WO2004064715A2 - Polyhydroxylated pyrrolizidine - Google Patents

Polyhydroxylated pyrrolizidine Download PDF

Info

Publication number
WO2004064715A2
WO2004064715A2 PCT/GB2004/000198 GB2004000198W WO2004064715A2 WO 2004064715 A2 WO2004064715 A2 WO 2004064715A2 GB 2004000198 W GB2004000198 W GB 2004000198W WO 2004064715 A2 WO2004064715 A2 WO 2004064715A2
Authority
WO
WIPO (PCT)
Prior art keywords
compound
cells
casuarine
cell
treatment
Prior art date
Application number
PCT/GB2004/000198
Other languages
French (fr)
Other versions
WO2004064715A3 (en
Inventor
Alison Ann Watson
Robert James Nash
Emma Louisa Evinson
Original Assignee
M N L Pharma Limited
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 M N L Pharma Limited filed Critical M N L Pharma Limited
Priority to AU2004206085A priority Critical patent/AU2004206085B2/en
Priority to EP04703841A priority patent/EP1587480B1/en
Priority to JP2006500223A priority patent/JP5289706B2/en
Priority to NZ541839A priority patent/NZ541839A/en
Priority to ES04703841T priority patent/ES2394072T3/en
Priority to CA2513881A priority patent/CA2513881C/en
Priority to US10/543,014 priority patent/US8383665B2/en
Publication of WO2004064715A2 publication Critical patent/WO2004064715A2/en
Publication of WO2004064715A3 publication Critical patent/WO2004064715A3/en
Priority to HK06104746.3A priority patent/HK1084576A1/en
Priority to US13/742,102 priority patent/US8557859B2/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/26Carbohydrates, e.g. sugar alcohols, amino sugars, nucleic acids, mono-, di- or oligo-saccharides; Derivatives thereof, e.g. polysorbates, sorbitan fatty acid esters or glycyrrhizin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • A61K31/407Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil condensed with other heterocyclic ring systems, e.g. ketorolac, physostigmine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7028Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/14Blood; Artificial blood
    • A61K35/15Cells of the myeloid line, e.g. granulocytes, basophils, eosinophils, neutrophils, leucocytes, monocytes, macrophages or mast cells; Myeloid precursor cells; Antigen-presenting cells, e.g. dendritic cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K36/00Medicinal preparations of undetermined constitution containing material from algae, lichens, fungi or plants, or derivatives thereof, e.g. traditional herbal medicines
    • A61K36/18Magnoliophyta (angiosperms)
    • A61K36/185Magnoliopsida (dicotyledons)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K36/00Medicinal preparations of undetermined constitution containing material from algae, lichens, fungi or plants, or derivatives thereof, e.g. traditional herbal medicines
    • A61K36/18Magnoliophyta (angiosperms)
    • A61K36/185Magnoliopsida (dicotyledons)
    • A61K36/61Myrtaceae (Myrtle family), e.g. teatree or eucalyptus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • 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
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P33/00Antiparasitic agents
    • 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
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H3/00Compounds containing only hydrogen atoms and saccharide radicals having only carbon, hydrogen, and oxygen atoms
    • C07H3/02Monosaccharides
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • the present invention relates to immunomodulatory polyhydroxylated pyrrolizidine compounds and to their use in medicine
  • the invention relates to the use of casua ⁇ ne and certain casua ⁇ ne analogues as immunomodulatory (immunostimulatory or immunosuppressive) drugs
  • the innate immune system serves as the first line of defence against invading pathogens, holding the pathogen in check while the adaptive responses are matured It is triggered within minutes of infection in an antigen- independent fashion, responding to broadly conserved patterns in the pathogens (though it is not non-specific, and can distinguish between self and pathogens) Crucially, it also generates the inflammatory and co- stimulatory milieu (sometimes referred to as the danger signal) that potentiates the adaptive immune system and steers (or polarizes it) towards the cellular or humoral responses most appropriate for combating the infectious agent (discussed in more detail below)
  • the adaptive response becomes effective over days or weeks, but ultimately provides the fine antigenic specificity required for complete elimination of the pathogen and the generation of immunologic memory It is mediated principally by T and B cells that have undergone germline gene rearrangement and are characterized by anaki specificity and long-lasting memory However, it also involves the recruitment of elements of the innate immune system, including professional phagocytes (macrophages, neutrophils etc ) and granulocytes (basophils, eosinophils etc ) that engulf bacteria and even relatively large protozoal parasites
  • the activated DCs then migrate to lymph no ⁇ es ⁇ nce mere, tney activate immune cent, of the adaptive response (principally naive B- and T-cells) by acting as antigen-presenting cells (APCs)
  • the activated cells then migrate to the sites of infection (guided by the "danger signal") and once there further amplify the response by recruiting cells of the innate immune system (including eosinophils, basophils, monocytes, NK cells and granulocytes)
  • This cellular trafficking is orchestrated by a large array of cytokines (particularly those of the chemokme subgroup) and involves immune cells of many different types and tissue sources (for a review, see Luster (2002), Current Opinion in Immunology 14 129-135)
  • the adaptive immune response is principally effected via two independent limbs cell-mediated (type 1) immunity and antibody-mediated or humoral (type 2) immunity
  • Type 1 immunity involves the activation of T-lymphocytes that either act upon infected cells bearing foreign antigens or stimulate other cells to act upon infected cells This bi anch of the immune system therefore effectively contains and kills cells that are cancerous or infected with pathogens (particularly viruses)
  • Type 2 immunity involves the generation of antibodies to foreign antigens by B-lymphocytes This antibody-mediated branch of the immune system attacks and effectively neutralizes extracellular foreign antigens
  • the type of immune response is just as important as its intensity or its duration
  • the balance of the typel/type 2 response also referred to as the Th1 Th2 response ratio/balance by reference to the distinct cytokine and effector cell subsets involved in the regulation of each response - see below
  • the balance of the typel/type 2 response may also play a role in determining the effectiveness (and repercussions) of the immune defence
  • the immune response is skewed heavily towa r ds a type 1 or type 2 response soon after exposure to antigen
  • the mechanism of this type1/type 2 skewing or polarization is not yet fully understood, but is known to involve a complex system of cell-mediated chemical messengers (cytokines, and particularly chemokines) in which the typel/type 2 polarization (or balance) is determined, at least in part, by the nature of the initial PRR-PAMP interaction when the DCs and macrophages of the innate immune system are first stimulated and subsequently by the cytokine milieu in which antigen priming of naive helper T cells occuis
  • cytokines in particular appear to have early roles in determining the path of the immune response lnterleuk ⁇ n-12 (IL-12), secreted by macrophages, drives the type 1 response by stimulating the differentiation of Th1 cells, the helper cells that oversee the type 1 response Another macrophage cytokine, ⁇ nterleuk ⁇ n- 0 (IL- 10) inhibits this response, instead driving a type 2 response
  • IL-12 lnterleuk ⁇ n-12
  • IL-12 macrophage cytokine
  • ⁇ nterleuk ⁇ n- 0 IL-10 inhibits this response, instead driving a type 2 response
  • the type 1 and type 2 responses can be distinguished inter alia on the basis of certain phenotypic changes attendant on priming and subsequent polarization of naive helper T cells These phenotypic changes are characterized, at least in part, by the nature of the cytokines secreted by the polarized helper T cells
  • Th1 cells produce so-called Th1 cytokines, which include one or more of TNF, IL-1 , IL-2, IFN-gamma, IL-12 and/or IL-18
  • the Th1 cytokines are involved in macrophage activation and Th1 cells orchestrate Type 1 responses
  • Th2 cells produce so-called Th2 cytokines, which include one or more of IL-4, I -5, IL- 10 and IL-13
  • the Th2 cytokines promote the production of various antibodies and can suppress the type 1 response
  • Th1 and Th2 cells and cytokines are used interchangeably herein
  • Th1 Th2 ratio is symptomatic of many immunological diseases and disorders, and the development of methods for altering the Th1 Th2 ratio is now a priority
  • alkaloid is used herein sensu stricto to define any basic, organic, nitrogenous compound which occurs naturally in an organism
  • alkaloid is also used herein sensu lato to define a broader grouping of compounds which include not only the naturally occurring alkaloids, but also their synthetic and semi-synthetic analogues and derivatives
  • alkaloids are phytochemicals, present as secondary metabolites in plant tissues (where they may play a role in defence), but some occur as secondary metabolites in the tissues of animals, microorganisms and fungi
  • the standard techniques for screening microbial cultures are inappropriate for detecting many classes of alkaloids (particularly highly polar alkaloids, see below) and that microbes (including bacteria and fungi, particularly the f'lamentous representatives) will prove to be an important source of alkaloids as screening techniques become more sophisticated
  • alkaloids are small molecules, with molecular weights below 250 Daltons
  • the skeletons may be derived from ammo acids, though some are derived from olher groups (such as steroids) Olhers can be consideied as sugar analogues It is becoming apparent (see Watson et al (2001 ) Phytochemistry 56 265-295) that the water soluble fractions of medicinal plants and microbial cultures contain many interesting novel polar alkaloids, including many carbohydrate analogues Such analogues include a rapidly growing number of polyhydroxylated alkaloids
  • alkaloids are classified structurally on the basis of the configuration of the N-heterocycle Examples of some important alkaloids and their structures are set out in Kutchan (1995) The Plant Cell 7 1059-1070 Watson et al (2001 ) Phytochemistry 56 265-295 have classified a comprehensive range of polyhydroxylated alkaloids inter alia as pipe ⁇ dine, pyrroline, pyrrolidme pyrrolizidine, indohzidine and nortropanes alkaloids (see Figs 1-7 of Watson et al (2001 ), the disclosure of which is incorporated herein by reference)
  • alkaloids are pharmacologically active, and humans have been using alkaloids (typically in the form of plant extracts) as poisons, narcotics stimulants and medicines for thousands of years
  • alkaloids typically in the form of plant extracts
  • narcotics stimulants and medicines for thousands of years
  • the therapeutic applications of polyhydroxylated alkaloids have been comprehensively reviewed in Watson et al (2001 ), ibidem applications include cancer therapy immune stimulation, the treatment of diabetes, the ti eatment of infections (especially viral infections) therapy of glycosphmgolipid lysosomal storage diseases and the treatment of autoimmune disorders (such as arthi itis and sclerosis)
  • the alexmes occur in all species of the genus Alexa and also in the related species Castanospermum australe Stereoiso ers of alexine, including 1 ,7a-d ⁇ ep ⁇ alex ⁇ ne (3) have also been isolated (Nash et al (1990) Phytochemistry (29) 111 ) and synthesised (Choi et al. (1991 ) Tetrahedron Letters (32) 5517 and Denmark and Cottell (2001 ) J. Org. Chem. (66) 4276-4284).
  • swainsonine (4) is a potent and specific inhibitor of ⁇ -mannosidase and is reported to have potential as an antimetastic, tumour anti- proliferative and immu ⁇ oregulatory agent (see e.g. US5650413, WO00/37465, WO93/09117).
  • Another compound, 1 ⁇ ,2 ⁇ ,6 ⁇ ,7 ⁇ ,7 ⁇ -1 ,2,6,7-tetrahydroxypyrrol ⁇ z ⁇ d ⁇ ne (7) is an analogue of 1 , diepiswainsonine and described as a ' useful" inhibitor of glycosidase enzymes in EP0417059
  • Casuarine, (I R ⁇ R.SR. ⁇ S S aRJ-S ⁇ hydroxymethy -l ⁇ .e ⁇ -tetrahydroxypyrrolizidine (8) is a highly oxygenated bicyc c pyrrolizidine alkaloid that can be regarded as a more highly oxygenated analogue of the 1 ,7a-d ⁇ ep ⁇ alex ⁇ ne (shown in 3) or as a C(3) hydroxymethyl-substituted analogue of the 1 ⁇ ,2 ⁇ ,6 ⁇ ,7 ⁇ J ⁇ -1 , 2,6,7- tetrahydroxypyn o zidine (shown in 7)
  • Casuarine can be isolated from sevei al botanical sources, including the bark of Casuanna equi ⁇ etifolia (Casuannaceae), the leaves and bark of Eugenia jambolana (Myrtaceae) and Syzygium guineense (Myrtaceae) (see e g Nash et al (1994) Tetrahedron Letters (35) 7849-7852) Epimers of casuarine, and probably casuarine itself, can be synthesised by sodium hydrogen telluride-induced cyclisation of azidodimesylates (Bell er a/ (1997) Tetrahedron Letters (38) 5869-5872) Casuanna equisetifolia wood, bark and leaves have been claimed to be useful against diarrhoea, dysentery and colic (Chopra ef al (1956) Glossary of Indian Medicinal Plants, Council of Scientific and Industrial Research (India), New Delhi, p 55) and a
  • casuar ⁇ ne-6- ⁇ -glucos ⁇ de casuar ⁇ ne-6- ⁇ -D-glucopyranose 9
  • casuar ⁇ ne-6- ⁇ -D-glucopyranose 9 has also been isolated from the bark and leaves of Eugenia jambolana (Wormald ef al (1996) Carbohydrate Letters (2) 169-174)
  • DCs Dendritic cells
  • Dendritic cells therefoi e act as highly specialized antigen-presenting cells (APCs) serving as "nature's adjuvants' , they potentiate adaptive T-cell dependent immunity as well as triggering the natural killer (NK and NKT) cells of the innate immune system Dendritic cells therefore play a fundamental and important regulatory role in the magnitude, quality and memory of the immune response As a result, there is now a growing interest in the use of dendritic cells in various immunomodulatory interventions, which are described in more detail below Dendritic cells can be classified into different subsets inter alia on the basis of their state of maturation (mature or immature) and their cellular developmental origin (ontogeny) Each of these subsets appear to play distinct roles in vivo, as described below
  • Immature (or resting) DCs are located in non-lymphoid tissue, such as the skin and mucosae, are highly phagocytic and readily internalize soluble and particulate antigens It is only when such antigen-loaded immature DCs are also subject to inflammatory stimuli (referred to as maturation stimuli) that they undergo a maturation process that transforms them from phagocytic and migratory cells into non-phagocytic, highly efficient stimulators of naive T cells
  • Immature DCs are characterized by high intracellular MHC II in the form of MIICs, the expression of CD1a, active endocytosis for certain particulates and proteins, presence of FcgR and active phagocytosis, deficient T cell sensitization in vi o, low/absent adhesive and costimulatory molecules (CD40/54/58/80/86), low/absent CD25, CD83, p55, DEC-205, 2A1 antigen, responsiveness to GM-CSF, but not M-CSF and G-CSF and a sensitivity to IL-10, which inhibits maturation
  • naive CD4 + T cells migrate from the non- lymphoid tissues to the lymph nodes or spleen, where they process the antigen load and present it to the resident naive CD4 + T cells and CD8 + cytotoxic T cells This latter interaction generates CTLs, the cellular arm of the adaptive immune response, and these cells eliminate virally infected cells and tumour cells
  • the naive CD4 + T cells differentiate into memory helper T cells, which support the differentiation and expansion of CD8 + CTLs and B cells
  • helper T cells exert anti-tumour activity indirectly through the activation of important effector cells such as macrophages and CTLs
  • Mature DC cells are characterized morphologically by motility and the presence of numerous processes (veils or dend ⁇ tes) They are competent for antigen capture and presentation (exhibiting high MHC class I and II expression) and express a wide range of molecules involved in T cell binding and costimulation, (e g CD40, CD54/ICAM-1 , CD58/LFA-3, CD80/B7-1 and CD86/B7-2) as well as various cytokines (including IL-12) They are phenotypically stable there is no reversion/conversion to macrophages or lymphocytes
  • mature DCs play an important role in T cell activation and cell-mediated immunity
  • immature DCs are involved in regulating and maintaining immunological tolerance (inducing antigen-specific T cell anergy)
  • Dendritic cells are not represented by a single ceil type, but rather comprise a heterogeneous collection of different classes of cells, each with a distinct ontogeny At least three different developmental pathways have been described, each emerging from unique progenitors and driven by particular cytokine combinations to DC subsets with distinct and specialized functions.
  • the primitive CD34 + DC progenitors are subject to various stimulatory signals. These signals can direct the progenitors along one of at least three different pathways, each differing with respect to intermediate stages, cytokine requirements, surface marker expression and biological function.
  • Lymphoid DCs are a distinct subset of DCs that are closely linked to the lymphocyte lineage. This lineage is characterized by the lack of the surface antigens CD 1b, CD13, CD14 and CD33. Lymphoid DCs share ancestry with T and natural killer (NK) cells, the progenitors for all being located in the thymus and in the T cell areas of secondary lymphoid tissues. The differentiation of lymphoid DCs is driven by interleukins 2, 3 and 15 (IL-3, IL-2 and IL-15), but not by granulocyte macrophage colony- stimulating factor (GM-CSF).
  • IL-3, IL-2 and IL-15 granulocyte macrophage colony- stimulating factor
  • lymphoid promote negative selection in the thymus (possibly by inducing fas-mediated apoptosis) and are costimulatory for CD4 + and CD8 + T cells. More recently, lymphoid-like DCs derived from human progenitors have also been shown to preferentially activate the Th2 response. Because of their capacity to induce apoptosis and their role in eliminating potentially self-reactive T cells, it has been suggested that lymphoid DCs primarily mediate regulatory rather than stimulatory immune effector functions.
  • Myeloid DCs are distinguished by a development stage in which there is expression of certain features associated with phagocytes. There appear to be at least two structurally and functionally distinct subsets. The first is defined antigenically as CD14 " , CD34 + , CD68 " and CD1a + and sometimes referred to as DCs of the Langerhans cell type. This subset appears to prime T cells to preferentially activate Th1 responses and IL-12 appears implicated in this process. The subset may also activate na ⁇ ve B cells to secrete IgM and may therefore be predominantly associated with an inflammatory Th1 . response.
  • a second myeloid DC subset is defined antigenically as CD14 + , CD68 + and CD1 a " and related to monocytes (as a result they are also referred to as monocyte-derived DCs or Mo-DCs).
  • DC cells are taken from a patient (for example by apheresis) and then pulsed (primed or spiked) with a particular antigen or antigens (for example, tumour antigen(s)). They are then re-administered as an autologous cellular vaccine to potentiate an appropriate immune response.
  • a particular antigen or antigens for example, tumour antigen(s)
  • the responding T cells include helper cells, especially Th1 CD4 + cells (which produce IFN- ⁇ ) and killer cells (especially CD8 + cytolytic T lymphocytes).
  • helper cells especially Th1 CD4 + cells (which produce IFN- ⁇ ) and killer cells (especially CD8 + cytolytic T lymphocytes).
  • the DCs may also mediate responses by other classes of lymphocytes (B, NK, and NKT cells) They may also elicit T cell memory, a critical goal of vaccination
  • Mo-DCs These cells are obtained by exposing monocytes to GM-CSF and IL-4 (or IL-13) to produce immature Mo-DCs, which are then matured by incubation in a maturation medium
  • Such media comprise one or more maturation stimulation factor(s), and typically comprise Toll-like receptor (TLR) ligands (e g microbial products such as lipopolysaccha ⁇ de and/or monophosphoryl pid), inflammatory cytokines (such as TNF- ⁇ ), CD40L, monocyte conditioned medium (MCM) or MCM mimic (which contains IL- I ⁇ , TNF- ⁇ , IL-6 and PGE 2 )
  • TLR Toll-like receptor
  • MCM monocyte conditioned medium
  • MCM mimic which contains IL- I ⁇ , TNF- ⁇ , IL-6 and PGE 2
  • MCM or MCM mimic currently represent a standard Mo-DCs matured using these media are homogenous, have a high viability, migrate well to chemotactic stimuli and induce CTLs both in vitro and in vivo
  • Dendritic cells for vaccination have also been prepared from CD34 + -der ⁇ ved DCs comprising a mixture of interstitial and DC s of the Langerhans cell type Some workeis believe that the latter DC subsel are moie potent than Mo-DCs when used as DC vaccines
  • the antigens can be xenoantigens or autoantigens
  • One or moi e defined neoant ⁇ gen(s) may be selected in the case of cancer treatment, the enoant ⁇ gen(s) may comprise a tumour-associated antigen
  • GMP manufactunng practice
  • Other approaches have employed antigens as immune complexes, which are delivered to Fc-receptor-bearing DCs and which results in the formation of both MHC class I and MHC class II peptide sequences. This offers the potential for inducing both CTLs and Th cells (Berlyn ef al. (2001) Clin Immunol 101 : 276-283).
  • Antigens can also be loaded by transfecting the DCs with encoding nucleic acid (e.g. by electroporation) such that the antigens are expressed by the DC, processed and presented at the cell surface.
  • This approach avoids the need for expensive GMP proteins and antibodies.
  • RNA is preferred for this purpose, since it produces only transient expression (albeit sufficient for antigen processing) and avoids the potential problems associated with the integration of DNA and attendant long-term expre ⁇ sion/mutagenesis.
  • Such transfection techniques also permit exploration of the whole antigenic repertoire of a target cell by use of ⁇ fptal or PCR-amplified tumour RNA.
  • helper proteins for example, keyhole limpet hemocyanin (KLH) and tetanus toxoid (TT)
  • KLH keyhole limpet hemocyanin
  • TT tetanus toxoid
  • the dose, frequency and route of DC vaccine administration have not yel been optimised in clinical trials.
  • the absolute number of cells administered will depend on the route of administration and effectiveness of migration after infusion. In this respect there are indications that intradermal or subcutaneous administration may be preferred for the development of Th1 responses, although direct intranodal delivery has been employed to circumvent the need for migration from the skin to the nodes (Nestle et al. (1998) Nat Med 4: 328-332).
  • DCs are targeted to a tumour and activated to elicit immune responses in situ without the need for ex vivo antigen loading.
  • In situ DC vaccination constitutes yet another distinct (but related) approach (Hawiger et al. (2001) J Exp Med 194: 769-779.
  • antigen is targeted to DCs in vivo which are expanded and induced to mature in situ.
  • This approach depends on efficient targeting of antigen to endogenous DCs (for example, using exosomes - see Thery et al. (2002) Nat Rev Immunol 2: 569-579) and the development of maturation stimulants that can effectively trigger maturation (preferably of defined DC subset(s)) in vivo.
  • Cytotoxic T lymphocytes can be administered to a patient in order to confer or supplement an immune response to a particular disease or infection (typically cancer).
  • a particular disease or infection typically cancer
  • tumour specific T cells can be extracted from a patient (e.g. by leukapheresis), selectively expanded (for example by tetramer-guided cloning - see Dunbar et al. (1999) J Immunol 162: 6959-6962) and then re-administered as an autologous cellular vaccine.
  • Dendritic cells are also involved in regulating and maintaining immunological tolerance: in the absence of maturation, the cells induce antigen-specific silencing or tolerance.
  • immature DCs are administered as part of an immunomodulatory intervention designed to combat autoimmune disorders.
  • the suppressive potential of the DCs has been enhanced by in vitro transfection with genes encoding cytokines.
  • Granucci describes the important role played by DC-derived IL-2 in mediating not only T cell activation but also that of NK cells and goes on to suggest that DC-derived IL-2 is a key factor regulating and linking innate and adaptive immunity.
  • casuarine and certain casuarine analogues have unexpected immunomodulatory activity, and that this activity may not be dependent on glycosidase inhibition
  • an isolated immunomodulatory (e g immunostimulatory) polyhydroxylated pyrrolizidine compound for use in therapy or prophylaxis having the formula
  • R is selected from the group comprising hydrogen, straight or branched, unsubstituted or substituted, saturated or unsaturated acyl, alkyl (e g cycloalkyl), alkenyl, alkynyl and aryl groups, or a pharmaceutically acceptable salt or derivative thereof
  • the compounds of the invention are alkaloids (as hereinbefore defined)
  • the compound of the invention preferably has the formula
  • R is selected from the gioup comprising hydrogen, straight or branched, unsubstituted or substituted, saturated or unsaturated acyl, alkyl (e g cycloalkyl), alkenyl, alkynyl and aryl groups, or a pharmaceutically acceptable salt or derivative thereof rv
  • R 2R,3R,6S,7S,7aR -3-(hydroxymethyl)-1 2,6,7-tetrahydroxypyrrol ⁇ z ⁇ d ⁇ ne (casuarine), wherein R is hydrogen and which has the formula
  • a casuarine glucoside or a pharmaceutically acceptable salt or derivative thereof.
  • a particularly preferred casuarine glucoside is casuarine-6- ⁇ -D-glucoside of the formula:
  • the invention contemplates diastereomers of the compounds of the invention.
  • diastereomers selected from 3,7-d/ep/-casuarine (10), 7-ep/-casuarine (11), 3,6,7-.r/ep/-casuarine (12), 6,7-d/ep/-casuarine (13) and 3-ep/-casuarine (14), as well as pharmaceutically acceptable salts and derivatives thereof.
  • Other preferred diastereomers include 7a epimers selected from 3,7,7a-tr/ep/-casuarine, 7,7a-diep/-ca ⁇ uarine, 3,6,7,7a-fefraep/-casuarine, 6,7,7a-triep/-casuarine and 3,7a-diep/-casuarine, as well as pharmaceutically acceptable salts and derivatives thereof.
  • the invention provides a method for immunomodulation (e.g. immunostimulation) comprising administering to a patient a composition comprising a polyhydroxylated pyrrolizidine compound having the formula:
  • R is selected from the group comprising hydrogen, straight or branched, unsubstituted or substituted, saturated or unsaturated acyl, alkyl (e.g. cycloalkyl), alkenyl, alkynyl and aryl groups, or a pharmaceutically acceptable salt or derivative thereof
  • the invention provides a method for chemoprotection comprising administering the compound of the invention to a patient undergoing chemotherapy.
  • the invention also contemplates the use of the polyhydroxylated pyrrolizidine compound of the invention for the manufacture of a medicament for use in immunostimulation and/or chemoprotection, as well as a process for the manufacture of a medicament for use in immunostimulation and/or chemoprotection, characterized in the use of the polyhydroxylated pyrrolizidine compound of the invention.
  • the invention contemplates a composition
  • a composition comprising the polyhydroxylated pyrrolizidine compound of the invention in combination with an immunostimulant and/or cytotoxic agent (e.g. AZT) and/or an antimicrobial (e.g. antibacterial) agent and/or an antiviral agent and/or a dendritic cell (e.g. a primed dendritic cell).
  • an immunostimulant and/or cytotoxic agent e.g. AZT
  • an antimicrobial agent e.g. antibacterial
  • an antiviral agent e.g. a dendritic cell
  • dendritic cell e.g. a primed dendritic cell
  • the invention contemplates a vaccine comprising the polyhydroxylated- pyrrolizidine compound of the invention in combination with an antigen, the compound being present in an amount sufficient to produce an adjuvant effect on vaccination.
  • kits of parts comprising the polyhydroxylated pyrrolizidine compound of the invention in combination with an immunostimulant and/or cytotoxic agent (e.g. 5' fluoro-uracil and ricin) and/or an antimicrobial (e.g. antibacterial) agent and/or an antiviral agent " (e.g. AZT).
  • cytotoxic agent e.g. 5' fluoro-uracil and ricin
  • an antimicrobial agent e.g. antibacterial
  • an antiviral agent e.g. AZT
  • the compounds of the invention have broad utility in therapy and prophylaxis, including treatments for increasing the Th1 :Th2 response ratio, for example in the treatment of Th1 -related diseases or disorders (e.g. proliferative disorders or microbial infection) and/or Th2-related diseases or disorders (for example allergies, e.g. asthma), as well as in haemorestoration, the alleviation of immunosuppression, in cytokine stimulation, in the treatment of proliferative disorders, vaccination (wherein the compound acts as an adjuvant), vaccination with dendritic cell vaccines (e.g.
  • adjunctive as applied to the use of the drugs of the invention in therapy
  • Such adjunctive therapies may comprise the concurrent, separate or sequential administration/application of the pyrrolizidine compound of the invention and the other treatment(s).
  • adjunctive use of the pyrrolizidine compound of the invention is reflected in the formulation of the pharmaceutical compositions of the invention.
  • adjunctive use may be reflected in a specific unit dosage or in formulations in which the pyrrolizidine compound of the invention is present in admixture with the other drug(s) with which it is to be used adjunctively (or else physically associated with the other drug(s) within a single unit dose)
  • adjunctive use of the pyrrolizidine compound of the invention may be reflected in the composition of the pharmaceutical kits of the invention, wherein the pyrrolizidine compound or the invention is co-pac ⁇ age ⁇ ⁇ e g as parr o ⁇ an array o ⁇ unit doses) with the other drug(s) with which it is to be used adjunctively
  • adjunctive use of the pyrrolizidine compound of the invention may be reflected in the content of the information and/or instructions co-packaged with the pyrrolizidine compound relating to formulation and/or posology
  • neoantigen is used herein to define any newly expressed antigenic determinant Neoantigens may arise upon conformational change in a protein as newly expressed determinants (especially on the surfaces of transformed or infected cells), as the result of complex formation of one or more molecules or as the result of cleavage of a molecule with a resultant display of new antigenic determinants
  • neoantigen covers antigens expressed upon infection (e g viral infection, protozoal infection or bacterial infection), in p ⁇ on-mediated diseases (e g BSE and CJD), an on cell transformation (cancer), in which latter case the neoantigen may be termed a tumour-associated antigen
  • tumour-associated antigen is used herein to define an antigen present in transformed (malignant or tumourous) cells which is absent (or present in lower amounts or in a different cellular compartment) in normal cells of the type from which the tumour originated Oncogenic viruses can also induce expression of tumour antigens which are often host proteins induced by the virus
  • herbal medicine is used herein to define a pharmaceutical composition in which at least one active principle is not chemically synthesized and is a phytochemical constituent of a plant In most cases, this non- synthetic active principle is not isolated (as defined herein), but present together with other phytochemicals with which it is associated in the source plant In some cases, however, the plant-de ⁇ ved bioactive pnnc ⁇ ple(s) may be in a concentrated fraction or isolated (sometimes involving high degrees of purification) In many cases, however, the herbal medicine comprises a more or less crude extract, infusion or fraction of a plant or even an unprocessed whole plant (or part thereof), though in such cases the plant (or plant part) is usually at least dried and/or milled
