiency virus, human or animal herpes viruses, such as gD or derivatives thereof or Immediate Early protein such as ICP27 from HSV1 of HSV2, cytomegalovirus (especially human) (such as gB or derivatives thereof), Varicella Zoster Virus (such as gpl, II or III), or from a hepatitis virus such as hepatitis B virus for example Hepatitis B Surface antigen or a derivative thereof, hepatitis A virus, hepatitis C virus and hepatitis E virus, of from other viral pathogens, such as Respiratory Syncytial virus (for example RSV F and G proteins or immunogenic fragments thereof disclosed in U.S. Pat. No. 5,149,650 or chimeric polypeptides containing immunogenic fragments from HSRV proteins F and G, eg GF glycoprotein disclosed in U.S. Pat. No. 5,194,595), antigens derived from meningitis strains such as meningitis A, B and C, Streptococcus Pneumonia, human papilloma virus, in particular from strains HPV6, 11, 16 and 18, Influenza virus, Haemophilus Influenza B (Hib), Epstein Barr Virus (EBV), or derived from bacterial pathogens such as Salmonella, Neisseria, Borrelia (for example OspA or OspB or derivatives thereof), or Chlamydia, or Bordetella for example P.69, PT and FHA, or derived from parasites such as plasmodium or toxoplasma. [0017]
  • The P815 tumour is a mastocytoma, induced in a D BA/2 mouse with methylcholanthrene and cultured as both an in vitro tumour and cell line. This represents an excellent model system for the human. [0018]
  • The model system described in this application is a murine system whereby a a murine tumour antigen, P1A, expressed in the mouse mastocytoma P815, is being tested for its ability to stimulate CTL in the mouse with and without adjuvant. The significance of this system is that P1A is a true tumour rejection antigen in that its gene is the same in both normal and tumour cells but the gene is silent in normal cells and only expressed in tumour cells. This is in comparison to other P815 antigens that were previously found and which are created by mutation of normal alleles. These are called tum- variants or tum- antigens. Mutations in the tumantigens create new antigenic peptides which can then be recognised by CTL. [0019]
  • Tum- antigens are likely to be tumour-specific whereas true tumour antigens will be shared between different tumours and patients and therefore the latter will be better candidates for vaccine formulations. Human tumour rejection antigens analogous to P1A include the MAGE, BAGE, GAGE etc. families as described earlier. These genes are found in both normal and tumour tissues but the corresponding proteins are expressed only in tumours and in normal testis. As the testis is an immune privileged site it is unlikely to be affected by any vaccine. [0020]
  • P1A is a true murine TRAs. Therefore, one can test a large number of adjuvants with a variety of different assays in mice giving a good indication as to which formulations should be used with human tumour rejection antigens in human clinical trials. [0021]
  • Preferred compositions of the invention are those forming a liposome structure. Compositions where the sterol/immunologically active saponin fraction forms an ISCOM structure also form an aspect of the invention. [0022]
  • The ratio of QS21 : sterol will typically be in the order of 1:100 to 1:1 weight to weight. Preferably excess sterol is present, the ratio of QS21: sterol being at least 1:2 w/w. Typically for human administration QS21 and sterol will be present in a vaccine in the range of about 1 ng to about 100μg, preferably about 10 μg to about 50 μg per dose. [0023]
  • The liposomes preferably contain a neutral lipid, for example phosphatidylcholine, which is preferably non-crystalline at room temperature, for example eggyolk phosphatidylcholine, dioleoyl phosphatidylcholine or dilauryl phosphatidylcholine. The liposomes may also contain a charged lipid which increases the stability of the lipsome-QS21 structure for liposomes composed of saturated lipids. In these cases the amount of charged lipid is preferably 1-20% w/w, most preferably 5-10%. The ratio of sterol to phospholipid is 1-50% (mol/mol), most preferably 20-25%. [0024]
  • The compositions of the invention also contain a 3 deacylated monophosphoryl lipid A derivative (3-de-[0025] 0-acylated monophosphoryl lipid A, also known as 3D-MPL) and is manufactured by Ribi Immunochem, Montana. A preferred form is disclosed in International Patent Application 92/116556.
  • Suitable compositions of the invention are those wherein liposomes are initially prepared without 3D-MPL, and 3D-MPL is then added, preferably as 100nm particles. The 3D-MPL is therefore not contained within the vesicle membrane (known as 3D-MPL out). Compositions where the 3D-MPL is contained within the vesicle membrane (known as 3D-MPL in) also form an aspect of the invention. The antigen can be contained within the vesicle membrane or contained outside the vesicle membrane or encapsulated. Preferably soluble antigens are outside and hydrophobic or lipidated antigens are either contained inside or outside the membrane. The 3D-MPL will be present in the range of about 1Ug to 100Ug and preferably about 10 to 50 μg per dose of human vaccine. [0026]
  • Often the vaccines of the invention will not require any specific carrier and formulated in an aqueous or other pharmaceutically acceptable buffer. In some cases it may be advantageous that the vaccines of the present invention will further contain alum.[0027]
  • Example 1:
  • a)—Immunisation of DBA/2 mice with peptide+3 D-MPL+QS21+Lipids ±IL12 [0028]
  • Human tumours express antigens that can be recognized by autologous CTL. These antigens constitute useful targets for cancer immunotherapy. We decided to evaluate in the P815 murine mastocytoma model the efficacy of an immunization method that could be applied to human patients. Syngeneic DBA/2 mice were an-injected with antigenic peptides mixed with adjuvant and murine IL12. [0029]
  • An adjuvant composition comprising QS21, lipids (DQ) and 3 de-o-acylated monophosphoryl lipid A (3D-MPL) was prepared. [0030]
  • Briefly a mixture of lipid (such as phosphatidyicholine either from egg-yolk or synthetic) and cholesterol in organic solvent, is dried down under vacuum (or alternatively under a stream of inert gas). An aqueous solution (such as phosphate buffered saline) is then added, and the vessel agitated until all the lipid is in suspension. This suspension is then microfluidised until the liposome size is reduced to [0031] 100 nm, and then sterile filtered through a 0.2 Um filter. Extrusion or sonication could replace this step.
  • The cholesterol phosphatidylcholine ratio is 1:4 (w/w), and the aqueous solution is added to give a final cholesterol concentration of 5 to 50 ng/ml. The liposomes have a defined size of 100 nm and are referred to as SUV (for small unilamellar vesicles). If this solution is repeatedly frozen and thawed the vesicles fuse to form large multilamellar structures (MLV) of size ranging from 500nm to 15μm. [0032]
  • QS21 in aqueous solution is added to the liposomes. This mixture is then added to 50 μg of P198 peptide (KYQAVTTTL) and 3D-MPL. [0033]
  • FIG. 1 Immunisation of DBA/2 mice with peptide p198±DQS21/3D-MPL +IL12 [0034]
  • DBA/2 mice were injected s.c. in the two footpads with 50μg of P198 peptide (KYQAVTTTL), corresponding to the antigen expressed by the P198 TUM—clone (Sibille et al., J. Exp. Med., 1990: 172, 3545), mixed with the DQS21/3DMPL adjuvant (adjuvant) 100μl final. For a second group of animals we added to the peptide and adjuvant solution 50μg (500U) murine IL12. This murine IL12 was purified from the supernatant of transfected P1HTR cells as described in Gajewski et al. (J. Immunol. 1995, 154: 5637-5648). On d1 and 2, we injected locally an additional dose of IL12 100ng (1000U) or PBS. On day 16, the mice were bled and stimulation of blood lymphocytes was performed by mixing 3×10[0035] 6 Ficoll purified lymphocytes with 105 irradiated stimulating cells (100 Gy) and 2×106 irradiated normal syngeneic spleen cells (30 Gy) as feeder cells. The stimulating cells were P1983 cells, an azaguanine-resistant variant derived from the P198 TUM-clone. The cells were incubated in 48-well plates in a final volume of 0,8ml MLTC medium described in Warnier et al. (Int. J. Cancer, 1996, 67, 303-310). Seven days later, CTL activity was measured in a standard chromium-release assay using 1,000 51Cr-labelled targets. Two targets were used: the P1983 cells or the P511 cells (azaguanine-resistant variant derived from the P815 TUM+cells) not expressing the P198 antigen. To eliminate non-specific lysis, 105 cold P511 cells were added as competitors. On day 26, the mice received a second injection of peptide, adjuvant and IL12 or PBS, followed by two local injections of 100ng (1000U) IL12 or PBS. A second bleeding of the mice was performed on day 41 to estimate CTL activity after two injections. Data are expressed in lytic units (LU)/106 lymphocytes as described in Brichard et al. (Eur. J. Inununol., 1995, 25: 664-671). Specific lytic units were calculated by subtracting the values obtained with the negative targets (usually less than 0,3 LU) from those obtained with the positive target. Mice were scored as±when the LU detected were comprised between 0,1 and 1;+when LU were comprised between 1 and 10; and++above 10 LU.
