US20210140972A1 - Mechanism of action - Google Patents

Mechanism of action Download PDF

Info

Publication number
US20210140972A1
US20210140972A1 US17/072,665 US202017072665A US2021140972A1 US 20210140972 A1 US20210140972 A1 US 20210140972A1 US 202017072665 A US202017072665 A US 202017072665A US 2021140972 A1 US2021140972 A1 US 2021140972A1
Authority
US
United States
Prior art keywords
compound
receptors
tnf
trimer
tnfα
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
US17/072,665
Inventor
James Philip O'CONNELL
John Robert Porter
Alastair Lawson
Boris Kroeplien
Stephen Edward Rapecki
Timothy John Norman
David James MCMILLAN
Graham John Warrellow
Daniel Christopher Brookings
Rikki Peter Alexander
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
UCB Pharma SA
UCB SA
UCB Biopharma SRL
Original Assignee
UCB Biopharma SRL
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=53784155&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US20210140972(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by UCB Biopharma SRL filed Critical UCB Biopharma SRL
Priority to US17/072,665 priority Critical patent/US20210140972A1/en
Assigned to UCB Biopharma SRL reassignment UCB Biopharma SRL CHANGE OF NAME (SEE DOCUMENT FOR DETAILS). Assignors: UCB BIOPHARMA SPRL
Assigned to UCB BIOPHARMA SPRL reassignment UCB BIOPHARMA SPRL ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: UCB PHARMA S.A.
Assigned to UCB PHARMA S.A. reassignment UCB PHARMA S.A. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: UCB S.A.
Assigned to CELLTECH R&D LIMITED reassignment CELLTECH R&D LIMITED ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: BROOKINGS, DANIEL CHRISTOPHER, LAWSON, ALASTAIR, MCMILLAN, David James, NORMAN, TIMOTHY JOHN, PORTER, JOHN ROBERT, RAPECKI, STEPHEN EDWARD, WARRELLOW, GRAHAM JOHN, ALEXANDER, RIKKI PETER, KROEPLIEN, Boris, O'CONNELL, James Philip
Assigned to UCB S.A. reassignment UCB S.A. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: CELLTECH R&D LIMITED
Publication of US20210140972A1 publication Critical patent/US20210140972A1/en
Pending legal-status Critical Current

Links

Images

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6803General methods of protein analysis not limited to specific proteins or families of proteins
    • G01N33/6845Methods of identifying protein-protein interactions in protein mixtures
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • 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
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/62Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being a protein, peptide or polyamino acid
    • A61K47/64Drug-peptide, drug-protein or drug-polyamino acid conjugates, i.e. the modifying agent being a peptide, protein or polyamino acid which is covalently bonded or complexed to a therapeutically active agent
    • A61K47/6425Drug-peptide, drug-protein or drug-polyamino acid conjugates, i.e. the modifying agent being a peptide, protein or polyamino acid which is covalently bonded or complexed to a therapeutically active agent the peptide or protein in the drug conjugate being a receptor, e.g. CD4, a cell surface antigen, i.e. not a peptide ligand targeting the antigen, or a cell surface determinant, i.e. a part of the surface of a cell
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • C07D213/04Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D213/60Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D213/72Nitrogen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D235/00Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, condensed with other rings
    • C07D235/02Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, condensed with other rings condensed with carbocyclic rings or ring systems
    • C07D235/04Benzimidazoles; Hydrogenated benzimidazoles
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/02Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings
    • C07D239/24Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members
    • C07D239/26Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members with only hydrogen atoms, hydrocarbon or substituted hydrocarbon radicals, directly attached to ring carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/525Tumour necrosis factor [TNF]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70575NGF/TNF-superfamily, e.g. CD70, CD95L, CD153, CD154
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/24Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against cytokines, lymphokines or interferons
    • C07K16/241Tumor Necrosis Factors
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6854Immunoglobulins
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6863Cytokines, i.e. immune system proteins modifying a biological response such as cell growth proliferation or differentiation, e.g. TNF, CNF, GM-CSF, lymphotoxin, MIF or their receptors
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/32Immunoglobulins specific features characterized by aspects of specificity or valency specific for a neo-epitope on a complex, e.g. antibody-antigen or ligand-receptor
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/34Identification of a linear epitope shorter than 20 amino acid residues or of a conformational epitope defined by amino acid residues
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/55Fab or Fab'
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/565Complementarity determining region [CDR]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/52Assays involving cytokines
    • G01N2333/525Tumor necrosis factor [TNF]
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/705Assays involving receptors, cell surface antigens or cell surface determinants
    • G01N2333/715Assays involving receptors, cell surface antigens or cell surface determinants for cytokines; for lymphokines; for interferons
    • G01N2333/7151Assays involving receptors, cell surface antigens or cell surface determinants for cytokines; for lymphokines; for interferons for tumor necrosis factor [TNF]; for lymphotoxin [LT]
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2500/00Screening for compounds of potential therapeutic value
    • G01N2500/02Screening involving studying the effect of compounds C on the interaction between interacting molecules A and B (e.g. A = enzyme and B = substrate for A, or A = receptor and B = ligand for the receptor)
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2500/00Screening for compounds of potential therapeutic value
    • G01N2500/04Screening involving studying the effect of compounds C directly on molecule A (e.g. C are potential ligands for a receptor A, or potential substrates for an enzyme A)