  • bioactive principle used he r ⁇ n to define a phytochemical which is necessary or sufficient for the pharmaceutical efficacy of the herbal medicament in which it is comprised
  • the bioactive principle com ⁇ nses the immunomodulatory compound of the invention (e g casuarine, casuarine glucoside or mixtures thereof)
  • the term standard specification is used herein to define a characteristic, or a phytochemical profile, which is correlated with an acceptable quality of the herbal medicine
  • quality is used to define the overall fitness of the herbal medicament for its intended use, and includes the presence of one or more of the bioactive principles (at an appropriate concentration) described above or else the presence of one or more bioactive markers or a phytochemical profile which correlates with the presence of one or more of the bioactive principles (at an appropriate concentration)
  • the term phytochemical profile is used herein to define a set of characteristics relating to different phytochemical constituents.
  • the term isolated as applied to the pyrrolizidine compounds of the invention is used herein to indicate that the compound exists in a physical milieu distinct from that in which it occurs in nature.
  • the isolated material may be substantially isolated (for example purified) with respect to the complex cellular milieu in which it naturally occurs.
  • the absolute level of purity is not critical and those skilled in the art can readily determine appropriate levels of purity according to the use to which the material is to be put. Preferred, however, are purity levels of 90% w/w, 99% w/w or higher.
  • the isolated compound forms part of a composition (for example a more or less crude extract containing many other substances) or buffer system, which may for example contain other components.
  • the isolated compound may be purified to essential homogeneity, for example as determined ⁇ pectrophotometrically, by NMR or by chromatography (for example GC-MS).
  • pharmaceutically acceptable derivative as applied to the pyrrolizidine compounds of the invention define compounds which are obtained (or obtainable) by chemical derivatization of the parent pyrrolizidine compounds of the invention.
  • the pharmaceutically acceptable derivatives are therefore suitable for administration to or use in contact with the tissues of humans without undue toxicity, irritation or allergic response (i.e. commensurate with a reasonable benefit/risk ratio).
  • Preferred derivatives are those obtained (or obtainable) by alkyiation, e ⁇ terification or acylation of the parent pyrrolizidine compounds of the invention.
  • the derivatives may be immunomodulatory perse, or may be inactive until processed in vivo. In the latter case, the derivatives of the invention act as pro-drugs.
  • Particularly preferred pro-drugs are ester derivatives which are esterified at one or more of the free hydroxyls and which are activated by hydrolysi ⁇ in vivo.
  • the pharmaceutically acceptable derivatives of the invention retain some or all of the immunomodulatory activity of the parent compound. In some cases, the immunomodulatory activity is increased by derivatization.
  • Derivatization may also augment other biological activities of the compound, for example bioavailability and/or glycosidase inhibitory activity and/or glycosidase inhibitory profile.
  • derivatization may increase glycosidase inhibitory potency and/or specificity.
  • pharmaceutically acceptable salt as applied to the pyrrolizidine compounds of the invention defines any non-toxic organic or inorganic acid addition salt of the free base compounds which are suitable for use in contact with the tissues of humans and lower animals without undue toxicity, irritation, allergic response and which are commensurate with a reasonable benefit/risk ratio. Suitable pharmaceutically acceptable salts are well known in the art.
  • Examples are the salts with inorganic acids (for example hydrochloric, hydrobromic, sulphuric and phosphoric, acids), organic carboxylic acids (for example acetic, propionic, glycolic, lactic, pyruvic, malonic, ⁇ uc nic, fumaric, malic, tartaric, citric, ascorbic, maleic, hydroxymaleic, dihydro/ymaleic, benzoic, phenylacetic, 4-arn ⁇ nobenzo ⁇ c, 4-hydroxyb zoic, anthranilic, cinnamic, salicylic, 2-phenoxybenzoic, 2- acetoxyben ⁇ oic and mandelic acid) and organic sulfonic acids (for example methanesulfonic acid and p- toluenesulfonic acid).
  • inorganic acids for example hydrochloric, hydrobromic, sulphuric and phosphoric, acids
  • organic carboxylic acids for example acetic, propionic, glyco
  • the drugs of the invention may also be converted into salts by reaction with an alkali metal halide, for example sodium chloride, sodium iodide or lithium iodide.
  • an alkali metal halide for example sodium chloride, sodium iodide or lithium iodide.
  • the pyrrolizidine compounds of the invention are converted into their salts by reaction with a stoichiometric amount of sodium chloride in the presence of a solvent such as acetone.
  • These salts and the free base compounds can exist in either a hydrated or a substantially anhydrous form.
  • Crystalline forms of the compounds of the invention are also contemplated and in general the acid addition salts of the pyrrolizidine compounds of the invention are crystalline materials which are ⁇ oluble in water and various hydrophilic organic solvents and which in comparison to their free base forms, demonstrate higher melting point ⁇ and an increased solubility.
  • the present invention contemplates all optical isomers, racemic forms and diastereomers of the pyrrolizidine compounds of the invention.
  • the pyrrolizidine compounds of the invention may exist and be synthesised and/or isolated in optically active and racemic forms.
  • references to the pyrrolizidine compounds of the present invention encompass the pyrrolizidine compounds as a mixture of diastereomers, as individual diastereomers, as a mixture of enantiomers as well as in the form of individual enantiomers.
  • the pre ⁇ ent invention contemplates all optical isomers and racemic forms thereof of the compounds of the invention, and unless indicated otherwise (e.g. by use of dash-wedge structural formulae) the compounds shown herein are intended to encompass all possible optical isomers of the compounds so depicted. In cases where the stereochemical form of the compound is important for pharmaceutical utility, the invention contemplates use of an isolated eutomer.
  • the immunomodulatory activity of the compounds of the invention may arise from the stimulation and/or suppression of cytokine secretion in vivo.
  • the immunomodulatory activity of the compounds of the invention arises from the stimulation of secretion of one or more cytokines (e.g. one or more Thl cytokines), including interleukins 2 and/or 12 (IL-2 and/or IL-12) and/or the suppression of secretion of one or more Th2 cytokines (e.g. IL-5).
  • cytokines e.g. one or more Thl cytokines
  • IL-2 and/or IL-12 interleukins 2 and/or 12
  • Th2 cytokines e.g. IL-5
  • the immunostimulatory activity of the compounds of the invention may arise from the stimulation of 11- 12 and IL-2 by dendritic cells.
  • the induced Th1 cells then produce IFN- ⁇ and IL-2.
  • the IL-2 then enhances further proliferation of Th1 cells and the differentiation of pathogen (e.g. tumour and virus) -specific CD8 + T cells.
  • the IL-2 also stimulates the cytolytic activity of NK cells of the innate immune system.
  • IL-12 is the primary mediator ot type- 1 immunity (the Th l response) It induces natural killer (NK) cells to produce IFN-y as part of the innate immune response and promotes the expansion of CD4 + Th 1 cells and cytotoxic CDS * cells which produce IFN-y It therefore increases T-cell invasion of tumours as well as the susceptibility of tumour cells to T-cell invasion.
  • NK natural killer
  • the compounds of the invention are preferably stimulators of cytokine secretion.
  • Particularly preferred are compounds which induce, potentiate, activate or stimulate the release one or more cytokines (for example Th1 cytokines, e.g. IL-12 and/or II-2, optionally together with one or more other cytokines) in vivo.
  • cytokines for example Th1 cytokines, e.g. IL-12 and/or II-2, optionally together with one or more other cytokines
  • This primary immunomodulatory activity of the compounds of the invention is particularly important in certain medical applications (discussed in detail infra). For example, increased production of IL-12 may overcome the suppression of innate and cellular immunities of HIV-1 -infected individuals and AIDS patients.
  • the cytokine stimulation exhibited by the compounds of the invention may be dependent, in whole or in part, on the presence of co-stimulatory agents.
  • co-stimulatory agents may include, for example, agents that stimulate the innate immune system, including Toll-like receptor (TLR) ligands.
  • TLR Toll-like receptor
  • ligands include microbial products such as lipopolysaccharide (LPS) and/or monophosphoryl lipid) as well as other molecules associated with microbial infection.
  • LPS lipopolysaccharide
  • monophosphoryl lipid monophosphoryl lipid
  • glycosidase inhibition may lead to any or all of the following in vivo:
  • tumour cell glycosylation e.g. tumour antigen glycosylation
  • This ancillary biological activity may therefore augment the primary immunomodulatory activity in some preferred embodiments of the invention. It may be particularly desirable in certain medical applications, including the treatment of proliferative disorders (such as cancer) or in applications where infection is attendant on immune suppression. For example, selective modification of virion antigen glycosylation may render an infecting virus less (or non-) infective and/or more susceptible to endogenous immune responses.
  • the compounds of the invention may alter the HIV viral envelope glycoprotein gp120 glycosylation patterns, hence inhibiting the entry of HIV into the host cell by interfering with the binding to cell surface receptors.
  • the compounds of the invention are preferably (but not necessarily) glycosidase inhibitors.
  • Particularly preferred are compounds which exhibit specificity of glycosidase inhibition, for example Glucosidase I rather than mannosidases.
  • Such preferred compounds can therefore be quite different in their glycosidase inhibitory profile to swainsonine and its analogues, since the latter are potent and specific inhibitors of mannosidase.
  • the invention finds broad application in medicine, for example in methods of therapy, prophylaxis and/or diagnosis.
  • the applications may be applied to any warm-blooded animal, including humans.
  • the applications include veterinary applications, wherein the pyrrolizidine compounds of the invention are administered to non- human animals including primates, dogs cats, horses, cattle and sheep
  • the pyrrolizidine compound ⁇ of the invention are immunomodulators Thus, they find general application in the treatment or prophylaxis of conditions in which stimulation augmentation or induction of the immune system is indicated and/or in which suppression or elimination of part or all of the immune response is indicated
  • the immune response comprises two distinct types the Th1 response (type-1 , cellular or cell mediated immunity) and Th2 response (type-2, humoral or antibody mediated immunity)
  • Th1 and Th2 responses are not mutually exclusive and in many circumstances occur in parallel In such circumstances the balance of the Th1 Th2 response determines the nature (and repercussions) of the immunological defence (as explained herein)
  • the Th1 Th2 balance (which can be expressed as the Th1 Th2 response ratio) is determined, at least in part, by the nature of the environment (and in particular the cytokine milieu) in which antigen priming of naive helper T cells occurs when the immune system is first stimulated
  • Th1 and Th2 response ⁇ are distinguished inter alia on the basi ⁇ of certain phenotypic change ⁇ attendant on priming and subsequent polarization of naive helper T cells These phenotypic changes are characterized, at least in part, by the nature of the cytokines secreted by the polarized helper T cells
  • Th1 cells produce so-called TM cytokines, which include one or more of IL-1 , TNF, IL-2, IFN-gamma, IL-12 and/or IL-18
  • TM cytokines include one or more of IL-1 , TNF, IL-2, IFN-gamma, IL-12 and/or IL-18
  • the Th1 cytokines are involved in macrophage activation and Th1 cells orchestrate cell-mediated defences (including cytotoxic T lymphocyte production) that form a k ⁇ > limb of the defence against bacte ⁇ ai and viral attack, as well as malignant cells
  • Th2 cells produce so-called Th2 ortokines, which include one oi more of IL-4 , IL-5, IL- I0 and IL- 13
  • the Th2 cytokines promote the production of various antibodies and can suppress the Th1 response
  • Th1 a cell that makes IFN-gamma and not IL-4
  • Th2 a CD4 + cell that expresses IL-4 and not IFN-gamma
  • Th1 or Th2 the phenotype of the T cell response
  • the immunomodulatory pyrrolizidine compounds of the invention can increase the Th1 Th2 response ratio in vivo (for example by preferentially promoting a Th1 response and/or preferentially suppressing a Th2 response)
  • the compounds of the invention find application in methods of therapy and/or prophylaxis which comprise increasing the Th1 Th2 response ratio (for example, by preferentially promoting a Th1 response and/or preferentially suppressing a Th2 response)
  • the medical applications contemplated herein therefore include any diseases, conditions or disorders in which an increase in the Th1 Th2 response ratio is indicated or desired
  • the medical applications contemplated include diseases, conditions or disorder ⁇ in which stimulation of a Th1 re ⁇ ponse and/or suppression of a Th2 respon ⁇ e is indicated or desired
  • the mechan ⁇ sm(s) by which the compounds of the invention increase the Thl Th2 response ratio are not yet fully understood It is likely that the activity is based, at least in part, on selective Th1 cytokine induction (since Th1 and Th2 cytokines exhibit mutual inhibition), for example in dendritic cells
  • the compounds of the invention may induce, potentiate, activate or stimulate (either directly or indirectly) the release and/or activity (in vitro and/or in vivo) of one or more Th1 cytokines (for example one or more cytokines selected from IFN-gamma, IL-12, IL-2 and IL-18)
  • Th1 cytokines for example one or more cytokines selected from IFN-gamma, IL-12, IL-2 and IL-18
  • Particularly preferred are compounds which induce, potentiate, activate or stimulate the release and/or activity (in vitro and/or in vivo) of IFN-gamma and/or IL-12 and/or IL-2
  • the compounds of the invention may also suppress or inactivate (either directly or indirecilyl the release and/or activity (in vif.ro and/or in ⁇ ivo) of one oi more Th2 cytokines (for ⁇ Aample one or more cytokines selected from IL-4, IL-5, IL-10 and IL- l ⁇ )
  • Th2 cytokines for ⁇ Aample one or more cytokines selected from IL-4, IL-5, IL-10 and IL- l ⁇
  • Particularly preferred are compounds which suppress or inactivate the release and/oi activity (in vitro and/or in vivo) of IL-5
  • compositions which exhibit a Th I cytokine stimulatory activity together with a complementary Th2 cytokine inhibitor,' activity
  • Th1 -related diseases are described in the following sections Th1 -related diseases
  • Th1 -related diseases are diseases, disorders, syndromes, conditions or infections in which Th1 cells are involved in preventing, curing or alleviating the effects of the disease, disorder, syndrome, condition or infection
  • Th1 -related diseases may also include diseases, disorders, syndromes, conditions or infections in which the Th1 component of the immune response is pathologically depressed or diseases, disorders, syndromes, conditions or infections in which stimulation of a Th1 response is indicated
  • tumour cells may inhibit dendritic cells, cause the expression of inhibitory receptors on T cells, down legulate MHC class I expression and induce the secretion of anti-inflammatory factors and immunosuppressive cytokines which deactivate or suppress immune cell cytotoxicity
  • Th 1 -related diseases include infectious diseases (particularly viral infe ions) and proliferative disorders (e g cancer)
  • the Th1 -related diseases include any malignant or pre-ma gnant condition, proliferative or hyper- proliferative condition or any disease arising or deriving from or associated with a functional or other disturbance or abnormality in the proliferative capacity or behaviour of any cells or tissues of the body
  • the invention finds application in the treatment or prophylaxis of breast cancer, colon cancer, lung cancer and prostate cancer It also finds application in the treatment or prophylaxis of cancers of the blood and lymphatic systems (including Hodgkin's Di ⁇ ease, leukemias, lymphomas, multiple myeloma, and Waldenstrom's disease), skin cancers (including malignant melanoma), cancers of the digestive tract (including head and neck cancers, cesophageal cancer, stomach cancer, cancer of the pancreas, liver cancer, colon and rectal cancer, anal cancer), cancers of the genital and urinary systems (including kidney cancer, bladder cancer, testis cancer, prostate cancer), cancers in women (including breast cancer, ovarian cancer, gynecological cancers and cho ⁇ ocarcincma) as well as ,n bra.n, bone carcmoid, na ⁇ opharyngeal, retropentoneal, thyroid and soft tissue tumours It also finds application in the treatment or prophylaxis of
  • the Th1-related infectious diseases include bacterial, p ⁇ on (e g BSE and CJD), vital, fungal, protozoan and metazoan infections
  • the Th l-related infectious diseases include infection with respiratory syncytial virus (RSV), hepatitis B virus (HBV), Epstein-Barr, hepatitis C virus (HCV), herpes simplex type 1 and 2, herpes genitalis herpes keratitis, herpes encephalitis, herpes zoster, human immunodeficiency virus (HIV), influenza A virus, hantann virus (hemorrhagic fever), human papilloma virus (HPV), tuberculosis, leprosy and measles
  • Particularly preferred Th1-related infectious diseases include those in which the pathogen occupies an intracellular compartment, including HIV/AIDS, leishmaniasis, trypanosomiasis, influenza, tuberculosis and malaria.
  • the compounds of the invention may also find application in the treatment of patients in which the Th1 immune response is defective.
  • patients may include neonates, juveniles in which the Th1 response is immature and not fully developed, as well as older patients in which the Th1 response has become senescent or compromised over time.
  • the compounds of the invention may be used prophylactically (as a generalized type 1 immune stimulant to reduce the risks of (e.g. viral) infections.
  • Th2-related diseases are diseases, disorders, syndromes, conditions or infections in which Th2 cells are implicated in (e.g. support, cause or mediate) the effects of the disease, disorder, syndrome, condition or infection.
  • the compounds of the invention find application in the treatment or prophylaxis of Th2-related diseases.
  • Th2-related diseases treatable with the compounds of the invention is allergic disease.
  • allergy is used to define a state of hypersensitivity induced by expo ⁇ ure to a particular antigen (allergen) resulting in harmful and/or uncomfortable immunologic reactions on sub ⁇ equent exposures to the allergen.
  • the harmful, uncomfortable and/or undesirable immunologic reactions present in allergy include a wide range of symptoms. Many different organs and tissues may be affected, including the gastrointestinal tract, the skin, the lungs, the nose and the central nervous system. The symptoms may include abdominal pain, abdominal bloating, disturbance of bowel function, vomiting, rashes, skin irritation, wheezing and shortness of breath, nasal running and nasal blockage, headache and mood changes. In severe cases the cardiovascular and respiratory systems are compromised and anaphylaetic shock leads in extreme cases to death.
  • the compounds of the invention may suppress or inactivate (either directly or indirectly) the release and/or activity (in vitro and/or in vivo) of one or more Th2 cytokines (for example one or more cytokines selected from IL-4, IL-5, IL-10 and IL-13).
  • Th2 cytokines for example one or more cytokines selected from IL-4, IL-5, IL-10 and IL-13.
  • the compounds of the invention may be used to effect a remedial or palliative modulation of the harmful and/or uncomfortable immunologic reaction ⁇ characteristic of allergic reactions by inhibiting, suppressing or eliminating the Th2 response to the allergen.
  • the compounds of the invention therefore find application in the treatment or prophylaxis of allergy.
  • Any allergy may be treated according to the invention, including atopic allergy, allergic rhinitis, allergic conjunctivitis, atopic dermatitis, hypereosinophilia, irritable bowel syndrome, allergen-induced migraine, bacterial allergy, bronchial allergy (asthma), contact allergy (dermatitis), delayed allergy, pollen allergy (hay fever), drug allergy, sting allergy, bite allergy, gastrointestinal or food allergy (including that associated with inflammatory bowel disease, including ulcerative colitis and Crohn's disease) and physical allergy.
  • Physical allergies include cold allergy (cold urticaria or angioedema), heat allergy (cholinergic urticaria) and photosensitivity.
  • the pyrrolizidine compounds of the invention increase splenic and bone marrow cell proliferation and can act as myeloproliferative agents. They therefore find application as haemorestoratives.
  • Haemorestoration may be indicated following immunosuppressant therapies (such as cyclosporine A, azathioprine or immunosuppressant radiotherapies), chemotherapy (including treatment with both cycle-specific and non-specific chemotherapeutic agents), steroid administration or other forms of surgical or medical intervention (including radiotherapy).
  • immunosuppressant therapies such as cyclosporine A, azathioprine or immunosuppressant radiotherapies
  • chemotherapy including treatment with both cycle-specific and non-specific chemotherapeutic agents
  • steroid administration including radiotherapy.
  • the use of the pyrrolizidine compounds of the invention as haemorestoratives may be adjunctive to other treatments which tend to depress splenic and bone marrow ceil populations.
  • adjunctive therapies according to the invention include the administration of an immunorestorative dose of the pyrrolizidine compound of the invention adjunctive to: (a) chemotherapy; and/or (b) radiotherapy; and/or (c) bone marrow transplantation; and/or (d) haemoablative immunotherapy.
  • the pyrrolizidine compounds of the invention may be used to alleviate, control or modify states in which the immune system is partially or completely suppressed or depressed. Such states may arise from congenital (inherited) conditions, be acquired (e.g. by infection or malignancy) or induced (e.g. deliberately as part of the management of transplants or cancers).
  • the pyrrolizidine compounds of the invention may find application as adjunctive immunomodulators (e.g. immunostimulants) in the treatment and/or management of various diseases (including certain cancers) or medical interventions (including radiotherapy, immunosuppressant therapy (such as the administration of cyclosporine A, azathioprine or immunosuppressant radiotherapies), chemotherapy and cytotoxic drug administration (for example the administration of ricin, cyclophosphamide, cortisone acetate, vinblastine, vincristine, adriamycin, 6-mercaptopurine, 5-fluorouracil, mitomycin C, chloramphenicol and other steroid-based therapies).
  • immunosuppressant therapy such as the administration of cyclosporine A, azathioprine or immunosuppressant radiotherapies
  • chemotherapy and cytotoxic drug administration for example the administration of ricin, cyclophosphamide, cortisone acetate, vinblastine, vincristine, a
  • the pyrrolizidine compounds of the invention may find application as immuno ⁇ timulants in the treatment or management of microbial infections which are associated with immune-suppressed states, including many viral infections (including HIV infection in AIDS) and in other situations where a patient has been immunocompromised (e g following infection with hepatitis C, or other viruses or infectious agents in uding bacteria, fungi, and parasites in patients undergoing bone marrow transplants, and in patients with chemical or tumor-induced immune suppression)
  • immunocompromised e g following infection with hepatitis C, or other viruses or infectious agents in uding bacteria, fungi, and parasites in patients undergoing bone marrow transplants, and in patients with chemical or tumor-induced immune suppression
  • diseases or disorders which may give rise to an immunosupressed state treatable according to the invention include ataxia-telangiectasia, DiGeorge syndrome, Chediak-Higashi syndrome, Job syndrome, leukocyte adhesion defects, panhypogammaglobulinemia (e g associated with Bruton disease or congenital agammaglobulinemia) selective deficiency of IgA, combined immunodeficiency disease, Wiscott-Ald ⁇ ch syndrome and complement deficiencies It may be associated with organ and/or tissue (e g bone marrow) transplantation or grafting, in which applications the pyrrolizidine compounds of the invention may be used adjunctively as part of an overall treatment regimen including surgery and post-operative management of immune status
  • the pyrrolizidine compounds of the invention may be used to induce, potentiate or activate various cytokine ⁇ in vivo, including va ⁇ ou ⁇ interleukms (including IL-2 and/or IL-12)
  • the pyrrolizidine compounds of the invention find general application in the treatment or prophylaxis of conditions in which the in vivo induction, polentiation or activation of one or more cytokines (e g IL-12 and/or II-2) is indicated
  • cytokines e g IL-12 and/or II-2
  • Such applications may be employed to stimulate particular elements of the cellular immunity system, including dendritic cells, macrophages (e g tissue-specific macrophages), CTL, NK, NKT, B and LAK cells
  • the compounds of the invention may be employed as an adjunct to gene therapies designed to increase the production of endogenous cytokines (for example IL-2)
  • the invention finds application in the treatment of proliferative disorders, including various cancers and cancer metastasis
  • the pyi rolizidine compounds of the invention may find particular application in the treatment of leukemias lymphomas melanomas adenomas sarcomas, carcinomas of solid tissues, melanoma (including melanoma of the eye) pancieatic cancer, cervico-ute ⁇ ne cancer, cancel s of the kidney, stomach, lung ovary, rectum, breast, prostate, bowel, gastric, liver thyroid, neck, cervix, salivary gland, leg, tongue, lip, bile duct, pelvis, mediastinum, urethra, lung, bladder, esophagus and colon, and Kapo ⁇ i's Sarcoma (e g when associated with AIDS)
  • tumour cell glycosylation e.g. tumour antigen glycosylation
  • viral protein glycosylation e.g. virion antigen glycosylation
  • cell-surface protein glycosylation in infected host cells and/or the modification of bacterial cell walls, hence promoting an increased immune response or inhibiting growth/infectivity directly.
  • the pyrrolizidine compounds of the invention find utility as vaccine adjuvants, in which embodiments they may promote, induce or enhance an immune response to antigens, particularly antigens having low intrinsic immunogenicity.
  • the pyrrolizidine compounds of the invention may augment vaccine immunogenicity by stimulating cytokine release, thereby promoting T-cell help for B cell and CTL responses. They may also change glycosylation of cancer or viral antigens and increase vaccine effectiveness.
  • the compounds of the invention may be administered concurrently, separately or sequentially with adminislration of the vaccine.
  • the invention finds application in any vaccine, but may be particularly as a subunit vaccine, a conjugate vaccine, a DNA vaccine, a recombinant vaccine or a mucosal vaccine.
  • the vaccine may be therapeutic or prophylactic. It may be used imrnunoprophylactically or immunotherapeutically in both human and non-human subjects. Preferred non-human subjects include mammals and birds. Particularly preferred are veterinary applications. Such applications include the treatment or prophylaxis of infection in domesticated animals (for example dogs and cats) and livestock (e.g. sheep, cows, pigs, horses, chickens and turkeys). '
  • the pyrrolizidine compound of the invention may be present in admixture with other vaccine component(s), or else co-packaged (e.g. as part of an array of unit doses) with the other vaccine components with which it is to be used as adjuvant.
  • the use of the pyrrolizidine compounds of the invention as adjuvant is simply reflected in the content of the information and/or instructions co-packaged with the vaccine components and relating to the vaccination procedure, vaccine formulation and/or posology.
  • the pyrrolizidine compounds of the invention may induce sustained and pronounced cytokine production (e.g sustained and pronounced IL-12 and/or IL-2 production) in dendritic cells.
  • the compounds of the invention find application in methods of therapy or prophylaxis comprising the induction of cytokine production in dendritic cells or in which the induction of cytokine production in dendritic cells is indicated or required.
  • the cells are pulsed (primed or spiked) with a particular antigen or antigens (for example, tumour antigen(s)) and then administered to promote a Th1 immune response.
  • the responding T ceils include helper cells, especially Th1 CD4 + cells (which produce IFN- ⁇ ) and killer cells (especially CD8 + cytolytic T lymphocytes).
  • the dendritic cells may also mediate responses by other classes of lymphocytes (B, NK, and NKT cells). They may also elicit T cell memory, a critical goal of vaccination.
  • antigen selection for use in the dendritic cell vaccine sof the invention, both defined and undefined antigens can be employed.
  • the antigens can be xenoantigens or autoantigens.
  • One or more defined neoantigen(s) may be selected: in the case of cancer treatment, the neoantigen(s) may comprise a tumour-associated antigen.
  • peptides for example, synthetic 9-11 amino acid peptides
  • Such peptides may comprise natural sequences.
  • they may be synthetic analogues designed for enhanced MHC binding.
  • the antigens used according to the invention are provided in the form of immune complexes. These are preferably delivered to Fc-receptor-bearing DCs so that both MHC class I and MHC class II peptide sequences are formed. In this way, dendritic cell vaccines can be used according to the invention for inducing both CTLs and Th cells.
  • DC-tumour cell hybrids in which the dendritic cells are treated with compound (thereby to induce the expression of IL-2) before or after hybridisation.
  • necrotic or apoptotic tumour cells or cell lysates for example lysates of infected cells or tumour cells.
  • Antigens derived from fresh tumour cells may also be employed.
  • the compounds of the invention be incorporated into cellular antigens by introducing them into the cellular membrane or into an intracellular compartment (as described for example in WO96017614, the contents of which are incorporated herein by reference).
  • antigen loading various techniques can be used to deliver the selected antigen(s) to the DCs (variously referred to in the art as antigen loading, pulsing, priming or spiking).
  • loading techniques which load the DCs internally: this can be achieved through the use of peptides linked to cell-penetrating moieties.
  • Antigens can also be loaded by transfecting the DCs with encoding nucleic acid (e.g. by electroporation) such that the antigens are expressed by the DC, processed and presented at the cell surface.
  • This approach avoids the need for expensive GMP proteins and antibodies.
  • RNA is preferred for this purpose, since it produces only transient expression (albeit sufficient for antigen processing) and avoids the potential problems associated with the integration of DNA and attendant long-term expression/mutagenesis.
  • Such transfection techniques also permit exploration of the whole antigenic repertoire of a target cell by use of total or PCR-amplified tumour RNA.
  • the dendritic ceil based vaccines of the invention find particular application in the treatment or prophylaxis of various proliferative disorders (including various cancers, as described below).