  • FIG. 2 CTL activity in mice injected with peptide P198±DQS21/3D-MPL±IL12 [0036]
  • After the first injection, no CTL activity was detected in the mice injected with the peptide and the adjuvant. When IL12 was added significant CTL activity was detected in all the animals. For the majority of the mice (13/15) the response was moderate since we measured less than 1 LU/10[0037] 6PBL. After the second immunization, two mice out of 15, injected with the peptide and adjuvant alone were positive. In the group injected with IL12, CTL activity had increased and half of the mice showed a very high response. The addition of IL12 to the peptide and the adjuvant increased strongly the number of responding mice and the level of CTL activity observed after only a few injections.
  • FIG. 3 Immunisation of DBA/2 mice with peptide P198±DQS21/3D-MPL±IL12 in footpads or flanks. [0038]
  • In this second experiment, we applied the immunisation protocol described before (FIG. 1) with some modifications. To determine the relative contribution of IL12 and adjuvant in CTL induction, we injected one group of mice in the footpads with P198 peptide and IL12 without adjuvant. To test a s.c. injection site that is applicable to humans, we also injected 2 groups of mice s.c. in the flank instead of the footpads; the first one receiving the peptide, the adjuvant and the IL12 and the second receiving only the peptide and the adjuvant. Four injections were performed and mice were bled after the first, the second and the fourth injection for CTL activity determinations. [0039]
  • FIG. 4 CTL activity in mice injected with peptide P198±DQS21/3D-MPL±IL12 in the footpads or the flanks. [0040]
  • After the first immunisation, we observed that 4 mice out of 10 injected with peptide, the adjuvant and IL12 in the footpads showed a significant CTL activity. In the group injected without [0041] adjuvant 3 mice also showed CTL activity but the response was lower. Nearly no response was obtained after injection into the flanks since only 2 mice receiving the peptide, the adjuvant and IL12 showed a weak CTL activity.
  • After the second immunisation, all the mice receiving the peptide, the adjuvant and IL12 combination in the footpads exhibited high CTL activity. All the mice injected with the peptide and the IL12 without adjuvant also showed a specific CTL activity, but much weaker. The same situation is observed for the mice injected in the flank with the peptide, the adjuvant and the IL12 while in the absence of IL12 no response is observed after injection in the flank. After the fourth injection, all the mice that received peptide, adjuvant and IL12 in the footpads had a CTL activity located in the high values. We also observed an increase in the average of CTL activity for the mice injected without adjuvant or receiving the peptide, the adjuvant and the IL12 in the flank. Even after [0042] 4 injections we did not observe any response in the mice injected in the flank without IL12.
  • We confirm in this experiment the potent effect of IL12 on the generation of CTL activity after immunisation with the P198 peptide. This effect is enhanced by the combination with the DQS21/3D-MPL adjuvant since the response is obtained earlier in all the mice and since the average level of response is higher. The effect of IL12 is also required to obtain CTL activity when the antigen is injected in the flank instead of in the footpads. [0043]
  • FIG. 5 IL12 dose curve [0044]
  • Mice were injected with the P198 peptide mixed with the DQS21/3D-MPL adjuvant. Different doses of murine IL12 3ng (30U), 10ng (100U), 30ng (300U), 100ng (1000U) were mixed with the peptide and the adjuvant and also repeated locally the two following days. The control group received the peptide and the adjuvant but no IL12. Mice were bled after each of the two immunisations to monitor the appearance and level of CTL activity. [0045]
  • FIG. 6 CTL activity in mice receiving peptide P198±DQS21/3D-MPL±various doses of IL12 [0046]
  • In the two preceding experiments we used a high dose of IL12 (1μg/mouse/day). Even if the IL12 was injected locally we saw a systemic toxicity with symptoms similar to those observed in a LPS shock. We decided to try decreasing doses of IL12. The effect of IL12 was nearly fully maintained when the dose/mouse/day was decreased to 10ng (100U). It disappeared when the mice were injected with only 30 ng IL12. At that dose, the systemic toxicity of IL12 was largely reduced but not totally absent. [0047]
  • FIG. 7 Immunisation of DBA/2 mice with peptide P1A±DQS21/3D-MPL±IL12 [0048]
  • After several experiments with the peptide P198 showing that high CTL activity were induced by injections of a combination of peptide, adjuvant and IL12, we decided to apply this protocol to the P1A peptide. This peptide presented by the Ld molecule constitutes the P815A antigen that is a major target for the immune rejection in vivo (Uyttenhove et al. J. Exp. Med., 1983, 157: 1040-1052). Gene P1A, which code for the P815A antigen is expressed in several mastocytoma tumour lines (Van den Eynde et al. J. Exp. Med., 1991, 173: 1373-1384). Like the MAGE, BAGE and GAGE genes, it is not expressed in adult normal tissues, with the exception of spermatogonia in the testis (Van den Eynde et al, 1991 and Uyttenhove et al, Int. J. Cancer, 1997, 70: 349-356). Accordingly, the P815A antigen represents a good mouse model for the human MAGE, BAGE and GAGE antigens. [0049]
  • Mice were injected s.c. in the two footpads with 50μg of P1A peptide (LPYLGWLVF described in Lethé et al. Eur. J. Immunol. 1992, 22: 2283-2288) mixed with the adjuvant DQS21/3D-MPL. For one group, 100ng (1000U) of IL-12 was added to the peptide and the adjuvant. These mice received additional doses of 100ng (1000U) IL-12 injected locally the two following days. This injection scheme was repeated four times and the mice were bled after the second and the fourth injection. The lymphocytes were restimulated in vitro for 7 days and the CTL activity was measured in a conventional[0050] 51Cr assay. We used L1210. P1A cells as stimulating cells. The syngeneic L1210 P1A transfectant cells expressing the antigen P815AB were generated as described in Uyttenhove et al. Int. J. Cancer (1997) 70: 349-356. As target cells we used P511 cells expressing all the P815 antigens and P1-204 cells, an antigen-loss variant not expressing the P815 AB antigen described in Uyttenhove et al (J. Exp. Med., 157, 1040-1052, 1983). To avoid problems of non specific lysis, cold P1-204 were added as competitors.
  • FIG. 8 CTL activity in mice injected with the P1A peptide±DQS21/13D-MPL ±IL12 [0051]
  • After two injections, 9 mice out of ten showed significant CTL activity specific for the P815A antigen when IL12 was added to the peptide and adjuvant. Half of those mice exhibited high CTL activity levels. In the group injected without IL12, a positive response was detected only in one mouse and this activity was rather low. [0052]
  • After the fourth injection, all the mice receiving IL12 were positive and the average of lytic units had increased. Without IL12 we detected a good CTL activity in four mice, one additional mouse showed a very low response at the limit of the significance threshold. In this system again, the addition of IL12 increased the number of responding mice and diminished the number of injections needed to obtain high and specific CTL activity. In this experiment we injected a high dose of IL12 (100ng (1000U)/mouse/day). Like in the previous experiments using the P198 peptide we observed systemic toxic effects of the IL12. [0053]
  • Conclusion: [0054]
  • The addition of IL12 to peptide and adjuvant combination is very effective at increasing the number of mice displaying high CTL responses after immunisation. In addition, CTL responses appear earlier in the presence of IL12. [0055]