Definitions

  • the present invention relates to methods for identifying compounds which modulate signalling of TNF superfamily member trimers through TNF receptors.
  • the invention relates to identification of new small molecule modulators.
  • the invention also relates to compounds identified by such methods and complexes of the compounds and trimers. The compounds and complexes may be used therapeutically.
  • TNF Tumour Necrosis Factor
  • TNF Tumour Necrosis Factor
  • a common core motif which consists of two antiparallel ⁇ -pleated sheets with antiparallel ⁇ -strands, forming a “jelly roll” ⁇ -structure.
  • Another common feature shared by members of the TNF superfamily is the formation of homo- or heterotrimeric complexes. It is these trimeric forms of the TNF superfamily members that bind to, and activate, specific TNF superfamily receptors.
  • TNF ⁇ is the archetypal member of the TNF superfamily. Dysregulation of TNF ⁇ production has been implicated in a number of pathological conditions of significant medical importance. For example, TNF ⁇ has been implicated in rheumatoid arthritis, inflammatory bowel diseases (including Crohn's disease), psoriasis, Alzheimer's disease (AD), Parkinson's disease (PD), pain, epilepsy, osteoporosis, asthma, systemic lupus erythematosus (SLE) and multiple sclerosis (MS). Other members of the TNF superfamily have also been implicated in pathological conditions, including autoimmune disease.
  • inflammatory bowel diseases including Crohn's disease
  • PD psoriasis
  • AD Alzheimer's disease
  • PD Parkinson's disease
  • SLE systemic lupus erythematosus
  • MS multiple sclerosis
  • Other members of the TNF superfamily have also been implicated in pathological conditions, including autoimmune disease.
  • TNF superfamily members are macromolecular and act by inhibiting the binding of the TNF superfamily member to its receptor.
  • conventional antagonists include anti-TNF ⁇ antibodies, particularly monoclonal antibodies, such as infliximab (Remicade®), adalimumab (Humira®) and certolizumab pegol (Cimzia®), or soluble TNF ⁇ receptor fusion proteins, such as etanercept (Enbrel®).
  • TNF trimer When receptors are present at an equivalent or excess concentration compared with TNF monomers (i.e. at a molar (concentration) ratio of at least 1:1 (receptors:monomers); at least 3:1 (receptors:trimers)), a TNF trimer will typically bind three receptors.
  • the present inventors have identified small molecular entities (SMEs) that modulate TNF signalling. These SME compounds act by binding to the trimeric form of TNF, and inducing and/or stabilising a conformational change in the trimer. Trimers with the compounds bound have an altered affinity for the requisite receptors, especially a reduced affinity for the second and third receptors, which decreases the number of receptors binding per trimer-compound complex. Accordingly, downstream signalling through the receptors is reduced. These compounds can therefore be used in the treatment of conditions mediated by TNF.
  • the present inventors have also developed methods that can identify compounds which are capable of modulating TNF signalling in this manner.
  • the present invention therefore provides a method of identifying a compound that is capable of binding to a trimeric protein that is a TNF superfamily member and modulating signalling of the trimeric protein through the requisite TNF superfamily receptor, the method comprising determining the average number of receptors bound per trimer-compound complex and thereby identifying whether the compound is capable of modulating signalling through the receptor.
  • the invention also provides:
  • FIG. 1 shows the interactions involved in TNF ⁇ /TNF-R1 signalling in the absence of compound.
  • the TNF ⁇ trimers bind the first two receptors with a K D of 93 pM, and bind the third receptor with a K D of approximately 1 ⁇ M. These trimer and receptor complexes (with three receptors bound) then form raft structures via dimerisation of the receptors, which result in downstream signalling.
  • FIG. 2 shows a crystal structure with only two receptor dimers interacting with a compound-stabilised TNF ⁇ trimer.
  • FIG. 3 shows the effects of two types of compounds (A and B) on TNF ⁇ /TNF-R1 signalling. Both compounds have no effect on the formation of receptor dimers, but induce and/or stabilise formation of trimers with distorted conformations. Trimers with the first type of compound (A) bind the first and second receptor, but have a reduced affinity for the third receptor. Accordingly, trimers form with only two receptors bound. Trimers with the second compound (B) bind the first receptor, but have a reduced affinity for the second and third receptors. Trimers therefore form with only one receptor bound. The decrease in the number of receptors binding per trimer interferes with raft formation.
  • FIGS. 4A, 4B, 4C, 4D, and 4E shows structures of compounds which are capable of modulating signalling of TNF superfamily member trimers through TNF receptors.
  • FIG. 4A shows the structure of a compound of formula (1)
  • FIG. 4B shows the structure of a compound of formula (2)
  • FIG. 4C shows the structure of a compound of formula (3)
  • FIG. 4D shows the structure of a compound of formula (4)
  • FIG. 4E shows the structure of a compound of formula (5).
  • FIG. 5 shows results of size exclusion chromatography (SEC) experiments using compound (2), TNF ⁇ and TNFR1 (in a 3.2 fold excess relative to the trimer-compound complex).
  • SEC size exclusion chromatography
  • results for controls comprising TNF ⁇ alone, and TNF ⁇ and TNFR1 are also presented.
  • TNF ⁇ and TNFR1 negative controls three receptors bind per trimer-compound complex. This is achieved by preincubating a 3.2 excess of TNFR1 over TNF ⁇ trimer. More data is presented in FIG. 6 .
  • FIG. 6 shows the control for the SEC experiments described in FIG. 5 .
  • TNF ⁇ was incubated with varying concentrations of TNFR1 (ranging from a 1.2-5 fold excess of receptors:trimers). As an increasing concentration of TNFR1 is added the molecular weight of the complex with TNF ⁇ increases (shifts to the left). Addition of a 5 fold excess of TNFR1 over the concentration of TNF ⁇ trimers does not increase the molecular weight of the complex over that using a 3.2 fold excess. This suggests that TNF ⁇ is saturated at a molar ratio of 3 TNFR1s to 3 TNF ⁇ monomers (3 TNFR1s per trimer).
  • FIG. 7 shows results of SEC experiments using compound (5), TNF ⁇ and TNFR1 (in a 3.5 fold excess relative to the concentration of the trimer-compound complex). Results are also presented for controls, the first of which is TNF ⁇ and receptors in the absence of compound. The second and third controls are still TNF ⁇ and receptors in the absence of compound, but the TNF ⁇ is mutated to disrupt interactions at the third, and third and second, receptor binding sites.
  • the control which is TNF ⁇ and receptors shows a peak indicating three receptors binding per trimer.
  • the control with a mutation at one receptor binding site shows a peak corresponding to two receptors binding per trimer, and the control with mutations at two sites shows a peak corresponding to one receptor binding per trimer.
  • the peak obtained in the presence of compound (5) is mid-way between the second and third controls, and therefore indicates trimers binding a mixture of two receptors and one receptor respectively.
  • FIGS. 8A-8D show the results of crystallography experiments, which reveal two receptors binding per trimer-compound complex in the presence of compound (1). Parts (A)-(D) are alternate views of the same crystal structure.
  • FIG. 9 shows results of FRET experiments with increasing concentrations of compound (3). Complete inhibition as would be observed with a blocking antibody would result in no receptor binding to TNF ⁇ i.e. complete inhibition of a FRET signal. In this instance the FRET signal is partially inhibited. At the highest concentrations of the compound, maximal inhibition is 29%, suggesting that one out of the three receptors is inhibited from binding to the TNF trimer.
  • FIG. 10 shows results of FRET experiments with increasing concentrations of compound (4). Again, the FRET signal is partially inhibited. At the highest concentrations of the compound, maximal inhibition is 36%. Similar to the observation described in FIG. 9 , this suggests that one out of the three receptors is inhibited from binding to the TNF trimer.
  • FIG. 11 shows an analysis of receptor binding stoichiometry by ion-mobility mass spectrometry.
  • control comprising TNF ⁇ and an excess of TNFR1
  • three receptors are shown to bind on average per trimer compound-complex.
  • the receptor stoichiometry is reduced and predominantly two receptors bind per trimer-compound complex.
  • FIG. 12 shows determination of dissociation constants in a control sample with TNF ⁇ and TNFR1.
  • TNFR1 As increasing concentrations of TNFR1 are added to TNF ⁇ different mass species appear and then disappear corresponding to the appearance of firstly 1 TNFR1 bound to TNF ⁇ , followed by 2 TNFR1 bound to TNF ⁇ , and finally 3 TNFR1 bound to TNF ⁇ .
  • FIG. 13 shows determination of dissociation constants in a sample with TNF ⁇ , TNFR1 and compound (3). This shows a significantly worse (lower affinity) TNFR1 interaction with TNF ⁇ of the third receptor (0.22 nM to 9.612 nM).
  • SEQ ID Nos: 1 and 2 show sequences used in the Examples.
  • SEQ ID NO: 3 shows the HCVR of C185_01974.0.
  • SEQ ID NO: 4 shows the LCVR of C185_01974.0.
  • SEQ ID NO: 5 shows the amino acid sequence of the mIgG1 heavy chain of C185_01974.0.
  • SEQ ID NO: 6 shows the amino acid sequence of the kappa light chain of C185_01974.0.
  • SEQ ID NO: 7 shows the HCVR of C185_01979.0.
  • SEQ ID NO: 8 shows the LCVR of C185_01979.0.
  • SEQ ID NO: 9 shows the amino acid sequence of the mIgG1 heavy chain of C185_01979.0.
  • SEQ ID NO: 10 shows the amino acid sequence of the kappa light chain of C185_01979.0.
  • the present invention relates to methods of (assays for) identifying compounds that are capable of binding to a trimeric protein that is a TNF superfamily member and modulating signalling of the trimeric protein through the requisite TNF superfamily receptor.
  • Compounds identified by the methods of the invention are therefore also known as modulators.
  • the compounds identified by the methods of the invention generally prevent or decrease (inhibit) signalling of TNF through the requisite receptors. Such compounds are antagonists of TNF signalling.
  • the methods of the invention may also be used to identify agonist compounds, which increase (enhance) signalling of TNF through the requisite receptors. In both cases, the compounds are capable of modulating TNF signalling without having to compete with the high affinity interaction between the TNF superfamily member and its receptor.
  • the compounds identified by the methods of the invention bind to the trimeric forms of TNF superfamily members.
  • the compounds are therefore allosteric modulators that bind to the natural agonists of the TNF superfamily receptors, i.e. to trimeric forms of TNF superfamily members. Methods of screening for compounds which are capable of binding to TNF trimers are discussed further below.
  • TNF superfamily members which are TNF ⁇ (TNFSF1A), TNF ⁇ (TNFSF1B), CD40L (TNFSF5), BAFF (TNFSF13B/BlyS), APRIL (TNFSF13), OX40L (TNFSF4), RANKL (TNFSF11/TRANCE), TWEAK (TNFSF12), TRAIL (TNFSF10), TL1A (TNFSF15), LIGHT (TNFSF14), Lymphotoxin, Lymphotoxin ⁇ (TNFSF3), 4-1BBL (TNFSF9), CD27L (TNFSF7), CD30L (TNFSF8), EDA (Ectodysplasin), EDA-A1 (Ectodysplasin A1), EDA-A2 (Ectodysplasin A2), FASL (TNFSF6), NGF and GITRL (TNFSF18).
  • TNF ⁇ TNF ⁇
  • TNFSF1B TNF ⁇
  • CD40L T
  • the methods of the invention may be used to identify compounds which modulate signalling of any TNF superfamily member, including the 22 known TNF superfamily members.
  • the compounds identified using the methods of the invention may bind specifically to the trimeric forms of one or more TNF superfamily members.
  • Compounds identified by the methods of the invention may bind specifically to only one of the TNF superfamily members, but not to any other TNF superfamily members.
  • Compounds identified by the methods of the invention may also bind specifically to two, three, four or up to all of the TNF superfamily members.
  • the compounds bind to the molecule or molecules of interest, in this case the trimeric form of the TNF superfamily member, with no significant cross-reactivity to any other molecule, which may include other members of the TNF superfamily.
  • Cross-reactivity may be assessed by any suitable method, for example surface plasmon resonance.
  • Cross-reactivity of a compound for the trimeric form of a TNF superfamily member with a molecule other than the trimeric form of that particular TNF superfamily member may be considered significant if the compound binds to the other molecule at least 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90% or 100% as strongly as it binds to the trimeric form of the TNF superfamily member of interest.
  • a compound that is specific for the trimeric form of a TNF superfamily member may bind to another molecule at less than 90%, 85%, 80%, 75%, 70%, 65%, 60%, 55%, 50%, 45%, 40%, 35%, 30%, 25% or 20% the strength that it binds to the trimeric form of the TNF superfamily member.
  • the compound binds to the other molecule at less than 20%, less than 15%, less than 10% or less than 5%, less than 2% or less than 1% the strength that it binds to the trimeric form of the TNF superfamily member
  • the TNF superfamily member is TNF ⁇ .
  • TNF ⁇ exists in both a soluble (TNF ⁇ s ) and membrane-bound form (TNF ⁇ m ).
  • TNF ⁇ is referred to herein this encompasses both the TNF ⁇ s and TNF ⁇ m forms.
  • TNF ⁇ is in the TNF ⁇ s form.
  • TNF receptors which are 4-1BB (TNFRSF9/CD137), NGF R (TNFRSF16), BAFF R (TNFRSF13C), Osteoprotegerin (TNFRSF11B), BCMA (TNFRSF17), OX40 (TNFRSF4), CD27 (TNFRSF7), RANK (TNFRSF11A), CD30 (TNFRSF8), RELT (TNFRSF19L), CD40 (TNFRSF5), TACI (TNFRSF13B), DcR3 (TNFRSF6B), TNFRH3 (TNFRSF26), DcTRAIL R1 (TNFRSF23), DcTRAIL R2 (TNFRSF22), TNF-R1 (TNFRSF1A), TNF-R2 (TNFRSF1B), DR3 (TNFRSF25), TRAIL R1 (TNFRSF10A), DR6 (TNFRSF21), TRAIL R2 (TNFRSF10B), EDAR, TRAIL R3 (TNFRSF10C),
  • 4-1BB
  • a requisite receptor is a receptor which acts in conjunction with a particular TNF superfamily member.
  • a requisite receptor is a receptor which is activated by a TNF superfamily member.
  • the TNF superfamily member trimers bind to the receptor and activation of the receptor results in downstream signalling. Combinations of TNF superfamily members and their requisite receptors are known in the art.
  • the methods of the invention are used to identify compounds which modulate signalling through TNF-R1 (TNFR1) and TNF-R2 (TNFR2).
  • TNF-R is referred to herein this encompasses both TNF-R1 and TNF-R2, including the extracellular domain (ECD) of TNF-R1 and TNF-R2.
  • the TNF superfamily member is TNF ⁇ and the TNF receptor is TNF-R1 or TNF-R2.
  • the TNF superfamily member is TNF ⁇ and the TNF receptor is TNF-R1.
  • the TNF superfamily member is TNF ⁇ s and the TNF receptor is TNF-R1.
  • the methods of the invention may be used to identify compounds which act by specifically modulating the signalling of TNF superfamily members through TNF-R1.
  • the compounds may act by modulating the signalling of TNF superfamily members through TNF-R1, but have no effect on signalling of TNF superfamily members through TNF-R2.
  • the TNF superfamily members and their receptors may be purified or present in mixtures, such as in cultured cells, tissue samples, body fluids or culture medium.
  • compounds are identified which modulate signalling of the trimeric protein through the requisite receptors. Modulation of signalling may refer to an increase (enhancement) in signalling through the requisite receptors.
  • Compounds which increase signalling are agonist compounds.
  • compounds identified using the methods of the invention generally prevent or decrease (inhibit) signalling through the requisite receptors. Such compounds are known as antagonists.
  • assays that measure the downstream effects of TNF superfamily receptor signalling can be performed.
  • a L929 murine fibrosarcoma cell-killing assay can be used to assess the stimulation of cell death by TNF.
  • Inhibition of TNF-induced IL-8 production by human monocytes may also be used to assess whether a test compound inhibits TNF signalling via its receptor.
  • Such assays are well known in the art.
  • the compounds identified by the methods of the invention may completely or partially inhibit signalling through a TNF receptor when a TNF superfamily member in the form of a compound-trimer complex binds to the receptor.
  • the compound may act to reduce signalling through a TNF superfamily receptor by at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or 100%. Any change in the level of signalling may be measured by an appropriate technique, including measuring reporter gene activity by alkaline phosphatase or luciferase, NF- ⁇ B translocation using machines such as the Cellomics Arrayscan, phosphorylation of downstream effectors, recruitment of signalling molecules, or cell death.
  • the compounds identified by the methods of the invention may modulate at least one of the downstream effects of signalling through a TNF receptor when a TNF superfamily member in the form of a compound-trimer complex binds to the receptor.
  • Standard techniques are known in the art for measuring the downstream effects of TNF superfamily members.
  • the compounds identified by the methods of the invention may modulate at least 1, 2, 3, 4, 5, 10 or up to all of the downstream effects of signalling through a TNF receptor.
  • the activity of the compounds identified by the methods of the invention may be quantified using standard terminology, such as IC 50 or half maximal effective concentration (EC 50 ) values.
  • IC 50 values represent the concentration of a compound that is required for 50% inhibition of a specified biological or biochemical function.
  • EC 50 values represent the concentration of a compound that is required for 50% of its maximal effect.
  • the compounds identified by the methods of the invention may have IC 50 or EC 50 values of 500 nM, 400 nM, 300 nM, 200 nM, 100 nM, 90 nM, 80 nM, 70 nM, 60 nM, 50 nM, 40 nM, 30 nM, 20 nM, 10 nM, 5 nM, 1 nM, 100 pM or less.
  • IC 50 and EC 50 values may be measured using any appropriate technique, for example cytokine production can be quantified using ELISA. IC 50 and EC 50 values can then be generated using a standard 4-parameter logistic model also known as the sigmoidal dose response model.
  • libraries of compounds may be screened in order to identify modulators of TNF superfamily members (i.e. using the methods disclosed herein).
  • Such libraries typically comprise at least 260 compounds.
  • such libraries comprise at least 300, at least 500 or even at least 1000 compounds.
  • the average number of receptors bound per trimer-compound complex is determined in order to identify compounds capable of modulating TNF signalling.
  • receptors are present at an equivalent or excess concentration compared with TNF monomers (at a molar ratio of greater than 1:1 (receptors:monomers); greater than 3:1 (receptors:trimers)) typically three receptors bind per TNF trimer.
  • trimer and receptor complexes then form rafts via the formation of receptor dimers. The rafts are responsible for downstream signalling.
  • FIG. 1 shows TNF trimer and receptor interactions, and the formation of rafts, involved in TNF ⁇ and TNF-R1 signalling.
  • the TNF ⁇ trimers bind the first and second requisite receptors with an affinity (K D ) of approximately 93 pM.
  • the trimers then bind the third and final receptor with a K D of approximately 1 ⁇ M.
  • the compounds identified using the methods of the present invention induce and/or stabilise conformational changes within the TNF trimers.
  • These trimers have altered affinities for receptors; especially for the second and third receptors where there is reduced affinity. This reduced affinity results in a decrease in the number of receptors binding per trimer-compound complex. For example, as shown in FIG. 2 only two receptor dimers may bind per trimer-compound complex (instead of the three receptors that would bind under normal conditions).
  • FIG. 3 shows the effects of two types of compounds on TNF ⁇ /TNF-R1 signalling. Both compounds have no effect on the formation of receptor dimers, but induce and/or stabilise formation of trimers with distorted conformations. Trimers with the first type of compound bind the first and second receptor, but have a reduced affinity for the third receptor. Accordingly, trimers form with only two receptors bound. Trimers with the second compound bind the first receptor, but have a reduced affinity for the second and third receptors. Trimers therefore form with only one receptor bound. Both types of compound thus interfere with the formation of signalling rafts due to the decrease in the number of receptors binding per trimer
  • the methods of the present invention involve determining the average number of receptors bound per trimer-compound complex in order to identify compounds which modulate TNF signalling.
  • the term “average” reflects the fact that a mixed population of trimers/receptors will almost certainly be present in a sample. For example, as some compounds reduce the affinity of trimers for receptor three, some trimers may still bind all three receptors in the presence of the compound, but the majority of trimers will only have two receptors bound.
  • the term “average” may refer to a modal value, i.e. the number of receptors bound per trimer-compound complex which occurs most frequently within a sample.
  • a modal value may be determined visually from experimental results. This is illustrated in the Examples section below.
  • the term “average” may also refer to a mean value.
  • the modal value will be two receptors bound per trimer.
  • the mean value will be 2.15 receptors bound per trimer ((5 ⁇ 1+75 ⁇ 2+20 ⁇ 3)/100).
  • a result of 0-0.4 is taken to indicate that on average zero receptors are bound per trimer
  • a result of 0.5-1.4 is taken to indicate that on average one receptor is bound per trimer
  • a result of 1.5-2.4 is taken to indicate that on average two receptors are bound per trimer
  • a result of greater than 2.5 is taken to indicate that on average three receptors are bound per trimer.
  • the average number of receptors bound per trimer-compound complex is typically determined in comparison to a control.
  • the control sample is treated in the same way as the sample with the test compound.
  • the control sample is subjected to the same experimental conditions as the sample comprising the test compound, including the same concentrations of reagents, trimers and receptors.
  • the average number of receptors bound per trimer for the control is determined using the same experimental method as for the test compound.
  • the average number of receptors bound per trimer is usually determined at the same time for the test sample and for the control. In other words, the experiments are run in parallel. However, values for the average number of receptors bound per trimer in a control may also be determined prior to performing experiments on the test sample. Such values may be recorded e.g. on a computer.
  • the average number of receptors bound per trimer for a control is calculated in the same way as for the test sample (i.e. modal values or mean values as discussed above).
  • the control sample may comprise TNF superfamily member trimers and the requisite receptors in the absence of compound (a negative control).
  • the control sample is identical to the test compound sample, except there is no test compound present.
  • the TNF superfamily member trimers and the requisite receptors may be any of those discussed above, but are the same in the control and test sample (and are present at the same concentrations).
  • a decrease/reduction in the average number of receptors bound per trimer-compound complex in the test sample in comparison to the control identifies that the compound is capable of modulating signalling through the receptor.
  • a test compound is identified as being capable of modulating signalling of the trimeric protein through the receptor if a lower number of receptors are identified as being bound on average per trimer in the sample with the test compound compared with the control sample.
  • a test compound when calculated using a modal value if the control is determined to have an average of three receptors bound per trimer, a test compound may be identified as being capable of modulating signalling if two or fewer receptors are determined to be bound on average per trimer-compound complex.
  • a negative control comprising TNF superfamily member trimers, and the requisite receptors, in the absence of compound should be found to bind an average of three receptors per trimer (when the receptors are present at an equivalent concentration or in an excess compared with the TNF monomers; at a molar ratio of at least 1:1 (receptors:monomers) or 3:1 (receptors:trimers)).
  • a test compound will be identified as being capable of modulating signalling if one or zero receptors are determined to be bound on average per trimer-compound complex.
  • a test compound will be identified as being capable of modulating signalling if zero receptors are determined to be bound on average per trimer-compound complex.
  • a decrease in the average number of receptors bound per trimer, relative to the negative control, can also simply be calculated based on the percentage of trimers in a sample binding three, two, one or zero receptors. In this case, it is first necessary to identify the percentage of trimers in both the control sample and the sample containing the test compound having three, two, one or zero receptors bound. A test compound is then identified as being capable of modulating signalling if the presence of the compound results in a change in the percentage of trimers having a certain number of receptors bound.
  • Such calculations are typically based on the percentage of trimers in a sample having three receptors bound, where a decreased percentage of trimers having three receptors bound would be indicative of an antagonist compound that modulates signalling (the percentage of trimers binding two, one or zero receptors must concurrently increase).
  • a negative control comprising receptors and trimers only (without compound) 90% of trimers may be found to bind three receptors and 10% of trimers may be found to bind two receptors.
  • a test compound may then be identified as being capable of modulating signalling if the compound results in less than 90% of trimers binding three receptors. If a lower percentage of trimers are binding three receptors, the percentage of trimers binding two, one or zero receptors must have increased. Accordingly, the average number of receptors bound per trimer-compound complex is decreased relative to the control.
  • a test compound is identified as being capable of modulating signalling if in the test compound sample the percentage of trimers having three receptors bound is decreased by at least 10% (i.e. at least 10% lower), at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, or at least 95% compared with the percentage of trimers having three receptors bound in the negative control sample (comprising the TNF superfamily member and receptors in the absence of compound).
  • control may comprise TNF superfamily member trimers, the requisite receptors, and a compound which is known to modulate signalling through the receptors (a so called “positive control”).
  • the compound which is known to modulate signalling through the receptor may be any compound known to decrease the average number of receptors bound per trimer to two, one, or zero (under conditions where the trimers would bind three receptors in the absence of any compound).
  • Such compounds can be identified using the methods described herein. Examples of compounds known to decrease the average number of receptors to two per trimer are compounds (1)-(4) and an example of a compound known to decrease the average number of receptors to closer to one per trimer is compound (5).
  • the positive control may comprise any one of these exemplary compounds.
  • the positive control sample is treated in the same way as the sample with the test compound and the same experimental conditions, methods and calculations are used for both the control sample and test compound sample.
  • the average number of receptors bound per trimer-compound complex is usually determined at the same time for the test compound sample and for the control, but could also be determined prior to performing experiments on the test compound sample.
  • the positive control sample comprises TNF superfamily member trimers, the requisite receptors, and a compound which is known to be capable of modulating signalling through the receptor
  • an equivalent average number of receptors bound per trimer-compound complex in the test sample in comparison with the control, or decrease in the average number of receptors bound per trimer-compound complex in the test sample in comparison with the control identifies that the compound is capable of modulating signalling through the receptor.
  • a test compound is identified as being capable of modulating signalling of the trimeric protein through the receptor if an identical or lower number of receptors are on average identified as being bound per trimer in the sample with the test compound compared with the positive control.
  • a test compound when calculated using a modal value (described above) if the positive control is determined to have an average of two receptors bound per trimer, a test compound will be identified as being capable of modulating signalling if two or fewer receptors (two receptors, one receptor or zero receptors) are determined to be bound on average per trimer-compound complex. Likewise, if the control is identified as having an average of one receptor bound per trimer, a test compound will be identified as being capable of modulating signalling if one or zero receptors are determined to be bound on average per trimer-compound complex. If the control is identified as having an average of zero receptors bound per trimer, a test compound will be identified as being capable of modulating signalling if zero receptors are also determined to be bound on average per trimer-compound complex.
  • An equivalent average number of receptors bound per trimer, or a decrease in the average number of receptors bound per trimer, in comparison with a positive control can again also be calculated using the proportions (percentages) of trimers in a sample binding three, two, one or zero receptors. As described above, it is first necessary to identify the percentage of trimers in both the control sample and the sample containing the test compound having three, two, one or zero receptors bound. A test compound is then identified as being capable of modulating signalling if the presence of the compound results in an equivalent percentage, or increased/higher percentage, of trimers having the desired number of receptors bound (or a lower number of receptors bound) in comparison with the control. Such calculations may be focused on the percentage of trimers in a sample having two or fewer receptors bound, one or fewer receptor bound, or zero receptors bound.
  • a compound may result in 30% of the trimers present binding three receptors and 70% of the trimers present binding two receptors.
  • a test compound may then be identified as being capable of modulating signalling if the compound results in at least 70% percent of the trimers present binding two or fewer (two, one or zero) receptors.
  • “An equivalent percentage” in this context typically refers to values which are within 10% or less of each other, preferably 5% or less. For example, a test compound which results in 70% of trimers binding two receptors can be seen as resulting in an equivalent percentage of trimers binding two receptors as a control where 75% of trimers bind two receptors.
  • a compound may also be identified as being capable of modulating signalling simply based on determination of the number of receptors binding per trimer-compound complex, without a direct comparison to a control.
  • a compound may be identified as being capable of modulating signalling of the trimeric protein through the receptor if an average of less than three receptors are determined to be bound per trimer-compound complex.
  • the average number of receptors bound per trimer-compound complex may be a mean value or a modal value as described above.
  • Compounds are preferably identified as being capable of modulating signalling of the trimeric protein through the receptor if the compound results in an average of two receptors binding per trimer. More preferably, a compound is identified as being capable of modulating signalling of the trimeric protein through the receptor if the compound results in an average of one receptor binding per trimer.
  • a compound may also be identified as being capable of modulating signalling of the trimeric protein through the receptor if the compound results in an average of zero receptors binding per trimer. Examples of such compounds are discussed in more detail below.
  • the average number of receptors binding per trimer-compound complex is typically determined at approximately an equivalent concentration (a 1:1 molar ratio) of receptors to TNF monomers.
  • a 1:1 ratio of receptors:momoners corresponds to a 3:1 ratio of receptors:trimers.
  • the concentration of receptors may be in a slight excess compared with the concentration of monomers.
  • Experiments are preferably conducted at a molar ratio of up to approximately 10:1 (receptors:trimers). Experiments may be conducted at any molar ratio within a range of between approximately 3:1 and 10:1 (receptors:trimers).
  • assays are conducted at ratios of approximately 3:1, 4:1, 5:1: 6:1, 7:1, 8:1, 9:1 or 10:1 (receptors:trimers).
  • assays are conducted at multiple concentrations of receptors:trimers e.g. approximately 3:1, 6:1 and 10:1. These titrations are illustrated in more detail in the Examples section.
  • experiments to determine the average number of receptors binding per trimer-compound complex are carried out when the compound is present at a concentration of compound that ensures complete occupancy of the trimers with compound.
  • occupancy of the trimers with compound may be determined using mass spectrometry.
  • the compound may be present at an equal concentration compared with the concentration of trimers (a 1:1 ratio of compound:trimers; a 1:3 ratio of compound:TNF monomers).
  • the compound is, however, typically present in an excess relative to the concentration of the TNF trimers.
  • the compound may be present in an excess of between 1.5 ⁇ and 500 ⁇ relative to the concentration of trimers.
  • the compound is present in an excess of between 5 ⁇ and 100 ⁇ relative to the concentration of trimers, more preferably in an excess of between 10 ⁇ and 50 ⁇ relative to the concentration of trimers.
  • the compound may be present at an excess of at least 1.5 ⁇ , at least 2 ⁇ , at least 3 ⁇ , at least 4 ⁇ , at least 5 ⁇ , at least 6 ⁇ , at least 7 ⁇ , at least 8 ⁇ , at least 9 ⁇ , at least 10 ⁇ , at least 20 ⁇ , at least 50 ⁇ , at least 100 ⁇ , at least 250 ⁇ or at least 500 ⁇ relative to the concentration of TNF trimers.
  • the average number of receptors binding per trimer may be determined using any suitable technique, for example using ion mobility mass spectrometry, size exclusion chromatography, an aggregation assay, Förster Resonance Energy Transfer and/or crystallography. These techniques may be used alone or, preferably, in combination in order to determine the average number of receptors binding per trimer. For example, any two three, four or all five of the techniques may be used together.
  • IMS-MS Ion mobility mass spectrometry
  • the test compound is incubated with TNF overnight at room temperature.
  • a native mass spectrum is first recorded to ensure that the compound has 100% occupancy of the TNF prior to addition of the compound.
  • the native mass spectrum ensures that the compound is binding to the trimers.
  • the compound is typically present in an excess relative to the concentration of trimers, as described above.
  • the requisite TNF receptors are then added and incubated before the ion mobility mass spectrum of the sample is collected.
  • Ion mobility mass spectrometry assays are conducted at any suitable ratio of receptors:trimers, typically at approximate ratios of between 3:1 and 10:1, for example 3:1, 6:1 and/or 10:1 (receptors:trimers). In many cases, ion mobility mass spectrometry assays are conducted at a number of concentration ratios for a test sample.
  • Ion mobility mass spectrometry can also be used to provide affinity data for binding of the three receptors to TNF trimers. This is illustrated in the Examples section below.
  • Size exclusion chromatography Another technique that can be used to determine the number of receptors binding on average per TNF trimer is size exclusion chromatography. Size exclusion chromatography methods are well known in the art, and involve separating components in a solution based on their size. Smaller components in the solution elute more slowly and require a larger elution volume in comparison to larger components.
  • a TNF trimer binding three receptors will be larger than a trimer binding only two receptors. Trimers binding three receptors are therefore eluted at a smaller volume compared with trimers binding two receptors or one receptor. The peaks at different elution volumes can thus be used to identify the average number of receptors bound per trimer. This is illustrated in more detail in the Examples section below.
  • the TNF trimers are incubated with an excess of compound. Occupancy of the TNF trimers with compound can be determined by IMS-MS, as described above. The samples are then incubated with receptors, and analysed by size exclusion HPLC.