  • the dendritic cells are preferably pulsed (primed or spiked) with one or more tumour antigens e* vivo and the compounds of the invention used to potentiate the dendritic cell component of the vaccine by contacting the dendritic cells with the compound either ex vivo (before or after pulsing of the cells) or in vivo (for example by co-administration, either concurrently, separately or sequentially, of the dendritic cells and the compound).
  • the dendritic cell based vaccines of the invention may be used in the treatment or prophylaxis of any malignant or pre-malignant condition, proliferative or hyper-proliferative condition or any disease arising or deriving from or associated with a functional or other disturbance or abnormality in the proliferative capacity or behaviour of any cells or tissues of the body.
  • the invention finds application in the treatment or prophylaxis of breast cancer, colon cancer, lung cancer and prostate cancer. It also finds application in the treatment or prophylaxis of cancers of the blood and lymphatic systems (including Hodgkin's Disea ⁇ e, leukemias, lymphomas, multiple myeloma, and Waldenstrom's disease), skin cancers (including malignant melanoma), cancers of the digestive tract (including head and neck cancers, cesophageal cancer, stomach cancer, cancer of the pancreas, liver cancer, colon and rectal cancer, anal cancer), cancers of the genital and urinary systems (including kidney cancer, bladder cancer, testis cancer, prostate cancer), cancers in women (including breast cancer, ovarian cancer, gynecological cancers and choriocarcinoma) as well as in brain, bone carcinoid, nasopharyngeal, retroperitoneal, thyroid and soft tissue tumours. It also finds application in the treatment or prophylaxis of cancers
  • the dendritic cell based vaccines of the invention also find application in the treatment or prophylaxis of various infections, including bacterial, viral, fungal, protozoan and metazoan infections.
  • the vaccines may be used in the treatment or prophylaxis of infection with respiratory syncytial virus (RSV), Epstein-Barr, hepatitis B virus (HBV), hepatitis G virus (HCV), herpes simplex type 1 and 2, herpes genitali ⁇ , herpes keraliti ⁇ , herpes encephalitis, herpes zoster, human immunodeficiency virus (HIV), influenza A virus, hantann virus (hemorrhagic fever), human papilloma virus (HPV), tuberculosis, leprosy and measles.
  • RSV respiratory syncytial virus
  • HBV hepatitis B virus
  • HCV hepatitis G virus
  • the treatment or prophylaxis of infections in which the pathogen occupies an intracellular compartment or causes the expression of neoantigens by host cells, including HIV/AIDS, leishmania, trypanosomiasis, influenza, tuberculosis and malaria.
  • the present invention also contemplates a more general approach to DC cell-based therapy which involves the stimulation of the dendritic cells with the compound of the invention irrespective of the antigens present and either with or without antigen priming.
  • the invention finds application in therapies in which dendritic cells exposed to the compound of the invention are targeted to diseased or infected tissue (for example injected directly into a tumour), where the cells can prime endogenous T cells extranodally.
  • the invention contemplates targeting of DCs to a tumour and their activation in situ to elicit immune responses without the need for ex vivo antigen loading.
  • the invention contemplates in situ DC vaccination where antigen is targeted to DCs in vivo which are then expanded and induced to mature in situ (by the co-administration of one or more DC maturation stimulants).
  • antigen is targeted to endogenous DCs by any convenient method, for example through the use of exosomes (as described in Thery et al. (2002) Nat Rev Immunol 2: 569-579).
  • the dendritic cells may be myeloid or lymphoid, or mixtures thereof.
  • the myeloid dendritic cells if used, may be of the Langerhans cell type or interstitial DCs. Alternatively, mixtures of these myeloid subsets may be used. Especially preferred is the use of monocyte-derived DCs (Mo-DCs).
  • Helper proteins may be used to potentiate the activity of the dendritic cell vaccines of the invention.
  • Dendritic cells are also involved in regulating and maintaining immunological tolerance: in the absence of maturation, the cells induce antigen-specific silencing or tolerance. Thus, in another dendritic cell-based treatment paradigm the cells are administered as part of an immunomodulatory intervention designed to combat autoimmune disorders.
  • dendritic cells have been enhanced by in vitro transfectlon with genes encoding cytokines.
  • gene therapy approaches are inherently dangerous and a more efficient and attractive approach would be to pulse dendritic cells in vitro with biologically active compounds which stimulate an appropriate cytokine secretion pattern in the dendritic cells.
  • the pyrrolizidine compounds of the invention can induce sustained and pronounced cytokine production in dendritic cells.
  • the compounds of the invention find application in the enhancement of the suppressive potential of dendritic cells.
  • the invention finds application in the treatment or prophylaxis of autoimmune disorders, including myasthenia gravis, rheumatoid arthritis, systemic lupus erythematosus, Sjogren syndrome, scleroderma, polymyositis and ' dermomyositis, ankylosing spondylitis, and rheumatic fever, insulin-dependent diabetes, thyroid diseases (including Grave's disease and Hashimoto thyroiditis), Addison's disease, multiple sclerosis, psoriasis, inflammatory bowel disease, ulcerative colitis and autoimmune male and female infertility.
  • autoimmune disorders including myasthenia gravis, rheumatoid arthritis, systemic lupus erythematosus, Sjogren syndrome, scleroderma, polymyositis and ' dermomyositis, ankylosing spondylitis, and rheumatic fever, insulin-dependent diabetes, thyroid
  • the pyrrolizidine compounds of the invention can reverse a Th2 type splenocyte response ex vivo in a normally non-healing infectious disease model.
  • Antigen specific splenocyte IFN-gamma can be significantly increased and IL-5 production significantly reduced in such models, indicative of a healing response ' .
  • the invention finds application in the treatment of wounds.
  • the invention finds application in the treatment or prophylaxis of wounds and lesions, for example those associated with post-operative healing, burns, infection (e.g. necrotic lesions), malignancy or trauma (e.g. associated with cardiovascular disorders such as stroke or induced as part of a surgical intervention).
  • the wound treatments may involve the selective suppression or elimination of a Th2 response (for example to eliminate or suppress an inappropriate or harmful inflammatory response).
  • the pyrrolizidine compounds of the present invention can be administered by oral or parenteral routes, including intravenous, intramuscular, intraperitoneal, subcutaneous, transdermal, airway (aerosol), rectal, vaginal and topical (including buccal and sublingual) administration.
  • oral or parenteral routes including intravenous, intramuscular, intraperitoneal, subcutaneous, transdermal, airway (aerosol), rectal, vaginal and topical (including buccal and sublingual) administration.
  • the amount of the pyrrolizidine compound administered can vary widely according to the particular dosage unit employed, the period of treatment, the age and sex of the patient treated, the nature and extent of the disorder treated, and the particular pyrrolizidine compound selected.
  • the pyrrolizidine compounds of the invention can be used in conjunction with other agents known to be useful in the treatment of diseases, disorders or infections where immunostimulation is indicated (as described infra) and in such embodiments the dose may be adjusted accordingly.
  • the effective amount of the pyrrolizidine compound administered will generally range from about 0.01 mg/kg to 500 mg/kg daily.
  • a unit dosage may contain from 0.05 to 500 mg of the pyrrolizidine compound, and can be taken one or more times per day.
  • the pyrrolizidine compound can be administered with a pharmaceutical carrier using conventional dosage unit forms either orally, parenterally, or topically, as described below.
  • a suitable dose will be in the range of 0.01 to 500 mg per kilogram body weight of the recipient per day, preferably in the range of 0.1 to 50 mg per kilogram body weight per day and most preferably in the range 1 to 5 mg per kilogram body weight per day.
  • the desired dose is preferably presented as a single dose for daily administration. However, two, three, four, five or six or more sub-doses administered at appropriate intervals throughout the day may also be employed. These sub-doses may be administered in unit dosage forms, for example, containing 0.001 to 100 mg, preferably 0.01 to 10 mg, and most preferably 0 5 to 1.0 mg of active ingredient per unit dosage form.
  • compositions of the invention comprise the pyrrolizidine compound of the invention, optionally together with a pharmaceutically acceptable excipient.
  • the pyrrolizidine compound of the invention may take any form. It may be synthetic, purified or isolated from natural sources (for example from Casuarina equisetifolia or Eugenia jambolana), using techniques described in the art (and referenced infra).
  • the pyrrolizidine compound of the invention When isolated from a natural source, the pyrrolizidine compound of the invention may be purified.
  • the compositions of the invention may take the form of herbal medicines, as hereinbefore defined. Such herbal medicines preferably are analysed to determine whether they meet a standard specification prior to use.
  • the herbal medicines for use according to the invention may be dried plant material.
  • the herbal medicine may be processed plant material, the processing involving physical or chemical pre-processing, for example powdering, grinding, freezing, evaporation, filtration, pressing, spray drying, extrusion, supercritical solvent extraction and tincture production.
  • the plant material may be dried prior to use. Any convenient form of drying may be used, including freeze-drying, spray drying or air-drying. '
  • any suitable excipient may be used, including for example inert diluents, disintegrating agents, binding agents, lubricating agents, sweetening agents, flavouring agents, colouring agents and preservatives.
  • suitable inert diluents include sodium and calcium carbonate, sodium and calcium phosphate, and lactose, while corn starch and alginic acid are suitable disintegrating agents.
  • Binding agents may include starch and gelatin, while the lubricating agent, if present, will generally be magnesium stearate, stearic acid or talc.
  • compositions may take any suitable form, and include for example tablets, elixirs, capsules, solutions, suspensions, powders, granules and aerosols.
  • the pharmaceutical composition may take the form of a kit of parts, which kit may comprise the composition of the invention together with instructions for use and/or a plurality of different components in unit dosage form.
  • Tablets for oral use may include the pyrrolizidine compound of the invention, either alone or together with other plant material associated with the botanical source(s) (in the case of herbal medicine embodiments).
  • the tablets may contain the pyrrolizidine compound of the invention mixed with pharmaceutically acceptable excipients, ⁇ uch as inert diluents, disintegrating agents, binding agents, lubricating agents, sweetening agents, flavouring agents, colouring agents and preservatives.
  • suitable inert diluents include sodium and calcium carbonate, sodium and calcium phosphate, and lactose, while corn starch and alginic acid are suitable disintegrating agents.
  • Binding agents may include starch and gelatin, while the lubricating agent, if present, will generally be magnesium stearate, stearic acid or talc. If desired, the tablets may be coated with a material such as ⁇ lvcervl monostearate or glyceryl distearate, to delay absorption in the gastrointestinal tract.
  • Capsules for oral use include hard gelatin capsules in which the pyrrolizidine compound of the invention is mixed with a solid diluent, and soft gelatin capsules wherein the active ingredient is mixed with water or an oil such as peanut oil, liquid paraffin or olive oil.
  • Formulations for rectal administration may be presented as a suppository with a suitable base comprising for example cocoa butter or a salicylate.
  • Formulations suitable for vaginal administration may be presented as pessaries, tampons, creams, gels, pastes, foams or spray formulations containing in addition to the active ingredient such carriers as are known in the art to be appropriate.
  • the compounds of the invention will generally be provided in sterile aqueous solutions or suspensions, buffered to an appropriate pH and . isotonicity.
  • Suitable aqueous vehicles include Ringer's solution and isotonic sodium chloride.
  • Aqueous suspensions according to the invention may include suspending agents such as cellulose derivatives, sodium alginate, polyvinylpyrrolidone and gum tragacanth, and a wetting agent such as lecithin.
  • Suitable preservatives for aqueous suspensions include ethyl and n-propyl p-hydroxybenzoate.
  • the compounds of the invention may also be presented as iiposome formulations.
  • the pyrrolizidine compound of the invention can be formulated into solid or liquid preparations such as capsules, pills, tablets, troches, lozenges, melts, powders, granules, solutions, suspensions, dispersions or emulsions (which solutions, suspensions dispersions or emulsions may be aqueous or non-aqueous).
  • the solid unit dosage forms can be a capsule which can be of the ordinary hard- or soft-shelled gelatin type containing, for example, surfactants, lubricants, and inert fillers such as lactose, sucrose, calcium phosphate, and comstarch.
  • the pyrrolizidine compounds of the invention are tableted with conventional tablet bases such as lactose, sucrose, and comstarch in combination with binders such as acacia, comstarch, or gelatin, disintegrating agents intended to assist the break-up and dissolution of the tablet following administration such as potato starch, alginic acid, corn starch, and guar gum, lubricants intended to improve the flow of tablet granulations and to prevent the adhesion of tablet material to the surfaces of the tablet dies and punches, for example, talc, stearic acid, or magnesium, calcium, or zinc stearate, dyes, coloring agents, and ' flavoring agents intended to enhance the aesthetic qualities of the tablets and make them more acceptable to the patient.
  • conventional tablet bases such as lactose, sucrose, and comstarch in combination with binders such as acacia, comstarch, or gelatin
  • disintegrating agents intended to assist the break-up and dissolution of the tablet following administration such as potato starch
  • Suitable excipients for use in oral liquid dosage forms include diluents such as water and alcohols, for example, ethanol, benzyl alcohol, and the polyethylene alcohols, either with or without the addition of a pharmaceutically acceptably surfactant, suspending agent or emulsifying agent.
  • the pyrrolizidine compounds of the invention may also be administered parenterally, that is, subcutaneously, intravenously, intramuscularly, or interperitoneally.
  • the pyrrolizidine compound is provided as injectable doses in a physiologically acceptable diluent together with a pharmaceutical carrier (which can be a sterile liquid or mixture of liquids).
  • a pharmaceutical carrier which can be a sterile liquid or mixture of liquids.
  • suitable liquids include water, saline, aqueous dextrose and related sugar solutions, an alcohol (such as ethanol, isopropanol, or hexadecyl alcohol), glycols (such as propylene glycol or polyethylene glycol), glycerol ketals (such as 2,2-dimethyl-1 ,3-dioxolane-4-methanol), ethers (such as poly(ethylene-glycol) 400), an oil, a fatty acid, a fatty acid ester or glyceride, or an acetylated fatty acid glyceride with or without the addition of a pharmaceutically acceptable surfactant (such as a soap or a detergent), suspending agent (
  • Suitable fatty acids include oleic acid, stearic acid, and isostearic acid.
  • Suitable fatty acid esters are, for example, ethyl oleate and isopropyl myristate.
  • Suitable soaps include fatty alkali metal, ammonium, and triethanolamine salts and suitable detergents include cationic detergents, for example, dimethyl dialkyl ammonium halides, alkyl pyridinium halides, and alkylamines acetates; anionic detergents, for example, alkyl, aryl, and olefin sulphonates, alkyl, olefin, ether, and monoglyceride sulphates, and sulphosuccinates; nonionic detergents, for example, fatty amine oxides, fatty acid alkanolamides, and polyoxyethylenepolypropylene copolymers; and amphoteric detergents, for example, alkyl-beta-aminopropionates, and 2-alkylimidazoline quartemary ammonium salts, as well as mixtures.
  • suitable detergents include cationic detergents, for example, dimethyl dialkyl ammonium halides, al
  • compositions of this invention will typically contain from about 0.5 to about 25% by weight of the pyrrolizidine compound of the invention in solution. Preservatives and buffers may also be used. In order to minimize or eliminate irritation at the site of injection, such compositions may contain a non-ionic surfactant having a hydrophile-lipophile balance (HLB) of from about 12 to about 17. The quantity of surfactant in such formulations ranges from about 5 to about 15% by weight.
  • the surfactant can be a single component having the above HLB or can be a mixture of two or more components having the desired HLB.
  • surfactants used in parenteral formulations are the class of polyethylene sorbitan fatty acid esters, for example, sorbitan monooleate and the high molecular weight adducts of ethylene oxide with a hydrophobic base, formed by the condensation of propylene oxide with propylene glycol.
  • the pyrrolizidine compounds of the invention may also be administered topically, and when done so the carrier may suitably comprise a solution, ointment or gel base.
  • the base for example, may comprise one or more of the following: petrolatum, lanolin, polyethylene glycols, bee wax, mineral oil, diluents such as water and alcohol, and emulsifiers and stabilizers Topical formulations may contain a concentration of the compound from about 0 1 to about 10% w/v (weight per unit volume)
  • the pyrrolizidine compounds of the invention may be formulated for use with one or more other ⁇ rug(s) in particular, the pyrrolizidine compounds of the invention may be used in combination with antitumor agents, antimicrobial agents, anti-inflammato ⁇ es, antiproliferative agents and/or other immunomodulatory (e g immunostimulatory) agent ⁇
  • the pyrrolizidine compound ⁇ of the invention may be used with anti-viral and/or anti-proliferative agents such as cytokines, including ⁇ nterleuk ⁇ ns-2 and 12, interferons and inducers thereof, tumor necrosis factor (TNF) and/or transforming growth factor (TGF), as well as with myelosuppressive agents and/or chemotherapeutic agents (such as doxorubicin, 5-fluorourac ⁇ l, cyclopho ⁇ phamide and methotrexate), isoniazid (e g in the prevention or treatment of peripheral neuropathy) and with anal
  • adiunctive use may be reflected in a specific unit dosage designed to be compatible (or to synergize) with the othei drug(s), or in formulations in which the pyrrolizidine compound is admixed with one or more antitumor agents, antimicrobial agents and/or anliinflammatones (or el ⁇ e physically associated with the other drug(s) within a single unit dose)
  • Adjunctive uses may also be reflected in the composition of the pharmaceutical kits of the invention, in which the pyrrolizidine compound of the invention is co-packaged (e g as part of an array of unit doses) with the antitumor agents, antimicrobial agents and/or antiinflammato ⁇ es
  • Adjunctive use may also be reflected in information and/or instructions relating to the co-administration of the pyrrolizidine compound with antitumor agents, antimicrobial agents and/or antiinflammato ⁇ es
  • Bone marrow was obtained from the femurs of mice The femurs were washed in 70% ethanol and placed in a clean pet ⁇ dish Dendritic cell (DC) medium (2 5% granulocyte-macrophage colony-stimulating factor GM- CSF), 10% heat and activated foetal calf serum, 1% L-glutamme, 1 % Penicillin/Streptomycin in RPMI-1640 medium) was injected into the bone marrow of the femur by a pumping action and the cells and medium were collected 1 ml of the cells in medium was added to a 75cm 2 flask with 15mls of DC medium The flasks were then incubated at 37°C, 5% CO 2 to allow DC growth and development. After 5 days an additional 10mls of DC medium was added.
  • DC Dendritic cell
  • the dendritic cells were harvested. This process was carried out in a tissue culture hood. The contents of the flasks were poured into centrifuge tubes to ensure collection of floating DCs. Approximately 10mls of cooled phosphate buffered saline (PBS) was added to each empty flask, the flasks gently agitated and the contents collected. This ensured recovery of adhesive DCs. The collected contents of the flasks were centrifuged for 5 minutes at 200g and the pellet resuspended in 2mls of DC medium without GM-CSF. A cell count was then carried out.
  • PBS phosphate buffered saline
  • Cells were counted using a haemocylometer. Approximately 20 ⁇ l of the resuspended cells was pipetted into the chamber of the haemocytometer, the cells were adjusted to the correct cell concentration (approx. 5 x 10 4 , and not less than 1 x 10 4 , per well) and then plated out for assay.
  • the plates were incubated overnight at 37°C with 5% CO2 and allowed to settle (harvesting stimulates them). The next day the compounds (50 ⁇ g/mi and 20 ⁇ g/ml) and controls were added then again incubated at 37°C with 5% CO 2 for 24 hrs (or 48 hrs). Harvesting and addition of the compounds was all done in a hood. The plates were then frozen to kill the cells and once defrosted the supernatant analysed as described below.
  • IL-12 concentration in the supernatants was measured. All reagents used in this assay were from Pharivlingen.
  • a 96-well flat-bottomed ELISA plate was coated with purified rat anti-mouse IL-12 (p40/p70) MAb (Cat no. 554478) at 2 ⁇ g/ml diluted in PBS pH 9.0 at 50 ⁇ l/well. The plate was then covered in cling film and incubated at 4°C. Following incubation the plate was washed 3 times in washing buffer and dried.
  • Biotin labelled anti-mouse IL-12 (p40/p70) MAb (Cat no. 18482D) at 1 ⁇ g/ml (diluted in blocking buffer) was added to each well at a volume of 100 ⁇ l/well. The plate was covered in cling film and incubated at 37°C for 1 hour. The plate was then washed 5 times, dried and the conjugate added. Streptavidin-AKP (Cat no. 13043E) at 100 ⁇ l/well was added at a dilution of 1/2000 in blocking buffer followed by incubation under cling film at 37°C for 45 minutes. • The plate was finally washed 6 times, dried and the substrate added! pN.PP (Sigma) in glycine buffer at 1mg/ml was added at 100 ⁇ l/well. The plate was then covered in tinfoil, incubated at 37°C and checked every 30 minutes for a colour change.
  • Example 2 Stimulation of IL-2 production by dendritic cells
  • Example 2 The protocols described in Example 1 above were carried out but the appropriate Mabs and standards for determination of II-2 were substituted. The results are shown in Table 2.1 , below.
  • mice BALB/c male and female mice bred and maintained at the University of Strathclyde under conventional conditions were used at varying age.
  • the mouse spleen was removed aseptically and placed in a sterile petri dish containing 5mls of complete medium (RPMI, 1 % L-Glutamine, 1% Penicillin/Streptomycin and 10% foetal calf serum 1 ).
  • Cells suspensions were prepared by using the end of a syringe and grinding the spleen through a wire mesh. The cell suspension was then centrifuged al lOOOrpm for 5 minutes. To remove the erythrocytes, the cell pellet was resuspended in Boyle's solution (Tris 0. 17M e Ammonium Chloride 0. 16M) and centrifuged again for 5 minutes. The pellet was then washed in medium a further two times, then resuspended in 3ml- medium. A cell count was then carried out.
  • All spleen cell experiments were carried out in 96-well tissue culture plates 100 ⁇ l aliquots of 5x10 s /well cells were added to all wells and each well had a final volume of 200 ⁇ l
  • Unstimulated wells contained 100 ⁇ l of cells and 10O ⁇ l of medium.
  • the stimulated wells contained 10O ⁇ l of cells plus 50 ⁇ l of LPS at 1 ⁇ g/ml or 50 ⁇ l anti- CD3 at 0 5 ⁇ g/ml and 50 ⁇ l of medium
  • the remaining wells contained 10O ⁇ l cells, 50 ⁇ l of MNLP compound and either 50 ⁇ l of ant ⁇ -CD3 or medium alone
  • mice were 3-4 weeks old female BALB/c Mice were inoculated with 10 p f u HSV- 1 (SC16) using the neck skin method This dose is sublethal but produces clinical symptoms including inflammation (measured by increase in ear pinna thickness)
  • mice were ⁇ administered (, 100 ml i p ) with one of two doces of casuarine ( ⁇ ) on day one and daily thereafter for 5 days
  • Group 1 received 15 mg/kg in PBS
  • group 2 received 150 mg/kg in PBS
  • a negative control group 3 were infected but received no casuarine
  • a positive contiol group 4 were administered with famciclovir (1 /a drinking water spiked at 1 mg/ml for the same time period)
  • mice were checked daily and sample ⁇ were obtained from mice killed on selected days The results are present ⁇ d in Tables 6 1 - 6 3, below Table 6.1 : Weight (% change)
  • mice (C57/bl6 under i/p ketamin ⁇ anae ⁇ thesia) w ⁇ re challeng ⁇ d i/v (tail v ⁇ in) with 5x10 4 B16-F10 tumour cells in a final volume of 100 ⁇ l per mouse on day 0.
  • Test compounds 50mg/kg in 200 ⁇ l sterile non-pyrogenic saline) were administer ⁇ d ⁇ /c (right flank) on days 2 and 4.
  • mice were sacrifices and the lungs dissected and stained in Indian ink solution (150ml bidistilled water, 30 ml India Ink, 4 drops NH 4 OH) for 10 minutes then fixed for at least 24 hr in Fakete's solution (90ml 37% formaldehyde, 900 ml 70% EtOH and 45ml glacial acetic acid).
  • Indian ink solution 150ml bidistilled water, 30 ml India Ink, 4 drops NH 4 OH
  • Fakete's solution 90ml 37% formaldehyde, 900 ml 70% EtOH and 45ml glacial acetic acid.
  • MCF-7 cells European Collection of Cell Cultures Ref. 86012803 were taken from liquid nitrogen stock, thawed at room temperature and transferred to 10mi Dulbeccos Modified Eagle's Medium with Hams F I2, 15mM Hepes and L-glutamine (DMEM: Cambrex Cat. Mo. BE12-719F) supplemented with 10% v/v foetal calf serum (FCS: BioWest Labs Cat. No. S02755, Lot. No. SI 800). The FCS was pre-filtered through a 0.2 ⁇ m ⁇ teril filter.
  • the cells were then centrifuged at 1 ,500 rpm in a Centaur bench-top centrifuge and the supernatant removed.
  • the cells were reconstituted in fresh media and seeded into two T75cm 3 Nunclon tissue culture flasks and allowed to settle overnight al 37°C in a 5% CO 2 incubator.
  • the flasks were wrapped in cling film to prevent cross-contamination and the following day the media was changed to include the antibiotics penicillin and streptomycin as a precautionary measure again ⁇ t infection (at concentrations of 1 mg/cm 3 and 5mg/cm 3 , r ⁇ spectively).
  • the cells were ⁇ allow ⁇ d to grow near confluence and then split at a 1 in 4.resuspen ⁇ ion.
  • the cells used for the experim ⁇ nts were of passage number 31. Two flasks of cells were ⁇ pr ⁇ pared in media containing 20% v/v FCS with 10% dimethylsulphoxide and bank ⁇ d down into liquid nitrog ⁇ n for later use if necessary.
  • the cells were harvest ⁇ d using a non-enzymatic method. At each of the time points the cells were viewed under the invert ⁇ d light microscope and the morphology evaluated. Before harvesting, the cells were washed with sterile PBS, theree times, 7cm 3 per wash. The cells were then scraped from the flasks using a sterile cell scraper and transferred to Grenier tubes. The cells were quickly passed through a 21 G2 gauge needle to disaggregate the cells. Cells were then pell ⁇ t ⁇ d by centrifugation at 1500g/5min and resusp ⁇ nded in a known volume of PBS.
  • the number of cells was then counted in a haemocytometer and cell viability evaluated by mixing 0.1 cm 3 of each cell suspension with a drop of trypan blue solution. Each of the cell pellets was frozen at -80°C until glycan rel ⁇ ase and analysis.
  • the cell pellets were placed in an iced water bath and allowed to thaw.
  • the pellets were ' then homogenized in a total of 4cm 3 (made up to volume with deionized water).
  • An Ultraturrax T25 homogeniser was used for this purpose, with the blade spe ⁇ d set to 22,500 rpm.
  • the samples were maintained on ice and 3 bursts, each of 10s ⁇ c, were applied with, a period of approximately 1 in between each homogenisation step to allow the froth the settle.
  • the blade was washed carefully between each of the samples to prevent sample cross- contamination.
  • the homogenates were stored in 1cm 3 aliquots at -80°C prior to the protein assay and glycan release.
  • Aft ⁇ r the incubation step, the sample ⁇ were loaded onto prewa ⁇ h ⁇ d and prim ⁇ d Ludg ⁇ r Clean E cartridge ⁇ (Cat. No. LC-E10-A6).
  • the glycan ⁇ were eluted according to th ⁇ manufacturer' ⁇ in ⁇ tructions and dried by centrifugal evaporation overnight.
  • the glycans were labelled by reductive amination, for 2 hours at 65°C, according to the method described by Bigge ef al. (1995) Anal. Biochem. 230(2): 229-238.
  • the incubation mixture was then "cleaned up” to remove any uneonjugated fluorophor ⁇ by spotting th ⁇ samples onto Whatman 3MM paper and running in a descending chr ⁇ rriatography tank with a mobile phase of 4:1 :1 butanol:ethanol:water overnight. Glycans were then eluted
  • the glycans were ⁇ eparated on a normal phase (hydrophilic interaction) HPLC column (LudgerSep N1 amide)
  • the buffer sy ⁇ tem u ⁇ ed was the high salt sy ⁇ tem, with acetonitril ⁇ as buff ⁇ r A and 0.25M ammonium format ⁇ pH4.4 as buffer B.
  • Flow rate wa ⁇ maintained at Q.3cm 3 /min throughout.
  • Example 10 Increasing the Th1 :Th2 response ratio in a non-healing leishmaniasis model
  • Leishmaniasis is a classic model of a Th l disease: non-healing cutaneous lesions arise from an undesirable polarization of the immune response Which becomes heavily Th2-skewed.
  • Th I :Th2 response ratio In order to study the ability of the compounds of the invention to increase the Th I :Th2 response ratio in this disease model (and so promote a healing Th1 response), spleen cells from Leishmania major infected BALB/c mice having a non-healing cutaneous infection were stimulated with parasite antigen (Table 10.1) or polyclonally with anti-CD3 (Table 10.2) in the pres ⁇ nce of 3,7-diep/-casuarine (10).
  • Table 10.1 Reversal of the inability of T-cells to produce IFN- ⁇ in a non-healing mouse model
  • Amberlite IR- 120, strongly acidic ion- ⁇ change resin was prepar ⁇ d by soaking the resin in 2M hydrochloric acid for at least two hours followed by elution with distilled water until the eluant reached pH 5.
  • Dowev 50WX8-100 was prepared by soaking the resin with 2M hydrochloric acid for at least two hours followed by elution with distilled water until neutral.
  • Infrared spectra were recorded on a Perkin-Elmer 1750 IR Fourier Transform ⁇ pectrophotometer using thin films on sodium chloride plates. Only characteristic peaks are recorded.
  • Optical rotations were measured on a Perkin-Elm ⁇ r 241 polarim ⁇ t ⁇ r with a path length of 1dm.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Organic Chemistry (AREA)
  • Natural Medicines & Medicinal Plants (AREA)
  • Epidemiology (AREA)
  • Engineering & Computer Science (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Biotechnology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Immunology (AREA)
  • Hematology (AREA)
  • Molecular Biology (AREA)
  • Mycology (AREA)
  • Alternative & Traditional Medicine (AREA)
  • Microbiology (AREA)
  • Botany (AREA)
  • Medical Informatics (AREA)
  • Virology (AREA)
  • Biochemistry (AREA)
  • Oncology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Communicable Diseases (AREA)
  • Cell Biology (AREA)
  • Zoology (AREA)
  • Developmental Biology & Embryology (AREA)
  • Biomedical Technology (AREA)
  • Genetics & Genomics (AREA)
  • Oil, Petroleum & Natural Gas (AREA)
  • Diabetes (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)