Claims (10)

1. A vaccine composition comprising a saponin adjuvant, monophosphoryl lipid A or derivative thereof, Interleukin 12, and an antigen.
2. A vaccine composition as claimed in claim 1 wherein the monophosphoryl lipid A is 3-O-deacylated monophosphoryl lipid A.
3. A vaccine composition as claimed in claim 1 or claim 2 wherein the saponin adjuvant is QS21.
4. A vaccine composition as claimed in claim 3 additionally comprising cholesterol.
5. A vaccine composition as claimed in claim 1 to 4, wherein the antigen is a tumour rejection antigen.
6. A vaccine composition as claimed herein for use in medicine.
7. Use of a saponin adjuvant, monophosphoryl lipid A or a derivative thereof and Interleukin 12 and an antigen in the manufacture of a vaccine for the treatment of prophylaxis of pathogenic infections or cancer.
8. A method of enhancing an immune response to an antigen comprising administering the antigen with an adjuvant composition as described in cairns 1 to 4:
9. A method of treating or preventing a pathogenic infection in a patient comprising administering a vaccine as claimed herein.
10. A method of producing a vaccine composition as claimed in claim 1, comprises admixing a saponin adjuvant, monophosphoryl lipid A or derivative thereof, Interleukin 12 and an antigen.
US09/445,903 1997-06-14 1998-06-09 Adjuvant compositions for vaccines Abandoned US20020160011A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
GBGB9712347.5A GB9712347D0 (en) 1997-06-14 1997-06-14 Vaccine
GB9712347.5 1997-06-14

Publications (1)

Publication Number Publication Date
US20020160011A1 true US20020160011A1 (en) 2002-10-31

Family

ID=10814118

Family Applications (2)

Application Number Title Priority Date Filing Date
US09/445,903 Abandoned US20020160011A1 (en) 1997-06-14 1998-06-09 Adjuvant compositions for vaccines
US09/445,841 Expired - Fee Related US6375945B1 (en) 1997-06-14 1998-06-09 Adjuvant compositions for vaccines

Family Applications After (1)

Application Number Title Priority Date Filing Date
US09/445,841 Expired - Fee Related US6375945B1 (en) 1997-06-14 1998-06-09 Adjuvant compositions for vaccines

Country Status (6)