  • size exclusion chromatography experiments are conducted at approximate ratios of between 3:1 and 10:1, for example 3:1, 6:1 and/or 10:1 receptors:trimers.
  • a compound is tested at a range of concentration ratios.
  • Controls to establish the migration (peak) positions for trimers with two receptors bound, or one receptor bound are illustrated in the Examples section below. These controls comprise mutant TNF ⁇ , which has impaired binding of either the third, or second and third, receptors.
  • Another technique which may be used to determine the average number of receptors binding per TNF trimer is an aggregation assay.
  • FRET Forster Resonance Energy Transfer
  • the receptors may be tagged with e.g. the donor fluorophore.
  • the trimers are then tagged with e.g. the acceptor fluorophore (possibly via a linker).
  • Experiments are typically conducted at a ratio of 1:1 receptors:monomers, but receptor titrations may also be performed.
  • experiments may be performed with the test compound present at a variety of concentrations. This is illustrated in the Examples below.
  • crystallography may be used in order to determine the average number of receptors binding per trimer. Crystallography techniques are well known in the art.
  • All of the methods of the invention involve providing an output that identifies that the compound is capable of modulating signalling through the receptor.
  • the output may be recording information e.g. in a laboratory notebook.
  • the output may also be recording information on a computer.
  • the present invention also relates to compounds that are capable of binding to a trimeric protein that is a TNF superfamily member and modulating signalling of the TNF superfamily member through the requisite receptor.
  • the compounds result in corresponding average number, or a change in the average number, of receptors bound per trimer-compound complex in comparison with a control.
  • Such compounds may be identified by the methods described above.
  • TNF superfamily members and requisite receptors may be any of those described above.
  • TNF trimer can readily be screened for binding to a TNF trimer using routine methods known in the art, such as mass spectrometry. Mass spectrometry can also be used to identify the presence of the TNF trimers themselves in a sample.
  • Mass spectrometric methods may include, for example, matrix-assisted laser desorption/ionization mass spectrometry (MALDI MS), surface-enhanced laser desorption/ionization mass spectrometry (SELDI MS), time of flight mass spectrometry (TOF MS) and liquid chromatography mass spectrometry (LC MS).
  • MALDI MS matrix-assisted laser desorption/ionization mass spectrometry
  • SELDI MS surface-enhanced laser desorption/ionization mass spectrometry
  • TOF MS time of flight mass spectrometry
  • LC MS liquid chromatography mass spectrometry
  • the compounds are not limited in terms of their chemical formula or structure.
  • the compounds are typically small molecular entities (SMEs) that have a molecular weight of 1000 Da or less, preferably 750 Da or less, more preferably 600 Da or less.
  • SMEs small molecular entities
  • the compounds may bind inside the central space present within the TNF superfamily member trimer (i.e. the core of the trimer). Binding of a compound within the core of the trimer can be detected using routine methods e.g. using crystallography.
  • the compounds may comprise a benzimidazole moiety or an isostere thereof.
  • the compounds bind to at least one TNF superfamily member and modulate the signalling of the TNF superfamily member through the requisite receptor. Modulation of signalling is described above in the context of the methods of the present invention.
  • a TNF trimer usually binds an average of three receptors.
  • the compounds of the present invention modulate signalling by resulting in an equivalent average number of receptors bound per trimer-compound complex, or a change in the average number of receptors bound per trimer-compound complex, in comparison with a control.
  • control may comprise TNF superfamily member trimers and receptors in the absence of the compound (a negative control).
  • TNF superfamily member trimers and receptors are the same for both the test compound and the control, and the test compound sample and control are subjected to the same experimental conditions (e.g. concentrations of reagents) and methods.
  • control may be run in parallel to the sample comprising the test compound. Alternatively, the control may be run prior to the test compound sample.
  • control comprises TNF superfamily member trimers and receptors in the absence of compound
  • three receptors would be expected to bind on average per trimer when assays are performed at an equivalent (1:1) molar ratio of TNF monomers:receptors, or when the receptors are present in an excess compared to the concentration of monomers.
  • An antagonist compound results in a decrease in the average number of receptors binding per trimer under such conditions. Methods of determining a decrease in the average number of receptors binding per trimer are discussed above.
  • control may comprise TNF superfamily member trimers, the requisite receptors, and a compound known to modulate signalling of the trimers through the receptors (a positive control).
  • TNF superfamily member trimers and receptors are the same for both the test compound and the control, and the test compound sample and control are subjected to the same experimental conditions (e.g. concentrations of reagents) and methods.
  • the control is run in parallel to the test compound. However, the control may also be run prior to the test compound sample.
  • the control comprises TNF superfamily member trimers, the requisite receptors, and a compound known to modulate signalling of the trimers through the receptors
  • a compound is identified as being capable of modulating signalling if it results in an equivalent average number of receptors bound per trimer-compound complex in comparison with the control, or decrease in the average number of receptors bound per trimer-compound complex in comparison with the control.
  • Antagonistic compounds result in an average of less than three receptors binding per trimer-compound complex (under conditions where in the absence of compound three receptors on average would bind per trimer-compound complex).
  • a test compound results in an average of two receptors binding per trimer-compound complex.
  • Examples of such compounds include compounds (1)-(4). These compounds can be used as positive control compounds, when evaluating whether another test compound is capable of modulating signalling.
  • a test compound results in an average of one receptor binding per trimer-compound complex.
  • examples of such compounds include compound (5), which results in a shift towards a single receptor binding per trimer.
  • these compounds can be used as positive control compounds when evaluating whether another test compound is capable of modulating signalling.
  • a test compound may also result in an average of zero receptors binding per trimer-compound complex.
  • the present invention also relates to a complex comprising a trimeric protein that is a TNF superfamily member and a compound.
  • the trimeric protein that is a TNF superfamily member, and the compound may be any of those described above.
  • the present inventors developed antibodies that bind selectively to complexes comprising compounds of the invention and a trimeric TNF superfamily member. These antibodies may be used to identify further compounds that are capable of inhibiting TNF.
  • CA185_01974 and CA185_01979 Two antibodies, termed CA185_01974 and CA185_01979, which were raised against human TNF ⁇ in complex with a compound of the invention.
  • the heavy chain variable region (HCVR) of CA185_01974 is shown in SEQ ID NO: 3 and the light chain variable region (LCVR) of CA185_01974 is shown in SEQ ID NO: 4.
  • the full length IgG1 heavy chain is shown in SEQ ID NO: 5 (1974 HC mIgG1 full) and the full length light chain (1974 LC kappa full) is shown in SEQ ID NO: 6.
  • the HCVR of CA185_01979 is shown in SEQ ID NO: 7 and the LCVR of CA185_01979 is shown in SEQ ID NO: 8.
  • the full length IgG1 heavy chain of CA185_01979 is shown in SEQ ID NO: 9 (1979 HC mIgG1 full) and the full length light chain in SEQ ID NO: 10 (1979 L C Kappa full).
  • Antibodies comprising the above HCVR/LCVR or full-length sequence pairs can readily be generated by the skilled person using standard techniques.
  • Methods of the invention for determining compounds which are capable of binding to a trimeric protein which is a TNF superfamily member and modulating signalling through the receptor may therefore involve identifying whether an antibody with a HCVR/LCVR pair of SEQ ID NOs: 3/4 or 7/8 binds the trimer-compound complex. Likewise, methods may involve identifying whether an antibody with a sequence pair of SEQ ID Nos: 5/6 or 9/10 binds the trimer compound complex.
  • Antibody assays may be used in addition to the other assays described herein.
  • Antibodies of the invention can be tested for binding to a compound-trimer complex by, for example, standard ELISA or Western blotting.
  • the binding selectivity of an antibody may also be determined by monitoring binding of the antibody to cells expressing the target protein, for example by flow cytometry.
  • a screening method of the invention may comprise the step of identifying an antibody that is capable of binding a compound-trimer complex by carrying out an ELISA or Western blot or by flow cytometry.
  • the antibodies described herein selectively (or specifically) recognise at least one compound-trimer complex, i.e. epitopes within a compound-trimer complex.
  • An antibody, or other compound “selectively binds” or “selectively recognises” a protein when it binds with preferential or high affinity to the protein for which it is selective but does not substantially bind, or binds with low affinity, to other proteins.
  • a compound-trimer complex may typically bind an antibody with a HCVR/LCVR pair of SEQ ID NOs: 3/4 or 7/8 (or with sequence pairs of SEQ ID NOs: 5/6 or 9/10) with an affinity of less than 1 nM.
  • the methods of the invention may involve determining that a compound is capable of binding to a trimeric protein which is a TNF superfamily member and modulating signalling through the receptor by identifying that an antibody with a HCVR/LCVR pair of SEQ ID NOs: 3/4 or 7/8 (or sequence pairs of SEQ ID NOs: 5/6 or 9/10) binds the trimer-compound complex with a KD-ab of less than 1 nM.
  • the KD-ab may be less than 500 pM, or less than 200 pM.
  • the affinity may be determined by surface plasmon resonance.
  • the TNF is typically human TNF ⁇ .
  • a complex of the invention may be a complex of a trimeric TNF superfamily member and a compound, wherein the compound-trimer complex binds an antibody with a HCVR/LCVR pair of SEQ ID NOs: 3/4 or 7/8 (or sequence pairs of SEQ ID Nos: 5/6 or 9/10).
  • the TNF is typically human TNF a
  • the binding affinity is typically less than 1 nM (or less than 500 pM/200 pM). Binding affinity is typically determined by surface plasmon resonance.
  • TNF ⁇ is the archetypal member of the TNF superfamily.
  • TNF ⁇ is a pleiotropic cytokine that mediates immune regulation and inflammatory responses.
  • TNF ⁇ is also known to be involved in responses to bacterial, parasitic and viral infections.
  • TNF ⁇ is known to have a role in rheumatoid arthritis (RA), inflammatory bowel diseases (including Crohn's disease), psoriasis, Alzheimer's disease (AD), Parkinson's disease (PD), pain, epilepsy, osteoporosis, asthma, sepsis, fever, Systemic lupus erythematosus (SLE) and Multiple Sclerosis (MS) and cancer.
  • RA rheumatoid arthritis
  • AD Alzheimer's disease
  • PD Parkinson's disease
  • SLE Systemic lupus erythematosus
  • MS Multiple Sclerosis
  • TNF ⁇ is also known to have a role in Amyotrophic Lateral Sclerosis (ALS), ischemic stroke, immune complex-mediated glomerulonephritis, lupus nephritis (LN), antineutrophil cytoplasmic antibodies (ANCA-) associated glomerulonephritis, minimal change disease, diabetic nephropathy (DN), acute kidney injury (AKI), obstructive uropathy, kidney allograft rejection, cisplatin-induced AKI and obstructive uropathy.
  • ALS Amyotrophic Lateral Sclerosis
  • ischemic stroke immune complex-mediated glomerulonephritis
  • LN lupus nephritis
  • ANCA- antineutrophil cytoplasmic antibodies
  • TNF superfamily Other members of the TNF superfamily are known to be involved in autoimmune disease and immune deficiencies.
  • members of the TNF superfamily are known to be involved in RA, SLE, cancer, MS, asthma, rhinitis, osteoporosis and multiple myeloma (MM).
  • TL1A is known to play a role in organ transplant rejection.
  • a compound identified using the methods of the invention, or a TNF-trimer compound complex may be used in a method of therapy of the human or animal body.
  • a compound or complex may be used to treat, prevent or ameliorate any condition that that can be treated, prevented or ameliorated by a conventional TNF superfamily member modulator.
  • the compound or complex may be used alone or in combination with a conventional TNF superfamily member modulator.
  • Pathogenic signalling through a TNF receptor by a TNF superfamily member includes increased signalling through a TNF receptor over and above the normal physiological level of signalling, signalling through a TNF receptor which is initiated normally, but which fails to stop in response to normal physiological signals and signalling through a TNF receptor that is within the normal physiological range of magnitude, but which is initiated by non-physiological means.
  • the invention relates to the treatment, prevention or amelioration of conditions mediated or influenced by TNF ⁇ .
  • the compounds that interact with TNF ⁇ are accordingly beneficial in the treatment and/or prevention of various human ailments. These include autoimmune and inflammatory disorders; neurological and neurodegenerative disorders; pain and nociceptive disorders; and cardiovascular disorders.
  • Inflammatory and autoimmune disorders include systemic autoimmune disorders, autoimmune endocrine disorders and organ-specific autoimmune disorders.
  • Systemic autoimmune disorders include systemic lupus erythematosus (SLE), psoriasis, vasculitis, polymyositis, scleroderma, multiple sclerosis, ankylosing spondylitis, rheumatoid arthritis and Sjögren's syndrome.
  • Autoimmune endocrine disorders include thyroiditis.
  • Organ-specific autoimmune disorders include Addison's disease, haemolytic or pernicious anaemia, glomerulonephritis (including Goodpasture's syndrome), Graves' disease, idiopathic thrombocytopenic purpura, insulin-dependent diabetes mellitus, juvenile diabetes, uveitis, inflammatory bowel disease (including Crohn's disease and ulcerative colitis), pemphigus, atopic dermatitis, autoimmune hepatitis, primary biliary cirrhosis, autoimmune pneumonitis, autoimmune carditis, myasthenia gravis, spontaneous infertility, osteoporosis, asthma and muscular dystrophy (including Duchenne muscular dystrophy).
  • Neurological and neurodegenerative disorders include Alzheimer's disease, Parkinson's disease, Huntington's disease, stroke, amyotrophic lateral sclerosis, spinal cord injury, head trauma, seizures and epilepsy.
  • Cardiovascular disorders include thrombosis, cardiac hypertrophy, hypertension, irregular contractility of the heart (e.g. during heart failure), and sexual disorders (including erectile dysfunction and female sexual dysfunction).
  • a compound or a complex may be used to treat or prevent inflammatory disorders, CNS disorders, immune disorders and autoimmune diseases, pain, osteoporosis, fever and organ transplant rejection.
  • a compound or a complex may be used to treat or prevent rheumatoid arthritis, inflammatory bowel diseases (including Crohn's disease), psoriasis, Alzheimer's disease, Parkinson's disease, epilepsy, asthma, sepsis, systemic lupus erythematosus, multiple sclerosis, asthma, rhinitis, cancer and osteoporosis.
  • a compound or a complex may be used to treat or prevent rheumatoid arthritis (RA), non specific inflammatory arthritis, erosive bone disease, chondritis, cartilage degeneration and/or destruction, juvenile inflammatory arthritis, Still's Disease (juvenile and/or adult onset), juvenile idiopathic arthritis, juvenile idiopathic arthritis (both oligoarticular and polyarticular forms), inflammatory bowel diseases (including Crohn's disease, ulcerative colitis, indeterminate colitis, pouchitis), psoriasis, psoriatic arthopathy, ankylosing spondylitis, Sjögren's Disease, Alzheimer's disease (AD), Behcet's Disease, Parkinson's disease (PD), amyotrophic lateral sclerosis (ALS), ischemic stroke, pain, epilepsy, osteoporosis, osteopenia, anaemia of chronic disease, cachexia, diabetes, dyslipidemia, metabolic syndrome, asthma, chronic obstructive airways (RA), r
  • Compounds identified using the methods of the invention and compound-trimer complexes will typically be formulated into pharmaceutical compositions, together with a pharmaceutically acceptable carrier.
  • “pharmaceutically acceptable carrier” includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like that are physiologically compatible.
  • the carrier may be suitable for parenteral, e.g. intravenous, intramuscular, intradermal, intraocular, intraperitoneal, subcutaneous, spinal or other parenteral routes of administration, for example by injection or infusion.
  • the carrier may be suitable for non-parenteral administration, such as a topical, epidermal or mucosal route of administration.
  • the carrier is suitable for oral administration.
  • the modulator may be coated in a material to protect the compound from the action of acids and other natural conditions that may inactivate the compound.
  • compositions of the invention may include one or more pharmaceutically acceptable salts.
  • a “pharmaceutically acceptable salt” refers to a salt that retains the desired biological activity of the parent compound and does not impart any undesired toxicological effects. Examples of such salts include acid addition salts and base addition salts.
  • Preferred pharmaceutically acceptable carriers comprise aqueous carriers or diluents.
  • suitable aqueous carriers that may be employed in the pharmaceutical compositions of the invention include water, buffered water and saline.
  • suitable aqueous carriers include ethanol, polyols (such as glycerol, propylene glycol, polyethylene glycol, and the like), and suitable mixtures thereof, vegetable oils, such as olive oil, and injectable organic esters, such as ethyl oleate.
  • isotonic agents for example, sugars, polyalcohols such as mannitol, sorbitol, or sodium chloride in the composition.
  • compositions typically must be sterile and stable under the conditions of manufacture and storage.
  • the composition can be formulated as a solution, microemulsion, liposome, or other ordered structure suitable to high drug concentration.
  • compositions of the invention may comprise additional active ingredients.
  • kits comprising compounds or complexes and instructions for use.
  • the kit may further contain one or more additional reagents, such as an additional therapeutic or prophylactic agent as discussed above.
  • the compounds and the compound-trimer complexes or formulations or compositions thereof may be administered for prophylactic and/or therapeutic treatments.
  • compounds and compound-trimer complexes are administered to a subject already suffering from a disorder or condition as described above, in an amount sufficient to cure, alleviate or partially arrest the condition or one or more of its symptoms.
  • Such therapeutic treatment may result in a decrease in severity of disease symptoms, or an increase in frequency or duration of symptom-free periods.
  • An amount adequate to accomplish this is defined as a “therapeutically effective amount”.
  • formulations are administered to a subject at risk of a disorder or condition as described above, in an amount sufficient to prevent or reduce the subsequent effects of the condition or one or more of its symptoms.
  • An amount adequate to accomplish this is defined as a “prophylactically effective amount”. Effective amounts for each purpose will depend on the severity of the disease or injury as well as the weight and general state of the subject.
  • a subject for administration may be a human or non-human animal.
  • non-human animal includes all vertebrates, e.g., mammals and non-mammals, such as non-human primates, sheep, dogs, cats, horses, cows, chickens, amphibians, reptiles, etc. Administration to humans is preferred.
  • a compound or a compound-trimer complex may be administered via one or more routes of administration using one or more of a variety of methods known in the art.
  • routes of administration for compounds or compound-trimer complexes of the invention include intravenous, intramuscular, intradermal, intraocular, intraperitoneal, subcutaneous, spinal or other parenteral routes of administration, for example by injection or infusion.
  • parenteral administration as used herein means modes of administration other than enteral and topical administration, usually by injection.
  • a compound identified by the methods of the invention or a compound-trimer complex of the present invention of the invention can be administered via a non-parenteral route, such as a topical, epidermal or mucosal route of administration.
  • a non-parenteral route such as a topical, epidermal or mucosal route of administration.
  • the compound identified by the methods of the invention or a compound-trimer complex of the invention is for oral administration.
  • a suitable dosage of a compound or a compound-trimer complex may be determined by a skilled medical practitioner. Actual dosage levels of the active ingredients in the pharmaceutical compositions of the present invention may be varied so as to obtain an amount of the active ingredient that is effective to achieve the desired therapeutic response for a particular patient, composition, and mode of administration, without being toxic to the patient.
  • the selected dosage level will depend upon a variety of pharmacokinetic factors including the activity of the particular compositions of the present invention employed, the route of administration, the time of administration, the rate of excretion of the particular compound being employed, the duration of the treatment, other drugs, compounds and/or materials used in combination with the particular compositions employed, the age, sex, weight, condition, general health and prior medical history of the patient being treated, and like factors well known in the medical arts.
  • a suitable dose may be, for example, in the range of from about 0.01 ⁇ g/kg to about 1000 mg/kg body weight, typically from about 0.1 ⁇ g/kg to about 100 mg/kg body weight, of the patient to be treated.
  • a suitable dosage may be from about 1 ⁇ g/kg to about 10 mg/kg body weight per day or from about 10 ⁇ g/kg to about 5 mg/kg body weight per day.
  • Dosage regimens may be adjusted to provide the optimum desired response (e.g., a therapeutic response). For example, a single dose may be administered, several divided doses may be administered over time or the dose may be proportionally reduced or increased as indicated by the exigencies of the therapeutic situation.
  • Dosage unit form as used herein refers to physically discrete units suited as unitary dosages for the subjects to be treated; each unit contains a predetermined quantity of active compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier.
  • Administration may be in single or multiple doses. Multiple doses may be administered via the same or different routes and to the same or different locations. Alternatively, doses can be via a sustained release formulation, in which case less frequent administration is required. Dosage and frequency may vary depending on the half-life of the antagonist in the patient and the duration of treatment desired.
  • compounds or compound-trimer complexes may be co-administered with one or other more other therapeutic agents.
  • the other agent may be an analgesic, anaesthetic, immunosuppressant or anti-inflammatory agent.
  • Combined administration of two or more agents may be achieved in a number of different ways. Both may be administered together in a single composition, or they may be administered in separate compositions as part of a combined therapy. For example, the one may be administered before, after or concurrently with the other.
  • Optical rotations were measured using an Optical Activity PolAAR 2001 polarimeter.
  • racemic title compound was prepared following the procedure described in patent application WO 2014/009295.
  • the racemic mixture thus prepared was separated into the constituent enantiomers by chiral chromatography as detailed below:
  • Compound (2) was added to 300 ⁇ M of fused TNF ⁇ trimer at a final concentration range from 90 ⁇ M to 690 ⁇ M compound with the DMSO concentration held constant at 1.0%. The sample of TNF ⁇ and compound were incubated overnight at a temperature of 4° C.
  • Receptor at a final concentration of 240 ⁇ M (3.2 fold excess over trimers) was added to 75 ⁇ M compound-trimer complex prepared as described above.
  • the final concentration of DMSO was 0.25%. The mixture was incubated for 1 hour at 22° C.
  • results are presented in FIG. 5 .
  • the numbers 1-3 refer to the number of receptors bound to TNF ⁇ trimers.
  • addition of increasing concentrations of compound (2) resulted in the number of receptors bound reducing from three on average per trimer to two on average per trimer.
  • a peak is observed showing three receptors binding per trimer (with a slight shoulder for two receptors binding per trimer).
  • the predominant peak corresponds to two receptors binding per trimer, with a slight shoulder corresponding to a peak for three receptors binding per trimer. Therefore, at this concentration of the compound the majority of trimer compound complexes bind two receptors.
  • Compound (5) was added to 20 ⁇ M of TNF ⁇ trimer at a final concentration of 200 ⁇ M (ratio of 1:10 trimers:compound) with DMSO concentration held constant at 2.0%.
  • the sample of TNF ⁇ and compound were incubated overnight at a temperature of 4° C.
  • Receptors at a final concentration of 35 ⁇ M were added to 10 ⁇ M trimer-compound complex (3.5 fold excess of receptors over the trimer-compound complex).
  • the final concentration of DMSO was 1.0%. The mixture was incubated for 1 hour at 22° C.
  • the conditions for analytical size exclusion using HPLC were as follows: injection volume: 50 ⁇ l; Superdex 200HR 10/300, L ⁇ I.D. 3 0 cm ⁇ 10 mm column, 13-15 ⁇ m particle size; and a buffer of 10 mM HEPES, pH 7.5, 150 mM NaCl.
  • the TNF ⁇ was a single polypeptide chain of trimeric human TNF ⁇ composed of human TNF ⁇ residues V77-L233 followed by two additional repeats of human TNF ⁇ residues D86-L233 linked together by Ser-Gly-Ser (sequences are based on UniProt P10375).
  • Human TNFR1 was (V43-N184)(N54D, C182S) based on sequence UniProt P19438.
  • FIG. 7 shows an overlay of the compound (5) trace with the control trace showing TNF ⁇ mutated to bind 1, 2 and 3 receptors. As compared to compound (2), the peak containing compound (5) has moved closer to 1 receptor bound.
  • mice TNF ⁇ The soluble form of mouse TNF ⁇ (VC 6535, UniProt P06804) was expressed as a fusion protein in E. coli and has the final sequence:
  • Cells were pre-cultured at 37° C. in rich media, induced with the addition of 0.1% arabinose and allowed to express overnight at 25° C. in vector pEMB54.
  • This vector introduces a cleavable N-terminal His 6 Smt-tag.
  • the cells were lysed and purified by Ni-NTA chelate chromatography.
  • the fusion protein was eluted with buffer containing imidazole and cleaved by the addition of protease.
  • the final cleaved TNF ⁇ protein was purified by a subtractive Ni chelate chromatography step to remove the fusion tag and further purified by size exclusion chromatography to remove the remaining impurities.
  • the final TNF ⁇ product was concentrated to 20.5 mg/ml and flash frozen in liquid nitrogen.
  • the extracellular domain of human TNFR1 (VC 5602, UniProt P19438) was expressed as a secreted protein in baculovirus infected insect cells and has the final sequence:
  • the fusion protein plasmid was cloned into the pEMB50 expression vector, which encodes a cleavable N-terminal secretion signal and His-tagged fusion protein.
  • Virus was generated using the baculovirus expression system. Infected insect cells secreted the fusion protein into the media.
  • the fusion protein was purified by Ni-NTA chelate chromatography and eluted from the Ni column using an imidazole gradient. The eluted protein was cleaved with protease to release the N-terminal His-fusion tag.
  • the cleaved TNFR1 was subsequently purified by a subtractive Ni chelate chromatography step and further purified by size exclusion chromatography. The final TNFR product was concentrated to 8.8 mg/ml and flash frozen in liquid nitrogen.
  • TNF ⁇ Purified mouse TNF ⁇ (20.5 mg/ml, VC 6535) was incubated with compound (1) (100 mM in DMSO) in 6 molar excess at 37° C. for 3 hours followed by overnight incubation at 4° C. The following day, human TNFR1 (8.8 mg/ml, VC 5602) was added for a final molar ratio of 3 TNF ⁇ monomers (equivalent to 1 trimer):3 TNFR1 receptor. The ternary complex (cytokine, ligand, receptor) was incubated for 1 hour prior to loading on Superdex 200 size exclusion column (23 ml) that was pre-equilibrated with 10 mM HEPES pH 7.5, 150 mM NaCl. The final purified ternary complex was concentrated to 18.5 mg/ml and immediately used in crystallization trials.
  • compound (1) 100 mM in DMSO
  • human TNFR1 8.8 mg/ml, VC 5602
  • the ternary complex cyto
  • the ternary complex was crystallized by sitting drop vapor diffusion by mixing 0.5 ⁇ l of complex with 0.5 ⁇ l of 800 mM sodium potassium tartrate, 0.5% PEG5000 MME, 100 mM Tris pH 8.5 over 100 ⁇ l of the same crystallization solution. Crystals were harvested for data collection approximately 2 months after initial set up. They were briefly soaked in paraffin oil and frozen directly in liquid nitrogen for data collection on Aug. 17, 2012 at Argonne Photon Source, beamline 21-IDF.
  • the structure of the mouse TNF ⁇ (VC 6535) and human TNFR (VC 5602) complex with compound (1) was solved by molecular replacement using Phaser with input models based upon a complexed human TNF ⁇ structure. Data were integrated in XDS and scaled using SCALA. Initial structure determination and refinement used data to 3.15 ⁇ resolution from a single crystal. Iterative manual model building using Coot (Emsley, P. and Cowtan, K. 2004. Coot: model-building tools for molecular graphics. Acta Crystallogr. D Biol. Crystallogr. December; 60(Pt 12 Pt 1):2126-32. PMID: 15572765) and in Refmac (Murshudov, G. N., Vagin, A.
  • the crystal structures, as presented in FIG. 8A-D show two receptors binding per trimer-compound complex.
  • TR-FRET Time Resolved Fluorescent Resonance Energy Transfer
  • Each of the labelled proteins was diluted to a final assay concentration of 7.5 nM (a 1:1 concentration ratio of TNF monomers:receptors) in a buffer solution (20 mM Tris, 150 mM NaCl, 0.05% Tween 20, pH 7.2). Compounds were tested in a 10 point titration with three fold dilution. The maximum compound concentration in the final assay was 25 ⁇ M. The final DMSO concentration of the assay was 5% by volume.
  • the plate was read using an LJL Analyst plate reader.
  • the samples were excited at 330 nm and fluorescence readings were taken at 495 nm and 520 nm, the emission wavelengths of the Terbium donor and AF488 acceptor, respectively.
  • a FRET ratio was calculated by dividing the acceptor counts by the donor counts and multiplying by 10,000.
  • TNFR1 In the absence of an interfering molecule TNFR1 will form a complex with the fused TNF ⁇ trimer generating a FRET signal. An interfering molecule will prevent TNFR1 from binding and subsequently will inhibit the FRET signal.
  • the inhibition of FRET may be either complete or partial, whereby there is a reduction in bound TNFR1 (ECD) indicating a reduction in TNF ⁇ -TNFR1 (ECD) stoichiometry. Complete inhibition as by an antibody is 100%.
  • FIGS. 9 and 10 represent a partial inhibition observed with compound (3) and compound (4). At the highest concentration of the compound the maximal inhibition is 29% and 36%, respectively. This corresponds to an average inhibition of one out of a possible three receptors binding (expected 33%)
  • TNF ⁇ Human TNF ⁇ was desalted and buffer exchanged into 20 mM ammonium acetate, pH 7.4 prior to use.
  • Compound was diluted from a 10 mM DMSO stock using 20 mM ammonium acetate, pH 7.4).
  • a DMSO-only control was also prepared where TNF ⁇ (20 ⁇ M) was added 1:1 (v:v) with buffer (20 mM, ammonium acetate, pH 7.4, 2% DMSO). Both solutions were incubated at room temperature overnight, after which the small molecule-containing sample was analysed by non-covalent time-of-flight mass spectrometry (Waters LCT Premier, equipped with Advion TriVersa NanoMate souce) for confirmation that the TNF ⁇ was fully bound.
  • Human TNFR1 (residues 41-184, C182S, de-glycosylated) was prepared for native MS analysis by buffer exchanging into 20 mM ammonium acetate, pH 7.4 using a zeba spin column (Thermo-Fisher, 7 kDa MWCO). Receptor was added 1:1 (v:v) to aliquots of the TNF ⁇ samples prepared previously, to give three samples per experiment containing 5, 10 and 23 ⁇ M TNFR (final TNF ⁇ concentration in each sample was 5 ⁇ M). Samples were incubated for 2 hours and analysed by ion-mobility mass spectrometry (Waters Synapt G2 Q-TOF mass spectrometer, equipped with Advion TriVersa NanoMate souce).
  • Receptor stoichiometry can be uniquely determined by mass spectrometry due to the significant mass differences obtained when 1, 2 or 3 receptors bind to TNF ⁇ . Problems are encountered however due to overlapping charge states on the mass-to-charge (m/z) scale used, e.g. the same m/z value of 2000 would be obtained from an analyte (MW 20,000 Da) with 20 charges, as an analyte (MW 32,000) with 16 charges. Ion mobility mass spectrometry is therefore required for these experiments due to the extra degree of separation obtained by measurement of both ‘drift time’ and mass-to-charge.
  • the drift time of an analyte is dependent on its mass, charge, and conformation, and is measured as the length of time taken for each analyte to traverse a gas-filled mobility cell inside the spectrometer.
  • the resulting two-dimensional plots of m/z versus drift time allow unambiguous assignment of receptor stoichiometry.
  • TNF was desalted using 2 ⁇ Zeba column followed by dialysis into 20 mM ammonium acetate pH 7.4.
  • TNFR was desalted using 2 ⁇ zeba column followed by dialysis into 20 mM ammonium acetate pH 7.4.
  • An A280 measurement was taken to confirm the final concentration of protein samples.
  • a DMSO-only control sample was also prepared.
  • hTNFR1 extracellular domain comprising residues 41 to 184 in the range of concentrations of 1000, 2000, 4000, 6000, 8000, 10000 and 23000 nM.
  • T0 represents the initial amount of TNF and Rmin, R, Rmax represent the initial concentration of Receptor R assayed starting from Rmin and ending at Rmax.
  • Fractions fobs are the molar fractions of species TNF, TNF+1R, TNF+2R and TNF+3R observed in equilibrium (ex. f TNFobs ) by native mass spectrometry measurements.
  • f calc are the molar fractions for each species (ex. f TNFcalc ) calculated by solving the equilibrium equations using the BioNetGen BNGL modelling tool (Blinov, M. L., Faeder, J. R., Goldstein, B., and Hlavacek, W. S.
  • BioNetGen software for rule-based modeling of signal transduction based on the interactions of molecular domains. Bioinformatics 20, 3289-3291) and taking as input the values of T0, R0 and K1, K2 and K3.
  • the Error function was minimised using the brute force minimisation utility implemented within the SciPy/NumPy framework (http://docs.scipy.org/doc/numpy/index.html).
  • the entire data processing analysis was implemented in the Python programming language (https://www.python.org/) calling BioNetGen routines when necessary.
  • Thermofluor which measures stabilisation of the thermal melting temperature of TNF ⁇ , did show a small stabilisation for C87.
  • C87 was preincubated with TNF ⁇ for 1 hour prior to the addition to HEK-293 cells stably transfected with SEAP under the control of NF ⁇ B.
  • An appropriate counter-screen was also tested in order to detect non-TNF related (off target) activity.
  • the assay was incubated overnight before inhibition was measured compared to 100% blocking by a control compound.
  • the maximum C87 concentration was 10,000 nM, with a 3-fold serial dilution.
  • TNF was immobilised using an avi-tag linker and C87 was passed over the chip.
  • C87 was passed over the chip.
  • a dose response of C87 from a highest concentration of 10 ⁇ M was performed. No binding was observed.
  • BIO8898 was tested against the immobilised soluble form of CD40L and the soluble form of TNF ⁇ by SPR on a Biacore 4000 machine.
  • a geomean IC 50 of 17 ⁇ M was determined for binding against CD40L whereas no binding was detected at a concentration of up to 100 ⁇ M for TNF ⁇ in this assay.
  • C87 was preincubated with TNF ⁇ for 1 hour prior to assay against the fluorescent compound (probe). Competition with the fluorescent compound either directly (binding at the same site) or indirectly (disrupting TNF) is detected by a reduction in FP.
  • Extrapolation of the inhibition curve produced an IC 50 of about 100 ⁇ M. Fluorescence quenching was, however, observed at the highest concentrations of inhibitor which, when subtracted, resulted in negligible inhibition of C87 in this assay.
  • Thermofluor measures the change of melting temperature (Tm) of TNF ⁇ due to compound either stabilising or disrupting the protein.
  • Tm melting temperature