Abstract

Isolated immunomodulatory (e.g. immunostimulatory) polyhydroxlated pyrrolizidine compounds having the formula (I), wherein R is selected from the group comprising hydrogen, straight or branched, unsubstituted or substituted, saturated or unsaturated acyl, alkyl (e.g. cycloalkyl), alkenyl, alkynyl and aryl groups, or a pharmaceutically acceptable salt or derivative thereof, are useful in therapy and prophylaxis, including increasing the Th1:Th2 response ratio, haemorestoration, alleviation of immunosuppression, cytokine stimulation, treatment of proliferative disorders (e.g. cancer), vaccination, stimulation of the innate immune response and boosting of the activity of endogenous NK cells.

Description

IMMUNOMODULATORY COMPOSITIONS
Field of the Invention
The present invention relates to immunomodulatory polyhydroxylated pyrrolizidine compounds and to their use in medicine In particular, the invention relates to the use of casuaπne and certain casuaπne analogues as immunomodulatory (immunostimulatory or immunosuppressive) drugs
Background to the Invention
Immunity
When the immune system is challenged by a foreign antigen it responds by launching a protective response This response is characterized by the coordinated interaction of both the innate and acquired immune systems These systems, once thought to be separate and independent, are now recognized as two interdependent parts that when integrated fulfil two mutually exclusive requirements speed (contributed by the innate system) and specificity (contributed by the adaptive system)
The innate immune system serves as the first line of defence against invading pathogens, holding the pathogen in check while the adaptive responses are matured It is triggered within minutes of infection in an antigen- independent fashion, responding to broadly conserved patterns in the pathogens (though it is not non-specific, and can distinguish between self and pathogens) Crucially, it also generates the inflammatory and co- stimulatory milieu (sometimes referred to as the danger signal) that potentiates the adaptive immune system and steers (or polarizes it) towards the cellular or humoral responses most appropriate for combating the infectious agent (discussed in more detail below)
The adaptive response becomes effective over days or weeks, but ultimately provides the fine antigenic specificity required for complete elimination of the pathogen and the generation of immunologic memory It is mediated principally by T and B cells that have undergone germline gene rearrangement and are characterized by an exquisite specificity and long-lasting memory However, it also involves the recruitment of elements of the innate immune system, including professional phagocytes (macrophages, neutrophils etc ) and granulocytes (basophils, eosinophils etc ) that engulf bacteria and even relatively large protozoal parasites Once an adaptive immune iβsponse has matured, subsequent exposure to the pathogen l esults in its rapid elimination (usually before symptoms of infection become manifest) because highly specific memory cells have been generated that are rapidly activated upon subsequent exposure to their cognate antigen
Interdependence of innate and adaptive responses
It is now thought that the earliest events following pathogen invasion are effected by cellular components of the innate immune system The response is initiated when resident tissue macrophages and dendritic cells (DCs) encounter pathogen and become activated by signals generated by interaction between pattern-recognition receptors (PRRs) and the pathogen-associated molecular patterns (PAMPs) shared by large groups of microorganisms The activated macrophages and DCs are stimulated to release various cytokines (including the chemokines IL-8, MIP-1α and MIP-1 β), which constitute the "danger signal" and triggers an influx of Natural Killer (NK) cells, macrophages, immature dendritic cells into the tissues
Loaded with antigen, the activated DCs then migrate to lymph noαes υnce mere, tney activate immune cent, of the adaptive response (principally naive B- and T-cells) by acting as antigen-presenting cells (APCs) The activated cells then migrate to the sites of infection (guided by the "danger signal") and once there further amplify the response by recruiting cells of the innate immune system (including eosinophils, basophils, monocytes, NK cells and granulocytes) This cellular trafficking is orchestrated by a large array of cytokines (particularly those of the chemokme subgroup) and involves immune cells of many different types and tissue sources (for a review, see Luster (2002), Current Opinion in Immunology 14 129-135)
Polarization of the adaptive immune response
The adaptive immune response is principally effected via two independent limbs cell-mediated (type 1) immunity and antibody-mediated or humoral (type 2) immunity
Type 1 immunity involves the activation of T-lymphocytes that either act upon infected cells bearing foreign antigens or stimulate other cells to act upon infected cells This bi anch of the immune system therefore effectively contains and kills cells that are cancerous or infected with pathogens (particularly viruses) Type 2 immunity involves the generation of antibodies to foreign antigens by B-lymphocytes This antibody-mediated branch of the immune system attacks and effectively neutralizes extracellular foreign antigens
Both limbs of the immune system are important in fighting disease and there is an increasing realization that the type of immune response is just as important as its intensity or its duration Moreover, since the type 1 and type 2 responses are not necessarily mutually exclusive (in many circumstances an effective immune response requires that both occur in parallel), the balance of the typel/type 2 response (also referred to as the Th1 Th2 response ratio/balance by reference to the distinct cytokine and effector cell subsets involved in the regulation of each response - see below) may also play a role in determining the effectiveness (and repercussions) of the immune defence
In many circumstances the immune response is skewed heavily towards a type 1 or type 2 response soon after exposure to antigen The mechanism of this type1/type 2 skewing or polarization is not yet fully understood, but is known to involve a complex system of cell-mediated chemical messengers (cytokines, and particularly chemokines) in which the typel/type 2 polarization (or balance) is determined, at least in part, by the nature of the initial PRR-PAMP interaction when the DCs and macrophages of the innate immune system are first stimulated and subsequently by the cytokine milieu in which antigen priming of naive helper T cells occuis
Two cytokines in particular appear to have early roles in determining the path of the immune response lnterleukιn-12 (IL-12), secreted by macrophages, drives the type 1 response by stimulating the differentiation of Th1 cells, the helper cells that oversee the type 1 response Another macrophage cytokine, ιnterleukιn- 0 (IL- 10) inhibits this response, instead driving a type 2 response The type 1 and type 2 responses can be distinguished inter alia on the basis of certain phenotypic changes attendant on priming and subsequent polarization of naive helper T cells These phenotypic changes are characterized, at least in part, by the nature of the cytokines secreted by the polarized helper T cells
Th1 cells produce so-called Th1 cytokines, which include one or more of TNF, IL-1 , IL-2, IFN-gamma, IL-12 and/or IL-18 The Th1 cytokines are involved in macrophage activation and Th1 cells orchestrate Type 1 responses In contrast, Th2 cells produce so-called Th2 cytokines, which include one or more of IL-4, I -5, IL- 10 and IL-13 The Th2 cytokines promote the production of various antibodies and can suppress the type 1 response
The involvement of Th1 and Th2 cells and cytokines in type 1 type 2 immune response polarization has given rise to the terms Th1 response and Th2 response being used to define the type 1 and type 2 immune responses, respectively Thus, these terms are used interchangeably herein
There is an increasing realization that the type of immune response is just as important in therapy and prophylaxis as its intensity or its duration For example, an excess Th1 response can result in autoimmune disease, inappropriate inflammatory responses and transplant rejection An excess Th2 response can lead to allergies and asthma Moreover, a perturbation in the Th1 Th2 ratio is symptomatic of many immunological diseases and disorders, and the development of methods for altering the Th1 Th2 ratio is now a priority
Alkaloids
' The term alkaloid is used herein sensu stricto to define any basic, organic, nitrogenous compound which occurs naturally in an organism The term alkaloid is also used herein sensu lato to define a broader grouping of compounds which include not only the naturally occurring alkaloids, but also their synthetic and semi-synthetic analogues and derivatives
Most known alkaloids are phytochemicals, present as secondary metabolites in plant tissues (where they may play a role in defence), but some occur as secondary metabolites in the tissues of animals, microorganisms and fungi There is growing evidence that the standard techniques for screening microbial cultures are inappropriate for detecting many classes of alkaloids (particularly highly polar alkaloids, see below) and that microbes (including bacteria and fungi, particularly the f'lamentous representatives) will prove to be an important source of alkaloids as screening techniques become more sophisticated
Structurally all aloids exhibit great diversity Many alkaloids are small molecules, with molecular weights below 250 Daltons The skeletons may be derived from ammo acids, though some are derived from olher groups (such as steroids) Olhers can be consideied as sugar analogues It is becoming apparent (see Watson et al (2001 ) Phytochemistry 56 265-295) that the water soluble fractions of medicinal plants and microbial cultures contain many interesting novel polar alkaloids, including many carbohydrate analogues Such analogues include a rapidly growing number of polyhydroxylated alkaloids
Most alkaloids are classified structurally on the basis of the configuration of the N-heterocycle Examples of some important alkaloids and their structures are set out in Kutchan (1995) The Plant Cell 7 1059-1070 Watson et al (2001 ) Phytochemistry 56 265-295 have classified a comprehensive range of polyhydroxylated alkaloids inter alia as pipeπdine, pyrroline, pyrrolidme pyrrolizidine, indohzidine and nortropanes alkaloids (see Figs 1-7 of Watson et al (2001 ), the disclosure of which is incorporated herein by reference)
Watson ef al (2001 ), ibidem also snow that a functional classification of at least some alkaloids is possible on the basis of their glycosidase inhibitory profile many polyhydroxylated alkaloids are potent and highly selective glycosidase inhibitors These alkaloids can mimic the number, position and configuration of hydroxyl groups present in pyranosyl or furanosyl moieties and so bind to the active site of a cognate glycosidase, thereby inhibiting it This area is reviewed in Legler (1990) Adv Carbohydr Chem Biochem 48 319-384 and in Asano et al (1995) J Med Chem 38 2349-2356
It has long been recognized that many alkaloids are pharmacologically active, and humans have been using alkaloids (typically in the form of plant extracts) as poisons, narcotics stimulants and medicines for thousands of years The therapeutic applications of polyhydroxylated alkaloids have been comprehensively reviewed in Watson et al (2001 ), ibidem applications include cancer therapy immune stimulation, the treatment of diabetes, the ti eatment of infections (especially viral infections) therapy of glycosphmgolipid lysosomal storage diseases and the treatment of autoimmune disorders (such as arthi itis and sclerosis)
Both natural and synthetic mono- and bi-cyclic nitrogen analogues of carbohydrates are known to have potential as chemotherapeutic agents Alexine (1 ) and australine (2) vvere the first pyrrolizidine alkaloids to be isolated with a carbon substituent at C-3, rather than the more common C-1 substituents characteristic of the necme family of pyrro zidines
Figure imgf000005_0001
CHoOH
Alexine (1)
Figure imgf000005_0002
Australine (2)
The alexmes occur in all species of the genus Alexa and also in the related species Castanospermum australe Stereoiso ers of alexine, including 1 ,7a-dιepιalexιne (3) have also been isolated (Nash et al (1990) Phytochemistry (29) 111 ) and synthesised (Choi et al. (1991 ) Tetrahedron Letters (32) 5517 and Denmark and Cottell (2001 ) J. Org. Chem. (66) 4276-4284).
Figure imgf000006_0001
1, 7a-diepialexine (3)
Because of the reported weak /n vitro antiviral properties of one 7,7a-diepialexine (subsequently defined as 1 ,7a-diepialexine), there has been some interest in the isolation of the natural products and the synthesis of analogues.
As an indolizidine alkaloid (and- so structurally distinct from the pyrrolizidine alexines), swainsonine (4) is a potent and specific inhibitor of α-mannosidase and is reported to have potential as an antimetastic, tumour anti- proliferative and immuπoregulatory agent (see e.g. US5650413, WO00/37465, WO93/09117).
Figure imgf000006_0002
Swainsonine (4)
The effect of variation in the size of the six-membered ring of swainsonine on its glycosidase inhibitory activity has been studied: pyrrolizidine derivatives (so-called "ring contracted swainsonines") have been synthesised. However, these synthetic derivatives (1S, 2R, 7R, 7aR)-1 ,2,7-trihydroxypyrroli∑idine (5) and the 7S-epimer (6)) were shown to have much weaker inhibitory activity relative to swainsonine itself (see US5075457).
Figure imgf000006_0003
1S, 2R, 7R, 7aR)-1, 2 , 7-trihydrox 'pyrrolizidine (5)
Figure imgf000007_0001
7S-epιmer (6)
Another compound, 1 α,2α,6α,7α,7αβ-1 ,2,6,7-tetrahydroxypyrrolιzιdιne (7) is an analogue of 1 , diepiswainsonine and described as a ' useful" inhibitor of glycosidase enzymes in EP0417059
Figure imgf000007_0002
1 a, 2a, 6a, 7 a, 7aβ- 1,2,6, 7-tetrahydroxypyι rolizidine (7)
Casuarine, (I R^R.SR.δS S aRJ-S^hydroxymethy -l ^.e^-tetrahydroxypyrrolizidine (8) is a highly oxygenated bicyc c pyrrolizidine alkaloid that can be regarded as a more highly oxygenated analogue of the 1 ,7a-dιepιalexιne (shown in 3) or as a C(3) hydroxymethyl-substituted analogue of the 1α,2α,6α,7αJαβ-1 , 2,6,7- tetrahydroxypyn o zidine (shown in 7)
Figure imgf000007_0003
CH OH
Casuarine ( B)
Casuarine can be isolated from sevei al botanical sources, including the bark of Casuanna equiεetifolia (Casuannaceae), the leaves and bark of Eugenia jambolana (Myrtaceae) and Syzygium guineense (Myrtaceae) (see e g Nash et al (1994) Tetrahedron Letters (35) 7849-7852) Epimers of casuarine, and probably casuarine itself, can be synthesised by sodium hydrogen telluride-induced cyclisation of azidodimesylates (Bell er a/ (1997) Tetrahedron Letters (38) 5869-5872) Casuanna equisetifolia wood, bark and leaves have been claimed to be useful against diarrhoea, dysentery and colic (Chopra ef al (1956) Glossary of Indian Medicinal Plants, Council of Scientific and Industrial Research (India), New Delhi, p 55) and a sample of bark has recently been prescribed in Western Samoa for the treatment of breast cancer An African plant containing casuarine (identified as Syzygium guineense) has been reported to be beneficial in the treatment of AIDS patients (see Wormald et al (1996) Carbohydrate Letters (2) 169-174)
The casuarιne-6-α-glucosιde (casuarιne-6-α-D-glucopyranose 9) has also been isolated from the bark and leaves of Eugenia jambolana (Wormald ef al (1996) Carbohydrate Letters (2) 169-174)
Figure imgf000008_0001
Casuarιne-6-α-D-glucopyranose (9)
Eugenia jambolana is a well known tree in India for the therapeutic value of its seeds, leaves and fruit against diabetes and bacterial infections Its fruit have been shown to reduce blood sugar levels in humans and aqueous extracts of the bark are claimed to affect glycogenolysis and glycogen storage in animals (Wormald et al (1996) Carbohydrate Letters (2) 169-174)
Dendritic Cells and their Immunotherapeutic Uses
(a) Introduction
Dendritic cells (DCs) are a heterogeneous cell population with distinctive morphology and a widespread tissue distribution (see Steinman (1991 ) Ann Rev Immunol 9 271-296) They play an important role in antigen presentation, capturing and piocessmg antigens into peptides and then presenting them (together with components of the MHC ) to T cells T cell adiva on may then be mediated by the expression of important cell surface molecules, such as high levels of MHC class I and II molecules, adhesion molecules, and costimulalory molecules
Dendritic cells therefoi e act as highly specialized antigen-presenting cells (APCs) serving as "nature's adjuvants' , they potentiate adaptive T-cell dependent immunity as well as triggering the natural killer (NK and NKT) cells of the innate immune system Dendritic cells therefore play a fundamental and important regulatory role in the magnitude, quality and memory of the immune response As a result, there is now a growing interest in the use of dendritic cells in various immunomodulatory interventions, which are described in more detail below Dendritic cells can be classified into different subsets inter alia on the basis of their state of maturation (mature or immature) and their cellular developmental origin (ontogeny) Each of these subsets appear to play distinct roles in vivo, as described below
(b) Dendritic Cell Maturation
Immature (or resting) DCs are located in non-lymphoid tissue, such as the skin and mucosae, are highly phagocytic and readily internalize soluble and particulate antigens It is only when such antigen-loaded immature DCs are also subject to inflammatory stimuli (referred to as maturation stimuli) that they undergo a maturation process that transforms them from phagocytic and migratory cells into non-phagocytic, highly efficient stimulators of naive T cells
Immature DCs are characterized by high intracellular MHC II in the form of MIICs, the expression of CD1a, active endocytosis for certain particulates and proteins, presence of FcgR and active phagocytosis, deficient T cell sensitization in vi o, low/absent adhesive and costimulatory molecules (CD40/54/58/80/86), low/absent CD25, CD83, p55, DEC-205, 2A1 antigen, responsiveness to GM-CSF, but not M-CSF and G-CSF and a sensitivity to IL-10, which inhibits maturation
Upon maturation, mature DCs, loaded with antigen and capable of priming T cells, migrate from the non- lymphoid tissues to the lymph nodes or spleen, where they process the antigen load and present it to the resident naive CD4+ T cells and CD8+ cytotoxic T cells This latter interaction generates CTLs, the cellular arm of the adaptive immune response, and these cells eliminate virally infected cells and tumour cells The naive CD4+ T cells differentiate into memory helper T cells, which support the differentiation and expansion of CD8+ CTLs and B cells Thus, helper T cells exert anti-tumour activity indirectly through the activation of important effector cells such as macrophages and CTLs
Having activated the T cells in this way, the mature DCs undergo apoptosis within 9-10 days
Mature DC cells are characterized morphologically by motility and the presence of numerous processes (veils or dendπtes) They are competent for antigen capture and presentation (exhibiting high MHC class I and II expression) and express a wide range of molecules involved in T cell binding and costimulation, (e g CD40, CD54/ICAM-1 , CD58/LFA-3, CD80/B7-1 and CD86/B7-2) as well as various cytokines (including IL-12) They are phenotypically stable there is no reversion/conversion to macrophages or lymphocytes
Thus, mature DCs play an important role in T cell activation and cell-mediated immunity In contrast, immature DCs are involved in regulating and maintaining immunological tolerance (inducing antigen-specific T cell anergy)
(c) Dendritic Cell Ontoqenic Subsets
Dendritic cells are not represented by a single ceil type, but rather comprise a heterogeneous collection of different classes of cells, each with a distinct ontogeny At least three different developmental pathways have been described, each emerging from unique progenitors and driven by particular cytokine combinations to DC subsets with distinct and specialized functions.
At present it is thought that the earliest DC progenitors/precursors common to all DCs originate in the bone marrow. These primitive progenitors are CD34'r, and they are released from the bone marrow to circulate through both the blood and lymphoid organs.
Once released from the bone marrow, the primitive CD34+ DC progenitors are subject to various stimulatory signals. These signals can direct the progenitors along one of at least three different pathways, each differing with respect to intermediate stages, cytokine requirements, surface marker expression and biological function.
* Lymphoid DCs are a distinct subset of DCs that are closely linked to the lymphocyte lineage. This lineage is characterized by the lack of the surface antigens CD 1b, CD13, CD14 and CD33. Lymphoid DCs share ancestry with T and natural killer (NK) cells, the progenitors for all being located in the thymus and in the T cell areas of secondary lymphoid tissues. The differentiation of lymphoid DCs is driven by interleukins 2, 3 and 15 (IL-3, IL-2 and IL-15), but not by granulocyte macrophage colony- stimulating factor (GM-CSF). Functionally, lymphoid promote negative selection in the thymus (possibly by inducing fas-mediated apoptosis) and are costimulatory for CD4+ and CD8+T cells. More recently, lymphoid-like DCs derived from human progenitors have also been shown to preferentially activate the Th2 response. Because of their capacity to induce apoptosis and their role in eliminating potentially self-reactive T cells, it has been suggested that lymphoid DCs primarily mediate regulatory rather than stimulatory immune effector functions.
• Myeloid DCs are distinguished by a development stage in which there is expression of certain features associated with phagocytes. There appear to be at least two structurally and functionally distinct subsets. The first is defined antigenically as CD14", CD34+, CD68" and CD1a+ and sometimes referred to as DCs of the Langerhans cell type. This subset appears to prime T cells to preferentially activate Th1 responses and IL-12 appears implicated in this process. The subset may also activate naϊve B cells to secrete IgM and may therefore be predominantly associated with an inflammatory Th1 . response. A second myeloid DC subset, sometimes referred to as interstitial DCs, is defined antigenically as CD14+, CD68+ and CD1 a" and related to monocytes (as a result they are also referred to as monocyte-derived DCs or Mo-DCs).
(d) Dendritic Cell Vaccines
In one dendritic cell-based treatment paradigm (reviewed in Schuler ef al. (2003) Current Opinion in Immunol 15: 138-147), DC cells are taken from a patient (for example by apheresis) and then pulsed (primed or spiked) with a particular antigen or antigens (for example, tumour antigen(s)). They are then re-administered as an autologous cellular vaccine to potentiate an appropriate immune response.
In this treatment paradigm, the responding T cells include helper cells, especially Th1 CD4+ cells (which produce IFN-γ) and killer cells (especially CD8+ cytolytic T lymphocytes). The DCs may also mediate responses by other classes of lymphocytes (B, NK, and NKT cells) They may also elicit T cell memory, a critical goal of vaccination
At present little is known about the identity of the DC subset(s) required for optimum effectiveness of DC vaccines, beyond the recognition that maturation is required and immature DCs are to be avoided (Dhodapkar and Steinman (2002) Blood 100 174-177)
Hsu ef al (1996) Nat Med 2 52-58 used rare DCs isolated ex vivo from blood These DCs were highly heterogeneous with respect to their ontogenic subsets but matured spontaneously during the isolation procedure However, the yields were very low
The yield problem has been addressed by the development of techniques for expanding the DCs ex vivo, for example with Flt3 ligand (Fong et al (2001 ) PNAS 98 8809-8814), but this is of limited effectiveness
However most studies have used Mo-DCs These cells are obtained by exposing monocytes to GM-CSF and IL-4 (or IL-13) to produce immature Mo-DCs, which are then matured by incubation in a maturation medium Such media comprise one or more maturation stimulation factor(s), and typically comprise Toll-like receptor (TLR) ligands (e g microbial products such as lipopolysacchaπde and/or monophosphoryl pid), inflammatory cytokines (such as TNF-α), CD40L, monocyte conditioned medium (MCM) or MCM mimic (which contains IL- I β, TNF- α, IL-6 and PGE2)
Although little is known at present about the influence of maturation medium on DC vaccine performance, MCM or MCM mimic currently represent a standard Mo-DCs matured using these media are homogenous, have a high viability, migrate well to chemotactic stimuli and induce CTLs both in vitro and in vivo
Techniques have been developed for generating large numbers of Mo-DCs (300 to 500 million mature DCs per apheresis) from adherent monocytes within semi-closed, multilayered communicating culture vessels offering a surface area large enough to cultivate one leukapheresis pioduct These so-called cell factories can be used to produce cryopreserved ahquots of antigen preloaded DCs which are highly viable on thawing, and optimised maturation and freezing procedures have been described (Berger et al (2002) J Immunol Melhods 268 131- 140 Tuyaerts et al (2002) J Immunol Methods 264 135-151 )
Dendritic cells for vaccination have also been prepared from CD34+-derιved DCs comprising a mixture of interstitial and DC s of the Langerhans cell type Some workeis believe that the latter DC subsel are moie potent than Mo-DCs when used as DC vaccines
With legard to antigen selection, various approaches have been used Both defined and undefined antigens can be employed The antigens can be xenoantigens or autoantigens One or moi e defined neoantιgen(s) may be selected in the case of cancer treatment, the enoantιgen(s) may comprise a tumour-associated antigen However, most popular are 9-11 ammo acid peptides containing defined antigens (either natural sequences or analogues designed for enhanced MHC binding) such antigens can be manufactured to good manufactunng practice (GMP) standard and are easily standardized Other approaches have employed antigens as immune complexes, which are delivered to Fc-receptor-bearing DCs and which results in the formation of both MHC class I and MHC class II peptide sequences. This offers the potential for inducing both CTLs and Th cells (Berlyn ef al. (2001) Clin Immunol 101 : 276-283).
Methods have also been developed for exploring the whole antigenic repertoire of any given tumour (or other target cell, such as a virally-infected cell). For example, DC-tumour cell hybrids have been successfully used to treat renal cell carcinoma (Kugler et al. (2000) 6: 332-336), but the hybrids are difficult to standardize and shortlived. Necrotic or apoptotic tumour cells have been used, as have various cellular lysates.
It appears that the selection of patient-specific antigens may be important in the treatment of at least some cancers, and antigens derived from fresh tumour cells rather than tumour cell lines or defined antigens may prove important (Dhodapkar et al. (2002) PNAS 99: 13009-13013).
As regards delivery of the selected antigen(s) to the DCs, various techniques are available. Since the number and quality of MHC-peptide complexes directly influences the immunogenicity of the DC, the antigen loading technique may prove critical to DC vaccine performance (van der Burg et al. (1996) J Immunol 156: 3308- 3314) It seems that prolonged presentation of MHC-peptide complexes by the DCs enhances immunogenicity and so loading techniques which promote prolonged presentation may be important. This has been achieved by loading the DCs internally through the use of peptides linked to cell-penetrating moieties (Wang and Wang (2002) Nat Biotechnol 20: 149-154).
Antigens can also be loaded by transfecting the DCs with encoding nucleic acid (e.g. by electroporation) such that the antigens are expressed by the DC, processed and presented at the cell surface. This approach avoids the need for expensive GMP proteins and antibodies. RNA is preferred for this purpose, since it produces only transient expression (albeit sufficient for antigen processing) and avoids the potential problems associated with the integration of DNA and attendant long-term expreεsion/mutagenesis. Such transfection techniques also permit exploration of the whole antigenic repertoire of a target cell by use of^fptal or PCR-amplified tumour RNA.
There is some evidence that helper proteins (for example, keyhole limpet hemocyanin (KLH) and tetanus toxoid (TT)) can provide unspecific help for CTL induction (Lanzavecchia (1998) Nature 393: 413-414) and it may prove advantageous to pulse DC with such helper proteins prior to vaccination.
With regard to posology, the dose, frequency and route of DC vaccine administration have not yel been optimised in clinical trials. Clearly, the absolute number of cells administered will depend on the route of administration and effectiveness of migration after infusion. In this respect there are indications that intradermal or subcutaneous administration may be preferred for the development of Th1 responses, although direct intranodal delivery has been employed to circumvent the need for migration from the skin to the nodes (Nestle et al. (1998) Nat Med 4: 328-332).
Quite distinct from the antigen-pulsed DC vaccine paradigm described above is an approach in which dendritic cells secreting various chemokines are injected directly into tumours where they have been shown to prime T cells extranodally (Kirk et al. (2001) Cancer Res 61 : 8794-8802). Thus, in another treatment paradigm, DCs are targeted to a tumour and activated to elicit immune responses in situ without the need for ex vivo antigen loading.
In situ DC vaccination constitutes yet another distinct (but related) approach (Hawiger et al. (2001) J Exp Med 194: 769-779. In this therapeutic paradigm, antigen is targeted to DCs in vivo which are expanded and induced to mature in situ. This approach depends on efficient targeting of antigen to endogenous DCs (for example, using exosomes - see Thery et al. (2002) Nat Rev Immunol 2: 569-579) and the development of maturation stimulants that can effectively trigger maturation (preferably of defined DC subset(s)) in vivo.
(e) Use of Dendritic Cells in Adoptive CTL Immunotherapy
Cytotoxic T lymphocytes (CTLs) can be administered to a patient in order to confer or supplement an immune response to a particular disease or infection (typically cancer). For example, tumour specific T cells can be extracted from a patient (e.g. by leukapheresis), selectively expanded (for example by tetramer-guided cloning - see Dunbar et al. (1999) J Immunol 162: 6959-6962) and then re-administered as an autologous cellular vaccine.
The clinical effectiveness, applicability and tractability of this type of passive immunotherapy can be greatly Increased by using dendritic cells to prime the T cells in vitro prior to administration.
(f) Dendritic Cell-based Approaches to the Treatment of Autoimmune Disorders
Dendritic cells are also involved in regulating and maintaining immunological tolerance: in the absence of maturation, the cells induce antigen-specific silencing or tolerance.
Thus, in another dendritic cell-based treatment paradigm, immature DCs are administered as part of an immunomodulatory intervention designed to combat autoimmune disorders. In such applications, the suppressive potential of the DCs has been enhanced by in vitro transfection with genes encoding cytokines.
(g) The Role of IL-2 in Dendritic Ceil Function
Granucci et al. (2002) Trends in Immunol. 23: 169-171 have reported transient upregulation of mRNA transcripts for IL-2 in dendritic cells following microbial stimulus. In WO03012078 Granucci describes the important role played by DC-derived IL-2 in mediating not only T cell activation but also that of NK cells and goes on to suggest that DC-derived IL-2 is a key factor regulating and linking innate and adaptive immunity.
Moreover, systemic, administration of IL-2 has recently been shown to enhance the therapeutic efficacy of a DC- vaccine (Shimizu ef al. (1999) PNAS 96: 2268-2273), while the presence of IL-2 was shown to be essential for specific peptide-mediated immunity mediated by dendritic, cells in at least some DC vaccination regimes (Eggert et al. (2002) Eur J Immunol 32: 122-127). In their recent review, Schuler ef al. (ibidem) conclude that "... it might be worthwhile to explore the combination of DC vaccination with IL-2 administration, as the T-cell responses induced by DC vaccination appear enhanced and therapeutically more effective.". It will be clear from the foregoing discussion that dendritic cells are now proven as valuable tools in immunotherapy (particularly in the treatment of cancer), but that DC vaccination is still at a relatively early stage Methods for preparing DCs are improving continuously and an increasing number of Phase I, II and III clinical trials are driving intense research and development in this area However, there is still a need to improve efficacy at the level ot DC Piology
The present inventors have now surprisingly discovered that casuarine and certain casuarine analogues have unexpected immunomodulatory activity, and that this activity may not be dependent on glycosidase inhibition
Summary of the Invention
According to the invention there is provided an isolated immunomodulatory (e g immunostimulatory) polyhydroxylated pyrrolizidine compound for use in therapy or prophylaxis having the formula