Country Link
US (2) US20020160011A1 (en)
EP (2) EP0977589A1 (en)
JP (2) JP2002504135A (en)
CA (2) CA2293614A1 (en)
GB (1) GB9712347D0 (en)
WO (2) WO1998057659A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100012927A1 (en) * 2001-07-25 2010-01-21 Nantero, Inc. Devices having vertically-disposed nanofabric articles and methods of making the same

Families Citing this family (179)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU1463097A (en) 1996-01-04 1997-08-01 Rican Limited Helicobacter pylori bacterioferritin
US6835721B2 (en) 1999-02-01 2004-12-28 Eisai Co., Ltd. Immunomodulatory compounds and methods of use thereof
US7915238B2 (en) 1999-02-01 2011-03-29 Eisai R & D Management Co., Ltd. Immunomodulatory compounds and methods of use thereof
US20040006242A1 (en) 1999-02-01 2004-01-08 Hawkins Lynn D. Immunomodulatory compounds and method of use thereof
GB9909077D0 (en) * 1999-04-20 1999-06-16 Smithkline Beecham Biolog Novel compositions
IL146382A0 (en) * 1999-05-13 2002-07-25 American Cyanamid Co Adjuvant combination formulations
EP1385541B1 (en) * 2000-04-13 2008-06-18 Corixa Corporation Immunostimulant compositions comprising an aminoalkyl glucosaminide phosphate and qs-21
AU2001270031B2 (en) * 2000-06-22 2005-11-03 Wyeth Holdings Corporation QS-21 and IL-12 as an adjuvant combination
WO2003026579A2 (en) * 2001-09-25 2003-04-03 The Regents Of The University Of California Combination therapies for immune mediated diseases
US7939087B2 (en) 2000-10-27 2011-05-10 Novartis Vaccines And Diagnostics, Inc. Nucleic acids and proteins from Streptococcus groups A & B
PL366031A1 (en) * 2000-11-10 2005-01-24 Wyeth Holdings Corporation Adjuvant combination formulations
EP1415005B1 (en) 2000-12-07 2012-11-21 Novartis Vaccines and Diagnostics, Inc. Endogenous retroviruses up-regulated in prostate cancer
GB0115176D0 (en) 2001-06-20 2001-08-15 Chiron Spa Capular polysaccharide solubilisation and combination vaccines
US8481043B2 (en) 2001-06-22 2013-07-09 Cpex Pharmaceuticals, Inc. Nasal immunization
JP4370161B2 (en) 2001-06-29 2009-11-25 ノバルティス バクシンズ アンド ダイアグノスティックス,インコーポレーテッド HCVE1E2 vaccine composition
GB0118249D0 (en) 2001-07-26 2001-09-19 Chiron Spa Histidine vaccines
GB0121591D0 (en) 2001-09-06 2001-10-24 Chiron Spa Hybrid and tandem expression of neisserial proteins
US20030129161A1 (en) * 2001-09-17 2003-07-10 Hsien-Jue Chu Interleukin-12 as a veterinary vaccine adjuvant
US7030094B2 (en) 2002-02-04 2006-04-18 Corixa Corporation Immunostimulant compositions comprising an aminoalkyl glucosaminide phosphate and QS-21
EP2572707A3 (en) 2002-02-20 2013-11-06 Novartis Vaccines and Diagnostics, Inc. Microparticles with adsorbed polypeptide-containing molecules
US6861410B1 (en) 2002-03-21 2005-03-01 Chiron Corporation Immunological adjuvant compositions
US8518694B2 (en) 2002-06-13 2013-08-27 Novartis Vaccines And Diagnostics, Inc. Nucleic acid vector comprising a promoter and a sequence encoding a polypeptide from the endogenous retrovirus PCAV
GB0220194D0 (en) 2002-08-30 2002-10-09 Chiron Spa Improved vesicles
DE60335477D1 (en) 2002-10-11 2011-02-03 Novartis Vaccines & Diagnostic POLYPEPTIDE FOR WIDE PROTECTION AGAINST HYPERVIRULENT MENINGOKOKKIN LINES
AU2003288660A1 (en) 2002-11-15 2004-06-15 Chiron Srl Unexpected surface proteins in neisseria meningitidis
GB0227346D0 (en) 2002-11-22 2002-12-31 Chiron Spa 741
EP2172213B1 (en) 2003-01-30 2013-04-03 Novartis AG Injectable vaccines against multiple meningococcal serogroups
CA2520124A1 (en) 2003-03-28 2004-10-14 Chiron Corporation Use of benzazole compounds for immunopotentiation
GB0308198D0 (en) 2003-04-09 2003-05-14 Chiron Srl ADP-ribosylating bacterial toxin
CN1798548B (en) 2003-06-02 2010-05-05 诺华疫苗和诊断公司 Immunogenic compositions based on microparticles comprising adsorbed toxoid and a polysaccharide-containing antigen
US20060035242A1 (en) 2004-08-13 2006-02-16 Michelitsch Melissa D Prion-specific peptide reagents
GB0323103D0 (en) 2003-10-02 2003-11-05 Chiron Srl De-acetylated saccharides
PL1961426T3 (en) 2003-10-02 2012-03-30 Gsk Vaccines S R L Combined meningitis vaccines
GB0323965D0 (en) * 2003-10-13 2003-11-19 Glaxosmithkline Biolog Sa Immunogenic compositions
GB0329146D0 (en) * 2003-12-16 2004-01-21 Glaxosmithkline Biolog Sa Vaccine
US20080254065A1 (en) 2004-03-09 2008-10-16 Chiron Corporation Influenza Virus Vaccines
GB0409745D0 (en) 2004-04-30 2004-06-09 Chiron Srl Compositions including unconjugated carrier proteins
GB0500787D0 (en) 2005-01-14 2005-02-23 Chiron Srl Integration of meningococcal conjugate vaccination
SI1740217T1 (en) 2004-04-30 2011-10-28 Novartis Ag Meningococcal conjugate vaccination
GB0410866D0 (en) 2004-05-14 2004-06-16 Chiron Srl Haemophilius influenzae
WO2006078294A2 (en) 2004-05-21 2006-07-27 Novartis Vaccines And Diagnostics Inc. Alphavirus vectors for respiratory pathogen vaccines
EP2277595A3 (en) 2004-06-24 2011-09-28 Novartis Vaccines and Diagnostics, Inc. Compounds for immunopotentiation
CA2571710A1 (en) 2004-06-24 2006-11-02 Nicholas Valiante Small molecule immunopotentiators and assays for their detection
CA2575548A1 (en) 2004-07-29 2006-07-27 John L. Telford Immunogenic compositions for gram positive bacteria such as streptococcus agalactiae