Landscapes

  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • Molecular Biology (AREA)
  • Engineering & Computer Science (AREA)
  • Immunology (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Cell Biology (AREA)
  • Genetics & Genomics (AREA)
  • Biomedical Technology (AREA)
  • Hematology (AREA)
  • Urology & Nephrology (AREA)
  • Physics & Mathematics (AREA)
  • Zoology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Toxicology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Pathology (AREA)
  • Analytical Chemistry (AREA)
  • Food Science & Technology (AREA)
  • Microbiology (AREA)
  • General Physics & Mathematics (AREA)
  • Biotechnology (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Bioinformatics & Computational Biology (AREA)
  • Epidemiology (AREA)
  • Peptides Or Proteins (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

The invention is in the field of TNF signalling. Compounds have been identified which are capable of modulating signalling of TNF trimers through receptors. Methods of identifying such compounds are therefore provided. The compounds themselves have utility in therapy.

Description

    FIELD OF THE INVENTION
  • The present invention relates to methods for identifying compounds which modulate signalling of TNF superfamily member trimers through TNF receptors. In particular, the invention relates to identification of new small molecule modulators. The invention also relates to compounds identified by such methods and complexes of the compounds and trimers. The compounds and complexes may be used therapeutically.
  • BACKGROUND OF THE INVENTION
  • The Tumour Necrosis Factor (TNF) superfamily is a family of proteins that share a primary function of regulating cell survival and cell death. Members of the TNF superfamily share a common core motif, which consists of two antiparallel β-pleated sheets with antiparallel β-strands, forming a “jelly roll” β-structure. Another common feature shared by members of the TNF superfamily is the formation of homo- or heterotrimeric complexes. It is these trimeric forms of the TNF superfamily members that bind to, and activate, specific TNF superfamily receptors.
  • TNFα is the archetypal member of the TNF superfamily. Dysregulation of TNFα production has been implicated in a number of pathological conditions of significant medical importance. For example, TNFα has been implicated in rheumatoid arthritis, inflammatory bowel diseases (including Crohn's disease), psoriasis, Alzheimer's disease (AD), Parkinson's disease (PD), pain, epilepsy, osteoporosis, asthma, systemic lupus erythematosus (SLE) and multiple sclerosis (MS). Other members of the TNF superfamily have also been implicated in pathological conditions, including autoimmune disease.
  • Conventional antagonists of TNF superfamily members are macromolecular and act by inhibiting the binding of the TNF superfamily member to its receptor. Examples of conventional antagonists include anti-TNFα antibodies, particularly monoclonal antibodies, such as infliximab (Remicade®), adalimumab (Humira®) and certolizumab pegol (Cimzia®), or soluble TNFα receptor fusion proteins, such as etanercept (Enbrel®).
  • SUMMARY OF THE INVENTION
  • When receptors are present at an equivalent or excess concentration compared with TNF monomers (i.e. at a molar (concentration) ratio of at least 1:1 (receptors:monomers); at least 3:1 (receptors:trimers)), a TNF trimer will typically bind three receptors. The present inventors have identified small molecular entities (SMEs) that modulate TNF signalling. These SME compounds act by binding to the trimeric form of TNF, and inducing and/or stabilising a conformational change in the trimer. Trimers with the compounds bound have an altered affinity for the requisite receptors, especially a reduced affinity for the second and third receptors, which decreases the number of receptors binding per trimer-compound complex. Accordingly, downstream signalling through the receptors is reduced. These compounds can therefore be used in the treatment of conditions mediated by TNF. The present inventors have also developed methods that can identify compounds which are capable of modulating TNF signalling in this manner.
  • The present invention therefore provides a method of identifying a compound that is capable of binding to a trimeric protein that is a TNF superfamily member and modulating signalling of the trimeric protein through the requisite TNF superfamily receptor, the method comprising determining the average number of receptors bound per trimer-compound complex and thereby identifying whether the compound is capable of modulating signalling through the receptor.
  • The invention also provides:
      • a compound that is capable of binding to a trimeric protein that is a TNF superfamily member and modulating signalling of the TNF superfamily member through the requisite receptor, wherein the compound results in an equivalent average number of receptors bound per trimer-compound complex, or a change in the average number of receptors bound per trimer-compound complex, in comparison with a control;
      • a compound of formula (5), or a pharmaceutically acceptable salt or solvate thereof;
      • a complex comprising a trimeric protein that is a TNF superfamily member and a compound as defined above;
      • a compound or complex as defined above for use in a method of therapy of the human or animal body; and
      • a pharmaceutical composition comprising the compound or complex as defined above and a pharmaceutically acceptable carrier.
    BRIEF DESCRIPTION OF THE FIGURES
  • FIG. 1 shows the interactions involved in TNFα/TNF-R1 signalling in the absence of compound. The TNFα trimers bind the first two receptors with a KD of 93 pM, and bind the third receptor with a KD of approximately 1 μM. These trimer and receptor complexes (with three receptors bound) then form raft structures via dimerisation of the receptors, which result in downstream signalling.
  • FIG. 2 shows a crystal structure with only two receptor dimers interacting with a compound-stabilised TNFα trimer.
  • FIG. 3 shows the effects of two types of compounds (A and B) on TNFα/TNF-R1 signalling. Both compounds have no effect on the formation of receptor dimers, but induce and/or stabilise formation of trimers with distorted conformations. Trimers with the first type of compound (A) bind the first and second receptor, but have a reduced affinity for the third receptor. Accordingly, trimers form with only two receptors bound. Trimers with the second compound (B) bind the first receptor, but have a reduced affinity for the second and third receptors. Trimers therefore form with only one receptor bound. The decrease in the number of receptors binding per trimer interferes with raft formation.
  • FIGS. 4A, 4B, 4C, 4D, and 4E shows structures of compounds which are capable of modulating signalling of TNF superfamily member trimers through TNF receptors. FIG. 4A shows the structure of a compound of formula (1), FIG. 4B shows the structure of a compound of formula (2), FIG. 4C shows the structure of a compound of formula (3), FIG. 4D shows the structure of a compound of formula (4) and FIG. 4E shows the structure of a compound of formula (5).
  • FIG. 5 shows results of size exclusion chromatography (SEC) experiments using compound (2), TNFα and TNFR1 (in a 3.2 fold excess relative to the trimer-compound complex). At low concentrations of the compound (90 μM) the predominant peak corresponds to three receptors binding per trimer-compound complex. This peak has a slight shoulder, indicating that some trimer-compound complexes bind two receptors. When the concentration of the compound is increased and is present in an excess (690 μM) relative to the concentration of the TNF trimers, the predominant peak corresponds to two receptors binding per trimer-compound complex. The slight shoulder on the peak does though indicate that some trimers still bind all three receptors. In the Figure, results for controls comprising TNFα alone, and TNFα and TNFR1 (negative controls in the absence of compound), are also presented. In the TNFα and TNFR1 negative controls, three receptors bind per trimer-compound complex. This is achieved by preincubating a 3.2 excess of TNFR1 over TNFα trimer. More data is presented in FIG. 6.
  • FIG. 6 shows the control for the SEC experiments described in FIG. 5. TNFα was incubated with varying concentrations of TNFR1 (ranging from a 1.2-5 fold excess of receptors:trimers). As an increasing concentration of TNFR1 is added the molecular weight of the complex with TNFα increases (shifts to the left). Addition of a 5 fold excess of TNFR1 over the concentration of TNFα trimers does not increase the molecular weight of the complex over that using a 3.2 fold excess. This suggests that TNFα is saturated at a molar ratio of 3 TNFR1s to 3 TNFα monomers (3 TNFR1s per trimer).
  • FIG. 7 shows results of SEC experiments using compound (5), TNFα and TNFR1 (in a 3.5 fold excess relative to the concentration of the trimer-compound complex). Results are also presented for controls, the first of which is TNFα and receptors in the absence of compound. The second and third controls are still TNFα and receptors in the absence of compound, but the TNFα is mutated to disrupt interactions at the third, and third and second, receptor binding sites. The control which is TNFα and receptors shows a peak indicating three receptors binding per trimer. The control with a mutation at one receptor binding site shows a peak corresponding to two receptors binding per trimer, and the control with mutations at two sites shows a peak corresponding to one receptor binding per trimer. The peak obtained in the presence of compound (5) is mid-way between the second and third controls, and therefore indicates trimers binding a mixture of two receptors and one receptor respectively.
  • FIGS. 8A-8D show the results of crystallography experiments, which reveal two receptors binding per trimer-compound complex in the presence of compound (1). Parts (A)-(D) are alternate views of the same crystal structure.
  • FIG. 9 shows results of FRET experiments with increasing concentrations of compound (3). Complete inhibition as would be observed with a blocking antibody would result in no receptor binding to TNFα i.e. complete inhibition of a FRET signal. In this instance the FRET signal is partially inhibited. At the highest concentrations of the compound, maximal inhibition is 29%, suggesting that one out of the three receptors is inhibited from binding to the TNF trimer.
  • FIG. 10 shows results of FRET experiments with increasing concentrations of compound (4). Again, the FRET signal is partially inhibited. At the highest concentrations of the compound, maximal inhibition is 36%. Similar to the observation described in FIG. 9, this suggests that one out of the three receptors is inhibited from binding to the TNF trimer.
  • FIG. 11 shows an analysis of receptor binding stoichiometry by ion-mobility mass spectrometry. In the control (comprising TNFα and an excess of TNFR1), three receptors are shown to bind on average per trimer compound-complex. In contrast, in the presence of compound (3) the receptor stoichiometry is reduced and predominantly two receptors bind per trimer-compound complex.
  • FIG. 12 shows determination of dissociation constants in a control sample with TNFα and TNFR1. As increasing concentrations of TNFR1 are added to TNFα different mass species appear and then disappear corresponding to the appearance of firstly 1 TNFR1 bound to TNFα, followed by 2 TNFR1 bound to TNFα, and finally 3 TNFR1 bound to TNFα.
  • FIG. 13 shows determination of dissociation constants in a sample with TNFα, TNFR1 and compound (3). This shows a significantly worse (lower affinity) TNFR1 interaction with TNFα of the third receptor (0.22 nM to 9.612 nM).
  • DESCRIPTION OF THE SEQUENCE LISTING
  • SEQ ID NOs: 1 and 2 show sequences used in the Examples.
  • SEQ ID NO: 3 shows the HCVR of C185_01974.0.
  • SEQ ID NO: 4 shows the LCVR of C185_01974.0.
  • SEQ ID NO: 5 shows the amino acid sequence of the mIgG1 heavy chain of C185_01974.0.
  • SEQ ID NO: 6 shows the amino acid sequence of the kappa light chain of C185_01974.0.
  • SEQ ID NO: 7 shows the HCVR of C185_01979.0.
  • SEQ ID NO: 8 shows the LCVR of C185_01979.0.
  • SEQ ID NO: 9 shows the amino acid sequence of the mIgG1 heavy chain of C185_01979.0.
  • SEQ ID NO: 10 shows the amino acid sequence of the kappa light chain of C185_01979.0.
  • DETAILED DESCRIPTION OF THE INVENTION
  • It is to be understood that different applications of the disclosed methods and products may be tailored to the specific needs in the art. It is also to be understood that the terminology used herein is for the purpose of describing particular embodiments of the invention only, and is not intended to be limiting.
  • In addition as used in this specification and the appended claims, the singular forms “a”, “an”, and “the” include plural referents unless the content clearly dictates otherwise.
  • All publications, patents and patent applications cited herein, whether supra or infra, are hereby incorporated by reference in their entirety.
  • Methods for Identifying Modulators of TNF Superfamily Members
  • The present invention relates to methods of (assays for) identifying compounds that are capable of binding to a trimeric protein that is a TNF superfamily member and modulating signalling of the trimeric protein through the requisite TNF superfamily receptor. Compounds identified by the methods of the invention are therefore also known as modulators.
  • As described further below, the compounds identified by the methods of the invention generally prevent or decrease (inhibit) signalling of TNF through the requisite receptors. Such compounds are antagonists of TNF signalling. However, the methods of the invention may also be used to identify agonist compounds, which increase (enhance) signalling of TNF through the requisite receptors. In both cases, the compounds are capable of modulating TNF signalling without having to compete with the high affinity interaction between the TNF superfamily member and its receptor.
  • The compounds identified by the methods of the invention bind to the trimeric forms of TNF superfamily members. The compounds are therefore allosteric modulators that bind to the natural agonists of the TNF superfamily receptors, i.e. to trimeric forms of TNF superfamily members. Methods of screening for compounds which are capable of binding to TNF trimers are discussed further below.
  • There are 22 TNF superfamily members currently known, which are TNFα (TNFSF1A), TNFβ (TNFSF1B), CD40L (TNFSF5), BAFF (TNFSF13B/BlyS), APRIL (TNFSF13), OX40L (TNFSF4), RANKL (TNFSF11/TRANCE), TWEAK (TNFSF12), TRAIL (TNFSF10), TL1A (TNFSF15), LIGHT (TNFSF14), Lymphotoxin, Lymphotoxin β (TNFSF3), 4-1BBL (TNFSF9), CD27L (TNFSF7), CD30L (TNFSF8), EDA (Ectodysplasin), EDA-A1 (Ectodysplasin A1), EDA-A2 (Ectodysplasin A2), FASL (TNFSF6), NGF and GITRL (TNFSF18).
  • The methods of the invention may be used to identify compounds which modulate signalling of any TNF superfamily member, including the 22 known TNF superfamily members. The compounds identified using the methods of the invention may bind specifically to the trimeric forms of one or more TNF superfamily members. Compounds identified by the methods of the invention may bind specifically to only one of the TNF superfamily members, but not to any other TNF superfamily members. Compounds identified by the methods of the invention may also bind specifically to two, three, four or up to all of the TNF superfamily members.
  • By specific, it will be understood that the compounds bind to the molecule or molecules of interest, in this case the trimeric form of the TNF superfamily member, with no significant cross-reactivity to any other molecule, which may include other members of the TNF superfamily. Cross-reactivity may be assessed by any suitable method, for example surface plasmon resonance. Cross-reactivity of a compound for the trimeric form of a TNF superfamily member with a molecule other than the trimeric form of that particular TNF superfamily member may be considered significant if the compound binds to the other molecule at least 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90% or 100% as strongly as it binds to the trimeric form of the TNF superfamily member of interest. A compound that is specific for the trimeric form of a TNF superfamily member may bind to another molecule at less than 90%, 85%, 80%, 75%, 70%, 65%, 60%, 55%, 50%, 45%, 40%, 35%, 30%, 25% or 20% the strength that it binds to the trimeric form of the TNF superfamily member. Preferably, the compound binds to the other molecule at less than 20%, less than 15%, less than 10% or less than 5%, less than 2% or less than 1% the strength that it binds to the trimeric form of the TNF superfamily member
  • Preferably, the TNF superfamily member is TNFα. TNFα exists in both a soluble (TNFαs) and membrane-bound form (TNFαm). When TNFα is referred to herein this encompasses both the TNFαs and TNFαm forms. Particularly preferably, TNFα is in the TNFαs form.
  • There are currently 34 known TNF receptors, which are 4-1BB (TNFRSF9/CD137), NGF R (TNFRSF16), BAFF R (TNFRSF13C), Osteoprotegerin (TNFRSF11B), BCMA (TNFRSF17), OX40 (TNFRSF4), CD27 (TNFRSF7), RANK (TNFRSF11A), CD30 (TNFRSF8), RELT (TNFRSF19L), CD40 (TNFRSF5), TACI (TNFRSF13B), DcR3 (TNFRSF6B), TNFRH3 (TNFRSF26), DcTRAIL R1 (TNFRSF23), DcTRAIL R2 (TNFRSF22), TNF-R1 (TNFRSF1A), TNF-R2 (TNFRSF1B), DR3 (TNFRSF25), TRAIL R1 (TNFRSF10A), DR6 (TNFRSF21), TRAIL R2 (TNFRSF10B), EDAR, TRAIL R3 (TNFRSF10C), Fas (TNFRSF6/CD95), TRAIL R4 (TNFRSF10D), GITR (TNFRSF18), TROY (TNFRSF19), HVEM (TNFRSF14), TWEAK R (TNFRSF12A), TRAMP (TNFRSF25), Lymphotoxin β R (TNFRSF3) and XEDAR.
  • A requisite receptor is a receptor which acts in conjunction with a particular TNF superfamily member. In particular, a requisite receptor is a receptor which is activated by a TNF superfamily member. The TNF superfamily member trimers bind to the receptor and activation of the receptor results in downstream signalling. Combinations of TNF superfamily members and their requisite receptors are known in the art.
  • Preferably, the methods of the invention are used to identify compounds which modulate signalling through TNF-R1 (TNFR1) and TNF-R2 (TNFR2). When TNF-R is referred to herein this encompasses both TNF-R1 and TNF-R2, including the extracellular domain (ECD) of TNF-R1 and TNF-R2. More preferably, the TNF superfamily member is TNFα and the TNF receptor is TNF-R1 or TNF-R2. Even more preferably, the TNF superfamily member is TNFα and the TNF receptor is TNF-R1. Most preferably, the TNF superfamily member is TNFαs and the TNF receptor is TNF-R1.
  • The methods of the invention may be used to identify compounds which act by specifically modulating the signalling of TNF superfamily members through TNF-R1. In particular, the compounds may act by modulating the signalling of TNF superfamily members through TNF-R1, but have no effect on signalling of TNF superfamily members through TNF-R2.
  • The TNF superfamily members and their receptors may be purified or present in mixtures, such as in cultured cells, tissue samples, body fluids or culture medium.
  • In the methods of the invention, compounds are identified which modulate signalling of the trimeric protein through the requisite receptors. Modulation of signalling may refer to an increase (enhancement) in signalling through the requisite receptors. Compounds which increase signalling are agonist compounds. However, compounds identified using the methods of the invention generally prevent or decrease (inhibit) signalling through the requisite receptors. Such compounds are known as antagonists.
  • To detect the level of signalling, assays that measure the downstream effects of TNF superfamily receptor signalling can be performed. For example, a L929 murine fibrosarcoma cell-killing assay can be used to assess the stimulation of cell death by TNF. Inhibition of TNF-induced IL-8 production by human monocytes may also be used to assess whether a test compound inhibits TNF signalling via its receptor. Such assays are well known in the art.
  • The compounds identified by the methods of the invention may completely or partially inhibit signalling through a TNF receptor when a TNF superfamily member in the form of a compound-trimer complex binds to the receptor. The compound may act to reduce signalling through a TNF superfamily receptor by at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or 100%. Any change in the level of signalling may be measured by an appropriate technique, including measuring reporter gene activity by alkaline phosphatase or luciferase, NF-κB translocation using machines such as the Cellomics Arrayscan, phosphorylation of downstream effectors, recruitment of signalling molecules, or cell death.
  • The compounds identified by the methods of the invention may modulate at least one of the downstream effects of signalling through a TNF receptor when a TNF superfamily member in the form of a compound-trimer complex binds to the receptor. Such effects are discussed herein and include TNF superfamily-induced IL-8, IL17A/F, IL2 and VCAM production, TNF superfamily-induced NF-κB activation and neutrophil recruitment. Standard techniques are known in the art for measuring the downstream effects of TNF superfamily members. The compounds identified by the methods of the invention may modulate at least 1, 2, 3, 4, 5, 10 or up to all of the downstream effects of signalling through a TNF receptor.
  • The activity of the compounds identified by the methods of the invention may be quantified using standard terminology, such as IC50 or half maximal effective concentration (EC50) values. IC50 values represent the concentration of a compound that is required for 50% inhibition of a specified biological or biochemical function. EC50 values represent the concentration of a compound that is required for 50% of its maximal effect. The compounds identified by the methods of the invention may have IC50 or EC50 values of 500 nM, 400 nM, 300 nM, 200 nM, 100 nM, 90 nM, 80 nM, 70 nM, 60 nM, 50 nM, 40 nM, 30 nM, 20 nM, 10 nM, 5 nM, 1 nM, 100 pM or less. IC50 and EC50 values may be measured using any appropriate technique, for example cytokine production can be quantified using ELISA. IC50 and EC50 values can then be generated using a standard 4-parameter logistic model also known as the sigmoidal dose response model.
  • In the present invention, libraries of compounds may be screened in order to identify modulators of TNF superfamily members (i.e. using the methods disclosed herein). Such libraries typically comprise at least 260 compounds. Preferably, such libraries comprise at least 300, at least 500 or even at least 1000 compounds.
  • In the methods of the invention, the average number of receptors bound per trimer-compound complex is determined in order to identify compounds capable of modulating TNF signalling. In the absence of any compound, when receptors are present at an equivalent or excess concentration compared with TNF monomers (at a molar ratio of greater than 1:1 (receptors:monomers); greater than 3:1 (receptors:trimers)) typically three receptors bind per TNF trimer. These trimer and receptor complexes then form rafts via the formation of receptor dimers. The rafts are responsible for downstream signalling.
  • This is illustrated in FIG. 1 which shows TNF trimer and receptor interactions, and the formation of rafts, involved in TNFα and TNF-R1 signalling. As shown in the Figure, the TNFα trimers bind the first and second requisite receptors with an affinity (KD) of approximately 93 pM. The trimers then bind the third and final receptor with a KD of approximately 1 μM.
  • The compounds identified using the methods of the present invention induce and/or stabilise conformational changes within the TNF trimers. These trimers have altered affinities for receptors; especially for the second and third receptors where there is reduced affinity. This reduced affinity results in a decrease in the number of receptors binding per trimer-compound complex. For example, as shown in FIG. 2 only two receptor dimers may bind per trimer-compound complex (instead of the three receptors that would bind under normal conditions).
  • FIG. 3 shows the effects of two types of compounds on TNFα/TNF-R1 signalling. Both compounds have no effect on the formation of receptor dimers, but induce and/or stabilise formation of trimers with distorted conformations. Trimers with the first type of compound bind the first and second receptor, but have a reduced affinity for the third receptor. Accordingly, trimers form with only two receptors bound. Trimers with the second compound bind the first receptor, but have a reduced affinity for the second and third receptors. Trimers therefore form with only one receptor bound. Both types of compound thus interfere with the formation of signalling rafts due to the decrease in the number of receptors binding per trimer
  • In view of this, the methods of the present invention involve determining the average number of receptors bound per trimer-compound complex in order to identify compounds which modulate TNF signalling. The term “average” reflects the fact that a mixed population of trimers/receptors will almost certainly be present in a sample. For example, as some compounds reduce the affinity of trimers for receptor three, some trimers may still bind all three receptors in the presence of the compound, but the majority of trimers will only have two receptors bound.
  • The term “average” may refer to a modal value, i.e. the number of receptors bound per trimer-compound complex which occurs most frequently within a sample. A modal value may be determined visually from experimental results. This is illustrated in the Examples section below.
  • It is also possible to resolve experimental data in order to identify a quantitative measurement of the proportion (percentage) of trimers in a sample with three, two, one or zero receptors bound. Such methods are routine in the art. For example, using size exclusion chromatography it is possible to resolve peaks at different elution volumes, each corresponding to trimers with a different number of receptors bound. Areas under the peaks can then be calculated and the areas used to determine the proportions (percentages) of trimers in a sample binding three, two, one or zero receptors. A modal average then refers to the number of receptors binding per trimer which occurs at the highest percentage.