Figure imgf000014_0001
wherein R is selected from the group comprising hydrogen, straight or branched, unsubstituted or substituted, saturated or unsaturated acyl, alkyl (e g cycloalkyl), alkenyl, alkynyl and aryl groups, or a pharmaceutically acceptable salt or derivative thereof
Preferably, the compounds of the invention are alkaloids (as hereinbefore defined)
The compound of the invention preferably has the formula
Figure imgf000014_0002
CH2OH wherein R is selected from the gioup comprising hydrogen, straight or branched, unsubstituted or substituted, saturated or unsaturated acyl, alkyl (e g cycloalkyl), alkenyl, alkynyl and aryl groups, or a pharmaceutically acceptable salt or derivative thereof rv
Particularly preferred is 1 R 2R,3R,6S,7S,7aR)-3-(hydroxymethyl)-1 2,6,7-tetrahydroxypyrrolιzιdιne (casuarine), wherein R is hydrogen and which has the formula
Figure imgf000015_0001
or a pharmaceutically acceptable derivative or salt thereof.
Particularly preferred is a casuarine glucoside, or a pharmaceutically acceptable salt or derivative thereof.
Other preferred compounds include 6-O-butanoylcasuarine of the formula:
Figure imgf000015_0002
CH2OH
or a pharmaceutically acceptable salt or derivative thereof.
A particularly preferred casuarine glucoside is casuarine-6-α-D-glucoside of the formula:
Figure imgf000015_0003
or a pharmaceutically acceptable salt or derivative thereof.
As mentioned infra, the invention contemplates diastereomers of the compounds of the invention. Particularly preferred are diastereomers selected from 3,7-d/ep/-casuarine (10), 7-ep/-casuarine (11), 3,6,7-.r/ep/-casuarine (12), 6,7-d/ep/-casuarine (13) and 3-ep/-casuarine (14), as well as pharmaceutically acceptable salts and derivatives thereof.
Figure imgf000016_0001
3,7-diepi-casuarine (10)
Figure imgf000016_0002
CH OH
7-epicasuarine (11)
Figure imgf000016_0003
CH2OH
3,6,7-triepi-casuarine (11
6,7-diepi-casuarine (13)
Figure imgf000017_0001
3-epi-casuarine (14)
Other preferred diastereomers are selected from 3,7-d/ep/-casuarine-6-α-D-glucoside (15), 7-ep/-casuarine-6-α- D-glucoside (16), 3,6,7-fr/ep/-casuarine-6-α-D-glucoside (17), 6,7-d/ep/-casuarine-6-α-D-glucoside (18) and 3- ep/-casuarine-6-α-D-glucoside (19), as well as pharmaceutically acceptable salts and derivatives thereof.
Figure imgf000017_0002
3, /-diepi-casuarine-6-α-D-glucoside (15)
Figure imgf000017_0003
7-epi-casuarine-6-α-D-glucoside (16)
Figure imgf000017_0004
3, 6, 7-triepi-casuarine-6-Q-D-glucoside (17)
Figure imgf000018_0001
6, 7-diepi-casuarine-6-a-D-glucoside (18)
Figure imgf000018_0002
3-epi-casuaπne-6-a-D-glucoside (19)
Other preferred diastereomers include 7a epimers selected from 3,7,7a-tr/ep/-casuarine, 7,7a-diep/-caεuarine, 3,6,7,7a-fefraep/-casuarine, 6,7,7a-triep/-casuarine and 3,7a-diep/-casuarine, as well as pharmaceutically acceptable salts and derivatives thereof.
In another aspect the invention provides a method for immunomodulation (e.g. immunostimulation) comprising administering to a patient a composition comprising a polyhydroxylated pyrrolizidine compound having the formula:
Figure imgf000018_0003
CH2UH wherein R is selected from the group comprising hydrogen, straight or branched, unsubstituted or substituted, saturated or unsaturated acyl, alkyl (e.g. cycloalkyl), alkenyl, alkynyl and aryl groups, or a pharmaceutically acceptable salt or derivative thereof
The immunostimulatory methods of the invention are described in more detail infra.
In another aspect, the invention provides a method for chemoprotection comprising administering the compound of the invention to a patient undergoing chemotherapy. The invention also contemplates the use of the polyhydroxylated pyrrolizidine compound of the invention for the manufacture of a medicament for use in immunostimulation and/or chemoprotection, as well as a process for the manufacture of a medicament for use in immunostimulation and/or chemoprotection, characterized in the use of the polyhydroxylated pyrrolizidine compound of the invention.
In another aspect, the invention contemplates a composition comprising the polyhydroxylated pyrrolizidine compound of the invention in combination with an immunostimulant and/or cytotoxic agent (e.g. AZT) and/or an antimicrobial (e.g. antibacterial) agent and/or an antiviral agent and/or a dendritic cell (e.g. a primed dendritic cell). Such compositions preferably further comprise a pharmaceutically acceptable excipient.
In another aspect the invention contemplates a vaccine comprising the polyhydroxylated- pyrrolizidine compound of the invention in combination with an antigen, the compound being present in an amount sufficient to produce an adjuvant effect on vaccination.
In another aspect the invention contemplates a pharmaceutical kit of parts comprising the polyhydroxylated pyrrolizidine compound of the invention in combination with an immunostimulant and/or cytotoxic agent (e.g. 5' fluoro-uracil and ricin) and/or an antimicrobial (e.g. antibacterial) agent and/or an antiviral agent "(e.g. AZT). Such kits preferably further comprise instructions for use in immunotherapy.
The compounds of the invention have broad utility in therapy and prophylaxis, including treatments for increasing the Th1 :Th2 response ratio, for example in the treatment of Th1 -related diseases or disorders (e.g. proliferative disorders or microbial infection) and/or Th2-related diseases or disorders (for example allergies, e.g. asthma), as well as in haemorestoration, the alleviation of immunosuppression, in cytokine stimulation, in the treatment of proliferative disorders, vaccination (wherein the compound acts as an adjuvant), vaccination with dendritic cell vaccines (e.g. with primed dendritic cell vaccines, wherein the dendritic cells are contacted with the compound), in the administration of dendritic cells in the treatment or prophylaxis of autoimmune disorders (wherein the dendritic cells are contacted with the compound) and in wound healing. These medical uses are described in more detail below.
Detailed Description of the Invention
Definitions
Where used herein and unless specifically indicated otherwise, the following terms are intended to have the following meanings in addition to any broader (or narrower) meanings the terms might enjoy in the art:
The term adjunctive (as applied to the use of the drugs of the invention in therapy) defines uses in which the pyrrolizidine compound is administered together with one or more other drugs, interventions, regimens or treatments (such as surgery and/or irradiation). Such adjunctive therapies may comprise the concurrent, separate or sequential administration/application of the pyrrolizidine compound of the invention and the other treatment(s). Thus, in some embodiments, adjunctive use of the pyrrolizidine compound of the invention is reflected in the formulation of the pharmaceutical compositions of the invention. For example, adjunctive use may be reflected in a specific unit dosage or in formulations in which the pyrrolizidine compound of the invention is present in admixture with the other drug(s) with which it is to be used adjunctively (or else physically associated with the other drug(s) within a single unit dose) In other embodiments, adjunctive use of the pyrrolizidine compound of the invention may be reflected in the composition of the pharmaceutical kits of the invention, wherein the pyrrolizidine compound or the invention is co-pacκageα <e g as parr oτ an array oτ unit doses) with the other drug(s) with which it is to be used adjunctively In yet other embodiments, adjunctive use of the pyrrolizidine compound of the invention may be reflected in the content of the information and/or instructions co-packaged with the pyrrolizidine compound relating to formulation and/or posology
The term neoantigen is used herein to define any newly expressed antigenic determinant Neoantigens may arise upon conformational change in a protein as newly expressed determinants (especially on the surfaces of transformed or infected cells), as the result of complex formation of one or more molecules or as the result of cleavage of a molecule with a resultant display of new antigenic determinants Thus as used herein, the term neoantigen covers antigens expressed upon infection (e g viral infection, protozoal infection or bacterial infection), in pπon-mediated diseases (e g BSE and CJD), an on cell transformation (cancer), in which latter case the neoantigen may be termed a tumour-associated antigen
The term tumour-associated antigen is used herein to define an antigen present in transformed (malignant or tumourous) cells which is absent (or present in lower amounts or in a different cellular compartment) in normal cells of the type from which the tumour originated Oncogenic viruses can also induce expression of tumour antigens which are often host proteins induced by the virus
The term herbal medicine is used herein to define a pharmaceutical composition in which at least one active principle is not chemically synthesized and is a phytochemical constituent of a plant In most cases, this non- synthetic active principle is not isolated (as defined herein), but present together with other phytochemicals with which it is associated in the source plant In some cases, however, the plant-deπved bioactive pnncιple(s) may be in a concentrated fraction or isolated (sometimes involving high degrees of purification) In many cases, however, the herbal medicine comprises a more or less crude extract, infusion or fraction of a plant or even an unprocessed whole plant (or part thereof), though in such cases the plant (or plant part) is usually at least dried and/or milled
The term bioactive principle 's used herειn to define a phytochemical which is necessary or sufficient for the pharmaceutical efficacy of the herbal medicament in which it is comprised In the case of the present invention the bioactive principle comμnses the immunomodulatory compound of the invention (e g casuarine, casuarine glucoside or mixtures thereof)
The term standard specification is used herein to define a characteristic, or a phytochemical profile, which is correlated with an acceptable quality of the herbal medicine In this context, the term quality is used to define the overall fitness of the herbal medicament for its intended use, and includes the presence of one or more of the bioactive principles (at an appropriate concentration) described above or else the presence of one or more bioactive markers or a phytochemical profile which correlates with the presence of one or more of the bioactive principles (at an appropriate concentration) The term phytochemical profile is used herein to define a set of characteristics relating to different phytochemical constituents.
The term isolated as applied to the pyrrolizidine compounds of the invention is used herein to indicate that the compound exists in a physical milieu distinct from that in which it occurs in nature. For example, the isolated material may be substantially isolated (for example purified) with respect to the complex cellular milieu in which it naturally occurs. When the isolated material is purified, the absolute level of purity is not critical and those skilled in the art can readily determine appropriate levels of purity according to the use to which the material is to be put. Preferred, however, are purity levels of 90% w/w, 99% w/w or higher. In some circumstances, the isolated compound forms part of a composition (for example a more or less crude extract containing many other substances) or buffer system, which may for example contain other components. In other circumstances, the isolated compound may be purified to essential homogeneity, for example as determined εpectrophotometrically, by NMR or by chromatography (for example GC-MS).
The term pharmaceutically acceptable derivative as applied to the pyrrolizidine compounds of the invention define compounds which are obtained (or obtainable) by chemical derivatization of the parent pyrrolizidine compounds of the invention. The pharmaceutically acceptable derivatives are therefore suitable for administration to or use in contact with the tissues of humans without undue toxicity, irritation or allergic response (i.e. commensurate with a reasonable benefit/risk ratio). Preferred derivatives are those obtained (or obtainable) by alkyiation, eεterification or acylation of the parent pyrrolizidine compounds of the invention. The derivatives may be immunomodulatory perse, or may be inactive until processed in vivo. In the latter case, the derivatives of the invention act as pro-drugs. Particularly preferred pro-drugs are ester derivatives which are esterified at one or more of the free hydroxyls and which are activated by hydrolysiε in vivo. The pharmaceutically acceptable derivatives of the invention retain some or all of the immunomodulatory activity of the parent compound. In some cases, the immunomodulatory activity is increased by derivatization.
Derivatization may also augment other biological activities of the compound, for example bioavailability and/or glycosidase inhibitory activity and/or glycosidase inhibitory profile. For example, derivatization may increase glycosidase inhibitory potency and/or specificity.
The term pharmaceutically acceptable salt as applied to the pyrrolizidine compounds of the invention defines any non-toxic organic or inorganic acid addition salt of the free base compounds which are suitable for use in contact with the tissues of humans and lower animals without undue toxicity, irritation, allergic response and which are commensurate with a reasonable benefit/risk ratio. Suitable pharmaceutically acceptable salts are well known in the art. Examples are the salts with inorganic acids (for example hydrochloric, hydrobromic, sulphuric and phosphoric, acids), organic carboxylic acids (for example acetic, propionic, glycolic, lactic, pyruvic, malonic, εuc nic, fumaric, malic, tartaric, citric, ascorbic, maleic, hydroxymaleic, dihydro/ymaleic, benzoic, phenylacetic, 4-arnιnobenzoιc, 4-hydroxyb zoic, anthranilic, cinnamic, salicylic, 2-phenoxybenzoic, 2- acetoxyben∑oic and mandelic acid) and organic sulfonic acids (for example methanesulfonic acid and p- toluenesulfonic acid). The drugs of the invention may also be converted into salts by reaction with an alkali metal halide, for example sodium chloride, sodium iodide or lithium iodide. Preferably, the pyrrolizidine compounds of the invention are converted into their salts by reaction with a stoichiometric amount of sodium chloride in the presence of a solvent such as acetone. These salts and the free base compounds can exist in either a hydrated or a substantially anhydrous form. Crystalline forms of the compounds of the invention are also contemplated and in general the acid addition salts of the pyrrolizidine compounds of the invention are crystalline materials which are εoluble in water and various hydrophilic organic solvents and which in comparison to their free base forms, demonstrate higher melting pointε and an increased solubility.
In its broadest aspect, the present invention contemplates all optical isomers, racemic forms and diastereomers of the pyrrolizidine compounds of the invention. Those skilled in the art will appreciate that, owing to the asymmetrically substituted carbon atoms present in the compounds of the invention, the pyrrolizidine compounds of the invention may exist and be synthesised and/or isolated in optically active and racemic forms. Thus, references to the pyrrolizidine compounds of the present invention encompass the pyrrolizidine compounds as a mixture of diastereomers, as individual diastereomers, as a mixture of enantiomers as well as in the form of individual enantiomers.
Therefore, the preεent invention contemplates all optical isomers and racemic forms thereof of the compounds of the invention, and unless indicated otherwise (e.g. by use of dash-wedge structural formulae) the compounds shown herein are intended to encompass all possible optical isomers of the compounds so depicted. In cases where the stereochemical form of the compound is important for pharmaceutical utility, the invention contemplates use of an isolated eutomer.
Biological activities of the compounds of the invention
Without wishing to be bound by any theory, it is thought that the immunomodulatory activity of the compounds of the invention may arise from the stimulation and/or suppression of cytokine secretion in vivo. In particular, it is thought that that the immunomodulatory activity of the compounds of the invention arises from the stimulation of secretion of one or more cytokines (e.g. one or more Thl cytokines), including interleukins 2 and/or 12 (IL-2 and/or IL-12) and/or the suppression of secretion of one or more Th2 cytokines (e.g. IL-5).
In particular, it is thought that the immunostimulatory activity of the compounds of the invention may arise from the stimulation of 11- 12 and IL-2 by dendritic cells. This leads to the stimulation of NK cells to produce IFN-γ and induces the development of CD + Th1 cells. The induced Th1 cells then produce IFN- γ and IL-2. The IL-2 then enhances further proliferation of Th1 cells and the differentiation of pathogen (e.g. tumour and virus) -specific CD8+ T cells. The IL-2 also stimulates the cytolytic activity of NK cells of the innate immune system.
IL-12 is the primary mediator ot type- 1 immunity (the Th l response) It induces natural killer (NK) cells to produce IFN-y as part of the innate immune response and promotes the expansion of CD4+ Th 1 cells and cytotoxic CDS* cells which produce IFN-y It therefore increases T-cell invasion of tumours as well as the susceptibility of tumour cells to T-cell invasion.
Thus, the compounds of the invention are preferably stimulators of cytokine secretion. Particularly preferred are compounds which induce, potentiate, activate or stimulate the release one or more cytokines (for example Th1 cytokines, e.g. IL-12 and/or II-2, optionally together with one or more other cytokines) in vivo. This primary immunomodulatory activity of the compounds of the invention is particularly important in certain medical applications (discussed in detail infra). For example, increased production of IL-12 may overcome the suppression of innate and cellular immunities of HIV-1 -infected individuals and AIDS patients.
The cytokine stimulation exhibited by the compounds of the invention may be dependent, in whole or in part, on the presence of co-stimulatory agents. Such co-stimulatory agents may include, for example, agents that stimulate the innate immune system, including Toll-like receptor (TLR) ligands. These ligands include microbial products such as lipopolysaccharide (LPS) and/or monophosphoryl lipid) as well as other molecules associated with microbial infection. In many applications, such co-stimulatory agents will be present in the patient to be treated at the time of administration of the compounds of the invention.
Without wishing to be bound by any theory, it is thought that at least some of the pharmacological activities of the compounds of the invention may be based on a secondary glycosidase inhibitory activity.
Such glycosidase inhibition may lead to any or all of the following in vivo:
β Modification of tumour cell glycosylation (e.g. tumour antigen glycosylation);
• Modification of viral protein glycosylation (e.g. virion antigen glycosylation); • Modification of cell-surface protein glycosylation in infected host cells;
• Modification of bacterial cell walls.
This ancillary biological activity may therefore augment the primary immunomodulatory activity in some preferred embodiments of the invention. It may be particularly desirable in certain medical applications, including the treatment of proliferative disorders (such as cancer) or in applications where infection is attendant on immune suppression. For example, selective modification of virion antigen glycosylation may render an infecting virus less (or non-) infective and/or more susceptible to endogenous immune responses. In particular, the compounds of the invention may alter the HIV viral envelope glycoprotein gp120 glycosylation patterns, hence inhibiting the entry of HIV into the host cell by interfering with the binding to cell surface receptors.
Thus, the compounds of the invention are preferably (but not necessarily) glycosidase inhibitors. Particularly preferred are compounds which exhibit specificity of glycosidase inhibition, for example Glucosidase I rather than mannosidases. Such preferred compounds can therefore be quite different in their glycosidase inhibitory profile to swainsonine and its analogues, since the latter are potent and specific inhibitors of mannosidase.
Medical applications of the compounds of the invention
The invention finds broad application in medicine, for example in methods of therapy, prophylaxis and/or diagnosis.
These medical applications may be applied to any warm-blooded animal, including humans. The applications include veterinary applications, wherein the pyrrolizidine compounds of the invention are administered to non- human animals including primates, dogs cats, horses, cattle and sheep
The pyrrolizidine compoundε of the invention are immunomodulators Thus, they find general application in the treatment or prophylaxis of conditions in which stimulation augmentation or induction of the immune system is indicated and/or in which suppression or elimination of part or all of the immune response is indicated
Particular medical uses of the pyrrolizidine compounds of the invention are described in detail below References to therapy and/or prophylaxis in the description or claims are to be interpreted accordingly and are intended to encompass inter alia the particular applications described below
1 Increasing the Th1 Th2 response ratio
General considerations
As explained earlier, the immune response comprises two distinct types the Th1 response (type-1 , cellular or cell mediated immunity) and Th2 response (type-2, humoral or antibody mediated immunity)
These Th1 and Th2 responses are not mutually exclusive and in many circumstances occur in parallel In such circumstances the balance of the Th1 Th2 response determines the nature (and repercussions) of the immunological defence (as explained herein)
The Th1 Th2 balance (which can be expressed as the Th1 Th2 response ratio) is determined, at least in part, by the nature of the environment (and in particular the cytokine milieu) in which antigen priming of naive helper T cells occurs when the immune system is first stimulated
The Th1 and Th2 responseε are distinguished inter alia on the basiε of certain phenotypic changeε attendant on priming and subsequent polarization of naive helper T cells These phenotypic changes are characterized, at least in part, by the nature of the cytokines secreted by the polarized helper T cells
Th1 cells produce so-called TM cytokines, which include one or more of IL-1 , TNF, IL-2, IFN-gamma, IL-12 and/or IL-18 The Th1 cytokines are involved in macrophage activation and Th1 cells orchestrate cell-mediated defences (including cytotoxic T lymphocyte production) that form a kε> limb of the defence against bacteπai and viral attack, as well as malignant cells
Th2 cells produce so-called Th2 ortokines, which include one oi more of IL-4 , IL-5, IL- I0 and IL- 13 The Th2 cytokines promote the production of various antibodies and can suppress the Th1 response
Accordingly, in the mouse, a cell that makes IFN-gamma and not IL-4 is classified as Th1 , whereas a CD4+ cell that expresses IL-4 and not IFN-gamma is classified as Th2 Although this distinction is less clear in humans (T cells that produce only Th1 or Th2 cytokines do not appear to exist in humans), the phenotype of the T cell response (Th1 or Th2) can still be distinguished in humans on the basis of the ratio of Th1 to Th2 cytokines expressed (usually, the ratio of IFN-gamma to IL-4 and/or IL-5) There is an increasing realization that the type of immune response is just as important in therapy and prophylaxis as its intensity or its duration For example, an excess Th1 response can result in autoimmune disease inappropriate inflammatory responses and transplant rejection An excess Th2 response can lead to allergies and asthma Moreover, a perturbation in the Th1 Th2 ratio is symptomatic of many immunological diseases and disorders, and the development of methods for altering the Th1 Th2 ratio is now a priority
It has now been discovered that the immunomodulatory pyrrolizidine compounds of the invention can increase the Th1 Th2 response ratio in vivo (for example by preferentially promoting a Th1 response and/or preferentially suppressing a Th2 response)
Thus, the compounds of the invention find application in methods of therapy and/or prophylaxis which comprise increasing the Th1 Th2 response ratio (for example, by preferentially promoting a Th1 response and/or preferentially suppressing a Th2 response)
The medical applications contemplated herein therefore include any diseases, conditions or disorders in which an increase in the Th1 Th2 response ratio is indicated or desired For example, the medical applications contemplated include diseases, conditions or disorderε in which stimulation of a Th1 reεponse and/or suppression of a Th2 responεe is indicated or desired
The mechanιsm(s) by which the compounds of the invention increase the Thl Th2 response ratio are not yet fully understood It is likely that the activity is based, at least in part, on selective Th1 cytokine induction (since Th1 and Th2 cytokines exhibit mutual inhibition), for example in dendritic cells
For example the compounds of the invention may induce, potentiate, activate or stimulate (either directly or indirectly) the release and/or activity (in vitro and/or in vivo) of one or more Th1 cytokines (for example one or more cytokines selected from IFN-gamma, IL-12, IL-2 and IL-18) Particularly preferred are compounds which induce, potentiate, activate or stimulate the release and/or activity (in vitro and/or in vivo) of IFN-gamma and/or IL-12 and/or IL-2
Particularly preferred are compounds that stimulate the release of IL-2 and IL-12 in dendritic cells
The compounds of the invention may also suppress or inactivate (either directly or indirecilyl the release and/or activity (in vif.ro and/or in \ ivo) of one oi more Th2 cytokines (for βAample one or more cytokines selected from IL-4, IL-5, IL-10 and IL- l ό) Particularly preferred are compounds which suppress or inactivate the release and/oi activity (in vitro and/or in vivo) of IL-5
Thus, particularly preferred are compounds which exhibit a Th I cytokine stimulatory activity together with a complementary Th2 cytokine inhibitor,' activity
Specific examples of applications falling within the geneial class of treatments based on increasing the Th1 Th2 response ratio are described in the following sections Th1 -related diseases
Th1 -related diseases are diseases, disorders, syndromes, conditions or infections in which Th1 cells are involved in preventing, curing or alleviating the effects of the disease, disorder, syndrome, condition or infection
Th1 -related diseases may also include diseases, disorders, syndromes, conditions or infections in which the Th1 component of the immune response is pathologically depressed or diseases, disorders, syndromes, conditions or infections in which stimulation of a Th1 response is indicated
Such conditions may arise, for example, from certain proliferative disorders (typically cancers) in which the proliferating (e g tumour) cells exert a suppressive effect on one or more components of the Th1 response For example, tumour cells may inhibit dendritic cells, cause the expression of inhibitory receptors on T cells, down legulate MHC class I expression and induce the secretion of anti-inflammatory factors and immunosuppressive cytokines which deactivate or suppress immune cell cytotoxicity
Thus the compounds of the invention find application in the treatment or prophylaxis of Th1-related diseaεeε
Examples of Th 1 -related diseases include infectious diseases (particularly viral infe ions) and proliferative disorders (e g cancer)
Thus, the Th1 -related diseases include any malignant or pre-ma gnant condition, proliferative or hyper- proliferative condition or any disease arising or deriving from or associated with a functional or other disturbance or abnormality in the proliferative capacity or behaviour of any cells or tissues of the body
Thus the invention finds application in the treatment or prophylaxis of breast cancer, colon cancer, lung cancer and prostate cancer It also finds application in the treatment or prophylaxis of cancers of the blood and lymphatic systems (including Hodgkin's Diεease, leukemias, lymphomas, multiple myeloma, and Waldenstrom's disease), skin cancers (including malignant melanoma), cancers of the digestive tract (including head and neck cancers, cesophageal cancer, stomach cancer, cancer of the pancreas, liver cancer, colon and rectal cancer, anal cancer), cancers of the genital and urinary systems (including kidney cancer, bladder cancer, testis cancer, prostate cancer), cancers in women (including breast cancer, ovarian cancer, gynecological cancers and choπocarcincma) as well as ,n bra.n, bone carcmoid, naεopharyngeal, retropentoneal, thyroid and soft tissue tumours It also finds application in the treatment or prophylaxis of cancers of unknown pi imary site
The Th1-related infectious diseases include bacterial, pπon (e g BSE and CJD), vital, fungal, protozoan and metazoan infections For example, the Th l-related infectious diseases include infection with respiratory syncytial virus (RSV), hepatitis B virus (HBV), Epstein-Barr, hepatitis C virus (HCV), herpes simplex type 1 and 2, herpes genitalis herpes keratitis, herpes encephalitis, herpes zoster, human immunodeficiency virus (HIV), influenza A virus, hantann virus (hemorrhagic fever), human papilloma virus (HPV), tuberculosis, leprosy and measles Particularly preferred Th1-related infectious diseases include those in which the pathogen occupies an intracellular compartment, including HIV/AIDS, leishmaniasis, trypanosomiasis, influenza, tuberculosis and malaria.
The compounds of the invention may also find application in the treatment of patients in which the Th1 immune response is defective. Such patients may include neonates, juveniles in which the Th1 response is immature and not fully developed, as well as older patients in which the Th1 response has become senescent or compromised over time. In such patient populations the compounds of the invention may be used prophylactically (as a generalized type 1 immune stimulant to reduce the risks of (e.g. viral) infections.
Th2-related diseases and allergy
Th2-related diseases are diseases, disorders, syndromes, conditions or infections in which Th2 cells are implicated in (e.g. support, cause or mediate) the effects of the disease, disorder, syndrome, condition or infection.
Thus, the compounds of the invention find application in the treatment or prophylaxis of Th2-related diseases.
One important class of Th2-related diseases treatable with the compounds of the invention is allergic disease.
It is well known that genetically predisposed individuals can become sensitised (allergic) to antigens originating from a variety of environmental sources. The allergic reaction occurs when a previously sensitised individual is re-exposed to the same or to a structurally similar or homologous allergen. Thus, as used herein the term allergy is used to define a state of hypersensitivity induced by expoεure to a particular antigen (allergen) resulting in harmful and/or uncomfortable immunologic reactions on subεequent exposures to the allergen.
The harmful, uncomfortable and/or undesirable immunologic reactions present in allergy include a wide range of symptoms. Many different organs and tissues may be affected, including the gastrointestinal tract, the skin, the lungs, the nose and the central nervous system. The symptoms may include abdominal pain, abdominal bloating, disturbance of bowel function, vomiting, rashes, skin irritation, wheezing and shortness of breath, nasal running and nasal blockage, headache and mood changes. In severe cases the cardiovascular and respiratory systems are compromised and anaphylaetic shock leads in extreme cases to death.
It is known that the harmful, undesirable and/or uncomfortable immunologic reactions characteristic of allergy have a ThZ response component.
As explained above, the compounds of the invention may suppress or inactivate (either directly or indirectly) the release and/or activity (in vitro and/or in vivo) of one or more Th2 cytokines (for example one or more cytokines selected from IL-4, IL-5, IL-10 and IL-13). Thus, the compounds of the invention may be used to effect a remedial or palliative modulation of the harmful and/or uncomfortable immunologic reactionε characteristic of allergic reactions by inhibiting, suppressing or eliminating the Th2 response to the allergen.
The compounds of the invention therefore find application in the treatment or prophylaxis of allergy. Any allergy may be treated according to the invention, including atopic allergy, allergic rhinitis, allergic conjunctivitis, atopic dermatitis, hypereosinophilia, irritable bowel syndrome, allergen-induced migraine, bacterial allergy, bronchial allergy (asthma), contact allergy (dermatitis), delayed allergy, pollen allergy (hay fever), drug allergy, sting allergy, bite allergy, gastrointestinal or food allergy (including that associated with inflammatory bowel disease, including ulcerative colitis and Crohn's disease) and physical allergy. Physical allergies include cold allergy (cold urticaria or angioedema), heat allergy (cholinergic urticaria) and photosensitivity.
, Particularly important is the treatment or prophylaxis of asthma.
2. Haemorestoration