GB0424092D0 (en) 2004-10-29 2004-12-01 Chiron Srl Immunogenic bacterial vesicles with outer membrane proteins
JP4993750B2 (en) 2005-01-27 2012-08-08 チルドレンズ ホスピタル アンド リサーチ センター アット オークランド Vesicular vaccine based on GNA1870 for broad protection against diseases caused by Neisseria meningitidis
GB0502095D0 (en) 2005-02-01 2005-03-09 Chiron Srl Conjugation of streptococcal capsular saccharides
SI2351772T1 (en) 2005-02-18 2016-11-30 Glaxosmithkline Biologicals Sa Proteins and nucleic acids from meningitis/sepsis-associated Escherichia coli
ES2385045T3 (en) 2005-02-18 2012-07-17 Novartis Vaccines And Diagnostics, Inc. Urotogenic Escherichia coli immunogens
JP2006296511A (en) * 2005-04-15 2006-11-02 Transcutaneous Technologies Inc External preparation, method for applying external preparation, iontophoresis device, and transdermal patch
WO2006116423A2 (en) 2005-04-26 2006-11-02 Eisai Co., Ltd Compositions and methods for cancer immunotherapy
EP1928539A1 (en) * 2005-09-30 2008-06-11 Tti Ellebeau, Inc. Functionalized microneedles transdermal drug delivery systems, devices, and methods
KR20080080087A (en) * 2005-09-30 2008-09-02 티티아이 엘뷰 가부시키가이샤 Transdermal drug delivery systems, devices, and methods employing novel pharmaceutical vehicles
WO2007041323A1 (en) * 2005-09-30 2007-04-12 Tti Ellebeau, Inc. Iontophoretic delivery of vesicle-encapsulated active agents
US20070078375A1 (en) * 2005-09-30 2007-04-05 Transcutaneous Technologies Inc. Iontophoretic delivery of active agents conjugated to nanoparticles
US20070135754A1 (en) * 2005-09-30 2007-06-14 Hidero Akiyama Electrode assembly for iontophoresis for administering active agent enclosed in nanoparticle and iontophoresis device using the same
KR20080066712A (en) * 2005-09-30 2008-07-16 티티아이 엘뷰 가부시키가이샤 Functionalized microneedles transdermal drug delivery systems, devices, and methods
EP2357000A1 (en) 2005-10-18 2011-08-17 Novartis Vaccines and Diagnostics, Inc. Mucosal and systemic immunizations with alphavirus replicon particles
US11707520B2 (en) 2005-11-03 2023-07-25 Seqirus UK Limited Adjuvanted vaccines with non-virion antigens prepared from influenza viruses grown in cell culture
CA2628152C (en) 2005-11-04 2016-02-02 Novartis Vaccines And Diagnostics S.R.L. Adjuvanted vaccines with non-virion antigens prepared from influenza viruses grown in cell culture
ES2514316T3 (en) 2005-11-22 2014-10-28 Novartis Vaccines And Diagnostics, Inc. Norovirus and Sapovirus virus-like particles (VLPs)
GB0524066D0 (en) 2005-11-25 2006-01-04 Chiron Srl 741 ii
TWI457133B (en) 2005-12-13 2014-10-21 Glaxosmithkline Biolog Sa Novel composition
US20080033398A1 (en) * 2005-12-29 2008-02-07 Transcutaneous Technologies Inc. Device and method for enhancing immune response by electrical stimulation
US7848801B2 (en) * 2005-12-30 2010-12-07 Tti Ellebeau, Inc. Iontophoretic systems, devices, and methods of delivery of active agents to biological interface
EP2357184B1 (en) 2006-03-23 2015-02-25 Novartis AG Imidazoquinoxaline compounds as immunomodulators
US20100015168A1 (en) 2006-06-09 2010-01-21 Novartis Ag Immunogenic compositions for streptococcus agalactiae
AU2007285484B2 (en) 2006-08-16 2013-05-02 Novartis Ag Immunogens from uropathogenic Escherichia coli
US20080193514A1 (en) * 2006-11-02 2008-08-14 Transcu Ltd. Compostions and methods for iontophoresis delivery of active ingredients through hair follicles
AU2007322424B2 (en) * 2006-11-20 2013-05-16 Duecom Use of lipid containing particles comprising quillaja saponins for the treatment of cancer
EP2123259A1 (en) * 2007-01-16 2009-11-25 Hokkaido University Liposome preparation for iontophoresis having antioxidant component encapsulated therein
RU2482841C2 (en) * 2007-05-18 2013-05-27 ТиТиАй Эллебо, Инк. Devices for transdermal delivery, providing improved release of active substance through biological surface
JP2010187707A (en) * 2007-06-12 2010-09-02 Hokkaido Univ Liposome preparation for iontophoresis comprising insulin encapsulated therein
GB0713880D0 (en) 2007-07-17 2007-08-29 Novartis Ag Conjugate purification
GB0714963D0 (en) 2007-08-01 2007-09-12 Novartis Ag Compositions comprising antigens
BRPI0816689B1 (en) 2007-09-12 2021-08-24 Novartis Ag VACCINE COMPOSITION, KIT AND METHOD FOR MAKING A VACCINE COMPOSITION FOR THE PREVENTION OR TREATMENT OF INFECTION BY STREPTOCOCCUS PYOGENES
US8815253B2 (en) 2007-12-07 2014-08-26 Novartis Ag Compositions for inducing immune responses
GB0818453D0 (en) 2008-10-08 2008-11-12 Novartis Ag Fermentation processes for cultivating streptococci and purification processes for obtaining cps therefrom
CA2709927C (en) 2007-12-21 2017-05-16 Novartis Ag Mutant forms of streptolysin o
AU2009215364B2 (en) 2008-02-21 2014-09-18 Glaxosmithkline Biologicals S.A. Meningococcal fHBP polypeptides
ES2557282T3 (en) 2008-03-10 2016-01-25 Children's Hospital & Research Center At Oakland Chimeric H-factor binding proteins (fHBP) containing a heterologous B domain, and methods of use
AU2009291764A1 (en) * 2008-09-10 2010-03-18 Transcu Ltd. Apparatus and method to dispense HPC-based viscous liquids into porous substrates, e.g., continuous web-based process
KR101644221B1 (en) 2008-12-09 2016-07-29 화이자 백신스 엘엘씨 IgE CH3 PEPTIDE VACCINE