  • The term “average” may also refer to a mean value.
  • To illustrate both modal and mean average values, if a method (such as those described below) identifies that 5% of trimers present in a sample have 1 receptor bound, 75% have two receptors bound, and 20% have three receptors bound, the modal value will be two receptors bound per trimer. The mean value will be 2.15 receptors bound per trimer ((5×1+75×2+20×3)/100).
  • When determining mean values in this way, a result of 0-0.4 is taken to indicate that on average zero receptors are bound per trimer, a result of 0.5-1.4 is taken to indicate that on average one receptor is bound per trimer, a result of 1.5-2.4 is taken to indicate that on average two receptors are bound per trimer and a result of greater than 2.5 is taken to indicate that on average three receptors are bound per trimer.
  • In the methods of the invention, the average number of receptors bound per trimer-compound complex is typically determined in comparison to a control. The control sample is treated in the same way as the sample with the test compound. In particular, the control sample is subjected to the same experimental conditions as the sample comprising the test compound, including the same concentrations of reagents, trimers and receptors. Furthermore, the average number of receptors bound per trimer for the control is determined using the same experimental method as for the test compound.
  • The average number of receptors bound per trimer is usually determined at the same time for the test sample and for the control. In other words, the experiments are run in parallel. However, values for the average number of receptors bound per trimer in a control may also be determined prior to performing experiments on the test sample. Such values may be recorded e.g. on a computer.
  • In order to allow an effective comparison between the results, the average number of receptors bound per trimer for a control is calculated in the same way as for the test sample (i.e. modal values or mean values as discussed above).
  • The control sample may comprise TNF superfamily member trimers and the requisite receptors in the absence of compound (a negative control). In other words, the control sample is identical to the test compound sample, except there is no test compound present. The TNF superfamily member trimers and the requisite receptors may be any of those discussed above, but are the same in the control and test sample (and are present at the same concentrations).
  • Preferably, when the control sample comprises TNF superfamily member trimers and the requisite receptors in the absence of compound, a decrease/reduction in the average number of receptors bound per trimer-compound complex in the test sample in comparison to the control identifies that the compound is capable of modulating signalling through the receptor. In other words, a test compound is identified as being capable of modulating signalling of the trimeric protein through the receptor if a lower number of receptors are identified as being bound on average per trimer in the sample with the test compound compared with the control sample.
  • For example, when calculated using a modal value if the control is determined to have an average of three receptors bound per trimer, a test compound may be identified as being capable of modulating signalling if two or fewer receptors are determined to be bound on average per trimer-compound complex. A negative control comprising TNF superfamily member trimers, and the requisite receptors, in the absence of compound should be found to bind an average of three receptors per trimer (when the receptors are present at an equivalent concentration or in an excess compared with the TNF monomers; at a molar ratio of at least 1:1 (receptors:monomers) or 3:1 (receptors:trimers)). Nevertheless, if the control is identified as having an average of two receptors bound per trimer, a test compound will be identified as being capable of modulating signalling if one or zero receptors are determined to be bound on average per trimer-compound complex. Finally, if the control is identified as having an average of one receptor bound per trimer, a test compound will be identified as being capable of modulating signalling if zero receptors are determined to be bound on average per trimer-compound complex.
  • The same reasoning applies when using mean values as the average, as calculated above.
  • A decrease in the average number of receptors bound per trimer, relative to the negative control, can also simply be calculated based on the percentage of trimers in a sample binding three, two, one or zero receptors. In this case, it is first necessary to identify the percentage of trimers in both the control sample and the sample containing the test compound having three, two, one or zero receptors bound. A test compound is then identified as being capable of modulating signalling if the presence of the compound results in a change in the percentage of trimers having a certain number of receptors bound. Such calculations are typically based on the percentage of trimers in a sample having three receptors bound, where a decreased percentage of trimers having three receptors bound would be indicative of an antagonist compound that modulates signalling (the percentage of trimers binding two, one or zero receptors must concurrently increase).
  • To illustrate, in a negative control comprising receptors and trimers only (without compound) 90% of trimers may be found to bind three receptors and 10% of trimers may be found to bind two receptors. A test compound may then be identified as being capable of modulating signalling if the compound results in less than 90% of trimers binding three receptors. If a lower percentage of trimers are binding three receptors, the percentage of trimers binding two, one or zero receptors must have increased. Accordingly, the average number of receptors bound per trimer-compound complex is decreased relative to the control.
  • Preferably, a test compound is identified as being capable of modulating signalling if in the test compound sample the percentage of trimers having three receptors bound is decreased by at least 10% (i.e. at least 10% lower), at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, or at least 95% compared with the percentage of trimers having three receptors bound in the negative control sample (comprising the TNF superfamily member and receptors in the absence of compound).
  • Alternatively, the control may comprise TNF superfamily member trimers, the requisite receptors, and a compound which is known to modulate signalling through the receptors (a so called “positive control”). The compound which is known to modulate signalling through the receptor may be any compound known to decrease the average number of receptors bound per trimer to two, one, or zero (under conditions where the trimers would bind three receptors in the absence of any compound). Such compounds can be identified using the methods described herein. Examples of compounds known to decrease the average number of receptors to two per trimer are compounds (1)-(4) and an example of a compound known to decrease the average number of receptors to closer to one per trimer is compound (5). The positive control may comprise any one of these exemplary compounds.
  • As described above, the positive control sample is treated in the same way as the sample with the test compound and the same experimental conditions, methods and calculations are used for both the control sample and test compound sample. The average number of receptors bound per trimer-compound complex is usually determined at the same time for the test compound sample and for the control, but could also be determined prior to performing experiments on the test compound sample.
  • Preferably, when the positive control sample comprises TNF superfamily member trimers, the requisite receptors, and a compound which is known to be capable of modulating signalling through the receptor, an equivalent average number of receptors bound per trimer-compound complex in the test sample in comparison with the control, or decrease in the average number of receptors bound per trimer-compound complex in the test sample in comparison with the control, identifies that the compound is capable of modulating signalling through the receptor. In other words, a test compound is identified as being capable of modulating signalling of the trimeric protein through the receptor if an identical or lower number of receptors are on average identified as being bound per trimer in the sample with the test compound compared with the positive control.
  • For example, when calculated using a modal value (described above) if the positive control is determined to have an average of two receptors bound per trimer, a test compound will be identified as being capable of modulating signalling if two or fewer receptors (two receptors, one receptor or zero receptors) are determined to be bound on average per trimer-compound complex. Likewise, if the control is identified as having an average of one receptor bound per trimer, a test compound will be identified as being capable of modulating signalling if one or zero receptors are determined to be bound on average per trimer-compound complex. If the control is identified as having an average of zero receptors bound per trimer, a test compound will be identified as being capable of modulating signalling if zero receptors are also determined to be bound on average per trimer-compound complex.
  • The same reasoning applies when using mean values as the average.
  • An equivalent average number of receptors bound per trimer, or a decrease in the average number of receptors bound per trimer, in comparison with a positive control can again also be calculated using the proportions (percentages) of trimers in a sample binding three, two, one or zero receptors. As described above, it is first necessary to identify the percentage of trimers in both the control sample and the sample containing the test compound having three, two, one or zero receptors bound. A test compound is then identified as being capable of modulating signalling if the presence of the compound results in an equivalent percentage, or increased/higher percentage, of trimers having the desired number of receptors bound (or a lower number of receptors bound) in comparison with the control. Such calculations may be focused on the percentage of trimers in a sample having two or fewer receptors bound, one or fewer receptor bound, or zero receptors bound.
  • To illustrate, in a positive control sample a compound may result in 30% of the trimers present binding three receptors and 70% of the trimers present binding two receptors. A test compound may then be identified as being capable of modulating signalling if the compound results in at least 70% percent of the trimers present binding two or fewer (two, one or zero) receptors.
  • “An equivalent percentage” in this context typically refers to values which are within 10% or less of each other, preferably 5% or less. For example, a test compound which results in 70% of trimers binding two receptors can be seen as resulting in an equivalent percentage of trimers binding two receptors as a control where 75% of trimers bind two receptors.
  • A compound may also be identified as being capable of modulating signalling simply based on determination of the number of receptors binding per trimer-compound complex, without a direct comparison to a control.
  • In this scenario, a compound may be identified as being capable of modulating signalling of the trimeric protein through the receptor if an average of less than three receptors are determined to be bound per trimer-compound complex. The average number of receptors bound per trimer-compound complex may be a mean value or a modal value as described above. Compounds are preferably identified as being capable of modulating signalling of the trimeric protein through the receptor if the compound results in an average of two receptors binding per trimer. More preferably, a compound is identified as being capable of modulating signalling of the trimeric protein through the receptor if the compound results in an average of one receptor binding per trimer. A compound may also be identified as being capable of modulating signalling of the trimeric protein through the receptor if the compound results in an average of zero receptors binding per trimer. Examples of such compounds are discussed in more detail below.
  • The average number of receptors binding per trimer-compound complex is typically determined at approximately an equivalent concentration (a 1:1 molar ratio) of receptors to TNF monomers. A 1:1 ratio of receptors:momoners corresponds to a 3:1 ratio of receptors:trimers.
  • The concentration of receptors may be in a slight excess compared with the concentration of monomers. Experiments are preferably conducted at a molar ratio of up to approximately 10:1 (receptors:trimers). Experiments may be conducted at any molar ratio within a range of between approximately 3:1 and 10:1 (receptors:trimers). Preferably, assays are conducted at ratios of approximately 3:1, 4:1, 5:1: 6:1, 7:1, 8:1, 9:1 or 10:1 (receptors:trimers). In some cases, assays are conducted at multiple concentrations of receptors:trimers e.g. approximately 3:1, 6:1 and 10:1. These titrations are illustrated in more detail in the Examples section.
  • Typically, experiments to determine the average number of receptors binding per trimer-compound complex are carried out when the compound is present at a concentration of compound that ensures complete occupancy of the trimers with compound. As discussed below, occupancy of the trimers with compound may be determined using mass spectrometry. The compound may be present at an equal concentration compared with the concentration of trimers (a 1:1 ratio of compound:trimers; a 1:3 ratio of compound:TNF monomers). The compound is, however, typically present in an excess relative to the concentration of the TNF trimers. For example, the compound may be present in an excess of between 1.5× and 500× relative to the concentration of trimers. Preferably, the compound is present in an excess of between 5× and 100× relative to the concentration of trimers, more preferably in an excess of between 10× and 50× relative to the concentration of trimers. The compound may be present at an excess of at least 1.5×, at least 2×, at least 3×, at least 4×, at least 5×, at least 6×, at least 7×, at least 8×, at least 9×, at least 10×, at least 20×, at least 50×, at least 100×, at least 250× or at least 500× relative to the concentration of TNF trimers.
  • Experiments may be carried out with a range of concentrations of compound, as shown in the Examples below.
  • The average number of receptors binding per trimer may be determined using any suitable technique, for example using ion mobility mass spectrometry, size exclusion chromatography, an aggregation assay, Förster Resonance Energy Transfer and/or crystallography. These techniques may be used alone or, preferably, in combination in order to determine the average number of receptors binding per trimer. For example, any two three, four or all five of the techniques may be used together.
  • Ion mobility mass spectrometry (IMS-MS) combines ion-mobility spectrometry and mass spectrometry in order to identify components within a test sample. Methods for conducting IMS-MS, and for resolving the obtained data, are well known in the art and are illustrated further in the Examples section below.
  • In an exemplary procedure, the test compound is incubated with TNF overnight at room temperature. A native mass spectrum is first recorded to ensure that the compound has 100% occupancy of the TNF prior to addition of the compound. In other words, the native mass spectrum ensures that the compound is binding to the trimers. The compound is typically present in an excess relative to the concentration of trimers, as described above. The requisite TNF receptors are then added and incubated before the ion mobility mass spectrum of the sample is collected.
  • Ion mobility mass spectrometry assays are conducted at any suitable ratio of receptors:trimers, typically at approximate ratios of between 3:1 and 10:1, for example 3:1, 6:1 and/or 10:1 (receptors:trimers). In many cases, ion mobility mass spectrometry assays are conducted at a number of concentration ratios for a test sample.
  • Ion mobility mass spectrometry can also be used to provide affinity data for binding of the three receptors to TNF trimers. This is illustrated in the Examples section below.
  • Another technique that can be used to determine the number of receptors binding on average per TNF trimer is size exclusion chromatography. Size exclusion chromatography methods are well known in the art, and involve separating components in a solution based on their size. Smaller components in the solution elute more slowly and require a larger elution volume in comparison to larger components.
  • A TNF trimer binding three receptors will be larger than a trimer binding only two receptors. Trimers binding three receptors are therefore eluted at a smaller volume compared with trimers binding two receptors or one receptor. The peaks at different elution volumes can thus be used to identify the average number of receptors bound per trimer. This is illustrated in more detail in the Examples section below.
  • In an exemplary size exclusion chromatography procedure, the TNF trimers are incubated with an excess of compound. Occupancy of the TNF trimers with compound can be determined by IMS-MS, as described above. The samples are then incubated with receptors, and analysed by size exclusion HPLC.
  • Typically, size exclusion chromatography experiments are conducted at approximate ratios of between 3:1 and 10:1, for example 3:1, 6:1 and/or 10:1 receptors:trimers. Preferably, a compound is tested at a range of concentration ratios.
  • Controls to establish the migration (peak) positions for trimers with two receptors bound, or one receptor bound, are illustrated in the Examples section below. These controls comprise mutant TNFα, which has impaired binding of either the third, or second and third, receptors.
  • Another technique which may be used to determine the average number of receptors binding per TNF trimer is an aggregation assay.
  • Another suitable assay for determining the average number of receptors bound per trimer is Forster Resonance Energy Transfer (FRET). FRET can be used to determine whether two fluorophores (donor and acceptor) are within close proximity to each other.
  • In the assay of the present invention, the receptors may be tagged with e.g. the donor fluorophore. The trimers are then tagged with e.g. the acceptor fluorophore (possibly via a linker). Experiments are typically conducted at a ratio of 1:1 receptors:monomers, but receptor titrations may also be performed. Likewise, experiments may be performed with the test compound present at a variety of concentrations. This is illustrated in the Examples below.
  • Finally, crystallography may be used in order to determine the average number of receptors binding per trimer. Crystallography techniques are well known in the art.
  • All of the methods of the invention involve providing an output that identifies that the compound is capable of modulating signalling through the receptor. The output may be recording information e.g. in a laboratory notebook. The output may also be recording information on a computer.
  • Compounds and Complexes
  • The present invention also relates to compounds that are capable of binding to a trimeric protein that is a TNF superfamily member and modulating signalling of the TNF superfamily member through the requisite receptor. The compounds result in corresponding average number, or a change in the average number, of receptors bound per trimer-compound complex in comparison with a control. Such compounds may be identified by the methods described above.
  • The TNF superfamily members and requisite receptors may be any of those described above.
  • Compounds can readily be screened for binding to a TNF trimer using routine methods known in the art, such as mass spectrometry. Mass spectrometry can also be used to identify the presence of the TNF trimers themselves in a sample.
  • Mass spectrometric methods may include, for example, matrix-assisted laser desorption/ionization mass spectrometry (MALDI MS), surface-enhanced laser desorption/ionization mass spectrometry (SELDI MS), time of flight mass spectrometry (TOF MS) and liquid chromatography mass spectrometry (LC MS).
  • The compounds are not limited in terms of their chemical formula or structure. The compounds are typically small molecular entities (SMEs) that have a molecular weight of 1000 Da or less, preferably 750 Da or less, more preferably 600 Da or less. The compounds may bind inside the central space present within the TNF superfamily member trimer (i.e. the core of the trimer). Binding of a compound within the core of the trimer can be detected using routine methods e.g. using crystallography. The compounds may comprise a benzimidazole moiety or an isostere thereof.
  • The compounds bind to at least one TNF superfamily member and modulate the signalling of the TNF superfamily member through the requisite receptor. Modulation of signalling is described above in the context of the methods of the present invention. At a 1:1 molar ratio of receptors:monomers, or in the presence of an excess of receptors, a TNF trimer usually binds an average of three receptors. The compounds of the present invention modulate signalling by resulting in an equivalent average number of receptors bound per trimer-compound complex, or a change in the average number of receptors bound per trimer-compound complex, in comparison with a control.
  • As described above, the control may comprise TNF superfamily member trimers and receptors in the absence of the compound (a negative control). The TNF superfamily member trimers and receptors are the same for both the test compound and the control, and the test compound sample and control are subjected to the same experimental conditions (e.g. concentrations of reagents) and methods.
  • The control may be run in parallel to the sample comprising the test compound. Alternatively, the control may be run prior to the test compound sample.
  • When the control comprises TNF superfamily member trimers and receptors in the absence of compound, three receptors would be expected to bind on average per trimer when assays are performed at an equivalent (1:1) molar ratio of TNF monomers:receptors, or when the receptors are present in an excess compared to the concentration of monomers. An antagonist compound results in a decrease in the average number of receptors binding per trimer under such conditions. Methods of determining a decrease in the average number of receptors binding per trimer are discussed above.
  • Alternatively, the control may comprise TNF superfamily member trimers, the requisite receptors, and a compound known to modulate signalling of the trimers through the receptors (a positive control). Again, the TNF superfamily member trimers and receptors are the same for both the test compound and the control, and the test compound sample and control are subjected to the same experimental conditions (e.g. concentrations of reagents) and methods. Typically, the control is run in parallel to the test compound. However, the control may also be run prior to the test compound sample.
  • When the control comprises TNF superfamily member trimers, the requisite receptors, and a compound known to modulate signalling of the trimers through the receptors, a compound is identified as being capable of modulating signalling if it results in an equivalent average number of receptors bound per trimer-compound complex in comparison with the control, or decrease in the average number of receptors bound per trimer-compound complex in comparison with the control. Methods of identifying compounds in this way are described above.
  • Antagonistic compounds result in an average of less than three receptors binding per trimer-compound complex (under conditions where in the absence of compound three receptors on average would bind per trimer-compound complex). Preferably, under such conditions a test compound results in an average of two receptors binding per trimer-compound complex. Examples of such compounds include compounds (1)-(4). These compounds can be used as positive control compounds, when evaluating whether another test compound is capable of modulating signalling.
  • More preferably, a test compound results in an average of one receptor binding per trimer-compound complex. Examples of such compounds include compound (5), which results in a shift towards a single receptor binding per trimer. Again, these compounds can be used as positive control compounds when evaluating whether another test compound is capable of modulating signalling.
  • A test compound may also result in an average of zero receptors binding per trimer-compound complex.
  • The present invention also relates to a complex comprising a trimeric protein that is a TNF superfamily member and a compound. The trimeric protein that is a TNF superfamily member, and the compound, may be any of those described above.
  • Antibodies for Identifying Trimer-Compound Complexes
  • The present inventors developed antibodies that bind selectively to complexes comprising compounds of the invention and a trimeric TNF superfamily member. These antibodies may be used to identify further compounds that are capable of inhibiting TNF.
  • In particular, the present inventors have identified two antibodies, termed CA185_01974 and CA185_01979, which were raised against human TNFα in complex with a compound of the invention. The heavy chain variable region (HCVR) of CA185_01974 is shown in SEQ ID NO: 3 and the light chain variable region (LCVR) of CA185_01974 is shown in SEQ ID NO: 4. The full length IgG1 heavy chain is shown in SEQ ID NO: 5 (1974 HC mIgG1 full) and the full length light chain (1974 LC kappa full) is shown in SEQ ID NO: 6.