The pyrrolizidine compounds of the invention increase splenic and bone marrow cell proliferation and can act as myeloproliferative agents. They therefore find application as haemorestoratives.
Haemorestoration may be indicated following immunosuppressant therapies (such as cyclosporine A, azathioprine or immunosuppressant radiotherapies), chemotherapy (including treatment with both cycle-specific and non-specific chemotherapeutic agents), steroid administration or other forms of surgical or medical intervention (including radiotherapy). Thus, the use of the pyrrolizidine compounds of the invention as haemorestoratives may be adjunctive to other treatments which tend to depress splenic and bone marrow ceil populations. Particularly;preferred adjunctive therapies according to the invention include the administration of an immunorestorative dose of the pyrrolizidine compound of the invention adjunctive to: (a) chemotherapy; and/or (b) radiotherapy; and/or (c) bone marrow transplantation; and/or (d) haemoablative immunotherapy.
3. Alleviation of immunosuppression
The pyrrolizidine compounds of the invention may be used to alleviate, control or modify states in which the immune system is partially or completely suppressed or depressed. Such states may arise from congenital (inherited) conditions, be acquired (e.g. by infection or malignancy) or induced (e.g. deliberately as part of the management of transplants or cancers).
Thus, the pyrrolizidine compounds of the invention may find application as adjunctive immunomodulators (e.g. immunostimulants) in the treatment and/or management of various diseases (including certain cancers) or medical interventions (including radiotherapy, immunosuppressant therapy (such as the administration of cyclosporine A, azathioprine or immunosuppressant radiotherapies), chemotherapy and cytotoxic drug administration (for example the administration of ricin, cyclophosphamide, cortisone acetate, vinblastine, vincristine, adriamycin, 6-mercaptopurine, 5-fluorouracil, mitomycin C, chloramphenicol and other steroid-based therapies). They may therefore be used as chemoprotectants in the management of various cancers and infections (including bacterial and viral infections, e.g. HIV infection) or to induce appropriate and complementary immunotherapeutic activity during conventional immunotherapy. In particular the pyrrolizidine compounds of the invention may find application as immunoεtimulants in the treatment or management of microbial infections which are associated with immune-suppressed states, including many viral infections (including HIV infection in AIDS) and in other situations where a patient has been immunocompromised (e g following infection with hepatitis C, or other viruses or infectious agents in uding bacteria, fungi, and parasites in patients undergoing bone marrow transplants, and in patients with chemical or tumor-induced immune suppression)
Other diseases or disorders which may give rise to an immunosupressed state treatable according to the invention include ataxia-telangiectasia, DiGeorge syndrome, Chediak-Higashi syndrome, Job syndrome, leukocyte adhesion defects, panhypogammaglobulinemia (e g associated with Bruton disease or congenital agammaglobulinemia) selective deficiency of IgA, combined immunodeficiency disease, Wiscott-Aldπch syndrome and complement deficiencies It may be associated with organ and/or tissue (e g bone marrow) transplantation or grafting, in which applications the pyrrolizidine compounds of the invention may be used adjunctively as part of an overall treatment regimen including surgery and post-operative management of immune status
4 Cytokine stimulation
The pyrrolizidine compounds of the invention may be used to induce, potentiate or activate various cytokineε in vivo, including vaπouε interleukms (including IL-2 and/or IL-12)
Accordingly, the pyrrolizidine compounds of the invention find general application in the treatment or prophylaxis of conditions in which the in vivo induction, polentiation or activation of one or more cytokines (e g IL-12 and/or II-2) is indicated Such applications may be employed to stimulate particular elements of the cellular immunity system, including dendritic cells, macrophages (e g tissue-specific macrophages), CTL, NK, NKT, B and LAK cells
In such applications, the compounds of the invention may be employed as an adjunct to gene therapies designed to increase the production of endogenous cytokines (for example IL-2)
5 τreatment of proliferative disorders
The invention finds application in the treatment of proliferative disorders, including various cancers and cancer metastasis For example, the pyi rolizidine compounds of the invention may find particular application in the treatment of leukemias lymphomas melanomas adenomas sarcomas, carcinomas of solid tissues, melanoma (including melanoma of the eye) pancieatic cancer, cervico-uteπne cancer, cancel s of the kidney, stomach, lung ovary, rectum, breast, prostate, bowel, gastric, liver thyroid, neck, cervix, salivary gland, leg, tongue, lip, bile duct, pelvis, mediastinum, urethra, lung, bladder, esophagus and colon, and Kapoεi's Sarcoma (e g when associated with AIDS)
In such applications the compounds of the invention may exhibit a εecondary glycosidase inhibitory activity The invention may therefore find application in methods of therapy or prophylaxis which comprise the modification of tumour cell glycosylation (e.g. tumour antigen glycosylation), the modification of viral protein glycosylation (e.g. virion antigen glycosylation), the modification of cell-surface protein glycosylation in infected host cells and/or the modification of bacterial cell walls, hence promoting an increased immune response or inhibiting growth/infectivity directly.
6. Use as adjuvant
The pyrrolizidine compounds of the invention find utility as vaccine adjuvants, in which embodiments they may promote, induce or enhance an immune response to antigens, particularly antigens having low intrinsic immunogenicity. Without wishing to be bound by any theory, the pyrrolizidine compounds of the invention may augment vaccine immunogenicity by stimulating cytokine release, thereby promoting T-cell help for B cell and CTL responses. They may also change glycosylation of cancer or viral antigens and increase vaccine effectiveness.
When used as adjuvant, the compounds of the invention may be administered concurrently, separately or sequentially with adminislration of the vaccine. The invention finds application in any vaccine, but may be particularly as a subunit vaccine, a conjugate vaccine, a DNA vaccine, a recombinant vaccine or a mucosal vaccine. The vaccine may be therapeutic or prophylactic. It may be used imrnunoprophylactically or immunotherapeutically in both human and non-human subjects. Preferred non-human subjects include mammals and birds. Particularly preferred are veterinary applications. Such applications include the treatment or prophylaxis of infection in domesticated animals (for example dogs and cats) and livestock (e.g. sheep, cows, pigs, horses, chickens and turkeys). '
Thus, in some embodiments, the pyrrolizidine compound of the invention may be present in admixture with other vaccine component(s), or else co-packaged (e.g. as part of an array of unit doses) with the other vaccine components with which it is to be used as adjuvant. In yet other embodiments, the use of the pyrrolizidine compounds of the invention as adjuvant is simply reflected in the content of the information and/or instructions co-packaged with the vaccine components and relating to the vaccination procedure, vaccine formulation and/or posology.
7. Dendritic Gell-based applications
As described above, it has now been found that the pyrrolizidine compounds of the invention may induce sustained and pronounced cytokine production (e.g sustained and pronounced IL-12 and/or IL-2 production) in dendritic cells. Thus, the compounds of the invention find application in methods of therapy or prophylaxis comprising the induction of cytokine production in dendritic cells or in which the induction of cytokine production in dendritic cells is indicated or required.
Dendritic cell vaccines
In one dendritic cell-based treatment paradigm, the cells are pulsed (primed or spiked) with a particular antigen or antigens (for example, tumour antigen(s)) and then administered to promote a Th1 immune response. The responding T ceils include helper cells, especially Th1 CD4+ cells (which produce IFN-γ) and killer cells (especially CD8+ cytolytic T lymphocytes). The dendritic cells may also mediate responses by other classes of lymphocytes (B, NK, and NKT cells). They may also elicit T cell memory, a critical goal of vaccination.
With regard to antigen selection for use in the dendritic cell vaccine sof the invention, both defined and undefined antigens can be employed. The antigens can be xenoantigens or autoantigens. One or more defined neoantigen(s) may be selected: in the case of cancer treatment, the neoantigen(s) may comprise a tumour-associated antigen.
However, most preferred for use according to the invention are peptides (for example, synthetic 9-11 amino acid peptides) containing defined antigens. Such peptides may comprise natural sequences. Alternatively, they may be synthetic analogues designed for enhanced MHC binding.
In other embodiments, the antigens used according to the invention are provided in the form of immune complexes. These are preferably delivered to Fc-receptor-bearing DCs so that both MHC class I and MHC class II peptide sequences are formed. In this way, dendritic cell vaccines can be used according to the invention for inducing both CTLs and Th cells.
In another approach to antigen selection for use according to the invention, the whole antigenic repertoire of any given tumour (or other target cell, such as a viraiiy-infected cell) is explored. Thus, in another embodiment of the invention there is provided DC-tumour cell hybrids in which the dendritic cells are treated with compound (thereby to induce the expression of IL-2) before or after hybridisation.
In yet other embodiments, necrotic or apoptotic tumour cells or cell lysates (for example lysates of infected cells or tumour cells) are used.
Antigens derived from fresh tumour cells (rather than tumour cell lines or defined antigens) may also be employed.
It is also contemplated that the compounds of the invention be incorporated into cellular antigens by introducing them into the cellular membrane or into an intracellular compartment (as described for example in WO96017614, the contents of which are incorporated herein by reference).
Various techniques can be used to deliver the selected antigen(s) to the DCs (variously referred to in the art as antigen loading, pulsing, priming or spiking). Preferred are loading techniques which load the DCs internally: this can be achieved through the use of peptides linked to cell-penetrating moieties.
Antigens can also be loaded by transfecting the DCs with encoding nucleic acid (e.g. by electroporation) such that the antigens are expressed by the DC, processed and presented at the cell surface. This approach avoids the need for expensive GMP proteins and antibodies. RNA is preferred for this purpose, since it produces only transient expression (albeit sufficient for antigen processing) and avoids the potential problems associated with the integration of DNA and attendant long-term expression/mutagenesis. Such transfection techniques also permit exploration of the whole antigenic repertoire of a target cell by use of total or PCR-amplified tumour RNA.
Current strategies for using dendritic cells in this way focus on identifying specific tumour antigens and defining antigenic peptideε that bind to the particular MHC alleles expresεed by each patient. However, a more general approach would involve the εtimulation of the dendritic cells in a manner appropriate for potentiating Th1 responses irrespective of the antigens present and either with or without antigen priming. Cytokine production by activated dendritic cells would then promote the appropriate Th1 response.
The dendritic ceil based vaccines of the invention find particular application in the treatment or prophylaxis of various proliferative disorders (including various cancers, as described below). In such applications, the dendritic cells are preferably pulsed (primed or spiked) with one or more tumour antigens e* vivo and the compounds of the invention used to potentiate the dendritic cell component of the vaccine by contacting the dendritic cells with the compound either ex vivo (before or after pulsing of the cells) or in vivo (for example by co-administration, either concurrently, separately or sequentially, of the dendritic cells and the compound).
The dendritic cell based vaccines of the invention may be used in the treatment or prophylaxis of any malignant or pre-malignant condition, proliferative or hyper-proliferative condition or any disease arising or deriving from or associated with a functional or other disturbance or abnormality in the proliferative capacity or behaviour of any cells or tissues of the body.
- Thus, the invention finds application in the treatment or prophylaxis of breast cancer, colon cancer, lung cancer and prostate cancer. It also finds application in the treatment or prophylaxis of cancers of the blood and lymphatic systems (including Hodgkin's Diseaεe, leukemias, lymphomas, multiple myeloma, and Waldenstrom's disease), skin cancers (including malignant melanoma), cancers of the digestive tract (including head and neck cancers, cesophageal cancer, stomach cancer, cancer of the pancreas, liver cancer, colon and rectal cancer, anal cancer), cancers of the genital and urinary systems (including kidney cancer, bladder cancer, testis cancer, prostate cancer), cancers in women (including breast cancer, ovarian cancer, gynecological cancers and choriocarcinoma) as well as in brain, bone carcinoid, nasopharyngeal, retroperitoneal, thyroid and soft tissue tumours. It also finds application in the treatment or prophylaxis of cancers of unknown primary site.
The dendritic cell based vaccines of the invention also find application in the treatment or prophylaxis of various infections, including bacterial, viral, fungal, protozoan and metazoan infections. For example, the vaccines may be used in the treatment or prophylaxis of infection with respiratory syncytial virus (RSV), Epstein-Barr, hepatitis B virus (HBV), hepatitis G virus (HCV), herpes simplex type 1 and 2, herpes genitaliε, herpes keralitiε, herpes encephalitis, herpes zoster, human immunodeficiency virus (HIV), influenza A virus, hantann virus (hemorrhagic fever), human papilloma virus (HPV), tuberculosis, leprosy and measles.
Particularly preferred is the treatment or prophylaxis of infections in which the pathogen occupies an intracellular compartment or causes the expression of neoantigens by host cells, including HIV/AIDS, leishmania, trypanosomiasis, influenza, tuberculosis and malaria. The present invention also contemplates a more general approach to DC cell-based therapy which involves the stimulation of the dendritic cells with the compound of the invention irrespective of the antigens present and either with or without antigen priming.
Thus, the invention finds application in therapies in which dendritic cells exposed to the compound of the invention are targeted to diseased or infected tissue (for example injected directly into a tumour), where the cells can prime endogenous T cells extranodally. In such embodiments, the invention contemplates targeting of DCs to a tumour and their activation in situ to elicit immune responses without the need for ex vivo antigen loading.
In yet another embodiment, the invention contemplates in situ DC vaccination where antigen is targeted to DCs in vivo which are then expanded and induced to mature in situ (by the co-administration of one or more DC maturation stimulants). In such embodiments, antigen is targeted to endogenous DCs by any convenient method, for example through the use of exosomes (as described in Thery et al. (2002) Nat Rev Immunol 2: 569-579).
Any class of dendritic cell may be used according to the invention. Thus, the dendritic cells may be myeloid or lymphoid, or mixtures thereof. The myeloid dendritic cells, if used, may be of the Langerhans cell type or interstitial DCs. Alternatively, mixtures of these myeloid subsets may be used. Especially preferred is the use of monocyte-derived DCs (Mo-DCs).
Helper proteins may be used to potentiate the activity of the dendritic cell vaccines of the invention.
I
Dendritic cell-based approaches to autoimmune disorders
Dendritic cells are also involved in regulating and maintaining immunological tolerance: in the absence of maturation, the cells induce antigen-specific silencing or tolerance. Thus, in another dendritic cell-based treatment paradigm the cells are administered as part of an immunomodulatory intervention designed to combat autoimmune disorders.
In such applications, the suppressive potential of dendritic cells has been enhanced by in vitro transfectlon with genes encoding cytokines. However, such gene therapy approaches are inherently dangerous and a more efficient and attractive approach would be to pulse dendritic cells in vitro with biologically active compounds which stimulate an appropriate cytokine secretion pattern in the dendritic cells.
As described above, it has now been discovered that the pyrrolizidine compounds of the invention can induce sustained and pronounced cytokine production in dendritic cells. Thus, the compounds of the invention find application in the enhancement of the suppressive potential of dendritic cells.
Thus, the invention finds application in the treatment or prophylaxis of autoimmune disorders, including myasthenia gravis, rheumatoid arthritis, systemic lupus erythematosus, Sjogren syndrome, scleroderma, polymyositis and'dermomyositis, ankylosing spondylitis, and rheumatic fever, insulin-dependent diabetes, thyroid diseases (including Grave's disease and Hashimoto thyroiditis), Addison's disease, multiple sclerosis, psoriasis, inflammatory bowel disease, ulcerative colitis and autoimmune male and female infertility.
8. Wound healing
The pyrrolizidine compounds of the invention can reverse a Th2 type splenocyte response ex vivo in a normally non-healing infectious disease model. Antigen specific splenocyte IFN-gamma can be significantly increased and IL-5 production significantly reduced in such models, indicative of a healing response'.
Thus, the invention finds application in the treatment of wounds. In particular, the invention finds application in the treatment or prophylaxis of wounds and lesions, for example those associated with post-operative healing, burns, infection (e.g. necrotic lesions), malignancy or trauma (e.g. associated with cardiovascular disorders such as stroke or induced as part of a surgical intervention).
The wound treatments may involve the selective suppression or elimination of a Th2 response (for example to eliminate or suppress an inappropriate or harmful inflammatory response).
Posolo y
The pyrrolizidine compounds of the present invention can be administered by oral or parenteral routes, including intravenous, intramuscular, intraperitoneal, subcutaneous, transdermal, airway (aerosol), rectal, vaginal and topical (including buccal and sublingual) administration.
The amount of the pyrrolizidine compound administered can vary widely according to the particular dosage unit employed, the period of treatment, the age and sex of the patient treated, the nature and extent of the disorder treated, and the particular pyrrolizidine compound selected.
Moreover, the pyrrolizidine compounds of the invention can be used in conjunction with other agents known to be useful in the treatment of diseases, disorders or infections where immunostimulation is indicated (as described infra) and in such embodiments the dose may be adjusted accordingly.
In general, the effective amount of the pyrrolizidine compound administered will generally range from about 0.01 mg/kg to 500 mg/kg daily. A unit dosage may contain from 0.05 to 500 mg of the pyrrolizidine compound, and can be taken one or more times per day. The pyrrolizidine compound can be administered with a pharmaceutical carrier using conventional dosage unit forms either orally, parenterally, or topically, as described below.
The preferred route of administration is oral administration. In general a suitable dose will be in the range of 0.01 to 500 mg per kilogram body weight of the recipient per day, preferably in the range of 0.1 to 50 mg per kilogram body weight per day and most preferably in the range 1 to 5 mg per kilogram body weight per day.
The desired dose is preferably presented as a single dose for daily administration. However, two, three, four, five or six or more sub-doses administered at appropriate intervals throughout the day may also be employed. These sub-doses may be administered in unit dosage forms, for example, containing 0.001 to 100 mg, preferably 0.01 to 10 mg, and most preferably 0 5 to 1.0 mg of active ingredient per unit dosage form.
Formulation
The compositions of the invention comprise the pyrrolizidine compound of the invention, optionally together with a pharmaceutically acceptable excipient.
The pyrrolizidine compound of the invention may take any form. It may be synthetic, purified or isolated from natural sources (for example from Casuarina equisetifolia or Eugenia jambolana), using techniques described in the art (and referenced infra).
When isolated from a natural source, the pyrrolizidine compound of the invention may be purified. However, the compositions of the invention may take the form of herbal medicines, as hereinbefore defined. Such herbal medicines preferably are analysed to determine whether they meet a standard specification prior to use.
The herbal medicines for use according to the invention may be dried plant material. Alternatively, the herbal medicine may be processed plant material, the processing involving physical or chemical pre-processing, for example powdering, grinding, freezing, evaporation, filtration, pressing, spray drying, extrusion, supercritical solvent extraction and tincture production. In cases where the herbal medicine is administered or sold in the form of a whole plant (or part thereof), the plant material may be dried prior to use. Any convenient form of drying may be used, including freeze-drying, spray drying or air-drying. '
In embodiments where the pyrrolizidine compound of the invention is formulated together with a pharmaceutically acceptable excipient, any suitable excipient may be used, including for example inert diluents, disintegrating agents, binding agents, lubricating agents, sweetening agents, flavouring agents, colouring agents and preservatives. Suitable inert diluents include sodium and calcium carbonate, sodium and calcium phosphate, and lactose, while corn starch and alginic acid are suitable disintegrating agents. Binding agents may include starch and gelatin, while the lubricating agent, if present, will generally be magnesium stearate, stearic acid or talc.
The pharmaceutical compositions may take any suitable form, and include for example tablets, elixirs, capsules, solutions, suspensions, powders, granules and aerosols.
The pharmaceutical composition may take the form of a kit of parts, which kit may comprise the composition of the invention together with instructions for use and/or a plurality of different components in unit dosage form.
Tablets for oral use may include the pyrrolizidine compound of the invention, either alone or together with other plant material associated with the botanical source(s) (in the case of herbal medicine embodiments). The tablets may contain the pyrrolizidine compound of the invention mixed with pharmaceutically acceptable excipients, εuch as inert diluents, disintegrating agents, binding agents, lubricating agents, sweetening agents, flavouring agents, colouring agents and preservatives. Suitable inert diluents include sodium and calcium carbonate, sodium and calcium phosphate, and lactose, while corn starch and alginic acid are suitable disintegrating agents. Binding agents may include starch and gelatin, while the lubricating agent, if present, will generally be magnesium stearate, stearic acid or talc. If desired, the tablets may be coated with a material such as αlvcervl monostearate or glyceryl distearate, to delay absorption in the gastrointestinal tract.
Capsules for oral use include hard gelatin capsules in which the pyrrolizidine compound of the invention is mixed with a solid diluent, and soft gelatin capsules wherein the active ingredient is mixed with water or an oil such as peanut oil, liquid paraffin or olive oil.
Formulations for rectal administration may be presented as a suppository with a suitable base comprising for example cocoa butter or a salicylate.
Formulations suitable for vaginal administration may be presented as pessaries, tampons, creams, gels, pastes, foams or spray formulations containing in addition to the active ingredient such carriers as are known in the art to be appropriate.
For intramuscular, intraperitoneal, subcutaneous and intravenous use, the compounds of the invention will generally be provided in sterile aqueous solutions or suspensions, buffered to an appropriate pH and . isotonicity.
Suitable aqueous vehicles include Ringer's solution and isotonic sodium chloride. Aqueous suspensions according to the invention may include suspending agents such as cellulose derivatives, sodium alginate, polyvinylpyrrolidone and gum tragacanth, and a wetting agent such as lecithin. Suitable preservatives for aqueous suspensions include ethyl and n-propyl p-hydroxybenzoate.
The compounds of the invention may also be presented as iiposome formulations.
For oral administration the pyrrolizidine compound of the invention can be formulated into solid or liquid preparations such as capsules, pills, tablets, troches, lozenges, melts, powders, granules, solutions, suspensions, dispersions or emulsions (which solutions, suspensions dispersions or emulsions may be aqueous or non-aqueous). The solid unit dosage forms can be a capsule which can be of the ordinary hard- or soft-shelled gelatin type containing, for example, surfactants, lubricants, and inert fillers such as lactose, sucrose, calcium phosphate, and comstarch.
In another embodiment, the pyrrolizidine compounds of the invention are tableted with conventional tablet bases such as lactose, sucrose, and comstarch in combination with binders such as acacia, comstarch, or gelatin, disintegrating agents intended to assist the break-up and dissolution of the tablet following administration such as potato starch, alginic acid, corn starch, and guar gum, lubricants intended to improve the flow of tablet granulations and to prevent the adhesion of tablet material to the surfaces of the tablet dies and punches, for example, talc, stearic acid, or magnesium, calcium, or zinc stearate, dyes, coloring agents, and ' flavoring agents intended to enhance the aesthetic qualities of the tablets and make them more acceptable to the patient. Suitable excipients for use in oral liquid dosage forms include diluents such as water and alcohols, for example, ethanol, benzyl alcohol, and the polyethylene alcohols, either with or without the addition of a pharmaceutically acceptably surfactant, suspending agent or emulsifying agent.
The pyrrolizidine compounds of the invention may also be administered parenterally, that is, subcutaneously, intravenously, intramuscularly, or interperitoneally.
In such embodiments, the pyrrolizidine compound is provided as injectable doses in a physiologically acceptable diluent together with a pharmaceutical carrier (which can be a sterile liquid or mixture of liquids). Suitable liquids include water, saline, aqueous dextrose and related sugar solutions, an alcohol (such as ethanol, isopropanol, or hexadecyl alcohol), glycols (such as propylene glycol or polyethylene glycol), glycerol ketals (such as 2,2-dimethyl-1 ,3-dioxolane-4-methanol), ethers (such as poly(ethylene-glycol) 400), an oil, a fatty acid, a fatty acid ester or glyceride, or an acetylated fatty acid glyceride with or without the addition of a pharmaceutically acceptable surfactant (such as a soap or a detergent), suspending agent (such as pectin, carhomers, methylcellulose, hydroxypropylmethylcellulose, or carboxymethylcellulose), or emulsifying agent and other pharmaceutically adjuvants. Suitable oils which can be used in the parenteral formulations of this invention are those of petroleum, animal, vegetable, or synthetic origin, for example, peanut oil, soybean oil, sesame oil, cottonseed oil, corn oil, olive oil, petrolatum, and mineral oil.
Suitable fatty acids include oleic acid, stearic acid, and isostearic acid. Suitable fatty acid esters are, for example, ethyl oleate and isopropyl myristate.
Suitable soaps include fatty alkali metal, ammonium, and triethanolamine salts and suitable detergents include cationic detergents, for example, dimethyl dialkyl ammonium halides, alkyl pyridinium halides, and alkylamines acetates; anionic detergents, for example, alkyl, aryl, and olefin sulphonates, alkyl, olefin, ether, and monoglyceride sulphates, and sulphosuccinates; nonionic detergents, for example, fatty amine oxides, fatty acid alkanolamides, and polyoxyethylenepolypropylene copolymers; and amphoteric detergents, for example, alkyl-beta-aminopropionates, and 2-alkylimidazoline quartemary ammonium salts, as well as mixtures.
The parenteral compositions of this invention will typically contain from about 0.5 to about 25% by weight of the pyrrolizidine compound of the invention in solution. Preservatives and buffers may also be used. In order to minimize or eliminate irritation at the site of injection, such compositions may contain a non-ionic surfactant having a hydrophile-lipophile balance (HLB) of from about 12 to about 17. The quantity of surfactant in such formulations ranges from about 5 to about 15% by weight. The surfactant can be a single component having the above HLB or can be a mixture of two or more components having the desired HLB. Illustrative of surfactants used in parenteral formulations are the class of polyethylene sorbitan fatty acid esters, for example, sorbitan monooleate and the high molecular weight adducts of ethylene oxide with a hydrophobic base, formed by the condensation of propylene oxide with propylene glycol.
The pyrrolizidine compounds of the invention may also be administered topically, and when done so the carrier may suitably comprise a solution, ointment or gel base. The base, for example, may comprise one or more of the following: petrolatum, lanolin, polyethylene glycols, bee wax, mineral oil, diluents such as water and alcohol, and emulsifiers and stabilizers Topical formulations may contain a concentration of the compound from about 0 1 to about 10% w/v (weight per unit volume)
When used adjunctively, the pyrrolizidine compounds of the invention may be formulated for use with one or more other αrug(s) in particular, the pyrrolizidine compounds of the invention may be used in combination with antitumor agents, antimicrobial agents, anti-inflammatoπes, antiproliferative agents and/or other immunomodulatory (e g immunostimulatory) agentε For example, the pyrrolizidine compoundε of the invention may be used with anti-viral and/or anti-proliferative agents such as cytokines, including ιnterleukιns-2 and 12, interferons and inducers thereof, tumor necrosis factor (TNF) and/or transforming growth factor (TGF), as well as with myelosuppressive agents and/or chemotherapeutic agents (such as doxorubicin, 5-fluorouracιl, cyclophoεphamide and methotrexate), isoniazid (e g in the prevention or treatment of peripheral neuropathy) and with analgesics (e g NSAIDs) for the prevention and treatment of gastroduodenal ulcers
Thus, adiunctive use may be reflected in a specific unit dosage designed to be compatible (or to synergize) with the othei drug(s), or in formulations in which the pyrrolizidine compound is admixed with one or more antitumor agents, antimicrobial agents and/or anliinflammatones (or elεe physically associated with the other drug(s) within a single unit dose) Adjunctive uses may also be reflected in the composition of the pharmaceutical kits of the invention, in which the pyrrolizidine compound of the invention is co-packaged (e g as part of an array of unit doses) with the antitumor agents, antimicrobial agents and/or antiinflammatoπes Adjunctive use may also be reflected in information and/or instructions relating to the co-administration of the pyrrolizidine compound with antitumor agents, antimicrobial agents and/or antiinflammatoπes
Exemplification
The invention will now be described with reference to specific Examples These are merely exemplary and for illustrative purposeε only they are not intended to be limiting in any way to the εcope of the monopoly claimed or to the invention described These examples constitute the best mode currently contemplated for practicing the invention
Example 1 Induction of IL-12 secretion in dendritic cells
Mice
BALB/c male and female mice bred and maintained at the University of Strathclyde under conventional conditions were used at 8 weeks old
Isolation of bone marrow and culture of dendritic cells
Bone marrow was obtained from the femurs of mice The femurs were washed in 70% ethanol and placed in a clean petπ dish Dendritic cell (DC) medium (2 5% granulocyte-macrophage colony-stimulating factor GM- CSF), 10% heat and activated foetal calf serum, 1% L-glutamme, 1 % Penicillin/Streptomycin in RPMI-1640 medium) was injected into the bone marrow of the femur by a pumping action and the cells and medium were collected 1 ml of the cells in medium was added to a 75cm2 flask with 15mls of DC medium The flasks were then incubated at 37°C, 5% CO2 to allow DC growth and development. After 5 days an additional 10mls of DC medium was added.