EP2385842A1 (en) 2009-01-12 2011-11-16 Novartis AG Cna_b domain antigens in vaccines against gram positive bacteria
PL2396032T3 (en) 2009-02-10 2017-05-31 Seqirus UK Limited Influenza vaccines with reduced amounts of squalene
CN102438650A (en) 2009-03-06 2012-05-02 诺华有限公司 Chlamydia antigens
CN109248313B (en) 2009-04-14 2023-01-17 葛兰素史密丝克莱恩生物有限公司 Compositions for immunization against staphylococcus aureus
WO2010125480A1 (en) 2009-04-30 2010-11-04 Coley Pharmaceutical Group, Inc. Pneumococcal vaccine and uses thereof
WO2010146414A1 (en) 2009-06-15 2010-12-23 National University Of Singapore Influenza vaccine, composition, and methods of use
ES2662716T3 (en) 2009-07-07 2018-04-09 Glaxosmithkline Biologicals Sa Preserved immunogens of Escherichia coli
MX2012000734A (en) 2009-07-16 2012-01-27 Novartis Ag Detoxified escherichia coli immunogens.
RU2518291C2 (en) 2009-07-30 2014-06-10 Пфайзер Вэксинс ЭлЭлСи Antigen tau-peptides and their application
AU2010288240B2 (en) 2009-08-27 2014-03-27 Novartis Ag Hybrid polypeptides including meningococcal fHBP sequences
MY163512A (en) 2009-09-03 2017-09-15 Pfizer Vaccines Llc Pcsk9 vaccine
EP2475385A1 (en) 2009-09-10 2012-07-18 Novartis AG Combination vaccines against respiratory tract diseases
GB0917003D0 (en) 2009-09-28 2009-11-11 Novartis Vaccines Inst For Global Health Srl Purification of bacterial vesicles
GB0917002D0 (en) 2009-09-28 2009-11-11 Novartis Vaccines Inst For Global Health Srl Improved shigella blebs
BR112012009014B8 (en) 2009-09-30 2022-10-04 Novartis Ag PROCESS FOR PREPARING S. AUREUS CAPSULAR POLYSACCHARIDE CONJUGATE TYPE 5 OR TYPE 8 AND CRM197 TRANSPORT MOLECULE, CONJUGATE AND IMMUNOGENIC COMPOSITION
CN102724988B (en) 2009-09-30 2014-09-10 诺华股份有限公司 Expression of meningococcal fHBP polypeptides
GB0918392D0 (en) 2009-10-20 2009-12-02 Novartis Ag Diagnostic and therapeutic methods
JP5960055B2 (en) 2009-10-27 2016-08-02 ノバルティス アーゲー Modified meningococcal fHBP polypeptide
GB0919690D0 (en) 2009-11-10 2009-12-23 Guy S And St Thomas S Nhs Foun compositions for immunising against staphylococcus aureus
RU2581020C2 (en) 2009-12-22 2016-04-10 Селлдекс Терапьютикс, Инк. Vaccine compositions
JP5781542B2 (en) 2009-12-30 2015-09-24 ノバルティス アーゲー E. polysaccharide immunogen conjugated to an E. coli carrier protein
GB201003333D0 (en) 2010-02-26 2010-04-14 Novartis Ag Immunogenic proteins and compositions
WO2011126863A1 (en) 2010-03-30 2011-10-13 Children's Hospital & Research Center Oakland Factor h binding proteins (fhbp) with altered properties and methods of use thereof
GB201005625D0 (en) 2010-04-01 2010-05-19 Novartis Ag Immunogenic proteins and compositions
WO2011127316A1 (en) 2010-04-07 2011-10-13 Novartis Ag Method for generating a parvovirus b19 virus-like particle
KR20130121699A (en) 2010-05-28 2013-11-06 테트리스 온라인, 인코포레이티드 Interactive hybrid asynchronous computer game infrastructure
CA2798837A1 (en) 2010-06-07 2011-12-15 Pfizer Inc. Her-2 peptides and vaccines
WO2011154878A1 (en) 2010-06-07 2011-12-15 Pfizer Vaccines Llc Ige ch3 peptide vaccine
GB201009861D0 (en) 2010-06-11 2010-07-21 Novartis Ag OMV vaccines
US9192661B2 (en) 2010-07-06 2015-11-24 Novartis Ag Delivery of self-replicating RNA using biodegradable polymer particles
WO2012006293A1 (en) 2010-07-06 2012-01-12 Novartis Ag Norovirus derived immunogenic compositions and methods
GB201101665D0 (en) 2011-01-31 2011-03-16 Novartis Ag Immunogenic compositions
GB201017519D0 (en) 2010-10-15 2010-12-01 Novartis Vaccines Inst For Global Health S R L Vaccines
WO2012072769A1 (en) 2010-12-01 2012-06-07 Novartis Ag Pneumococcal rrgb epitopes and clade combinations
NZ611176A (en) 2010-12-02 2015-07-31 Bionor Immuno As Peptide scaffold design
US20130315959A1 (en) 2010-12-24 2013-11-28 Novartis Ag Compounds
EP3338798A1 (en) 2011-01-06 2018-06-27 Bionor Immuno AS Multimeric peptide
US20140004142A1 (en) 2011-03-02 2014-01-02 Pfizer Inc. Pcsk9 vaccine
EP2736921B1 (en) 2011-07-25 2018-06-27 GlaxoSmithKline Biologicals SA Compositions and methods for assessing functional immunogenicity of parvovirus vaccines
GB201114923D0 (en) 2011-08-30 2011-10-12 Novartis Ag Immunogenic proteins and compositions
EP2755994A2 (en) 2011-09-14 2014-07-23 Novartis AG Escherichia coli vaccine combination
WO2013038375A2 (en) 2011-09-14 2013-03-21 Novartis Ag Methods for making saccharide-protein glycoconjugates
ES2704069T3 (en) 2011-12-08 2019-03-14 Glaxosmithkline Biologicals Sa Clostridium difficile toxin-based vaccine
WO2013108272A2 (en) 2012-01-20 2013-07-25 International Centre For Genetic Engineering And Biotechnology Blood stage malaria vaccine
CN104321335A (en) 2012-02-24 2015-01-28 诺华股份有限公司 Pilus proteins and compositions
US10279026B2 (en) 2012-04-26 2019-05-07 Glaxosmithkline Biologicals Sa Antigens and antigen combinations
RU2727476C2 (en) 2012-04-26 2020-07-21 Новартис Аг Antigens and antigen compositions
EP2659906A1 (en) 2012-05-01 2013-11-06 Affiris AG Compositions
EP2659907A1 (en) 2012-05-01 2013-11-06 Affiris AG Compositions
EP2659908A1 (en) 2012-05-01 2013-11-06 Affiris AG Compositions
PE20142406A1 (en) 2012-05-04 2015-01-23 Pfizer ANTIGENS ASSOCIATED WITH PROSTATE AND VACCINE-BASED IMMUNOTHERAPY REGIMES
US10124051B2 (en) 2012-05-22 2018-11-13 Glaxosmithkline Biologicals Sa Meningococcus serogroup X conjugate