  • The HCVR of CA185_01979 is shown in SEQ ID NO: 7 and the LCVR of CA185_01979 is shown in SEQ ID NO: 8. The full length IgG1 heavy chain of CA185_01979 is shown in SEQ ID NO: 9 (1979 HC mIgG1 full) and the full length light chain in SEQ ID NO: 10 (1979 L C Kappa full).
  • Antibodies comprising the above HCVR/LCVR or full-length sequence pairs can readily be generated by the skilled person using standard techniques.
  • Methods of the invention for determining compounds which are capable of binding to a trimeric protein which is a TNF superfamily member and modulating signalling through the receptor may therefore involve identifying whether an antibody with a HCVR/LCVR pair of SEQ ID NOs: 3/4 or 7/8 binds the trimer-compound complex. Likewise, methods may involve identifying whether an antibody with a sequence pair of SEQ ID Nos: 5/6 or 9/10 binds the trimer compound complex. Antibody assays may be used in addition to the other assays described herein.
  • Antibodies of the invention can be tested for binding to a compound-trimer complex by, for example, standard ELISA or Western blotting. The binding selectivity of an antibody may also be determined by monitoring binding of the antibody to cells expressing the target protein, for example by flow cytometry. Thus, a screening method of the invention may comprise the step of identifying an antibody that is capable of binding a compound-trimer complex by carrying out an ELISA or Western blot or by flow cytometry.
  • The antibodies described herein selectively (or specifically) recognise at least one compound-trimer complex, i.e. epitopes within a compound-trimer complex. An antibody, or other compound, “selectively binds” or “selectively recognises” a protein when it binds with preferential or high affinity to the protein for which it is selective but does not substantially bind, or binds with low affinity, to other proteins.
  • In the present instance, a compound-trimer complex may typically bind an antibody with a HCVR/LCVR pair of SEQ ID NOs: 3/4 or 7/8 (or with sequence pairs of SEQ ID NOs: 5/6 or 9/10) with an affinity of less than 1 nM. In other words, the methods of the invention may involve determining that a compound is capable of binding to a trimeric protein which is a TNF superfamily member and modulating signalling through the receptor by identifying that an antibody with a HCVR/LCVR pair of SEQ ID NOs: 3/4 or 7/8 (or sequence pairs of SEQ ID NOs: 5/6 or 9/10) binds the trimer-compound complex with a KD-ab of less than 1 nM. In some instances, the KD-ab may be less than 500 pM, or less than 200 pM. The affinity may be determined by surface plasmon resonance. The TNF is typically human TNFα.
  • Likewise, a complex of the invention may be a complex of a trimeric TNF superfamily member and a compound, wherein the compound-trimer complex binds an antibody with a HCVR/LCVR pair of SEQ ID NOs: 3/4 or 7/8 (or sequence pairs of SEQ ID Nos: 5/6 or 9/10). Again, the TNF is typically human TNF a, and the binding affinity is typically less than 1 nM (or less than 500 pM/200 pM). Binding affinity is typically determined by surface plasmon resonance.
  • Therapeutic Indications
  • TNFα is the archetypal member of the TNF superfamily. TNFα is a pleiotropic cytokine that mediates immune regulation and inflammatory responses. In vivo, TNFα is also known to be involved in responses to bacterial, parasitic and viral infections. In particular, TNFα is known to have a role in rheumatoid arthritis (RA), inflammatory bowel diseases (including Crohn's disease), psoriasis, Alzheimer's disease (AD), Parkinson's disease (PD), pain, epilepsy, osteoporosis, asthma, sepsis, fever, Systemic lupus erythematosus (SLE) and Multiple Sclerosis (MS) and cancer. TNFα is also known to have a role in Amyotrophic Lateral Sclerosis (ALS), ischemic stroke, immune complex-mediated glomerulonephritis, lupus nephritis (LN), antineutrophil cytoplasmic antibodies (ANCA-) associated glomerulonephritis, minimal change disease, diabetic nephropathy (DN), acute kidney injury (AKI), obstructive uropathy, kidney allograft rejection, cisplatin-induced AKI and obstructive uropathy.
  • Other members of the TNF superfamily are known to be involved in autoimmune disease and immune deficiencies. In particular, members of the TNF superfamily are known to be involved in RA, SLE, cancer, MS, asthma, rhinitis, osteoporosis and multiple myeloma (MM). TL1A is known to play a role in organ transplant rejection.
  • A compound identified using the methods of the invention, or a TNF-trimer compound complex, may be used in a method of therapy of the human or animal body. A compound or complex may be used to treat, prevent or ameliorate any condition that that can be treated, prevented or ameliorated by a conventional TNF superfamily member modulator. The compound or complex may be used alone or in combination with a conventional TNF superfamily member modulator.
  • Any condition that results, partially or wholly, from pathogenic signalling through a TNF receptor by a TNF superfamily member or from a deficiency in signalling through a TNF receptor by a TNF superfamily member may in principle be treated, prevented or ameliorated according to the present invention. Pathogenic signalling through a TNF receptor by a TNF superfamily member includes increased signalling through a TNF receptor over and above the normal physiological level of signalling, signalling through a TNF receptor which is initiated normally, but which fails to stop in response to normal physiological signals and signalling through a TNF receptor that is within the normal physiological range of magnitude, but which is initiated by non-physiological means. In a preferred embodiment, the invention relates to the treatment, prevention or amelioration of conditions mediated or influenced by TNFα.
  • The compounds that interact with TNFα are accordingly beneficial in the treatment and/or prevention of various human ailments. These include autoimmune and inflammatory disorders; neurological and neurodegenerative disorders; pain and nociceptive disorders; and cardiovascular disorders.
  • Inflammatory and autoimmune disorders include systemic autoimmune disorders, autoimmune endocrine disorders and organ-specific autoimmune disorders. Systemic autoimmune disorders include systemic lupus erythematosus (SLE), psoriasis, vasculitis, polymyositis, scleroderma, multiple sclerosis, ankylosing spondylitis, rheumatoid arthritis and Sjögren's syndrome. Autoimmune endocrine disorders include thyroiditis. Organ-specific autoimmune disorders include Addison's disease, haemolytic or pernicious anaemia, glomerulonephritis (including Goodpasture's syndrome), Graves' disease, idiopathic thrombocytopenic purpura, insulin-dependent diabetes mellitus, juvenile diabetes, uveitis, inflammatory bowel disease (including Crohn's disease and ulcerative colitis), pemphigus, atopic dermatitis, autoimmune hepatitis, primary biliary cirrhosis, autoimmune pneumonitis, autoimmune carditis, myasthenia gravis, spontaneous infertility, osteoporosis, asthma and muscular dystrophy (including Duchenne muscular dystrophy).
  • Neurological and neurodegenerative disorders include Alzheimer's disease, Parkinson's disease, Huntington's disease, stroke, amyotrophic lateral sclerosis, spinal cord injury, head trauma, seizures and epilepsy.
  • Cardiovascular disorders include thrombosis, cardiac hypertrophy, hypertension, irregular contractility of the heart (e.g. during heart failure), and sexual disorders (including erectile dysfunction and female sexual dysfunction).
  • In particular, a compound or a complex may be used to treat or prevent inflammatory disorders, CNS disorders, immune disorders and autoimmune diseases, pain, osteoporosis, fever and organ transplant rejection. In a preferred embodiment, a compound or a complex may be used to treat or prevent rheumatoid arthritis, inflammatory bowel diseases (including Crohn's disease), psoriasis, Alzheimer's disease, Parkinson's disease, epilepsy, asthma, sepsis, systemic lupus erythematosus, multiple sclerosis, asthma, rhinitis, cancer and osteoporosis. In another preferred embodiment, a compound or a complex may be used to treat or prevent rheumatoid arthritis (RA), non specific inflammatory arthritis, erosive bone disease, chondritis, cartilage degeneration and/or destruction, juvenile inflammatory arthritis, Still's Disease (juvenile and/or adult onset), juvenile idiopathic arthritis, juvenile idiopathic arthritis (both oligoarticular and polyarticular forms), inflammatory bowel diseases (including Crohn's disease, ulcerative colitis, indeterminate colitis, pouchitis), psoriasis, psoriatic arthopathy, ankylosing spondylitis, Sjögren's Disease, Alzheimer's disease (AD), Behcet's Disease, Parkinson's disease (PD), amyotrophic lateral sclerosis (ALS), ischemic stroke, pain, epilepsy, osteoporosis, osteopenia, anaemia of chronic disease, cachexia, diabetes, dyslipidemia, metabolic syndrome, asthma, chronic obstructive airways (or pulmonary) disease, sepsis, fever, respiratory distress syndrome, systemic lupus erythematosus (SLE), multiple sclerosis (MS) immune complex-mediated glomerulonephritis, lupus nephritis (LN), antineutrophil cytoplasmic antibodies (ANCA-) associated glomerulonephritis, minimal change disease, diabetic nephropathy (DN), acute kidney injury (AKI), obstructive uropathy, kidney allograft rejection, cisplatin-induced AKI and obstructive uropathy, eye diseases (including diabetic retinopathy, diabetic macular oedema, retinopathy of prematurity, age related macular degeneration, macular oedema, proliferative and/or non proliferative retinopathy, corneal vascularisation including neovascularization, retinal vein occlusion, various forms of uveitis and keratitis), thryoiditis, fibrosing disorders including various forms of hepatic fibrosis, various forms of pulmonary fibrosis, systemic sclerosis, scleroderma, cancer and cancer associated complications (including skeletal complications, cachexia and anaemia).
  • Pharmaceutical Compositions, Dosages and Dosage Regimes
  • Compounds identified using the methods of the invention and compound-trimer complexes will typically be formulated into pharmaceutical compositions, together with a pharmaceutically acceptable carrier.
  • As used herein, “pharmaceutically acceptable carrier” includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like that are physiologically compatible. The carrier may be suitable for parenteral, e.g. intravenous, intramuscular, intradermal, intraocular, intraperitoneal, subcutaneous, spinal or other parenteral routes of administration, for example by injection or infusion. Alternatively, the carrier may be suitable for non-parenteral administration, such as a topical, epidermal or mucosal route of administration. In a preferred embodiment the carrier is suitable for oral administration. Depending on the route of administration, the modulator may be coated in a material to protect the compound from the action of acids and other natural conditions that may inactivate the compound.
  • The pharmaceutical compositions of the invention may include one or more pharmaceutically acceptable salts. A “pharmaceutically acceptable salt” refers to a salt that retains the desired biological activity of the parent compound and does not impart any undesired toxicological effects. Examples of such salts include acid addition salts and base addition salts.
  • Preferred pharmaceutically acceptable carriers comprise aqueous carriers or diluents. Examples of suitable aqueous carriers that may be employed in the pharmaceutical compositions of the invention include water, buffered water and saline. Examples of other carriers include ethanol, polyols (such as glycerol, propylene glycol, polyethylene glycol, and the like), and suitable mixtures thereof, vegetable oils, such as olive oil, and injectable organic esters, such as ethyl oleate. In many cases, it will be preferable to include isotonic agents, for example, sugars, polyalcohols such as mannitol, sorbitol, or sodium chloride in the composition.
  • Therapeutic compositions typically must be sterile and stable under the conditions of manufacture and storage. The composition can be formulated as a solution, microemulsion, liposome, or other ordered structure suitable to high drug concentration.
  • Pharmaceutical compositions of the invention may comprise additional active ingredients.
  • Also within the scope of the present invention are kits comprising compounds or complexes and instructions for use. The kit may further contain one or more additional reagents, such as an additional therapeutic or prophylactic agent as discussed above.
  • The compounds and the compound-trimer complexes or formulations or compositions thereof may be administered for prophylactic and/or therapeutic treatments.
  • In therapeutic applications, compounds and compound-trimer complexes are administered to a subject already suffering from a disorder or condition as described above, in an amount sufficient to cure, alleviate or partially arrest the condition or one or more of its symptoms. Such therapeutic treatment may result in a decrease in severity of disease symptoms, or an increase in frequency or duration of symptom-free periods. An amount adequate to accomplish this is defined as a “therapeutically effective amount”.
  • In prophylactic applications, formulations are administered to a subject at risk of a disorder or condition as described above, in an amount sufficient to prevent or reduce the subsequent effects of the condition or one or more of its symptoms. An amount adequate to accomplish this is defined as a “prophylactically effective amount”. Effective amounts for each purpose will depend on the severity of the disease or injury as well as the weight and general state of the subject.
  • A subject for administration may be a human or non-human animal. The term “non-human animal” includes all vertebrates, e.g., mammals and non-mammals, such as non-human primates, sheep, dogs, cats, horses, cows, chickens, amphibians, reptiles, etc. Administration to humans is preferred.
  • A compound or a compound-trimer complex may be administered via one or more routes of administration using one or more of a variety of methods known in the art. As will be appreciated by the skilled artisan, the route and/or mode of administration will vary depending upon the desired results. Examples of routes of administration for compounds or compound-trimer complexes of the invention include intravenous, intramuscular, intradermal, intraocular, intraperitoneal, subcutaneous, spinal or other parenteral routes of administration, for example by injection or infusion. The phrase “parenteral administration” as used herein means modes of administration other than enteral and topical administration, usually by injection. Alternatively, a compound identified by the methods of the invention or a compound-trimer complex of the present invention of the invention can be administered via a non-parenteral route, such as a topical, epidermal or mucosal route of administration. In a preferred embodiment the compound identified by the methods of the invention or a compound-trimer complex of the invention is for oral administration.
  • A suitable dosage of a compound or a compound-trimer complex may be determined by a skilled medical practitioner. Actual dosage levels of the active ingredients in the pharmaceutical compositions of the present invention may be varied so as to obtain an amount of the active ingredient that is effective to achieve the desired therapeutic response for a particular patient, composition, and mode of administration, without being toxic to the patient. The selected dosage level will depend upon a variety of pharmacokinetic factors including the activity of the particular compositions of the present invention employed, the route of administration, the time of administration, the rate of excretion of the particular compound being employed, the duration of the treatment, other drugs, compounds and/or materials used in combination with the particular compositions employed, the age, sex, weight, condition, general health and prior medical history of the patient being treated, and like factors well known in the medical arts.
  • A suitable dose may be, for example, in the range of from about 0.01 μg/kg to about 1000 mg/kg body weight, typically from about 0.1 μg/kg to about 100 mg/kg body weight, of the patient to be treated. For example, a suitable dosage may be from about 1 μg/kg to about 10 mg/kg body weight per day or from about 10 μg/kg to about 5 mg/kg body weight per day.
  • Dosage regimens may be adjusted to provide the optimum desired response (e.g., a therapeutic response). For example, a single dose may be administered, several divided doses may be administered over time or the dose may be proportionally reduced or increased as indicated by the exigencies of the therapeutic situation. Dosage unit form as used herein refers to physically discrete units suited as unitary dosages for the subjects to be treated; each unit contains a predetermined quantity of active compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier.
  • Administration may be in single or multiple doses. Multiple doses may be administered via the same or different routes and to the same or different locations. Alternatively, doses can be via a sustained release formulation, in which case less frequent administration is required. Dosage and frequency may vary depending on the half-life of the antagonist in the patient and the duration of treatment desired.
  • As mentioned above, compounds or compound-trimer complexes may be co-administered with one or other more other therapeutic agents. For example, the other agent may be an analgesic, anaesthetic, immunosuppressant or anti-inflammatory agent.
  • Combined administration of two or more agents may be achieved in a number of different ways. Both may be administered together in a single composition, or they may be administered in separate compositions as part of a combined therapy. For example, the one may be administered before, after or concurrently with the other.
  • The following Examples illustrate the invention.
  • EXAMPLES Example 1(A)—Synthesis of the Compounds of Formulae (1), (2), (3) and (4)
  • Synthesis of compound (1) is disclosed in WO 2013/186229 (Example 490).
  • Synthesis of compound (2) is disclosed in WO 2013/186229 (Example 2).
  • Synthesis of compound (3) is disclosed in WO 2014/009295 (Example 4).
  • Synthesis of compound (4) is disclosed in WO 2013/186229 (Example 89).
  • Example 1(B)—Synthesis of the Compound of Formula (5)
  • Nomenclature
  • Compounds were named with the aid of ACD/Name Batch (Network) ver. 12.0 or Accelyrs Draw 4.0
  • Abbreviations
  • DCM: Dichloromethane EtOAc: Ethyl acetate
    DMF: N,N-Dimethylformamide MeOH: Methanol
    DMSO: Dimethylsulfoxide SiO2: Silica
    Et2O: Diethyl ether h: Hour
    THF: Tetrahydrofuran RT: retention time
    r.t.: Room temperature MeCN: Acetonitrile
    br.: Broad M: Mass
    Brine: Saturated aqueous sodium chloride solution
    HPLC: High Performance Liquid Chromatography
    LCMS: Liquid Chromatography Mass Spectrometry
    ES+: Electrospray Positive Ionisation
    TEA: Triethylamine
    TLC: thin layer chromatography
  • Analytical Conditions
  • All NMRs were obtained either at 300 MHz or 400 MHz.
  • All reactions involving air or moisture-sensitive reagents were performed under a nitrogen atmosphere using dried solvents and glassware.
  • All compound LCMS data were determined by using the method below.
  • Method 1:
  • Waters Acquity-SQD, Waters Acquity UPLC BEH C18, 2.1×50 mm, 1.7 μm column
  • Mobile phase A: 10 mM Ammonium Formate+0.1% Ammonia
  • Mobile phase B: 95 MeCN+5% H2O+0.1% Ammonia
  • Gradient program (Flow Rate 1.0 mL/min, Column Temperature 40° C.):
  • Time A % B %
    0.00 95 5
    0.50 95 5
    1.75 5 95
    2.00 5 95
    2.25 95 5
  • It will be apparent to the one skilled in the art that different retention times (RT) may be obtained for LCMS data if different analytical conditions are used.
  • Optical rotations were measured using an Optical Activity PolAAR 2001 polarimeter.
  • Intermediate 1
  • Figure US20210140972A1-20210513-C00001
  • (6-Bromo-7-fluoro-2-methylimidazo[1,2-a]pyridin-3-yl)[2-(difluoromethoxy)phenyl]-methanol—Enantiomer A
  • The racemic title compound was prepared following the procedure described in patent application WO 2014/009295. The racemic mixture thus prepared was separated into the constituent enantiomers by chiral chromatography as detailed below:
  • The title compound was isolated by purification of racemic (6-Bromo-7-fluoro-2-methylimidazo[1,2-a]pyridin-3-yl)[2-(difluoromethoxy)phenyl]-methanol under LC conditions on Chiralpak AD (100*500 mm*mm, flow 300 mL/min, 30° C., 2-PrOH/heptane 1/9, injection of 230 mL solution at a concentration of 7.5 g/L). The first eluting enantiomer (RT 27 min) was collected and the fractions were evaporated to yield enantiomer A. [α]−12.8°. The second eluting enantiomer (RT 50 min) was collected and the fractions were evaporated to yield enantiomer B. [α]+12.7°
  • Intermediate 2
  • Figure US20210140972A1-20210513-C00002
  • 3-(trifluoromethyl)azetidin-3-ol
  • To a solution of 1-boc-3-azetidinone (11.3 g, 58.4 mmol) and (trifluoromethyl)trimethylsilane (9.22 g, 64.3 mmol) in THF (100 mL) cooled to ˜−5° C. on an ice/brine bath was added portion wise caesium fluoride (9.77 g, 64.3 mmol). The resultant mixture was allowed to stir at r.t, TLC analysis after 4 hours at indicated complete consumption of starting material and a less polar component. The reaction was quenched by the addition of saturated aqueous ammonium chloride solution (100 mL) and the aqueous phase extracted with EtOAc (3×100 mL). The organic phase was separated, dried over sodium sulphate, filtered and the volatiles were removed in vacuo to give a crude oil. The oil thus obtained was dissolved in DCM (100 mL) and trifluoroacetic acid (40 mL) added. The mixture was stirred at ambient temperature for 4 hr. The volatiles were removed in vacuo and the residue azeotroped with toluene (3×150 mL) to give the title compound trifluoroacetate salt as a brown solid (15 g). 1H NMR (400 MHz, d6 DMSO): δ/ppm 9.48 (s, 2H), 7.95 (d, J 0.3 Hz, 1H), 4.28 (d, J 13.1 Hz, 2H), 4.06 (m, 2H).
  • The compound thus obtained was used in the subsequent reaction without further purification.
  • Intermediate 3
  • Figure US20210140972A1-20210513-C00003
  • 1-[5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyrimidin-2-yl]-3-(trifluoromethyl)azetidin-3-ol
  • To a solution of Intermediate 2 (12 g) in acetonitrile (150 mL) was added TEA (30 mL) and 2-chloro-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyrimidine (16 g) and the reaction stirred at 65° C. for 18 hours. The solvents were removed in vacuo and the solid residue triturated and washed with distilled water to give a beige solid and dried under high vacuum to give the title compound as a beige solid (18.5 g). 1H NMR (300 MHz, d6 DMSO): δ/ppm 8.53 (2H, s), 7.46 (1H, s), 4.33-4.31 (2H, m), 4.10-4.08 (2H, m), 1.29 (12H, s). LCMS (ES+) RT 1.14 min, 346.0 (M+H)+.
  • Compound (5)
  • Figure US20210140972A1-20210513-C00004
  • 1-[5-[3-[(S)-[2-(difluoromethoxy)phenyl]-hydroxy-methyl]-7-fluoro-2-methyl-imidazo[1,2-a]pyridin-6-yl]pyrimidin-2-yl]-3-(trifluoromethyl)azetidin-3-ol (enantiomer A)
  • A mixture of Intermediate 1 (0.7 g, 2 mmol), Intermediate 3 (0.7 g, 2 mmol), 1,1′-bis(diphenylphosphino)ferrocene-palladium(II)dichloride dichloromethane complex (36 mg, 0.044 mmol) and 2 M sodium carbonate (2 mL) in dioxane (12 mL) was de-gassed and refluxed for 3 h. The cooled reaction mixture was diluted with EtOAc, washed twice with brine, the organic layer was dried (MgSO4) and concentrated in vacuo. The residue was columned flash column chromatography (SiO2, 0-90% EtOAc/heptane), yielding the title compound as a cream solid (500 mg, 50%). 1H NMR (300 MHz, DMSO-d6): δ 8.51 (m, 3H), 7.95 (dd, J1 2.3 Hz, J2 6.7 Hz, 1H), 7.46 (m, 2H), 7.36 (m, 2H), 7.12 (m, 2H), 6.42 (d, J 4.4 Hz, 1H), 6.18 (d, J 4.4 Hz, 1H), 4.35 (m, 2H), 4.13 (d, J 10.2 Hz, 2H), 2.12 (s, 3H). LCMS (ES+) RT 1.34 min, 540.0 (M+H)+. [α]+39.7°.
  • Example 2—Analytical Size Exclusion Chromatography (SEC) of TNFα/TNFR1/Compound Complexes
  • Size exclusion chromatography was used to determine the number of TNFR1 receptors bound to TNFα in the absence or presence of different compounds. Compound (2) was tested under the conditions described in protocol 1 below. Compound (5) was tested as described in protocol 2.
  • Protocol 1
  • Compound (2) was added to 300 μM of fused TNFα trimer at a final concentration range from 90 μM to 690 μM compound with the DMSO concentration held constant at 1.0%. The sample of TNFα and compound were incubated overnight at a temperature of 4° C.
  • Receptor at a final concentration of 240 μM (3.2 fold excess over trimers) was added to 75 μM compound-trimer complex prepared as described above. The final concentration of DMSO was 0.25%. The mixture was incubated for 1 hour at 22° C.
  • Conditions for analytical size exclusion using HPLC were as follows: injection volume: 50 μl; TSK G3000SW L×I.D. 30 cm×7.5 mm column, 10 μm particle size; buffer of 10 mM HEPES, pH 7.5, 150 mM NaCl. For the proteins, a single polypeptide chain of trimeric human TNFα composed of human TNFα residues V77-L233 followed by two additional repeats of human TNFα residues D86-L233 linked together by Ser-Gly-Ser (sequence based on UniProt P10375). Human TNFR1 (V43-N184)(N54D, C182S) was based on sequence P19438 (UniProt).
  • Results are presented in FIG. 5. In the Figure, the numbers 1-3 refer to the number of receptors bound to TNFα trimers. As shown in this Figure, addition of increasing concentrations of compound (2) resulted in the number of receptors bound reducing from three on average per trimer to two on average per trimer. In particular, at low concentrations of the compound (90 μM), a peak is observed showing three receptors binding per trimer (with a slight shoulder for two receptors binding per trimer). At an excess concentration of the compound (690 μM) the predominant peak corresponds to two receptors binding per trimer, with a slight shoulder corresponding to a peak for three receptors binding per trimer. Therefore, at this concentration of the compound the majority of trimer compound complexes bind two receptors.