Harvesting of dendritic cells
After 10 days of incubation of bone marrow with GM-CSF, the dendritic cells were harvested. This process was carried out in a tissue culture hood. The contents of the flasks were poured into centrifuge tubes to ensure collection of floating DCs. Approximately 10mls of cooled phosphate buffered saline (PBS) was added to each empty flask, the flasks gently agitated and the contents collected. This ensured recovery of adhesive DCs. The collected contents of the flasks were centrifuged for 5 minutes at 200g and the pellet resuspended in 2mls of DC medium without GM-CSF. A cell count was then carried out.
Cell count and assay conditions
Cells were counted using a haemocylometer. Approximately 20μl of the resuspended cells was pipetted into the chamber of the haemocytometer, the cells were adjusted to the correct cell concentration (approx. 5 x 104, and not less than 1 x 104, per well) and then plated out for assay.
The plates were incubated overnight at 37°C with 5% CO2 and allowed to settle (harvesting stimulates them). The next day the compounds (50μg/mi and 20μg/ml) and controls were added then again incubated at 37°C with 5% CO2 for 24 hrs (or 48 hrs). Harvesting and addition of the compounds was all done in a hood. The plates were then frozen to kill the cells and once defrosted the supernatant analysed as described below.
Measurement of IL-12
Using an enzyme linked immunosorbent assay (ELISA) IL-12 concentration in the supernatants was measured. All reagents used in this assay were from Pharivlingen. A 96-well flat-bottomed ELISA plate was coated with purified rat anti-mouse IL-12 (p40/p70) MAb (Cat no. 554478) at 2μg/ml diluted in PBS pH 9.0 at 50μl/well. The plate was then covered in cling film and incubated at 4°C. Following incubation the plate was washed 3 times in washing buffer and dried. 200μl of blocking buffer (10% foetal calf serum in PBS pH 7.0) was added to each well then covered in cling film and incubated at 37°C for 45 minutes. The plate was washed 3 times and dried. Recombinant mouse 11-12 standard was added at 30μl in duplicate wells, starting at lOng/mi then 5, 2.5, 1.25, 0.625, 0.31 , 0.156, 0.078, 0.039, 0.020, 0.010, 0.005ng/ml. Standards were diluted in blocking buffer. The supernatant samples were added in at 50μl/well. The plate was then covered in cling film and incubated for 2 hours at 37°C. The plate was then washed 4 times, dried and the secondary antibody added.
Biotin labelled anti-mouse IL-12 (p40/p70) MAb (Cat no. 18482D) at 1 μg/ml (diluted in blocking buffer) was added to each well at a volume of 100μl/well. The plate was covered in cling film and incubated at 37°C for 1 hour. The plate was then washed 5 times, dried and the conjugate added. Streptavidin-AKP (Cat no. 13043E) at 100μl/well was added at a dilution of 1/2000 in blocking buffer followed by incubation under cling film at 37°C for 45 minutes. • The plate was finally washed 6 times, dried and the substrate added! pN.PP (Sigma) in glycine buffer at 1mg/ml was added at 100μl/well. The plate was then covered in tinfoil, incubated at 37°C and checked every 30 minutes for a colour change.
' The plate was then read at 405nm using a SPECTRAmax 90 spectrometer. The results are shown in Figures 1 and 2, in which LPS is lipopolysaccharide, IFN-g is interferon gamma, 462a is casuarine (8), 462b is casuarine-6-α-D-glucopyranose (9), 23 is 7-ep/casuarine (11) and 24 is 3,7-diep/-casuarine (10).
When tested at 50μg/ml in the same assay, swainsonine (4) failed to induce IL-12 secretion. Similar studies with other compounds for comparative purposes are shown in Table 1.1 , below.
Figure imgf000040_0001
Figure imgf000041_0001
Figure imgf000042_0001
Example 2: Stimulation of IL-2 production by dendritic cells
The protocols described in Example 1 above were carried out but the appropriate Mabs and standards for determination of II-2 were substituted. The results are shown in Table 2.1 , below.
Figure imgf000042_0002
Example 3: Cytokine modulation in εpleen cellε
Mice BALB/c male and female mice bred and maintained at the University of Strathclyde under conventional conditions were used at varying age.
Isolation of Spleen cells and culture of Spleen cells
The mouse spleen was removed aseptically and placed in a sterile petri dish containing 5mls of complete medium (RPMI, 1 % L-Glutamine, 1% Penicillin/Streptomycin and 10% foetal calf serum1). Cells suspensions were prepared by using the end of a syringe and grinding the spleen through a wire mesh. The cell suspension was then centrifuged al lOOOrpm for 5 minutes. To remove the erythrocytes, the cell pellet was resuspended in Boyle's solution (Tris 0. 17M e Ammonium Chloride 0. 16M) and centrifuged again for 5 minutes. The pellet was then washed in medium a further two times, then resuspended in 3ml- medium. A cell count was then carried out.
Experimental Protocol
All spleen cell experiments were carried out in 96-well tissue culture plates 100μl aliquots of 5x10s/well cells were added to all wells and each well had a final volume of 200μl Unstimulated wells contained 100μl of cells and 10Oμl of medium. The stimulated wells contained 10Oμl of cells plus 50μl of LPS at 1 μg/ml or 50μl anti- CD3 at 0 5μg/ml and 50μl of medium The remaining wells contained 10Oμl cells, 50μl of MNLP compound and either 50μl of antι-CD3 or medium alone
Measurement of IL-12, IL-2, IL-5 and IFN-y
The appropriate Mabs and standards were used according to the protocol described for IL-12 (described in Example 1 , above) The results are shown in Tables 3 1-3 3, below
Table 3.1 : Promotion of activated splenocyte (T-cell) IFN-γ production
Figure imgf000043_0001
Table 3 2: Effect of castanospermine on splenocyte IFN-γ production
Figure imgf000043_0002
As can be seen from the results shown in Tables 3 1 and 3 2, compounds according to the invention stimulate IFN-y secretion/production in splenocytes, whereas castanospermine inhibits the production of this cytokine in such assays Similar tests carried out with 1-Deoxynojιrιmycιn (DNJ) (21) showed that this imino sugar also inhibited IFN-y secretion/production in splenocyteε (data not shown)
Example 4 Inhibition of glycosidase activity
All enzymes were purchased from Sigma, as weie the appropriate p-nitrophenyl substrates Assays were carried out in microtitie plates Enzymes were assayed in 0 IM citric acιd/0 2M di-sodium hydrogen phosphate (Mcllvaine) buffeis at the optimum pH lor the enzyme /Ml assays were earned out at 20°C Foi screening assays the incubation assay consisted of 10 μl of enzyme solution, I0 μl of inhibitor solution (made up in water) and 50 μl of Ihe appropriate 5 mM p-nitiophenyl substrate (3 57mM final cone ) made up in Mcllvaine buffer at the optimum pH for the enzyme
The reactions were stopped with 0 4M glycine (pH 10 4) during the exponential phase of the reaction, which was determined at the beginning of the assay using blanks with water, which were incubated for a range of time periods to measure the reaction rate using 5 mM substrate solution Endpomt absorbances were read at 405nm with a Biorad microtitre plate reader (Benchmark) Water was substituted for the inhibitors in the blanks The enzymes tested are shown in Table 4.1 , below.
Figure imgf000044_0002
The compounds tested are shown in Table 4.2, below.
Figure imgf000044_0001
Figure imgf000045_0001
The results (% inhibition) for a number of different compounds (all at 1mg/ml) are shown in Table 4 3, below
Figure imgf000045_0002
Figure imgf000046_0001
The resultε εhow that the profile of inhibition for the compoundε of the invention ιε quite different from that of caεtanospermine None inhibits mannosidaεe significantly (seε alεo further data below) Some of the compoundε teεted (e g 3,7-dιep -casuaπne) do not significantly inhibit any of the enzymes tested
Further studieε showed that the K, for casuarine (8) with yeast α-D-glucosidase was 217μM (castanosperminε not being inhibitory at a concentration of 800μM) The K, for castanospermine (20) with almond β-D-glucosidaεe waε 9μM (caεuaπne not being inhibitory at 800μM) Moreover casuarine also inhibited rabbit gut mucosa α-D- glucosidase with an IC50 value of 210μM as compared with an IC50 value of 8μM for caεtanoεpermine Both casuarine and castanospermine inhibited rabbit small intestine sucrase at a concentration of 700μM Castanospermine also inhibited rabbit small intestine lactase and trehalase by over 50% at this concentration
Example 5 Differential inhibition of mannosidaεe and glucosidase
The glycosidase inhibitory profiles of swainsonine (4), casuaπne (8) and casuarine glucoεide (9) with reεpect to a mannoεidase and a glucosidase were compared The resultε (all at <0 1 mg/ml) are shown in Table 5 1 , below
Figure imgf000046_0002
Example 6 Treatment of muπne HSV-1 infεction
Mice were 3-4 weeks old female BALB/c Mice were inoculated with 10 p f u HSV- 1 (SC16) using the neck skin method This dose is sublethal but produces clinical symptoms including inflammation (measured by increase in ear pinna thickness)
Mice werε administered (, 100 ml i p ) with one of two doces of casuarine (δ) on day one and daily thereafter for 5 days Group 1 received 15 mg/kg in PBS, group 2 received 150 mg/kg in PBS A negative control group 3 were infected but received no casuarine A positive contiol group 4 were administered with famciclovir (1 /a drinking water spiked at 1 mg/ml for the same time period)
Mice were checked daily and sampleε were obtained from mice killed on selected days The results are presentεd in Tables 6 1 - 6 3, below Table 6.1 : Weight (% change)
Figure imgf000047_0001
Table 6.2: Group mεan weight (g)
Figure imgf000047_0002
Table 6.3: Ear pinna thickness (mm" )
Figure imgf000047_0003
Figure imgf000048_0001
The results show the expectεd pattεrn of ear pinna thicknesε increase, peaking at day 4. Famvir almost completely negated the ear thickness responεe. Caεuarine at both dosεs tested also produced a reduction in ear thicknεss.
Examplε 7: Control of lung metastasiε in mice
Mice (C57/bl6 under i/p ketaminε anaeεthesia) wεre challengεd i/v (tail vεin) with 5x104 B16-F10 tumour cells in a final volume of 100μl per mouse on day 0. Test compounds (50mg/kg in 200μl sterile non-pyrogenic saline) were administerεd ε/c (right flank) on days 2 and 4. On day 14 the mice were sacrifices and the lungs dissected and stained in Indian ink solution (150ml bidistilled water, 30 ml India Ink, 4 drops NH4OH) for 10 minutes then fixed for at least 24 hr in Fakete's solution (90ml 37% formaldehyde, 900 ml 70% EtOH and 45ml glacial acetic acid). The metastaεεs in the stained and fixed lungs could then be visualized, counted and photographed.
The results are shown below in Table 7.1 , below.
Figure imgf000048_0002
Example 8: Effect on glycosylation of breast cancer cells
Cell culture
MCF-7 cells (European Collection of Cell Cultures Ref. 86012803) were taken from liquid nitrogen stock, thawed at room temperature and transferred to 10mi Dulbeccos Modified Eagle's Medium with Hams F I2, 15mM Hepes and L-glutamine (DMEM: Cambrex Cat. Mo. BE12-719F) supplemented with 10% v/v foetal calf serum (FCS: BioWest Labs Cat. No. S02755, Lot. No. SI 800). The FCS was pre-filtered through a 0.2μm εteril filter.
The cells were then centrifuged at 1 ,500 rpm in a Centaur bench-top centrifuge and the supernatant removed. The cells were reconstituted in fresh media and seeded into two T75cm3 Nunclon tissue culture flasks and allowed to settle overnight al 37°C in a 5% CO2 incubator. The flasks were wrapped in cling film to prevent cross-contamination and the following day the media was changed to include the antibiotics penicillin and streptomycin as a precautionary measure againεt infection (at concentrations of 1 mg/cm3 and 5mg/cm3, rεspectively). The cells werε allowεd to grow near confluence and then split at a 1 in 4.resuspenεion. The cells used for the experimεnts were of passage number 31. Two flasks of cells werε prεpared in media containing 20% v/v FCS with 10% dimethylsulphoxide and bankεd down into liquid nitrogεn for later use if necessary.
A total of 16 T25cmJ flasks werε used. Each flask was seeded with 8.5x10s cells/cm3 and 4cm3 mεdia addεd. Thε cεlls were allowed to adherε to the culture flask overnight. The following morning the flasks were observed under the light microscope and the cells appeared 50-60% confluent. The cells from two of the flasks werε harvεstεd (sεe below) for the t=0 time point.
The remaining 14 flasks were available for testing with casuarine (8). Sevεn of these (untreated group) had their media changed to 7cm3 of fresh media containing 10% FCS, penicillin and streptomycin (as before), whilst the remaining seven werε incubatεd with frεsh mεdia supplεmented with 0.75mM caεuarine (trεatεd group).
Cells werε harvested at t=1.5 hours, t=28 hours, t=62 hours and t=86 hours.
Harvesting of cεlls and cell counting
The cells were harvestεd using a non-enzymatic method. At each of the time points the cells werε viewed under the invertεd light microscope and the morphology evaluated. Before harvesting, the cells were washed with sterile PBS, theree times, 7cm3 per wash. The cells were then scraped from the flasks using a sterile cell scraper and transferred to Grenier tubes. The cells were quickly passed through a 21 G2 gauge needle to disaggregate the cells. Cells were then pellεtεd by centrifugation at 1500g/5min and resuspεnded in a known volume of PBS. The number of cells was then counted in a haemocytometer and cell viability evaluated by mixing 0.1 cm3 of each cell suspension with a drop of trypan blue solution. Each of the cell pellets was frozen at -80°C until glycan relεase and analysis.
Homogenisation
The cell pellets were placed in an iced water bath and allowed to thaw. The pellets were' then homogenized in a total of 4cm3 (made up to volume with deionized water). An Ultraturrax T25 homogeniser was used for this purpose, with the blade speεd set to 22,500 rpm. The samples were maintained on ice and 3 bursts, each of 10sεc, were applied with, a period of approximately 1 in between each homogenisation step to allow the froth the settle. The blade was washed carefully between each of the samples to prevent sample cross- contamination. The homogenates were stored in 1cm3 aliquots at -80°C prior to the protein assay and glycan release.
Protein asεav
Evaluated using the BioRad protein assay according to the manufacturer's instructions. BSA was used as standard. Each of the homogenate samplεs was tested in duplicate using lOOμl aliquots from each time point. Glycan release
For the time points of 62 hourε and 86 hourε the equivalent of 25μg of protein waε taken and dried for 3 hourε on a centrifugal evaporator (without heating). For the eariier time pointε, whoεe protein concentration could not 5 be aεεessed with the protein asεay, 200μl waε taken and dried down ready for glycan reieaεe. κeιease was confirmed using 25μg of fetuin from foεtal calf sεrum,
Glycans wεrε incubated at 37°C overnight with N-glycosidase F (Rochε Bioεciεncεs Cat. No. 1365185, Lot. No. 9280212/31) at a final concεntration of 5U εnzymε in 25μl of samplε all in 20mM sodium phoεphatε buffεr 10 pH7.2. Aftεr the incubation step, the sampleε were loaded onto prewaεhεd and primεd Ludgεr Clean E cartridgeε (Cat. No. LC-E10-A6). The glycanε were eluted according to thε manufacturer'ε inεtructions and dried by centrifugal evaporation overnight.
Glycan labelling'
15
The glycans were labelled by reductive amination, for 2 hours at 65°C, according to the method described by Bigge ef al. (1995) Anal. Biochem. 230(2): 229-238. The incubation mixture was then "cleaned up" to remove any uneonjugated fluorophorε by spotting thε samples onto Whatman 3MM paper and running in a descending chrόrriatography tank with a mobile phase of 4:1 :1 butanol:ethanol:water overnight. Glycans were then eluted
20 with 0.5cm3 methanol and 2 x 1 cm3 HPLC grade water then filtered through a 0.2μm syringe top filter.
Analysis using normal phase HPLC
The glycans were εeparated on a normal phase (hydrophilic interaction) HPLC column (LudgerSep N1 amide)
25 4.6 x 25 cm in size. '
The basis of the separation is described in Guile ef al. (1996) Anal. Biochem. 240(2): 210-226. The column was fitted to a Dionex BioLC system with autosampler and switching pump heads and in-line mixer. The column was maintained at 30°C and the glycans detected using a Perkin Elmεr LS30 fluorimeter with excitation
30 λ=330nm and emisεion λ=420nm, the gain was set to 2. The buffer syεtem uεed was the high salt syεtem, with acetonitrilε as buffεr A and 0.25M ammonium formatε pH4.4 as buffer B. Flow rate waε maintained at Q.3cm3/min throughout.
The protocol used iε summarized below in Table 8. 1 , below. jb
Figure imgf000050_0001
An 80μl aliquot of each of the glycan mixtureε was loaded onto the column and the elution position compared, with reference to a hydrolyεate of dextran.
Summary of results and conclusions
At the initial harvest point and the 28 hour time point, there was no obvious differεncε bεtween the glycans released from the treated and untreated cells (data not shown). However, at the 62 and 86 hour time points, the untreated cells showed a marked preponderance of larger N-linked glycans than their treated counterparts (data not shown). In addition, the overall εignal (amount of fluoreεcently labelled glycan) waε greatεr in thε untreated group.
The resultε show that casuarine can inhibit glycan syntheεiε and/or N-linked glycosylation in breast cancer cells.
Example 9: Effect on glucose transport
The effect of casuarine (8) and castanospermine (20) on the initial rate of Na+ -dependent D-glucose uptake into ovine intestinal brush border membrane veεicles was examined in a competition assay with labelled D- giucosε. The rεsults arε εhown in Tablε 9.1 , below:
Figure imgf000051_0001
It can be seen that glucose transport was slightly inhibited by castanospermine but slightly stimulated by casuarine.
Example 10: Increasing the Th1 :Th2 response ratio in a non-healing leishmaniasis model
Leishmaniasis is a classic model of a Th l disease: non-healing cutaneous lesions arise from an undesirable polarization of the immune response Which becomes heavily Th2-skewed.
In order to study the ability of the compounds of the invention to increase the Th I :Th2 response ratio in this disease model (and so promote a healing Th1 response), spleen cells from Leishmania major infected BALB/c mice having a non-healing cutaneous infection were stimulated with parasite antigen (Table 10.1) or polyclonally with anti-CD3 (Table 10.2) in the presεnce of 3,7-diep/-casuarine (10). Table 10.1 : Reversal of the inability of T-cells to produce IFN-γ in a non-healing mouse model
Figure imgf000052_0001
Table 10.2: Downregulation of Th2 cytokine response in a non-healing mouse model
Figure imgf000052_0002
It can be seen that the presence of 3,7-diep/-casuarine (10) enhances IFN-γ (asεociated with a healing Th1 reεponse) whilst suppressing the Th2 responεe (via downregulation of the Th2 cytokine IL-5). The Th2-εkewed immune response profile associated with a non-healing disease was clearly reversed ex vivo by 3,7-diep/- caεuarinε (10).
Examplε 11 : Synthesis of 3,7-diep/'-casuarine (10)
General Experimental
All reactions were carried out under an atmoεphere of argon at room temperature uεing anhydrous solvents uπlesε otherwise stated. Anhydrous solvents were purchased from Fluka Chemicals and were used as supplied. Reagents were supplied from Aldrich, Fluka and Fiεher and were uεed aε εupplied. Thin layer chromatography (Tic) was performed on aluminium sheets pre-coatεd with Merck 60 F254 silica gel and were visualised under ultra-violet light and staining using 6% phosphomolybdic acid in ethanol. Silica gel chromatography was carried out using Sorbsil C60 40/60 silica gel under a positive atmosphere. Amberlite IR- 120, strongly acidic ion-ε change resin was preparεd by soaking the resin in 2M hydrochloric acid for at least two hours followed by elution with distilled water until the eluant reached pH 5. Dowev 50WX8-100 was prepared by soaking the resin with 2M hydrochloric acid for at least two hours followed by elution with distilled water until neutral. Infrared spectra were recorded on a Perkin-Elmer 1750 IR Fourier Transform εpectrophotometer using thin films on sodium chloride plates. Only characteristic peaks are recorded. Optical rotations were measured on a Perkin-Elmεr 241 polarimεtεr with a path length of 1dm. Concentrations are quoted in g/100mL. Nuclear magnetic reεonancε spectra were recorded on a Bruker DQX 400 spectrometer in the stated deuteratεd solvent. All spectra were recorded at ambient temperature. Chemical shifts (δ) are quoted in ppm and are relative to residual solvent as standard. Proton spectra (5H) were recorded at 400 MHz and carbon spectra (δc) at 100 MHz. 2,3 5.6 7,8-Trι-0-ιsopropylιdene-D-erytf7ro-L-ta/o-octono-1 ,4-lactone (Qc) 5,6 7,8-Dι-0-i8θpropylιdene-D-ervf/7ro-L-σa/acfo-octono-1 ,4-lactone (Qb)
Sodium cyanide (7 02 g, 142 mmol) waε added to a εtirred εolution of D-g/ycero-D-gu/o-heptose (Qa, 21 g, 100 mmol) in watεr (300 ml) Thε reaction mixture was stirred at room temperature for 48 h, heatεd at reflux for 48 h and pasεed through a column containing Amberlite IR-120 (εtrongly acidic lon-exchangε reεm, 300 ml) The eluent waε concεntratεd under reducεd preεεure and the residue dried in vacuo for 24 hours The resulting foam waε treated with acetone (500 ml) and sulphuric acid (5 4 ml) in the presence of anhydrous copper sulphate (10 g, 62 mmol) at room tempεrature for 48 h T i c analysis indicatεd the presεnce of two major products (ethyl acetate cyclohexane, 1 1 , R 0 72, 0 18) The reaction mixture was filtered and the filtrate was treated with sodium bicarbonate (50 g) for 24 h at room temperature Solid residues werε removed by filtration and the filtrate was concentrated under reduced pressure The resulting crude yellow syrup was purified by silica gel chromatography providing 2,3 5,6 7, δ-tπ-O-isopropylidεnε-D-e/yfftro-L-fa/o-octono-l ,4-lactone Qc as a colourless syrup (Re 0 72, 7 672 g, 21 %,) and 5,6 7,8-dι-0-ιsopropylιdenε-D-e Wτo-L- ;a/acfo-octono- 1 ,4- lactonε Qb as a clear oil (Rf 0 18, 8 105 g, 25 %) 2,3 5,6 7,8-trι-0-ιsopropylιdene-D-er /7/t>-L-fa/o-octono-1 ,4- lactone Qc δH (CDCI3) 1 29, 1 33, 1 35, I 38, I 42, 1 48 (6 x ε, 18H, 3 x C(CH3)z), 3 93-3 99 (m, 2H, H-8a, H- 7), 4 03-4 07 (m, 2H, H-5, H-6), 4 15 (dd, 1 H, J8a 8b 8 7 J8.7 6 I , H-8b), 4 75-4 78 (m, 3H, H-2, H-3, H-4), δc (CDCb) 25 23, 25 51 , 26 00, 26 71 , 26 73, 27 16 (3 x C(CH3)2), 67 93, 74 93, 76 33, 76 69, 78 65, 79 40, 80 06, 109 95, 110 72, 113 19, 174 27, vmaχ (film) 1793 5,6 7,8-dι-0-ιsopropylιdene-D-er tήro-L-ga/ac.o-octono-1 ,4- lactone Qb δH (de-acetone) 1 28, I 32, 1 34, 1 35 (4s, I2H, 2 x C(CH3)2), 3 92 (1 H, m, H-8a), 3 98 (m, 1 H, H- 7), 4 14 (m, 2H, H-5, H-8b), 4 23-4 25 (m, 2H, H-4, H-6), 4 35-440 (m, 2H, H-2, H-3), δc (cfe-acetonε) 25 31 , 25 87, 26 72, 27 31 , 68 06, 75 15, 75 23, 77 51 , 78 05, 78 41 , 79 01 , 110 06, 110 31 , 174 25, vmaλ (film) 1793, 3541
2,3 5,6-Dι-0-ιsopropylιdene-D-e/yfr?ro-L-fa/o-octono-1 ,4-lactone Qd
A solution of 2,3 5,6 7,8-trι-0-ιεopropylιdene-D-eryfΛro-L-fa/o-octono-1 ,4-lactone (Qc, 3 8 g, 10 6 mmol) waε treated with acetic acid water (2 3, 100 ml) at 50 °C for 2 h T i c analyεis (ethyl acetate cyclohexane, 1 1) indicated the disappearance of the starting material (Rf 0 72) and the presence of a more polar compound (Rf 0 15) The solvent was removεd under reduced pressure and the residue was purified by silica gel chromatography (ethyl acetae cyclohexane, 1 1 to 3 1) yielding 2,3 5,6-dι-O-ιsopropylιdene-D-ery.ftro-L-fa/o- oclono- 1 ,4-lactone Qd as a clear oil (3 23 g, 94 %) δH (CD3OD) I 28, I 38, , 1 43 (3 x s, I2H, 2 x C(GH3V.), 3 59 (dd, I H, -/8a7 5 40 8a 8b 11 41 , H-8a), 3 66-3 69 (m, I H, H-7), 3 7-) (dd, I H, J8_ 7 2 90 Hz, H-Sb), 4 0 I (app t, I H, 6 7 7 62 Hz, H-6), 4 24 (dd, I H, J5β S IT Hz J5 4 0 89 Hz, H-5), 4 79-4 81 (m, 2H, H-3, H-4), 4 89-4 91 (m, I H, H-2), δc (C D3OD) 24 62 25 42, 26 05, 26 49, 63 86, 73 81 , 75 40, 75 9 1 , 79 18, 79 90, 80 78, 1 10 53, 113 09, 175 76, vma (film) 1791 , 3478, [α]D -35 7 (c I , CHCI3)
6-0-ferf-Butyldιmethylsιlyl-2,3 5,6-dι-0-i8opropylιdene-D-e/yf/7ro-L-fa/o-oetono-1 ,4-lactone Qe
To a solution of 2,3 5,6-dι-0-ιsopropylιdene-D-er)'f/7ro-L-fa/o-octono-1 ,4-lactone (Qd, 3 18 g, 10 mmol) in N,N- dimethylformamide (40 ml) was added ferf-butyldimethylsilyl chloπdε (1 808 g, 12 mmol) and imidazole (1 361 g, 20 mmol) The reaction mixture was stirred at room temperature for 16 h after which 1 1 c analysis (ethyl acetate cyclohexanε, 1 1) showεd no starting material (Rf 0 15) and the formation of one major product (Rf 0.63). The solvent was removed under reducεd prεssurε and the residue was partitioned between ethyl acetate and brine. The aqueous layer was extractεd with ethyl acetate and the combined organic layers werε dried (MgS04), filtered and the solvent removed. The reεulting pale oil waε purified by silica gel chromatography (ethyl acetate:cyclohexane, 0:1 to 1 :2) to give 8-0-fe/t-butyldimethylsilyl-2,3:5,6-di-0-i8opropylidene-D-e/yffVo- L-ta/o-octono-1 ,4-lactone Qe as a clear oil (3.612 g, 85%): δH (UL>u3) U.U4 or s, 6H, 2 x CH3), 0.86 (s, 9H, C(CH3)3), 1.23, 1.30, 1.32, 1.41 (4 x s, 12H, 2 x C(CH3)2), 3.63-3.67 (m, 2H, H-8a, H-7), 3.76 (br d, 1 H, H-8b), 3.96 (app t, J6l7 8.21 J6,5 7.98, H-6), 4.08 (br d, 1 H, H-5), 4.72 (br s, 2H, H-2, H-3), 4.78 (br s, 1H, H-4); δc (CDC ) -5.52, -5.45, 18.25, 25.51 , 25.80, 25.93, 26 68, 27.18, 63.95, 72.97, 74.88, 74.93, 78.71 , 79.63, 79.87, 110.34, 113.00, 174.42; vmax (film) 1794, 3570; [α]D -20.1 (c 1 , CHCI3).
7-Azido-8-0-feff-butyldimethylsilyl-7-deoxy-2,3:5,6-dι-0-isopropylidenε-L-f/7reo-L-fa/o-octono-1 ,4-lactone Qf
A solution of 8-0-ferf-butyldimethylsιlyl-2, 3:5, 6-di-O-isopropylidene-D-e 'fftro-L-fa/o-octono-l ,4-lactone (Qe, 3.5 g, 8.2 mmol) in a pyridine:dichloromethanε mixture (1 :4, 25 ml) was cooled to -30 °C. Trifluoromethanesulfonic anhydride (3.5 g, 2.09 ml, 1 .4 mmol) was added portion-wise and the mixture was stirred for 2 h. T.l.c analysis (ethyl acetate yclohexane, 1 :3) indicated the disappearance of εtarting material (Rf 0.38) and the presence of a less polar product (Rf 0 48). The reaction mixture was concentrated under reduced pressure and the residue was partitioned between ethyl acetate and 0.5 M hydrochloric acid. The organic layer was washed with brine, dried (MgS04), filtered and concentrated under reduced pressure. The resulting crude pale orange residue waε treated with sodium a∑ide (807 mg, 12.4 mmol) in /V,Λ/-dimethylformamide (25 ml) for 16 h. T.l.c. analysis (ethyl acetatexyclohexane, 1 :4) indicated the disappearance of the intermediate triflate (Rf 0.42) and the presence of a more polar compound (Rf 0.40). The reaction solvent was removεd in vacua and thε residue was partitioned between ethyl acetate and brine. The aqueous layer waε extracted with ethyl acetate and the combined organic layers were dried (MgS04), filtered and concentrated in vacua. The resulting crude residue was purified by silica gel chromatography (ethyl acetate:cyclohexanε, 0:1 to 1 :4) providing 7-azido-8-0-.erf-butyldimεthylsilyl-7- deoxy-2,3.5,6-di-0-isopropylideπe-L-fftreo-L-fa/o-octono-1 ,4-lactone Qf as a colourless oil (3.026 g, 81%): 5H (CDCI3) 0.11 (2 x ε, 6H, 2 x CH3), 0.91 (s, 9H, C(CH3)3), 1.30, 1.38, 1.41 , 1.47 (4 x s, 12H, 2 x C(CH3)2), 3.41- 3,45 (m, 1 H, H-7), 3.87 (dd, 1 H, J8a,7 5.37 Hz J8a,8b 10.81 Hz, H-8a), 3.92 (dd, 1 H, Jsb 7.32 Hz, H-8b), 4.19-4.24 (m, 2H, H-5, H-6), 4.61 (br s, 1 H, H-4), 4.75-4.79 (m, 2H, H-2, H-3); δc (CDCI3) -5.59, -5.56, 18.14, 25.54, 25.73, 26.09, 26.71 , 26.98, 61.61 , 63.19, 67.94, 74.84, 74.94, 75.47, 78.36, 78.66, 110.90, 113.37, 174.02; vma (film) 1796, 2111 ; [ ]D +36.7 (c I , CHCI3).
7-Azιdo-8-0-ferf-butyldimethylsilyl-7-deoxy-2,3:5,6-di-Q-isopropylidene-L-ff7reo-L-fa/o-octitol Qq
7-azido-8-0-ferf-butyldimelhylsilyl-7-deo<y-2,3:5,6-di-G-isopropylidene-L-f/7reo-L-fa/o-octono- 1 ,4-lactone (Qf, 3 00 g, 6.6 mmol) was dissolved in tetrahydrofuran (40 ml) and was cooled to 0 °C. Lithium borohydride (216 mg, 9.9 mmol) was added and the mixture waε stirred at 0 °C to room temperature for 24 h. T.l.c. analysiε (ethyl acetatexyclohexane, 1 :1) indicated the disappearance of the starting material (Rf 0.76) and the presence of a more polar compound (Rf 0.45). The reaction was quenched through the addition of ammonium chloride (sat. aq.) and the partitioned between ethyl acetate and brine. The aqueous layer was extracted with ethyl acetate (2 x) and the combined organic layers were dried (MgS04), filtered and the solvent removed. The resulting crude residue was purified by silica gel chromatography (ethyl acetate yclohexanε, 1 :3 to 1 :1) affording 7-azido-8-0-fert-butyldimεthylsilyl-7-deoxy-2,3:5,6-di-0-isopropylidene-L-ff//-eo-L-fa/o-octitol Qg as a colourlεss syrup (2 476 g, 82 %) δH (CDCI3) 0 10 (s, 6H, 2 x CH3), 0 91 (s, 9H, C(CH3)3), 1 36, 1 41 , 1 42, 1 48 (4 x s, 12H, 2 x C(CH3)2), 3 43-3 47 (m, 1 H, H-7), 3 66 (br d, 1 H, H-4), 3 79-3 92 (m, 4H, H-1 , H-1a, H-8, H-8a), 4 10-4 14 (m, 2H, H-2, H-3), 4 30-4 38 (m, 2H, H-5, H-6), δc (CDCI3) -5 61 , -5 51 , 18 14, 25 18, 25 71 , 26 87, 27 07, 27 86, 60 65, 62 39 63 66 67 62, 75 90 76 91 , 77 18, 77 49 108 63 110 16, \ (film) 2109 3536 [α]D +46 6 (c i , UIΪ 3)
7-Azιdo-8-0-fer.-butyldιmethylsιlyl-7-dεoxy-2,3 5,6-dι-0-ιsopropylιdene-1 ,4-dι-0-methanesulphonyl-L-fr)reo-L- fa/o-octιtol Qh
7-Azιdo-8-0-ferf-butyldιmethylsιlyl-7-deoxy-2,3 5,6-dι-0-ιsopropylιdene-L-f/7reo-L-fa/o-octιtol (Qg, 2 4 g, 5 3 mmol) was dissolved in pyπdine (20 ml) and was added to a solution of 4-dιmethylamιno pyπdine (64 mg, 0 53 mmol) and methanεsulfonyl chloπdε (4 814 g, 3 253 ml, 42 mmol) in pyπdine (20 ml) and stirred for 2 h T i c analysis (ethyl acetate cyclohexane, 1 2, double elution) revealed the disappearance of starting material (Rf 0 33) and the presence of a more hydrophobic product (Rf 0 43) The solvent was removεd under educed preεsure and the residue was partitioned betweεn εthyl acetate and brine The aqueous layer was extracted with ethyl acetate and the combined organic layers were dried (MgS04), filtered and concentrated under reduced pressure The resulting crude residue was purified by silica gel chromatography (ethyl acetate cyclohexane, 1 2) giving 7-azιdo-8-0-ferf-butyldιmethylsιlyl-7-dεoxy-2,3 5,6-dι-0-ιsopropylιdεne-1 ,4-dι- O-methanesulfonyl-L-fhreo-L-fa/o-octitol Qh as a colourlesε oil (2 973 g, 92 %) bπ (CDCI3) 0 11 , 0 12 (2 x ε, 6H, 2 x CH3), 0 91 (s, 9H, C(CH3)s), 1 41 , 1 44, 1 46, 1 56 (4 x s, 12H, 2 x C(CH3)2), 3 08 (s, 3H, SO2CH3), 3 21 (ε, 3H, S02CH3), 3 49 (ddd, I H, J 5 2 82 Hz, J7 8 5 46 Hz, J7 Ba 7 94 Hz, H-7), 3 87-3 97 (m, 2H, H-8, H-8a), 4 19 (dd, 1 H, J652 30 Hz, H-6), 424-4 31 (m, 2H, H-1 , H-5), 4 36 (dd, I H, J342 96 Hz, J326 62 Hz, H-3),
4 49-4 53 (m, 1 H, H-2), 4 69 (dd, 1 H, Jιa 2 2 39 Hz, J1a 1 10 83 Hz, H-1 a), 5 11 (app t, 1 H, H-4), δc (CDCI3) -
5 56, 18 18, 25 76, 26 24, 26 78, 26 89, 27 56, 37 75, 39 02, 60 90, 63 57, 70 44, 76 00, 76 07, 76 46, 77 18, 77 32, 109 01 , 110 68, vmaλ (film) 2113, [α]D -16 2 (c 1 , CHCI3)
7-Azιdo-7-deoxy-1 ,4-dι-0-methanesulphonyl-L-ff?reo-L-fa/o-octιtol Qi
7-A∑ιdo-8-0-fert-butyldιmεthylειlyl-7-deoλy-2,3 5,6-dι-0-i8opropylιdene-1 ,4-dι-0-methaneεulfonyl-L-f/?reo-L- fa/o-oclιtol (Qh, 2 90 g, 4 7 mmol) waε treated with a tπfluroacetic acid watεr mixture (1 1 , 40 ml) for 3 h T i c analysis (ethyl acetate) showed the disappearance of starting material (Rf 0 9) and the presence of a more polar product (Rf 0 12) The solvent was lemovεd under reduced pressure and the residue was co-evaporated with toluene and dried under vacuum Purification by silica gel chromatography (ethyl acetate cylcohexane, I I to i 0) yielded 7-azido-7-deoxy- 1 ,4-dι-0-methanesulphonyl-L-f/?reo-L-fa/o-octιtol Qi as a colourless oil ( I 677 g, 85 %) δH (CD3OD) 3 12 (3, 3H, S02C H3), 3 21 (ε, 3H, SO2CH3), 3 61-3 71 (m, 2H, H-7, H-8), 3 78-3 82 ( , 2H, H-6, H-8a), 3 98-4 05 (m, 2H, H-2, H-3), 4 1 1-4 13 (m, I H, H-5), 4 34 (dd, I H, J1 2 4 87 Hz, J 1a I 0 44 Hz, H- l), 445 (dd, 1H, 1a 2 1 87 Hz, H-1 a), 5 00 (dd, IH, J 3 1 91 Hz, J45 6 15 Hz, H-4), δc (CD3OD) 36 17, 38 I I , 61 84, 66 62, 69 09, 70 33, 70 45, 71 08, 72 55, 86 41 , vmaλ (film) 2113, [α]D -9 1 (c 1 , H20) (1 R,2R,3S,6S,7R,7aR)-3-(Hvdroxymethyl)-1 ,2,6,7-tetrahvdroxypyrrolιzιdιne Qi r3,7-d;ep;-Casuarιne1
7-Azιdo-7-deoxy-1 ,4-dι-0-methanεsulphonyl-L-f/7reo-L-fa/o-octιtol (Qi, 1 6 g, 3 78 mmol) was disεolvεd in watεr (30 ml) and was treated with 10 % palladium on carbon (400 mg) under an atmosphere of hydrogen for 16 h T i c analysis (ethyl acetate methanol, 9 1) indicatεd the disappearancε of starting material (Rf 0 75) and the presence of a more polar product (Rf 0 05) Palladium waε removεd by filtration and thε filtrate was treated with sodium acetate (930 mg, 11 34 mmol) at 60 °C for 16 h The reaction mixture was cooled and the solvent removed in vacua The crude brown oil was purified by lon-exchangε chromatography (Dowex 50WX8-100, eluting with 2M ammonium hydroxide) to afford (1 R,2R,3S,6S,7R,7aR)-3-(hydroxymethyl)-1 ,2,6,7- tetrahydroxypyrrolizidine [3,7-d/ep;-Casuaπne] Qj aε a brown glass (671 mg, 87%) δH (D20) 2 81-2 92 (m, 2H, H-5, H-5a), 3 16 (dd, 1 H, J2 5 91 Hz, J3 S 10 74 Hz, H-3), 3 30 (app t, 1 H, J 3 78 Hz, H-7a), 3 76 (dd, 1 H, J8 8a 6 35 Hz, H-8), 3 87 (dd, I H, H-8a), 4 01 (d, 1 H, J2ι 3 55 Hz, H-2), 4 04-4 12 (m, 2H, H-6, H-7), 4 29 (app t, I H, H- 1), δc (D20) 49 32, 57 29, 63 78, 70 41 , 72 59, 72 65, 74 47, 78 25, [ ]D -2 I 1 (c 0 5, H20)
Equivalents
The foregoing description details presently preferred embodiments of the present invention Numerous modifications and variations in practice thereof are expected to occur to those skilled in the art upon consideration of these descriptions Those modifications and variations are intended to be encompassed within the claims appended hereto