IN2014KN02769A (en) 2012-06-06 2015-05-08 Bionor Immuno As
RU2662970C2 (en) 2012-09-18 2018-07-31 Новартис Аг Outer membrane vesicles
EP2903650B1 (en) 2012-10-02 2018-04-04 GlaxoSmithKline Biologicals SA Nonlinear saccharide conjugates
ES2848048T3 (en) 2012-10-03 2021-08-05 Glaxosmithkline Biologicals Sa Immunogenic compositions
US20210145963A9 (en) 2013-05-15 2021-05-20 The Governors Of The University Of Alberta E1e2 hcv vaccines and methods of use
IL296681B2 (en) 2014-01-21 2024-09-01 Pfizer Immunogenic compositions comprising conjugated capsular saccharide antigens and uses thereof
US11160855B2 (en) 2014-01-21 2021-11-02 Pfizer Inc. Immunogenic compositions comprising conjugated capsular saccharide antigens and uses thereof
WO2015123291A1 (en) 2014-02-11 2015-08-20 The Usa, As Represented By The Secretary, Dept. Of Health And Human Services Pcsk9 vaccine and methods of using the same
CA3212723A1 (en) 2014-07-23 2016-01-28 Peter T. Beernink Factor h binding protein variants and methods of use thereof
CN107427568B (en) 2015-01-15 2021-12-14 辉瑞公司 Immunogenic compositions for use in pneumococcal vaccines
AU2016271857B2 (en) 2015-06-03 2020-05-28 Affiris Ag IL-23-P19 vaccines
CN107849119A (en) 2015-07-07 2018-03-27 阿费里斯股份公司 For the vaccine for the disease for treating and preventing IgE mediations
PE20240927A1 (en) 2015-07-21 2024-04-30 Pfizer IMMUNOGENIC COMPOSITIONS COMPRISING CONJUGATED CAPSULAR SACCHARIDE ANTIGENS, KITS THAT COMPRISE THEM AND THEIR USES
WO2017085586A1 (en) 2015-11-20 2017-05-26 Pfizer Inc. Immunogenic compositions for use in pneumococcal vaccines
RU2718663C2 (en) 2016-01-19 2020-04-13 Пфайзер Инк. Anticancer vaccines
PL3570879T3 (en) 2017-01-20 2022-06-20 Pfizer Inc. Immunogenic compositions for use in pneumococcal vaccines
US11633471B2 (en) 2018-03-06 2023-04-25 Unm Rainforest Innovations Compositions and methods for reducing serum triglycerides
JP2021534761A (en) 2018-08-23 2021-12-16 グラクソスミスクライン バイオロジカルズ ソシエテ アノニム Immunogenic proteins and compositions
US11260119B2 (en) 2018-08-24 2022-03-01 Pfizer Inc. Escherichia coli compositions and methods thereof
CA3120922A1 (en) 2018-12-12 2020-06-18 Pfizer Inc. Immunogenic multiple hetero-antigen polysaccharide-protein conjugates and uses thereof
JP7239509B6 (en) 2019-02-22 2023-03-28 ファイザー・インク Method for purifying bacterial polysaccharides
CA3136278A1 (en) 2019-04-10 2020-10-15 Pfizer Inc. Immunogenic compositions comprising conjugated capsular saccharide antigens, kits comprising the same and uses thereof
WO2021084429A1 (en) 2019-11-01 2021-05-06 Pfizer Inc. Escherichia coli compositions and methods thereof
EP4093873A4 (en) 2020-01-24 2024-07-10 Aim Immunotech Inc Methods, compositions, and vaccines for treating a virus infection
WO2021160887A1 (en) 2020-02-14 2021-08-19 Immunor As Corona virus vaccine
BR112022015796A2 (en) 2020-02-21 2022-10-11 Pfizer PURIFICATION OF SACCHARIDES
WO2021165928A2 (en) 2020-02-23 2021-08-26 Pfizer Inc. Escherichia coli compositions and methods thereof
JP2023514825A (en) 2020-02-26 2023-04-11 ヴェルシテック リミテッド PD-1-based vaccine against coronavirus infection
AU2021368151B2 (en) 2020-10-27 2024-09-05 Pfizer Inc. Escherichia coli compositions and methods thereof
US20240000912A1 (en) 2020-11-04 2024-01-04 Pfizer Inc. Immunogenic compositions for use in pneumococcal vaccines
JP2023549736A (en) 2020-11-10 2023-11-29 ファイザー・インク Immunogenic compositions comprising conjugated capsular saccharide antigens and uses thereof
US20220202923A1 (en) 2020-12-23 2022-06-30 Pfizer Inc. E. coli fimh mutants and uses thereof
WO2022147373A1 (en) 2020-12-31 2022-07-07 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Antibody-guided pcsk9-mimicking immunogens lacking 9-residue sequence overlap with human proteins
CA3211240A1 (en) 2021-02-19 2022-08-25 Sanofi Pasteur Inc. Meningococcal b recombinant vaccine
EP4346892A2 (en) 2021-05-28 2024-04-10 Pfizer Inc. Immunogenic compositions comprising conjugated capsular saccharide antigens and uses thereof
WO2022249107A2 (en) 2021-05-28 2022-12-01 Pfizer Inc. Immunogenic compositions comprising conjugated capsular saccharide antigens and uses thereof
AU2022391752A1 (en) 2021-11-18 2024-06-13 Matrivax, Inc. Immunogenic fusion protein compositions and methods of use thereof
IL314243A (en) 2022-01-13 2024-09-01 Pfizer Immunogenic compositions comprising conjugated capsular saccharide antigens and uses thereof
WO2023161817A1 (en) 2022-02-25 2023-08-31 Pfizer Inc. Methods for incorporating azido groups in bacterial capsular polysaccharides
WO2023218322A1 (en) 2022-05-11 2023-11-16 Pfizer Inc. Process for producing of vaccine formulations with preservatives
WO2024110827A1 (en) 2022-11-21 2024-05-30 Pfizer Inc. Methods for preparing conjugated capsular saccharide antigens and uses thereof
US20240181028A1 (en) 2022-11-22 2024-06-06 Pfizer Inc. Immunogenic compositions comprising conjugated capsular saccharide antigens and uses thereof
WO2024116096A1 (en) 2022-12-01 2024-06-06 Pfizer Inc. Pneumococcal conjugate vaccine formulations
WO2024166008A1 (en) 2023-02-10 2024-08-15 Pfizer Inc. Immunogenic compositions comprising conjugated capsular saccharide antigens and uses thereof