  • In a separate experiment to observe the expected migration of one, two and three receptors bound to TNFα, a range of TNFR1 concentrations (1.2, 2.2, 3.2 and 5 fold excess relative to the concentration of TNFα trimer) were added to TNFα. Results are presented in FIG. 6. As shown in this Figure, increasing the concentration of receptors increases the molecular weight of the complex (shifts left) suggesting that the number of receptors binding per trimer shifts from one, through to three. Three receptors maximally occupies the TNFα trimer since increasing the TNFR1 concentration further has no effect.
  • Protocol 2
  • Compound (5) was added to 20 μM of TNFα trimer at a final concentration of 200 μM (ratio of 1:10 trimers:compound) with DMSO concentration held constant at 2.0%. The sample of TNFα and compound were incubated overnight at a temperature of 4° C. Receptors at a final concentration of 35 μM were added to 10 μM trimer-compound complex (3.5 fold excess of receptors over the trimer-compound complex). The final concentration of DMSO was 1.0%. The mixture was incubated for 1 hour at 22° C.
  • The conditions for analytical size exclusion using HPLC were as follows: injection volume: 50 μl; Superdex 200HR 10/300, L×I.D. 3 0 cm×10 mm column, 13-15 μm particle size; and a buffer of 10 mM HEPES, pH 7.5, 150 mM NaCl.
  • The TNFα was a single polypeptide chain of trimeric human TNFα composed of human TNFα residues V77-L233 followed by two additional repeats of human TNFα residues D86-L233 linked together by Ser-Gly-Ser (sequences are based on UniProt P10375). Human TNFR1 was (V43-N184)(N54D, C182S) based on sequence UniProt P19438.
  • To establish markers for the expected migration of one, two and three receptors bound to TNFα, point mutants of human TNFα that disrupt interactions at one, two and three receptor binding sites were added to 3.5× receptors (a 3.5 excess of receptors to trimers) in buffer containing the same final concentration of 1.0% DMSO.
  • FIG. 7 shows an overlay of the compound (5) trace with the control trace showing TNFα mutated to bind 1, 2 and 3 receptors. As compared to compound (2), the peak containing compound (5) has moved closer to 1 receptor bound.
  • Example 3—Crystallography Showing the Ternary Complex of Murine TNFα-TNFR1-Compound (1)
  • The soluble form of mouse TNFα (VC 6535, UniProt P06804) was expressed as a fusion protein in E. coli and has the final sequence:
  • (SEQ ID NO: 1)
    DKPVAHVVANHQVEEQLEWLSQRANALLANGMDLKDNQLVVPADGLYLVY
    SQVLFKGQGCPDYVLLTHTVSRFAISYQEKVNLLSAVKSPCPKDTPEGAE
    LKPWYEPIYLGGVFQLEKGDQLSAEVNLPKYLDFAESGQVYFGVIAL.
  • Cells were pre-cultured at 37° C. in rich media, induced with the addition of 0.1% arabinose and allowed to express overnight at 25° C. in vector pEMB54. This vector introduces a cleavable N-terminal His6Smt-tag. The cells were lysed and purified by Ni-NTA chelate chromatography. The fusion protein was eluted with buffer containing imidazole and cleaved by the addition of protease. The final cleaved TNFα protein was purified by a subtractive Ni chelate chromatography step to remove the fusion tag and further purified by size exclusion chromatography to remove the remaining impurities. The final TNFα product was concentrated to 20.5 mg/ml and flash frozen in liquid nitrogen.
  • The extracellular domain of human TNFR1 (VC 5602, UniProt P19438) was expressed as a secreted protein in baculovirus infected insect cells and has the final sequence:
  • (SEQ ID NO: 2)
    GSVCPQGKYIHPQDNSICCTKCHKGTYLYNDCPGPGQDTDCRECESGSFT
    ASENHLRHCLSCSKCRKEMGQVEISSCTVDRDTVCGCRKNQYRHYWSENL
    FQCFNCSLCLNGTVHLSCQEKQNTVCTCHAGFFLRENECVSSSN
  • The fusion protein plasmid was cloned into the pEMB50 expression vector, which encodes a cleavable N-terminal secretion signal and His-tagged fusion protein. Virus was generated using the baculovirus expression system. Infected insect cells secreted the fusion protein into the media. The fusion protein was purified by Ni-NTA chelate chromatography and eluted from the Ni column using an imidazole gradient. The eluted protein was cleaved with protease to release the N-terminal His-fusion tag. The cleaved TNFR1 was subsequently purified by a subtractive Ni chelate chromatography step and further purified by size exclusion chromatography. The final TNFR product was concentrated to 8.8 mg/ml and flash frozen in liquid nitrogen.
  • Purified mouse TNFα (20.5 mg/ml, VC 6535) was incubated with compound (1) (100 mM in DMSO) in 6 molar excess at 37° C. for 3 hours followed by overnight incubation at 4° C. The following day, human TNFR1 (8.8 mg/ml, VC 5602) was added for a final molar ratio of 3 TNFα monomers (equivalent to 1 trimer):3 TNFR1 receptor. The ternary complex (cytokine, ligand, receptor) was incubated for 1 hour prior to loading on Superdex 200 size exclusion column (23 ml) that was pre-equilibrated with 10 mM HEPES pH 7.5, 150 mM NaCl. The final purified ternary complex was concentrated to 18.5 mg/ml and immediately used in crystallization trials.
  • The ternary complex was crystallized by sitting drop vapor diffusion by mixing 0.5 μl of complex with 0.5 μl of 800 mM sodium potassium tartrate, 0.5% PEG5000 MME, 100 mM Tris pH 8.5 over 100 μl of the same crystallization solution. Crystals were harvested for data collection approximately 2 months after initial set up. They were briefly soaked in paraffin oil and frozen directly in liquid nitrogen for data collection on Aug. 17, 2012 at Argonne Photon Source, beamline 21-IDF.
  • The structure of the mouse TNFα (VC 6535) and human TNFR (VC 5602) complex with compound (1) was solved by molecular replacement using Phaser with input models based upon a complexed human TNFα structure. Data were integrated in XDS and scaled using SCALA. Initial structure determination and refinement used data to 3.15 Å resolution from a single crystal. Iterative manual model building using Coot (Emsley, P. and Cowtan, K. 2004. Coot: model-building tools for molecular graphics. Acta Crystallogr. D Biol. Crystallogr. December; 60(Pt 12 Pt 1):2126-32. PMID: 15572765) and in Refmac (Murshudov, G. N., Vagin, A. A., and Dodson, E. J. 1997. Refinement of macromolecular structures by the maximum-likelihood method. May 1; 53(Pt3):240-55. PMID: 15299926) continued until R and Rfree reached R=0.222, Rfree=0.272. Model quality was validated using Coot and MolProbity (Lovell, S. C., Davis, I. W., Arendall W. B., de Bakker, P. I., Word, J. M., Prisant, M. G., Richardson, J. S., and Richardson, D. C. 2003. Structure validation by Dalpha geometry: phi, psi and Cbeta deviation. Proteins. February 15; 50(3):437-50. PMID: 12557186). Final data processing and refinement statistics are listed in Table 1.
  • TABLE 1
    Data collection and refinement statistics.
    Data collection Dataset 1
    Crystal ID 234879c07
    Beamline APS 21-IDF
    Collection date Aug. 17, 2012
    Oscillation width (°) 1.0
    Frames 60
    Exposure (sec) 3
    Distance (mm) 300
    Wavelength (Å) 0.97872
    Data processing (outer shell)
    Space Group P 41 21 2
    Unit cell (Å, °) a = b = 133.577, c = 141.445;
    α = β = γ = 90
    Resolution (Å) 3.15
    I/σ 8.3 (1.8)
    Completeness (%) 99.9 (100)
    Rpim 0.095 (0.45)
    Rmerge (%) 0.19 (0.90)
    Reflections (unique) 18,167 (1,314)
    Multiplicity 4.8 (5.0)
    Refinement statistics
    Rwork/Rfreeoverall 22.2/27.2
    RMSD bonds (Å) 0.011
    RMSD angles (°) 1.222
    Ramachandran outliers (%) 0.9
    Ramachandran favored (%) 95.9
    Molprobity score 1.91; 100th percentile*
    (N = 2048, 3.15 Å ± 0.25 Å)
    Peer Reviewed by: David Fox
    *100th percentile is the best among structures of comparable resolution; 0th percentile is the worst.
  • The crystal structures, as presented in FIG. 8A-D, show two receptors binding per trimer-compound complex.
  • Example 4—TNFα TNFR1 TR-FRET
  • A homogenous Time Resolved Fluorescent Resonance Energy Transfer (TR-FRET) assay was developed to measure compound mediated reduction in TNF Receptor 1 (TNFR1 extracellular domain (ECD)) binding to a fused TNFα trimer.
  • Terbium labelled Streptavidin, in complex with a biotinylated fused TNFα trimer, formed the donor portion of the FRET pair. Alexa Fluor 488 (AF488) conjugated TNFR1 (ECD) was used as the FRET acceptor.
  • TABLE 2
    Protein constructs and labels for FRET assays
    Protein Construct Label
    TNFR1(ECD)-6xLysine -Human Amine reactive Alexa Fluor 488 5-TFP
    single chain Fc (Life technologies, A30005)
    TNFα (fused trimer) - AVI Tag Biotin molecule on AVI Tag (Avidity,
    Biotin-protein ligase, EC 6.3.4.15)
    Streptavidin LanthaScreen Amine Reactive Terbium
    Chelate (Life Technologies, PV3581)
  • Each of the labelled proteins was diluted to a final assay concentration of 7.5 nM (a 1:1 concentration ratio of TNF monomers:receptors) in a buffer solution (20 mM Tris, 150 mM NaCl, 0.05% Tween 20, pH 7.2). Compounds were tested in a 10 point titration with three fold dilution. The maximum compound concentration in the final assay was 25 μM. The final DMSO concentration of the assay was 5% by volume.
  • After incubating for twenty hours the plate was read using an LJL Analyst plate reader. The samples were excited at 330 nm and fluorescence readings were taken at 495 nm and 520 nm, the emission wavelengths of the Terbium donor and AF488 acceptor, respectively. A FRET ratio was calculated by dividing the acceptor counts by the donor counts and multiplying by 10,000.
  • In the absence of an interfering molecule TNFR1 will form a complex with the fused TNFα trimer generating a FRET signal. An interfering molecule will prevent TNFR1 from binding and subsequently will inhibit the FRET signal.
  • The inhibition of FRET may be either complete or partial, whereby there is a reduction in bound TNFR1 (ECD) indicating a reduction in TNFα-TNFR1 (ECD) stoichiometry. Complete inhibition as by an antibody is 100%.
  • FIGS. 9 and 10 represent a partial inhibition observed with compound (3) and compound (4). At the highest concentration of the compound the maximal inhibition is 29% and 36%, respectively. This corresponds to an average inhibition of one out of a possible three receptors binding (expected 33%)
  • Example 5—Analysis of TNF Receptor Binding Stoichiometry by Ion-Mobility Mass Spectrometry
  • Human TNFα was desalted and buffer exchanged into 20 mM ammonium acetate, pH 7.4 prior to use. A combination of zeba spin columns (Thermo-Fisher, 7 kDa MWCO) followed by micro-dialysis (Thermo slide-a-lyzer mini dialysis units, 10 kDa MWCO) ensured that the protein was fully desalted and produced well-resolved signals by native mass spectrometry. TNFα (20 μM) was added 1:1 (v:v) with a small molecule TNFα inhibitor (compound (3)), 200 μM, 2% DMSO. Compound was diluted from a 10 mM DMSO stock using 20 mM ammonium acetate, pH 7.4). A DMSO-only control was also prepared where TNFα (20 μM) was added 1:1 (v:v) with buffer (20 mM, ammonium acetate, pH 7.4, 2% DMSO). Both solutions were incubated at room temperature overnight, after which the small molecule-containing sample was analysed by non-covalent time-of-flight mass spectrometry (Waters LCT Premier, equipped with Advion TriVersa NanoMate souce) for confirmation that the TNFα was fully bound.
  • Human TNFR1 (residues 41-184, C182S, de-glycosylated) was prepared for native MS analysis by buffer exchanging into 20 mM ammonium acetate, pH 7.4 using a zeba spin column (Thermo-Fisher, 7 kDa MWCO). Receptor was added 1:1 (v:v) to aliquots of the TNFα samples prepared previously, to give three samples per experiment containing 5, 10 and 23 μM TNFR (final TNFα concentration in each sample was 5 μM). Samples were incubated for 2 hours and analysed by ion-mobility mass spectrometry (Waters Synapt G2 Q-TOF mass spectrometer, equipped with Advion TriVersa NanoMate souce).
  • Receptor stoichiometry can be uniquely determined by mass spectrometry due to the significant mass differences obtained when 1, 2 or 3 receptors bind to TNFα. Problems are encountered however due to overlapping charge states on the mass-to-charge (m/z) scale used, e.g. the same m/z value of 2000 would be obtained from an analyte (MW 20,000 Da) with 20 charges, as an analyte (MW 32,000) with 16 charges. Ion mobility mass spectrometry is therefore required for these experiments due to the extra degree of separation obtained by measurement of both ‘drift time’ and mass-to-charge. The drift time of an analyte is dependent on its mass, charge, and conformation, and is measured as the length of time taken for each analyte to traverse a gas-filled mobility cell inside the spectrometer. The resulting two-dimensional plots of m/z versus drift time allow unambiguous assignment of receptor stoichiometry.
  • In the control experiment, when the molar excess of added TNFR was greater than three-times the concentration of TNFα, three receptors were observed binding per TNFα trimer. In the presence of a small molecule TNFα inhibitor such as compound (3), the receptor stoichiometry was reduced and predominantly two receptors were bound per TNFα trimer (see FIG. 11).
  • Example 6—Mass Spectrometry Analysis Measuring the Effects of Compound (3) on TNFR1 Affinity for TNFα
  • Compound Stock:
  • 2 μl of 10 mM DMSO stock, plus 2 μl DMSO were added to 96 μl of 20 mM ammonium acetate buffer to give 100 μl 200 μM compound (3), 4% DMSO.
  • Protein Stocks:
  • TNF was desalted using 2× Zeba column followed by dialysis into 20 mM ammonium acetate pH 7.4. TNFR was desalted using 2× zeba column followed by dialysis into 20 mM ammonium acetate pH 7.4. An A280 measurement was taken to confirm the final concentration of protein samples.
  • Compounds were added 1:1 (v:v) to TNF (40 μM) and incubated overnight at room temperature. (Final DMSO concentration=2%)
  • A DMSO-only control sample was also prepared.
  • For each sample, 5 μL TNF plus compound or TNF plus DMSO-only was added to 5 μL TNFR at each concentration specified. Final [TNF] was 5 μM.
  • Samples were incubated with TNFR for 2 hours prior to analysis.
  • Solutions containing 5000 nM hTNFα and 5000 nM hTNFα in the presence of compound (3) were titrated with hTNFR1 (extracellular domain comprising residues 41 to 184) in the range of concentrations of 1000, 2000, 4000, 6000, 8000, 10000 and 23000 nM.
  • Ion Mobility Mass Spectral analysis was performed on Advion Nanomate—Waters Synapt G2 instrument. The following instrument parameters were utilised.
  • Cone=50V
  • Source temp=20C
  • Trap/transfer collision energy=off
  • Trap gas flow=0.4 mL/min
  • Helium cell=180 mL/min,
  • IMS (N2)=90 mL/min
  • Trap DC bias=40V
  • Mobility trapping manual release—not enabled
  • IMS wave delay=450 us
  • IMS wave velocity=750 m/s
  • IMS wave height 40 V
  • Backing=6.21 mbar
  • trap 2.05e-2 mbar
  • IMS 3.47 mbar
  • TOF 1.2e-6 mbar
  • Quad Profile:
  • 4000, 5000, 6000 (dwell 30, ramp 30)
  • Range 500-8000
  • Data was analysed by extracting mass spectra for each species within driftscope software. The resulting spectra were smoothed (50/5) and peak heights summed over all charge states.
  • The ion counts of peaks corresponding to species TNF (no receptor bound), TNF+1R (one receptor bound), TNF+2R (two receptors bound) and TNF+3R (three receptors bound) were measured. Normalised ion counts were calculated as the fraction of ions of each species divided by the total amount of ions counted. These values were used as equivalent to molar fraction of each species in equilibrium. Data from the two experiments are summarised in the tables below:
  • TABLE 3
    Initial Concentration
    hTNFα hTNFR1 Normalised Ion Count
    Trimer [nM] [nM] TNF TNF + 1R TNF + 2R TNF + 3R
    5000 1000 0.973 0.027 0.000 0.000
    5000 2000 0.767 0.233 0.000 0.000
    5000 4000 0.421 0.525 0.054 0.000
    5000 6000 0.121 0.547 0.332 0.000
    5000 8000 0.000 0.331 0.669 0.000
    5000 10000 0.000 0.000 0.720 0.280
    5000 23000 0.000 0.000 0.000 1.000
  • TABLE 4
    Initial Concentration
    hTNFα
    Trimer [nM] + hTNFR1 Normalised Ion Count
    compound (3) [nM] TNF TNF + 1R TNF + 2R TNF + 3R
    5000 1000 0.869 0.131 0.000 0.000
    5000 2000 0.789 0.211 0.000 0.000
    5000 4000 0.396 0.558 0.046 0.000
    5000 6000 0.113 0.665 0.222 0.000
    5000 8000 0.034 0.490 0.476 0.000
    5000 10000 0.000 0.050 0.950 0.000
    5000 23000 0.000 0.000 0.713 0.287
  • In order to derive equilibrium constants from these data, the system in equilibrium was represented by the transformations:
  • Figure US20210140972A1-20210513-C00005
  • To calculate the set of dissociation constants K1, K2 and K3 in best agreement with native mass spectrometry data, values of K1, K2, K3 that produce molar fractions of the species TNF, TNF+1R, TNF+2R and TNF+3R closest to the measured molar fractions of those species were obtained by minimisation of the function:
  • Error ( K 1 , K 2 , K 3 ) = T 0 , Rmin T 0 , Rmax [ ( f TNF calc - f TNF obs ) 2 + ( f TNF + 1 R calc - f TNF + 1 R obs ) 2 + ( f TNF + 2 R calc - f TNF + 2 R obs ) 2 + ( f TNF + 3 R calc - f TNF + 3 R obs ) 2 ] T 0 , R 1 / 2
  • where T0 represents the initial amount of TNF and Rmin, R, Rmax represent the initial concentration of Receptor R assayed starting from Rmin and ending at Rmax. Fractions fobs are the molar fractions of species TNF, TNF+1R, TNF+2R and TNF+3R observed in equilibrium (ex. fTNFobs) by native mass spectrometry measurements. fcalc are the molar fractions for each species (ex. fTNFcalc) calculated by solving the equilibrium equations using the BioNetGen BNGL modelling tool (Blinov, M. L., Faeder, J. R., Goldstein, B., and Hlavacek, W. S. (2004) BioNetGen: software for rule-based modeling of signal transduction based on the interactions of molecular domains. Bioinformatics 20, 3289-3291) and taking as input the values of T0, R0 and K1, K2 and K3. The Error function was minimised using the brute force minimisation utility implemented within the SciPy/NumPy framework (http://docs.scipy.org/doc/numpy/index.html). The entire data processing analysis was implemented in the Python programming language (https://www.python.org/) calling BioNetGen routines when necessary.
  • After data analysis, three equilibrium constants (K1, K2 and K3) corresponding to the three receptor binding events were calculated for the mixtures of TNF and receptor with and without compound (3). Data were visualised by plotting on the “Y” axis the molar fractions of all species in equilibrium and on the “X” axis, the concentration of receptor added to a fixed initial concentration of TNF. Symbols represent the observed molar fractions of species measured in the native mass spectrometry experiment and traces correspond to the expected concentrations calculated from the equilibrium constants K1, K2 and K3. These graphs are presented in FIGS. 12 and 13.
  • TABLE 5
    K1 K2 K3
    Sample [nM] [nM] [nM]
    TNF + Receptor 0.01 0.02 0.22
    TNF + Receptor + 0.04 0.19 9612
    compound (3)
  • Example 7—Compounds and Complexes of Ma et al (2014) and Silvian et al (2011) have Different Characteristics to Those of the Present Invention
  • As described on page 12458 of Ma et al. (2014) JBC 289:12457-12466, C87 was discovered through virtual screening by attempting to find molecules which fit the space occupied by a 7 amino-acid peptide from loop2/domain2 of TNFR1 in its interaction with the external surface of TNFβ. The C87 compound from Ma et al. and the B108898 compound from Silvian et al. (2011) ACS Chemical Biology 6:636-647 were tested by the present inventors.
  • Summary of Findings
  • The Biacore observations described in Ma et al. for C87 could not be repeated.
  • No evidence of TNF specific inhibition in cells was observed.
  • Additionally C87 was not observed to bind by mass spectrometry, which is sensitive to millimolar affinities.
  • Extensive crystallography trials only produced apo-TNF (TNF without compound).
  • In the fluorescence polarisation (FP) assay, C87 showed no significant inhibition above the interference level of the compound with the fluorescent read-out.
  • Thermofluor, which measures stabilisation of the thermal melting temperature of TNFα, did show a small stabilisation for C87.
  • In summary, no evidence was found that C87 binds in the centre of the trimer. The overwhelming majority of the data suggested no direct interaction with TNFα. BIO8898 was also found not to bind to TNFα.
  • Cells—TNF Induced HEK NFKB Reporter Gene Assay
  • C87 was preincubated with TNFα for 1 hour prior to the addition to HEK-293 cells stably transfected with SEAP under the control of NFκB. An appropriate counter-screen was also tested in order to detect non-TNF related (off target) activity. The assay was incubated overnight before inhibition was measured compared to 100% blocking by a control compound. The maximum C87 concentration was 10,000 nM, with a 3-fold serial dilution.
  • No inhibitory effect could be detected that could not be attributed to off-target activity.
  • Biacore
  • TNF was immobilised using an avi-tag linker and C87 was passed over the chip. In one experiment, a dose response of C87 from a highest concentration of 10 μM was performed. No binding was observed.
  • In a second experiment, the flow rate of C87 passing over the chip was reduced. A small shift was observed but overall binding was negligible.
  • The binding of C87 to TNF described in Ma et al was likely to be super-stoichiometric based on the RU value on the Y-axis. At standard TNF density on the chip this value was in the region of thirty times higher than expected for simple 1:1 binding.
  • In another experiment, BIO8898 was tested against the immobilised soluble form of CD40L and the soluble form of TNFα by SPR on a Biacore 4000 machine. A geomean IC50 of 17 μM was determined for binding against CD40L whereas no binding was detected at a concentration of up to 100 μM for TNFα in this assay.
  • Mass Spectrometry
  • There was no evidence of C87 binding to human TNFα (20 μM) at a concentration of 400 μM. A species of lower molecular weight (˜473 Da appears to bind at less than 5% occupancy). C87 has a molecular weight of 503 Da. Based on the occupancy at a concentration of 400 μM, an affinity of the low molecular weight species in excess of 1 mM is predicted.
  • Crystallography
  • Overall a large effort was put into crystallising C87 with TNFα, including testing conditions that routinely work with compounds described in the present application. This comprised setting up a large number of crystallization trials at different ligand concentrations, different protein concentrations, and different soaking times. A few crystals were observed that, on analysis, proved to be salt or TNF with no compound.
  • Fluorescent Polarization (FP)
  • C87 was preincubated with TNFα for 1 hour prior to assay against the fluorescent compound (probe). Competition with the fluorescent compound either directly (binding at the same site) or indirectly (disrupting TNF) is detected by a reduction in FP.
  • Extrapolation of the inhibition curve produced an IC50 of about 100 μM. Fluorescence quenching was, however, observed at the highest concentrations of inhibitor which, when subtracted, resulted in negligible inhibition of C87 in this assay.
  • Thermofluor
  • Thermofluor measures the change of melting temperature (Tm) of TNFα due to compound either stabilising or disrupting the protein. A stabilization effect of 3.8° C. was observed at a concentration of 500 μM C87, suggesting the possibility of weak binding, which may not be specific.
  • Sequence listing
    SEQ ID NO: 1
    DKPVAHVVANHQVEEQLEWLSQRANALLANGMDLKDNQLVVPADGLYLVY
    SQVLFKGQGCPDYVLLTHTVSRFAISYQEKVNLLSAVKSPCPKDTPEGAE
    LKPWYEPIYLGGVFQLEKGDQLSAEVNLPKYLDFAESGQVYFGVIAL
    SEQ ID NO: 2
    GSVCPQGKYIHPQDNSICCTKCHKGTYLYNDCPGPGQDTDCRECESGSFT
    ASENHLRHCLSCSKCRKEMGQVEISSCTVDRDTVCGCRKNQYRHYWSENL
    FQCFNCSLCLNGTVHLSCQEKQNTVCTCHAGFFLRENECVSSSN
    (HCVR of 1974)
    SEQ ID NO: 3
    DVQLVESGGGLVQPGRSLKLSCAASGFTFSAYYMAWVRQAPTKGLEWVAS
    INYDGANTFYRDSVKGRFTVSRDNARSSLYLQMDSLRSEDTATYYCTTEA
    YGYNSNWFGYWGQGTLVTVSS
    (LCVR of 1974)
    SEQ ID NO: 4
    DIQMTQSPASLPASPEEIVTITCQASQDIGNWLSWYQQKPGKSPQLLIYG
    ATSLADGVPSRFSASRSGTQYSLKISRLQVEDFGIFYCLQGQSTPYTFGA
    GTKLELK
    (1974 HC mIgG1 full)
    SEQ ID NO: 5
    DVQLVESGGGLVQPGRSLKLSCAASGFTFSAYYMAWVRQAPTKGLEWVAS
    INYDGANTFYRDSVKGRFTVSRDNARSSLYLQMDSLRSEDTATYYCTTEA
    YGYNSNWFGYWGQGTLVTVSSAKTTPPSVYPLAPGSAAQTNSMVTLGCLV
    KGYFPEPVTVTWNSGSLSSGVHTFPAVLQSDLYTLSSSVTVPSSTWPSET
    VTCNVAHPASSTKVDKKIVPRDCGCKPCICTVPEVSSVFIFPPKPKDVLT
    ITLTPKVTCVVVDISKDDPEVQFSWFVDDVEVHTAQTQPREEQFNSTFRS
    VSELPIMHQDWLNGKEFKCRVNSAAFPAPIEKTISKTKGRPKAPQVYTIP
    PPKEQMAKDKVSLTCMITDFFPEDITVEWQWNGQPAENYKNTQPIMDTDG
    SYFVYSKLNVQKSNWEAGNTFTCSVLHEGLHNHHTEKSLSHSPGK
    (1974 LC kappa full)
    SEQ ID NO: 6
    DIQMTQSPASLPASPEEIVTITCQASQDIGNWLSWYQQKPGKSPQLLIYG
    ATSLADGVPSRFSASRSGTQYSLKISRLQVEDFGIFYCLQGQSTPYTFGA
    GTKLELKRTDAAPTVSIFPPSSEQLTSGGASVVCFLNNFYPKDINVKWKI
    DGSERQNGVLNSWTDQDSKDSTYSMSSTLTLTKDEYERHNSYTCEATHKT
    STSPIVKSFNRNEC
    (HCVR of 1979)
    SEQ ID NO: 7
    EVHLVESGPGLVKPSQSLSLTCSVTGYSITNSYWDWIRKFPGNKMEWMGY
    INYSGSTGYNPSLKSRISISRDTSNNQFFLQLNSITTEDTATYYCARGTY
    GYNAYHFDYWGRGVMVTVSS
    (LCVR of 1979)
    SEQ ID NO: 8
    DIQMTQSPASLSASLEEIVTITCQASQDIGNWLSWYQQKPGKSPHLLIYG
    TTSLADGVPSRFSGSRSGTQYSLKISGLQVADIGIYVCLQAYSTPFTFGS
    GTKLEIK
    (1979 HC mIgG1 full)
    SEQ ID NO: 9
    EVHLVESGPGLVKPSQSLSLTCSVTGYSITNSYWDWIRKFPGNKMEWMGY
    INYSGSTGYNPSLKSRISISRDTSNNQFFLQLNSITTEDTATYYCARGTY
    GYNAYHFDYWGRGVMVTVSSAKTTPPSVYPLAPGSAAQTNSMVTLGCLVK
    GYFPEPVTVTWNSGSLSSGVHTFPAVLQSDLYTLSSSVTVPSSTWPSETV
    TCNVAHPASSTKVDKKIVPRDCGCKPCICTVPEVSSVFIFPPKPKDVLTI
    TLTPKVTCVVVDISKDDPEVQFSWFVDDVEVHTAQTQPREEQFNSTFRSV
    SELPIMHQDWLNGKEFKCRVNSAAFPAPIEKTISKTKGRPKAPQVYTIPP
    PKEQMAKDKVSLTCMITDFFPEDITVEWQWNGQPAENYKNTQPIMDTDGS
    YFVYSKLNVQKSNWEAGNTFTCSVLHEGLHNHHTEKSLSHSPGK
    (1979 LC Kappa full)
    SEQ ID NO: 10
    DIQMTQSPASLSASLEEIVTITCQASQDIGNWLSWYQQKPGKSPHLLIYG
    TTSLADGVPSRFSGSRSGTQYSLKISGLQVADIGIYVCLQAYSTPFTFGS
    GTKLEIKRTDAAPTVSIFPPSSEQLTSGGASVVCFLNNFYPKDINVKWKI
    DGSERQNGVLNSWTDQDSKDSTYSMSSTLTLTKDEYERHNSYTCEATHKT
    STSPIVKSFNRNEC