Claims

CLAIMS:
1. An isolated immunomodulatory (e.g. immunostimulatory) polyhydroxylated pyrrolizidine compound for use in therapy or prophylaxis having the formula:
Figure imgf000057_0001
CH OH
wherein R is selεctεd from the group comprising hydrogen, straight or branched, unsubstituted or substituted, saturated or unsaturated acyl, alkyl (e.g. cycloalkyl), alkenyl, alkynyl and aryl groups, or a pharmaceutically acceptablε salt or dεrivative thereof.
The compound of claim 1 having the formula:
Figure imgf000057_0002
wherein R is selected from the group comprising hydrogen, straight or branched, unsubεtituted or εubstituted, saturated or unsaturated acyl, alkyl (e.g. cycloalkyl), alkenyl, alkynyl and aryl groups, or a pharmaceutically acceptable salt or derivative thereof.
3. The compound of claim 1 or claim 2 which induces, potentiates or activates one or more cytokines (e.g. Thl cytokines) in vivo and/or suppresses one or more cytokines (e.g. Th2 cytokinε(s)) in vivo.
4. The compound of claim 3 wherein the one or more cytokines comprises one or more inlerleukinε.
5. The compound of claim A wherein the one or more interleukins induced, potentiated or activated comprises IL-12 and/or 11-2 (e.g. in dendritic cells).
6. The compound of any one of the preceding claims which is a glycosidase inhibitor.
7. The compound of claim 6 which inhibits glucosidase.
8. The compound of any one of the preceding claims which does not inhibit mannosidase.
9. The compound of any one of the precεding claims which:
(a) modifies tumour cell glycosylation (e.g. tumour antigen glycosylation); and/or
(b) modifies viral protein glycosylation (e.g. virion antigen glycosylation); and/or
(c) modifies cell-surface protein glycosylation in infected host cells; and/or
(d) modifies bacterial cell walls, when administered in vivo.
10. The compound of any one of the preceding claimε which is an acyl derivative.
1 1. The compound of claim 10 which is:
(a) peracylated; or
(b) acylated at C-3 hydroxymethyl; or
(c) acylated at C-6; (d) acylated at C-3 hydroxymethyl and C-6.
12. The compound of claim 10 or claim 1 1 wherein the acyl derivative is alkanoyl or aroyl.
13. The compound of claim 12 wherein the acyl derivative is an alkanoyl selectεd from acetyl, propanoyl or butanoyl.
14. The compound of any one of claimε 1 to 13 wherein R is a saccharide moiety.
15. The compound of claim 14 wherein R is a glucoside or arabinoside moiety.
16. The compound of claim 2 which is 1 R,2R,3R,6S,7S,7aR)-3-(hydroxymethyl)-1 ,2,6,7- tetrahydroxypyrroli∑idine (casuarine), wherein R is hydrogen and having the formula:
Figure imgf000058_0001
CH OH
or a pharmaceutically acceptable salt or derivative thereof.
17. The compound of claim 2 which is a casuarine glycoside, or a pharmaceutically acceptable salt or derivative thereof.
18. The compound of claim 17 which is casuariπe-6-α-D-glucoside of the formula:
Figure imgf000059_0001
or a pharmacεutically acceptable salt or derivative thereof.
19. The compound of claim 2 which is 6-O-butanoylcasuarine, or a pharmaceutically acceptablε salt or dεrivative thereof.
20. The compound of claim 1 which is selεcted from: (a) 3,7-d/ep/-casuarine;
(b) 7-ep/-casuarine;
(c) 3,6,7-friep/-casuarine;
(d) 6,7-d/ep/-casuarinε;
(e) 3-ep/-casuarine; (f) 3,7-d/ep/-casuarine-6-α-D-glucoside;
(g) 7-ep/-casuarine-6-α-D-glucoside;
(h) 3,6,7-fπ'ep/-Gaεuarine-6-α-D-glucoside;
(i) 6,7-d/ep/-caεuarine-6-α-D-glucosidε; and
(j) 3-ep/-casuarine-6-α-D-glucoside, or a pharmaceutically acceptable salt or derivative thereof.
21. A method for immunomodulation (e.g. immunostimulation) comprising administering to a patient a composition comprising a polyhydroxylated pyrrolizidine compound having the formula:
Figure imgf000059_0002
wherein R is selεctεd from the group comprising hydrogen, straight or branched, unsubstituted or substituted, saturated or unsaturated acyl, alkyl (e.g. cycloalkyl), alkenyl, alkynyl and aryl groups, or a pharmaceutically acceptable salt or derivative thereof.
22. The method of claim 21 wherein the compound has the formula:
Figure imgf000060_0001
wherein R is selεctεd from the group comprising hydrogen, straight or branched, unsubstituted or substituted, saturated or unsaturated acyl, alkyl (e.g. cycloalkyl), alkenyl, alkynyl and aryl groupε, or a pharmaceutically acceptablε salt or derivative thereof.
23. The method of claim 21 or claim 22 wherein the compound is as defined in any one of claimε 1 to 20.
24. The method of any one of claimε 21 to 23 wherein the composition comprises an isolated compound or a combination of one or more of the compounds as defined in any one of claims (for example wherein the composition comprises a combination of casuarine and caεuarine-6-α-D-glucoside).
25. The method of any one of claims 21 to 24 wherein the composition is a herbal medicinε.
26. Thε mεthod of claim 25 whεrεin the botanic source of the herbal medicine compriseε one or more plant speciεs sεlεctεd from:
(a) a member of the taxon Myrtaceae (for example Myrtus spp. (e.g. M. communis), Syzygium spp. (e.g. S. guineense) or Eugenia spp. (e.g. E. jambolana); or (b) a member of the taxon Casuannaceae;
(c) a combination of two or more plant speciεs selectεd from both of the taxons of (a) and (b).
27. The method of any one of claims 21 to 26 which comprises haemorestoration.
28. The method of claim 27 wherεin the haemorestoration is adjunctive to:
(a) chemotherapy; and/or
(b) radiotherapy; and/or
(c) bone marrow transplantation; and/or
(d) haemoablative immunotherapy.
29. The method of any one of claimε 21 to 28 which comprises the alleviation of immunosuppression.
30. The method of claim 29 wherein the immunosuppression is congenital, acquired (e.g. by infection or malignancy) or induced (e.g. deliberately as part of the managemεnt of tranεplantε or cancerε).
31. The method of any one of claims 21 to 30 which comprises induction, potentiation or activation of one or more cytokines (for exarriplε IL- 12 and/or IL-2) in vivo.
32. The method of any one of claims 21 to 30 which comprises the treatmεnt of proliferative disorderε, for example proliferativε disordεrs selected from cancer and cancer metastasis.
33. A method for chemoprotεction comprising administεring thε compound as dεfined in any one of claims 1 to 20 or composition as defined in any one ot claims 21 to 32 to a patient undergoing chemotherapy.
34. Use of the polyhydroxylated pyrrolizidine compound as defined in any one of claims 1 to 20 or composition aε definεd in any onε of claimε 21 to 32 for the manufacture of a medicamεnt for uεε in immunomodulation (e.g. immunoεtimulation) and/or chemoprotection.
35. A proceεs for the manufacture of a medicament for uεe in immunomodulation (e.g. immunostimulation) and/or chemoprotection, characterized in the use of the polyhydroxylated pyrrolizidine compound as defined in any one of claims 1 to 20 or composition as defined in any one of claimε 21 to 32.
36. The use of claim 34 or process of claim 35 wherein the immunomodulation and/or chemoprotection is as defined in any one of claims 21 to 33.
37. A composition comprising a polyhydroxylated pyrrolizidine compound as defined in any one of the preceding claimε in combination with: (a) an immunoεtimuiant; and/or
(b) a cytotoxic agent (e.g. cyclophoεphamide, cortisone acetatε, vinblastinε, vincristine, adriamycin, 6-mercaptopurine, 5-fluorouracil, mitomycin C or chloramphenicol); and/or
(c) an antimicrobial (e.g. antibacterial) agent; and/or
(d) an antiviral agent (e.g. AZT) (e) a dendritic cell (e.g. a primed dendritic cell).
38. The composition of claim 37 further comprising a pharmaceutically acceptable excipient.
39. A vaccine comprising a polyhydroxylated pyrrolizidine compound as definεd in any one of claimε 1 to 20 or composition as defined in any one of claims 21 to 32 in combination with an antigen, the compound being preεent in an amount εufficient to produce an adjuvant effect on vaccination.
40. A pharmaceutical kit of parts comprising a polyhydroxylated pyrrolizidine compound aε defined in any one of claims 1 to 20 or composition aε defined in any one of claims 21 to 32 in combination with any or all of the adjunctive therapeutic agents defined in claim 37(a)-(d).
4 I . The kit of claim 40 further comprising instructions for use.
42. The compound of any one of claims 1 to 20 for use in therapy or prophylaxis, wherein the therapy or prophylaxis comprises:
(a) Increasing the Th1 :Th2 reεponse ratio;
(b) Haemorestoration; (c) Alleviation of immunosuppression;
(d) Cytokine stimulation;
(e) Treatment of proliferativε disorders (e.g. cancer);
(f) Vaccination, wherεin the compound acts as an adjuvant;
(g) Vaccination with a dendritic cell vaccine (e.g. a primed dendritic cell vaccine), wherεin the utsnuuui- cells are contacted with the compound;
(h) Adminiεtration of dendritic cellε in thε treatment or prophylaxiε of autoimmune disorders, wherein the dendritic cells are contacted with the compound; and/or (i) Wound healing; (j) Stimulating the innate immune response;
(k) Boosting the activity of endogenouε NK cεlls.
43. Thε compound of any one of claims 1 to 20 wherεin the therapy or prophylaxis comprises:
(a) the treatment of Th1-related diseaεeε or disorders;
(b) the treatment of Th2-related disεaεεε or disorderε (for example allergies, e.g. asthma);
(c) the treatment of bacterial infectionε;
(d) the treatment of viral infectionε;
(e) the treatment of prion (e.g. BSE and CJD), fungal, protozoan or metazoan infectionε; (f) the treatment of diseases associated with intracellular pathogens (e.g. leishmaniasis, trypanosomiasiε or malaria).
' 44. The compound of claim 43 (d) wherein the viral infection is selected from respiratory syncytial virus (RSV), hεpatitis B virus (HBV), Epstein-Barr, hepatitis C virus (HCV), herpes simplex type 1 and 2, herpes genitalis, herpes keratitis, herpeε encephalitis, herpes zoster, human immunodeficiency virus (HIV), influenza A virus, hantann virus (hemorrhagic fever), human papilloma virus (HPV) and measleε.
PCT/GB2004/000198 2003-01-23 2004-01-21 Polyhydroxylated pyrrolizidine WO2004064715A2 (en)

Priority Applications (9)

Application Number Priority Date Filing Date Title
AU2004206085A AU2004206085B2 (en) 2003-01-23 2004-01-21 Polyhydroxylated pyrrolizidine
EP04703841A EP1587480B1 (en) 2003-01-23 2004-01-21 Polyhydroxylated pyrrolizidine
JP2006500223A JP5289706B2 (en) 2003-01-23 2004-01-21 Immunomodulatory composition
NZ541839A NZ541839A (en) 2003-01-23 2004-01-21 Polyhydroxylated pyrrolizidine
ES04703841T ES2394072T3 (en) 2003-01-23 2004-01-21 Polyhydroxylated Pyrrolizidine
CA2513881A CA2513881C (en) 2003-01-23 2004-01-21 Immunomodulatory compositions
US10/543,014 US8383665B2 (en) 2003-01-23 2004-01-21 Immunomodulatory compositions
HK06104746.3A HK1084576A1 (en) 2003-01-23 2006-04-20 Polyhydroxylated pyrrolizidine
US13/742,102 US8557859B2 (en) 2003-01-23 2013-01-15 Immunomodulatory compositions

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
GB0301554.2 2003-01-23
GBGB0301554.2A GB0301554D0 (en) 2003-01-23 2003-01-23 Immunostimulatory compositions

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US10/543,014 A-371-Of-International US8383665B2 (en) 2003-01-23 2004-01-21 Immunomodulatory compositions
US13/742,102 Division US8557859B2 (en) 2003-01-23 2013-01-15 Immunomodulatory compositions

Publications (2)

Publication Number Publication Date
WO2004064715A2 true WO2004064715A2 (en) 2004-08-05
WO2004064715A3 WO2004064715A3 (en) 2004-12-23

Family

ID=9951664

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/GB2004/000198 WO2004064715A2 (en) 2003-01-23 2004-01-21 Polyhydroxylated pyrrolizidine

Country Status (11)

Country Link
US (2) US8383665B2 (en)
EP (1) EP1587480B1 (en)
JP (1) JP5289706B2 (en)
CN (1) CN1761666A (en)
AU (1) AU2004206085B2 (en)
CA (1) CA2513881C (en)
ES (1) ES2394072T3 (en)
GB (1) GB0301554D0 (en)
HK (1) HK1084576A1 (en)
NZ (1) NZ541839A (en)
WO (1) WO2004064715A2 (en)

Cited By (57)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005070415A1 (en) * 2004-01-21 2005-08-04 M N L Pharma Limited Adjuvant compositions
WO2005070418A1 (en) * 2004-01-21 2005-08-04 M N L Pharma Limited Immunomodulatory alkaloids
WO2006008493A1 (en) * 2004-07-23 2006-01-26 Mnl Pharma Limited Synthesis of polyhydroxylated alkaloids
WO2006067419A2 (en) * 2004-12-21 2006-06-29 Mnl Pharma Limited Myrtaceous honey and the use thereof as immunomodulator
WO2006077427A2 (en) * 2005-01-21 2006-07-27 Mnl Pharma Limited Antiviral drug combinations
WO2008020335A2 (en) 2006-06-09 2008-02-21 Novartis Ag Immunogenic compositions for streptococcus agalactiae
WO2007075605A3 (en) * 2005-12-19 2008-06-26 Abbott Lab Use of beta-hydroxy-beta-methylbutyrate to modulate the imbalance in type and type 2 cytokine production
WO2008012555A3 (en) * 2006-07-27 2008-09-25 Isis Innovation Epitope reduction therapy
WO2010049678A2 (en) * 2008-10-31 2010-05-06 Summit Corporation Plc Treatment of energy utilization diseases
WO2010100632A2 (en) 2009-03-06 2010-09-10 Novartis Ag Chlamydia antigens
WO2011004263A2 (en) 2009-07-07 2011-01-13 Novartis Ag Conserved escherichia coli immunogens
WO2011008974A2 (en) 2009-07-15 2011-01-20 Novartis Ag Rsv f protein compositions and methods for making same
WO2011007257A1 (en) 2009-07-16 2011-01-20 Novartis Ag Detoxified escherichia coli immunogens
EP2298795A1 (en) 2005-02-18 2011-03-23 Novartis Vaccines and Diagnostics, Inc. Immunogens from uropathogenic escherichia coli
WO2011048561A1 (en) 2009-10-20 2011-04-28 Novartis Ag Diagnostic and therapeutic methods for rheumatic heart disease based upon group a streptococcus markers
WO2011058302A1 (en) 2009-11-10 2011-05-19 Guy's And St Thomas's Nhs Foundation Trust Bacteremia-associated antigen from staphylococcus aureus
WO2011080595A2 (en) 2009-12-30 2011-07-07 Novartis Ag Polysaccharide immunogens conjugated to e. coli carrier proteins
EP2351772A1 (en) 2005-02-18 2011-08-03 Novartis Vaccines and Diagnostics, Inc. Proteins and nucleic acids from meningitis/sepsis-associated Escherichia coli
EP2357184A1 (en) 2006-03-23 2011-08-17 Novartis AG Imidazoquinoxaline compounds as immunomodulators
EP2368572A2 (en) 2005-11-04 2011-09-28 Novartis Vaccines and Diagnostics S.r.l. Adjuvanted vaccines with non-virion antigens prepared from influenza viruses grown in cell culture
EP2368573A2 (en) 2005-11-04 2011-09-28 Novartis Vaccines and Diagnostics S.r.l. Influenza vaccines including combinations of particulate adjuvants and immunopotentiators
EP2382988A1 (en) 2006-03-31 2011-11-02 Novartis AG Combined mucosal and parenteral immunization against HIV
EP2382987A1 (en) 2006-03-24 2011-11-02 Novartis Vaccines and Diagnostics GmbH Storage of influenza vaccines without refrigeration
WO2011149564A1 (en) 2010-05-28 2011-12-01 Tetris Online, Inc. Interactive hybrid asynchronous computer game infrastructure
WO2011161551A2 (en) 2010-06-11 2011-12-29 Novartis Ag Omv vaccines
WO2012006359A1 (en) 2010-07-06 2012-01-12 Novartis Ag Delivery of self-replicating rna using biodegradable polymer particles
US8217077B2 (en) 2004-03-26 2012-07-10 Abbott Laboratories HMB uses thereof
EP2478916A1 (en) 2006-01-27 2012-07-25 Novartis Vaccines and Diagnostics GmbH Influenza vaccines containing hemagglutinin and matrix proteins
WO2012103361A1 (en) 2011-01-26 2012-08-02 Novartis Ag Rsv immunization regimen
EP2484377A1 (en) 2007-06-27 2012-08-08 Novartis AG Low-additive influenza vaccines
EP2497495A2 (en) 2006-09-11 2012-09-12 Novartis AG Making influenza virus vaccines without using eggs
EP2510947A1 (en) 2009-04-14 2012-10-17 Novartis AG Compositions for immunising against Staphylococcus aureus
EP2514437A1 (en) 2006-07-20 2012-10-24 Novartis AG Frozen stockpiling of influenza vaccines
WO2012158613A1 (en) 2011-05-13 2012-11-22 Novartis Ag Pre-fusion rsv f antigens
EP2532362A1 (en) 2006-12-06 2012-12-12 Novartis AG Vaccines including antigen from four strains of influenza virus
WO2013009564A1 (en) 2011-07-08 2013-01-17 Novartis Ag Tyrosine ligation process
EP2548895A1 (en) 2007-01-11 2013-01-23 Novartis AG Modified saccharides
EP2572726A1 (en) 2007-08-01 2013-03-27 Novartis AG Compositions comprising pneumococcal antigens
EP2586790A2 (en) 2006-08-16 2013-05-01 Novartis AG Immunogens from uropathogenic Escherichia coli
WO2013068949A1 (en) 2011-11-07 2013-05-16 Novartis Ag Carrier molecule comprising a spr0096 and a spr2021 antigen
EP2614835A1 (en) 2007-11-26 2013-07-17 Novartis AG Vaccination with multiple clades of H5 influenza A virus
EP2614709A1 (en) 2005-07-18 2013-07-17 Novartis AG Small animal model for HCV replication
WO2013108272A2 (en) 2012-01-20 2013-07-25 International Centre For Genetic Engineering And Biotechnology Blood stage malaria vaccine
USH2283H1 (en) 2009-04-27 2013-09-03 Novartis Ag Vaccines for protecting against influenza
EP2659912A2 (en) 2007-07-17 2013-11-06 Novartis AG Conjugate purification
WO2014053521A2 (en) 2012-10-02 2014-04-10 Novartis Ag Nonlinear saccharide conjugates
US8916217B2 (en) 2010-01-29 2014-12-23 Abbott Laboratories Aseptically packaged nutritional liquids comprising HMB
WO2015068129A1 (en) 2013-11-08 2015-05-14 Novartis Ag Salmonella conjugate vaccines
EP2889042A2 (en) 2008-03-18 2015-07-01 Novartis AG Improvements in preparation of influenza virus vaccine antigens
EP2891498A1 (en) 2007-12-20 2015-07-08 Novartis AG Fermentation processes for cultivating streptococci and purification processes for obtaining CPS therefrom
US9241508B2 (en) 2010-01-29 2016-01-26 Abbott Laboratories Nutritional emulsions comprising calcium HMB
US9521859B2 (en) 2010-06-10 2016-12-20 Normanella T. Dewille Substantially clear nutritional liquids comprising calcium HMB and soluble protein
US9693577B2 (en) 2010-01-29 2017-07-04 Abbott Laboratories Method of preparing a nutritional powder comprising spray dried HMB
EP3345617A1 (en) 2012-11-30 2018-07-11 GlaxoSmithKline Biologicals S.A. Pseudomonas antigens and antigen combinations
US10085952B2 (en) 2012-11-27 2018-10-02 Avexxin As Dermatitis treatment
US10953004B2 (en) 2016-03-14 2021-03-23 Avexxin As Combination therapy for proliferative diseases
US11351127B2 (en) 2016-09-21 2022-06-07 Avexxin As Pharmaceutical composition

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102659787B (en) * 2011-03-29 2015-04-01 中国科学院化学研究所 Fluoro polyhydroxy pyrrole pyrrolizidine, and preparation method and application thereof
US20150306139A1 (en) * 2011-04-21 2015-10-29 Jeffrey Ravetch Anti-inflammatory agents
CN103435619B (en) * 2013-09-04 2015-08-05 中国科学院化学研究所 A kind of Pochonicine analogue or its pharmacy acceptable salt and application thereof

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2000072770A (en) * 1998-08-31 2000-03-07 Yasunori Momose New pentahydroxyindolizidine compound and alpha glucosidase inhibitor

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
None

Cited By (90)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005070418A1 (en) * 2004-01-21 2005-08-04 M N L Pharma Limited Immunomodulatory alkaloids
WO2005070415A1 (en) * 2004-01-21 2005-08-04 M N L Pharma Limited Adjuvant compositions
US8217077B2 (en) 2004-03-26 2012-07-10 Abbott Laboratories HMB uses thereof
US8778994B2 (en) 2004-03-26 2014-07-15 Abbott Laboratories Method of using beta-hydroxy-beta-methylbutyrate and fatty acids for treating disease-associated wasting
US8778993B2 (en) 2004-03-26 2014-07-15 Abbott Laboratories Method of using β-hydroxy-β-methylbutyrate for the treatment of disease conditions
US8785495B2 (en) 2004-03-26 2014-07-22 Abbott Laboratories Compositions including beta-hydroxy-beta-methylbutyrate
US8785496B2 (en) 2004-03-26 2014-07-22 Abbott Laboratories Method of using beta-hydroxy-beta-methylbutyrate for treating disease-associated wasting
US8609725B2 (en) 2004-03-26 2013-12-17 Abbott Laboratories Method of using beta-hydroxy-beta-methylbutyrate for reducing tumor growth rate
WO2006008493A1 (en) * 2004-07-23 2006-01-26 Mnl Pharma Limited Synthesis of polyhydroxylated alkaloids
WO2006067419A2 (en) * 2004-12-21 2006-06-29 Mnl Pharma Limited Myrtaceous honey and the use thereof as immunomodulator
WO2006067419A3 (en) * 2004-12-21 2007-03-29 Mnl Pharma Ltd Myrtaceous honey and the use thereof as immunomodulator
WO2006077427A3 (en) * 2005-01-21 2006-09-14 Mnl Pharma Ltd Antiviral drug combinations
WO2006077427A2 (en) * 2005-01-21 2006-07-27 Mnl Pharma Limited Antiviral drug combinations
EP2298795A1 (en) 2005-02-18 2011-03-23 Novartis Vaccines and Diagnostics, Inc. Immunogens from uropathogenic escherichia coli
EP2351772A1 (en) 2005-02-18 2011-08-03 Novartis Vaccines and Diagnostics, Inc. Proteins and nucleic acids from meningitis/sepsis-associated Escherichia coli
EP2614709A1 (en) 2005-07-18 2013-07-17 Novartis AG Small animal model for HCV replication
EP3714900A1 (en) 2005-11-04 2020-09-30 Seqirus UK Limited Adjuvanted vaccines with non-virion antigens prepared from influenza viruses grown in cell culture
EP2368573A2 (en) 2005-11-04 2011-09-28 Novartis Vaccines and Diagnostics S.r.l. Influenza vaccines including combinations of particulate adjuvants and immunopotentiators
EP2368572A2 (en) 2005-11-04 2011-09-28 Novartis Vaccines and Diagnostics S.r.l. Adjuvanted vaccines with non-virion antigens prepared from influenza viruses grown in cell culture
US8796333B2 (en) 2005-12-19 2014-08-05 Abbott Laboratories Method of using β-hydroxy-β-methylbutyrate to treat a condition
WO2007075605A3 (en) * 2005-12-19 2008-06-26 Abbott Lab Use of beta-hydroxy-beta-methylbutyrate to modulate the imbalance in type and type 2 cytokine production
RU2469719C2 (en) * 2005-12-19 2012-12-20 Абботт Лаборэтриз METHOD OF TREATING ALLERGY, METHOD OF TREATING ASTHMA, METHOD OF REDUCING RISK OF DEVELOPING INFECTION, AND METHOD OF TREATING CONDITION CHARACTERISED BY CYTOKINE TYPE 1 AND 2 CONTENT IMBALANCE BY β-HYDROXY-β-METHYLBUTYRATE
US8778992B2 (en) 2005-12-19 2014-07-15 Abbott Laboratories Method of using beta-hydroxy-beta-methylbutyrate to treat allergies and asthma
EP3753574A1 (en) 2006-01-27 2020-12-23 Seqirus UK Limited Influenza vaccines containing hemagglutinin and matrix proteins
EP2478916A1 (en) 2006-01-27 2012-07-25 Novartis Vaccines and Diagnostics GmbH Influenza vaccines containing hemagglutinin and matrix proteins
EP2357184A1 (en) 2006-03-23 2011-08-17 Novartis AG Imidazoquinoxaline compounds as immunomodulators
EP2382987A1 (en) 2006-03-24 2011-11-02 Novartis Vaccines and Diagnostics GmbH Storage of influenza vaccines without refrigeration
EP2382988A1 (en) 2006-03-31 2011-11-02 Novartis AG Combined mucosal and parenteral immunization against HIV
WO2008020335A2 (en) 2006-06-09 2008-02-21 Novartis Ag Immunogenic compositions for streptococcus agalactiae
EP2514437A1 (en) 2006-07-20 2012-10-24 Novartis AG Frozen stockpiling of influenza vaccines
WO2008012555A3 (en) * 2006-07-27 2008-09-25 Isis Innovation Epitope reduction therapy
EP2586790A2 (en) 2006-08-16 2013-05-01 Novartis AG Immunogens from uropathogenic Escherichia coli
EP2497495A2 (en) 2006-09-11 2012-09-12 Novartis AG Making influenza virus vaccines without using eggs
EP3456348A1 (en) 2006-09-11 2019-03-20 Seqirus UK Limited Making influenza virus vaccines without using eggs
EP2532362A1 (en) 2006-12-06 2012-12-12 Novartis AG Vaccines including antigen from four strains of influenza virus
EP2679240A1 (en) 2006-12-06 2014-01-01 Novartis AG Vaccines including antigen from four strains of influenza virus
EP2548895A1 (en) 2007-01-11 2013-01-23 Novartis AG Modified saccharides
EP2484377A1 (en) 2007-06-27 2012-08-08 Novartis AG Low-additive influenza vaccines
EP2659912A2 (en) 2007-07-17 2013-11-06 Novartis AG Conjugate purification
US9463250B2 (en) 2007-07-17 2016-10-11 Glaxosmithkline Biologicals Sa Conjugate purification
EP2572726A1 (en) 2007-08-01 2013-03-27 Novartis AG Compositions comprising pneumococcal antigens
EP2614835A1 (en) 2007-11-26 2013-07-17 Novartis AG Vaccination with multiple clades of H5 influenza A virus
EP2891498A1 (en) 2007-12-20 2015-07-08 Novartis AG Fermentation processes for cultivating streptococci and purification processes for obtaining CPS therefrom
EP3459563A1 (en) 2008-03-18 2019-03-27 Seqirus UK Limited Improvements in preparation of influenza virus vaccine antigens
EP2889042A2 (en) 2008-03-18 2015-07-01 Novartis AG Improvements in preparation of influenza virus vaccine antigens
WO2010049678A2 (en) * 2008-10-31 2010-05-06 Summit Corporation Plc Treatment of energy utilization diseases
WO2010049678A3 (en) * 2008-10-31 2010-08-26 Summit Corporation Plc Treatment of energy utilization diseases
WO2010100632A2 (en) 2009-03-06 2010-09-10 Novartis Ag Chlamydia antigens
EP3549602A1 (en) 2009-03-06 2019-10-09 GlaxoSmithKline Biologicals S.A. Chlamydia antigens
EP3263128A2 (en) 2009-04-14 2018-01-03 GlaxoSmithKline Biologicals S.A. Compositions for immunising against staphylococcus aureus
EP2510947A1 (en) 2009-04-14 2012-10-17 Novartis AG Compositions for immunising against Staphylococcus aureus
USH2284H1 (en) 2009-04-27 2013-09-03 Novartis Ag Vaccines for protecting against influenza
USH2283H1 (en) 2009-04-27 2013-09-03 Novartis Ag Vaccines for protecting against influenza
WO2011004263A2 (en) 2009-07-07 2011-01-13 Novartis Ag Conserved escherichia coli immunogens
EP3988115A2 (en) 2009-07-15 2022-04-27 GlaxoSmithKline Biologicals S.A. Rsv f protein compositions and methods for making same
EP4183412A1 (en) 2009-07-15 2023-05-24 GlaxoSmithKline Biologicals S.A. Rsv f protein compositions and methods for making same
EP3178490A2 (en) 2009-07-15 2017-06-14 GlaxoSmithKline Biologicals S.A. Rsv f protein compositions and methods for making same
EP4218799A1 (en) 2009-07-15 2023-08-02 GlaxoSmithKline Biologicals S.A. Rsv f protein compositions and methods for making same
WO2011008974A2 (en) 2009-07-15 2011-01-20 Novartis Ag Rsv f protein compositions and methods for making same
EP4218800A1 (en) 2009-07-15 2023-08-02 GlaxoSmithKline Biologicals S.A. Rsv f protein compositions and methods for making same
EP2837386A1 (en) 2009-07-16 2015-02-18 Novartis AG Detoxified Escherichia coli immunogens
WO2011007257A1 (en) 2009-07-16 2011-01-20 Novartis Ag Detoxified escherichia coli immunogens
WO2011048561A1 (en) 2009-10-20 2011-04-28 Novartis Ag Diagnostic and therapeutic methods for rheumatic heart disease based upon group a streptococcus markers
WO2011058302A1 (en) 2009-11-10 2011-05-19 Guy's And St Thomas's Nhs Foundation Trust Bacteremia-associated antigen from staphylococcus aureus
WO2011080595A2 (en) 2009-12-30 2011-07-07 Novartis Ag Polysaccharide immunogens conjugated to e. coli carrier proteins
US8916217B2 (en) 2010-01-29 2014-12-23 Abbott Laboratories Aseptically packaged nutritional liquids comprising HMB
US9241508B2 (en) 2010-01-29 2016-01-26 Abbott Laboratories Nutritional emulsions comprising calcium HMB
US9693577B2 (en) 2010-01-29 2017-07-04 Abbott Laboratories Method of preparing a nutritional powder comprising spray dried HMB
WO2011149564A1 (en) 2010-05-28 2011-12-01 Tetris Online, Inc. Interactive hybrid asynchronous computer game infrastructure
US9521859B2 (en) 2010-06-10 2016-12-20 Normanella T. Dewille Substantially clear nutritional liquids comprising calcium HMB and soluble protein
WO2011161551A2 (en) 2010-06-11 2011-12-29 Novartis Ag Omv vaccines
EP3399021A1 (en) 2010-06-11 2018-11-07 GlaxoSmithKline Biologicals S.A. Omv vaccines
EP3611269A1 (en) 2010-07-06 2020-02-19 GlaxoSmithKline Biologicals SA Delivery of self-replicating rna using biodegradable polymer particles
WO2012006359A1 (en) 2010-07-06 2012-01-12 Novartis Ag Delivery of self-replicating rna using biodegradable polymer particles
EP4144368A1 (en) 2011-01-26 2023-03-08 GlaxoSmithKline Biologicals S.A. Rsv immunization regimen
EP4159232A1 (en) 2011-01-26 2023-04-05 GlaxoSmithKline Biologicals S.A. Rsv immunization regimen
EP3527224A1 (en) 2011-01-26 2019-08-21 GlaxoSmithKline Biologicals S.A. Rsv immunization regimen
WO2012103361A1 (en) 2011-01-26 2012-08-02 Novartis Ag Rsv immunization regimen
WO2012158613A1 (en) 2011-05-13 2012-11-22 Novartis Ag Pre-fusion rsv f antigens
EP3275892A2 (en) 2011-05-13 2018-01-31 GlaxoSmithKline Biologicals S.A. Pre-fusion rsv f antigens
WO2013009564A1 (en) 2011-07-08 2013-01-17 Novartis Ag Tyrosine ligation process
WO2013068949A1 (en) 2011-11-07 2013-05-16 Novartis Ag Carrier molecule comprising a spr0096 and a spr2021 antigen
WO2013108272A2 (en) 2012-01-20 2013-07-25 International Centre For Genetic Engineering And Biotechnology Blood stage malaria vaccine
WO2014053521A2 (en) 2012-10-02 2014-04-10 Novartis Ag Nonlinear saccharide conjugates
US10758499B2 (en) 2012-11-27 2020-09-01 Avexxin As Dermatitis treatment
US10085952B2 (en) 2012-11-27 2018-10-02 Avexxin As Dermatitis treatment
EP3345617A1 (en) 2012-11-30 2018-07-11 GlaxoSmithKline Biologicals S.A. Pseudomonas antigens and antigen combinations
WO2015068129A1 (en) 2013-11-08 2015-05-14 Novartis Ag Salmonella conjugate vaccines
US10953004B2 (en) 2016-03-14 2021-03-23 Avexxin As Combination therapy for proliferative diseases
US11351127B2 (en) 2016-09-21 2022-06-07 Avexxin As Pharmaceutical composition

Also Published As

Publication number Publication date
US20130209516A1 (en) 2013-08-15
NZ541839A (en) 2009-02-28
US8383665B2 (en) 2013-02-26
ES2394072T3 (en) 2013-01-16
US20070155814A1 (en) 2007-07-05
EP1587480B1 (en) 2012-06-27
HK1084576A1 (en) 2006-08-04
JP2006515357A (en) 2006-05-25
WO2004064715A3 (en) 2004-12-23
CA2513881C (en) 2013-04-02
CN1761666A (en) 2006-04-19
AU2004206085B2 (en) 2011-03-24
US8557859B2 (en) 2013-10-15
JP5289706B2 (en) 2013-09-11
AU2004206085A1 (en) 2004-08-05
CA2513881A1 (en) 2004-08-05
EP1587480A2 (en) 2005-10-26
GB0301554D0 (en) 2003-02-26

Similar Documents

Publication Publication Date Title
US8557859B2 (en) Immunomodulatory compositions
AU2005205962B2 (en) Immunomodulatory alkaloids
AU2009268381B2 (en) Alpha-galactosyl ceramide analogs and their use as immunotherapies, adjuvants, and antiviral, antibacterial, and anticancer agents
US10730871B2 (en) Immunomodulators and immunomodulator conjugates
EP1711174B1 (en) Adjuvant compositions
KR101487935B1 (en) A pharmaceutical composition comprising extract or fraction of Salvia plebeia R. Br. for preventing or treating STAT3-mediated disease
WO2007010266A1 (en) Pyrrolidine compositions
WO2006067419A2 (en) Myrtaceous honey and the use thereof as immunomodulator
KR20210063935A (en) Immune-boosting composition and Health functional food containing the same
WO2023027527A1 (en) Immunopotentiation composition comprising artemisia gmelinii extract
KR101509225B1 (en) Novel triterpenoids from acanthopanax sessiliflorus and an anti-inflammatory composition containing it
Pauwels Synthesis of new α-GalCer analogues as iNKT cell targeting agents
JP2002212079A (en) Endotoxin inhibitor containing diacylglycerol derivative as active ingredient
GB2461656A (en) Alpha-galactosyl ceramide analogues and their use as immunotherapies, adjuvants, and antiviral, antibacterial and anticancer agents

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BW BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE EG ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NA NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): BW GH GM KE LS MW MZ SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IT LU MC NL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

DPEN Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed from 20040101)
121 Ep: the epo has been informed by wipo that ep was designated in this application
WWE Wipo information: entry into national phase

Ref document number: 2004703841

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 3195/DELNP/2005

Country of ref document: IN

WWE Wipo information: entry into national phase

Ref document number: 2513881

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 2006500223

Country of ref document: JP

WWE Wipo information: entry into national phase

Ref document number: 2004206085

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 541839

Country of ref document: NZ

ENP Entry into the national phase

Ref document number: 2004206085

Country of ref document: AU

Date of ref document: 20040121

Kind code of ref document: A

WWP Wipo information: published in national office

Ref document number: 2004206085

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 20048074086

Country of ref document: CN

WWP Wipo information: published in national office

Ref document number: 2004703841

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 10543014

Country of ref document: US

WWP Wipo information: published in national office

Ref document number: 10543014

Country of ref document: US