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP3755890B2 (en) * 1992-06-25 2006-03-15 スミスクライン・ビーチャム・バイオロジカルス(ソシエテ・アノニム) Adjuvant-containing vaccine composition
GB9326253D0 (en) * 1993-12-23 1994-02-23 Smithkline Beecham Biolog Vaccines
WO1996010423A1 (en) * 1994-09-30 1996-04-11 Ludwig Institute For Cancer Research COMPOSITIONS CONTAINING A p53 DERIVED PROTEIN OR PEPTIDE, AN ADJUVANT, AND INTERLEUKIN-12 AND USES THEREOF
MX9702396A (en) * 1994-10-05 1997-12-31 Univ Vanderbilt Interleukin-12 as an adjuvant for paramyxoviridae vaccines.
GB9513261D0 (en) * 1995-06-29 1995-09-06 Smithkline Beecham Biolog Vaccines

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100012927A1 (en) * 2001-07-25 2010-01-21 Nantero, Inc. Devices having vertically-disposed nanofabric articles and methods of making the same

Also Published As

Publication number Publication date
WO1998057660A1 (en) 1998-12-23
CA2293614A1 (en) 1998-12-23
US6375945B1 (en) 2002-04-23
JP2002504135A (en) 2002-02-05
WO1998057659A1 (en) 1998-12-23
CA2294051A1 (en) 1998-12-23
EP0977589A1 (en) 2000-02-09
EP1001808A1 (en) 2000-05-24
GB9712347D0 (en) 1997-08-13
JP2002504136A (en) 2002-02-05

Similar Documents

Publication Publication Date Title
US6375945B1 (en) Adjuvant compositions for vaccines
JP4620251B2 (en) Composition of CPG and saponin adjuvant and method thereof
JP3755890B2 (en) Adjuvant-containing vaccine composition
EP0855184A1 (en) Pharmaceutical composition comprising a polynucleotide and an antigen especially for vaccination
US20040247662A1 (en) Systemic immune activation method using nucleic acid-lipid complexes
EP1146858A1 (en) Non-invasive vaccination through the skin
KR20060131749A (en) Immunogenic compositions
KR100850473B1 (en) Pharmaceutical compositions enhancing the immunogenicity of poorly immunogenic antigens
EP1159967B1 (en) Tumor vaccines
KR20060124669A (en) Immunostimulant composition comprising at least one toll-like receptor 7 or toll-like receptor 8 agonist and a toll-like receptor 4 agonist
LT5435B (en) Vaccine composition admixed with an alkylphosphatidylcholine
JP2019515005A (en) Aluminum hydroxide gel-sodium chloride complex immunological adjuvant, method for preparing it and use thereof
EP0422164B1 (en) Large multivalent immunogen
EP1377275B1 (en) Vaccine for modulating between t1 and t2 immune responses
JP2002526420A (en) Substantially non-toxic, biologically active mucosal adjuvants in vertebrates
US20040022813A1 (en) Shed antigen vaccine with dendritic cells adjuvant
US20040191214A1 (en) Nucleoside vaccine adjuvants
US20040081686A1 (en) Use of particle vectors in immunomodulation
US20090081156A1 (en) Bioactive molecular matrix and methods of use in the treatment of disease
Donnelly New developments in adjuvants
WO2000013704A1 (en) Formulation of nucleic acids and acemannan
EP1716866A1 (en) Proteoliposomes and derivatives thereof as cytotoxic response-inducing adjuvants and resulting formulations
Canbolat 14 Vaccine Adjuvants in Immunotoxicology
US20070259006A1 (en) Shed antigen vaccine with dendritic cells adjuvant
Egilmez Cytokines as vaccine adjuvants

Legal Events

Date Code Title Description
AS Assignment

Owner name: SMITHKLINE BEECHAM BIOLOGICALS S.A., BELGIUM

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:BOON, THIERRY;SILLA, SILVIA;UYTTENHOVE, CATHERINE;REEL/FRAME:010594/0086

Effective date: 19991223

Owner name: LUDWIG INSTITUTE FOR CANCER RESEARCH, NEW YORK

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:BOON, THIERRY;SILLA, SILVIA;UYTTENHOVE, CATHERINE;REEL/FRAME:010594/0086

Effective date: 19991223

STCB Information on status: application discontinuation

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