Claims (10)

1-20. (canceled)
21. A compound of formula (5), or a pharmaceutically acceptable salt or solvate thereof:
Figure US20210140972A1-20210513-C00006
22. A complex comprising a trimeric protein that is a tumor necrosis factor (TNF) superfamily member and the compound of claim 21.
23. (canceled)
24. A method of treating and/or preventing a disorder selected from an autoimmune, an inflammatory, a neurological, a neurodegenerative, a pain, a nociceptive, and a cardiovascular disorders in a subject in need thereof, comprising administering the compound of claim 21 to the subject.
25. The method of claim 24, wherein rheumatoid arthritis, Crohn's disease, psoriasis, systemic lupus erythematosus, Alzheimer's disease, Parkinson's disease or epilepsy is treated and/or prevented.
26. A pharmaceutical composition comprising the compound of claim 21 and a pharmaceutically acceptable carrier.
27. A method of treating and/or preventing a disorder selected from an autoimmune, an inflammatory, a neurological, a neurodegenerative, a pain, a nociceptive, and a cardiovascular disorder in a subject in need thereof, comprising administering the complex of claim 22 to the subject.
28. A pharmaceutical composition comprising the complex of claim 22 and a pharmaceutically acceptable carrier.
29. The method of claim 27, wherein rheumatoid arthritis, Crohn's disease, psoriasis, systemic lupus erythematosus, Alzheimer's disease, Parkinson's disease or epilepsy is treated and/or prevented.
US17/072,665 2015-06-18 2020-10-16 Mechanism of action Pending US20210140972A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US17/072,665 US20210140972A1 (en) 2015-06-18 2020-10-16 Mechanism of action

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
GBGB1510758.4A GB201510758D0 (en) 2015-06-18 2015-06-18 Novel TNFa structure for use in therapy
GB1510758.4 2015-06-18
PCT/EP2015/074490 WO2016202411A1 (en) 2015-06-18 2015-10-22 Mechanism of action
US201715736336A 2017-12-14 2017-12-14
US17/072,665 US20210140972A1 (en) 2015-06-18 2020-10-16 Mechanism of action

Related Parent Applications (2)

Application Number Title Priority Date Filing Date
US15/736,336 Division US10883996B2 (en) 2015-06-18 2015-10-22 Methods of identifying signaling modulators of the trimeric TNFa
PCT/EP2015/074490 Division WO2016202411A1 (en) 2015-06-18 2015-10-22 Mechanism of action

Publications (1)

Publication Number Publication Date
US20210140972A1 true US20210140972A1 (en) 2021-05-13

Family

ID=53784155

Family Applications (11)

Application Number Title Priority Date Filing Date
US15/736,336 Active US10883996B2 (en) 2015-06-18 2015-10-22 Methods of identifying signaling modulators of the trimeric TNFa
US15/736,535 Active US10969393B2 (en) 2015-06-18 2015-10-22 Complexes between anti-TNF antibodies, trimeric TNF proteins and organic molecules binding them
US15/736,614 Active US10775385B2 (en) 2015-06-18 2015-10-22 Treatment of autoimmune and inflammatory disorders with asymmetric TNF alpha trimers
US15/736,520 Active US10705094B2 (en) 2015-06-18 2015-10-22 TNF receptor signaling modulator assay
US15/736,558 Active US11022614B2 (en) 2015-06-18 2015-10-22 Antibodies binding to trimeric TNF alpha epitopes
US16/883,859 Active US11448655B2 (en) 2015-06-18 2020-05-26 Method for identifying a modulator of the TNFα or CD40L interaction with their cognate receptors
US17/013,326 Active 2036-10-10 US11674967B2 (en) 2015-06-18 2020-09-04 Method of identifying potential inhibitors of APO TNFα trimers
US17/072,665 Pending US20210140972A1 (en) 2015-06-18 2020-10-16 Mechanism of action
US17/143,719 Abandoned US20210132079A1 (en) 2015-06-18 2021-01-07 Antibody
US17/325,036 Pending US20220074947A1 (en) 2015-06-18 2021-05-19 Antibody epitope
US17/823,356 Pending US20230194541A1 (en) 2015-06-18 2022-08-30 Modulator assay

Family Applications Before (7)

Application Number Title Priority Date Filing Date
US15/736,336 Active US10883996B2 (en) 2015-06-18 2015-10-22 Methods of identifying signaling modulators of the trimeric TNFa
US15/736,535 Active US10969393B2 (en) 2015-06-18 2015-10-22 Complexes between anti-TNF antibodies, trimeric TNF proteins and organic molecules binding them
US15/736,614 Active US10775385B2 (en) 2015-06-18 2015-10-22 Treatment of autoimmune and inflammatory disorders with asymmetric TNF alpha trimers
US15/736,520 Active US10705094B2 (en) 2015-06-18 2015-10-22 TNF receptor signaling modulator assay
US15/736,558 Active US11022614B2 (en) 2015-06-18 2015-10-22 Antibodies binding to trimeric TNF alpha epitopes
US16/883,859 Active US11448655B2 (en) 2015-06-18 2020-05-26 Method for identifying a modulator of the TNFα or CD40L interaction with their cognate receptors
US17/013,326 Active 2036-10-10 US11674967B2 (en) 2015-06-18 2020-09-04 Method of identifying potential inhibitors of APO TNFα trimers

Family Applications After (3)

Application Number Title Priority Date Filing Date
US17/143,719 Abandoned US20210132079A1 (en) 2015-06-18 2021-01-07 Antibody
US17/325,036 Pending US20220074947A1 (en) 2015-06-18 2021-05-19 Antibody epitope
US17/823,356 Pending US20230194541A1 (en) 2015-06-18 2022-08-30 Modulator assay

Country Status (16)

Country Link
US (11) US10883996B2 (en)
EP (8) EP3960759A1 (en)
JP (12) JP6781718B2 (en)
KR (2) KR102599907B1 (en)
CN (6) CN107810419B (en)
AU (2) AU2015398985A1 (en)
CA (5) CA2988516A1 (en)
CL (2) CL2017003189A1 (en)
EA (4) EA201890078A1 (en)
ES (5) ES2895552T3 (en)
GB (1) GB201510758D0 (en)
IL (2) IL256099B2 (en)
MX (2) MX2017015481A (en)
PL (4) PL3311170T3 (en)
PT (4) PT3311168T (en)
WO (5) WO2016202415A1 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11174311B2 (en) 2016-12-21 2021-11-16 UCB Biopharma SRL Antibody against trimeric TNFα complex
US11448655B2 (en) 2015-06-18 2022-09-20 UCB Biopharma SRL Method for identifying a modulator of the TNFα or CD40L interaction with their cognate receptors

Families Citing this family (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EA035638B1 (en) 2015-03-31 2020-07-20 ВиЭйчСКВЕАРД ЛИМИТЕД Polypeptides
UY36629A (en) 2015-04-17 2016-11-30 Abbvie Inc INDAZOLONAS AS MODULATORS OF THE TNF SIGNALING
EP3286195A1 (en) 2015-04-17 2018-02-28 AbbVie Inc. Indazolones as modulators of tnf signaling
UY36630A (en) 2015-04-17 2016-11-30 Abbvie Inc TRICYCLIC MODULATORS OF TNF SIGNALING
GB201522394D0 (en) 2015-12-18 2016-02-03 Ucb Biopharma Sprl Antibodies
EP3519438A1 (en) 2016-09-30 2019-08-07 VHsquared Limited Compositions
AU2018208458B2 (en) * 2017-01-10 2020-07-16 ETH Zürich Cell-protective compounds and their use
CN114514243A (en) 2019-06-21 2022-05-17 索瑞索制药公司 Polypeptides
CN114466864A (en) 2019-06-21 2022-05-10 索瑞索制药公司 Polypeptides

Family Cites Families (124)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE3888224T2 (en) 1987-04-24 1994-07-21 Teijin Ltd Determination of tumor necrosis factor; monoclonal antibody and composition.
US5223409A (en) 1988-09-02 1993-06-29 Protein Engineering Corp. Directed evolution of novel binding proteins
ATE151110T1 (en) 1988-09-02 1997-04-15 Protein Eng Corp PRODUCTION AND SELECTION OF RECOMBINANT PROTEINS WITH DIFFERENT BINDING SITES
GB8823869D0 (en) 1988-10-12 1988-11-16 Medical Res Council Production of antibodies
US4908372A (en) 1988-10-13 1990-03-13 Merrell Dow Pharmaceuticals Inc. Antihistaminic piperidinyl benzimidazoles
US5750373A (en) 1990-12-03 1998-05-12 Genentech, Inc. Enrichment method for variant proteins having altered binding properties, M13 phagemids, and growth hormone variants
DE3843534A1 (en) 1988-12-23 1990-07-12 Basf Ag NEW TNF POLYPEPTIDES
US6451983B2 (en) 1989-08-07 2002-09-17 Peptech Limited Tumor necrosis factor antibodies
GB8928874D0 (en) 1989-12-21 1990-02-28 Celltech Ltd Humanised antibodies
US5780225A (en) 1990-01-12 1998-07-14 Stratagene Method for generating libaries of antibody genes comprising amplification of diverse antibody DNAs and methods for using these libraries for the production of diverse antigen combining molecules
WO1991010737A1 (en) 1990-01-11 1991-07-25 Molecular Affinities Corporation Production of antibodies using gene libraries
AU633698B2 (en) 1990-01-12 1993-02-04 Amgen Fremont Inc. Generation of xenogeneic antibodies
US5427908A (en) 1990-05-01 1995-06-27 Affymax Technologies N.V. Recombinant library screening methods
GB9015198D0 (en) 1990-07-10 1990-08-29 Brien Caroline J O Binding substance
EP0542810A1 (en) 1990-08-02 1993-05-26 B.R. Centre Limited Methods for the production of proteins with a desired function
WO1992003918A1 (en) 1990-08-29 1992-03-19 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
US5661016A (en) 1990-08-29 1997-08-26 Genpharm International Inc. Transgenic non-human animals capable of producing heterologous antibodies of various isotypes
US5625126A (en) 1990-08-29 1997-04-29 Genpharm International, Inc. Transgenic non-human animals for producing heterologous antibodies
US5633425A (en) 1990-08-29 1997-05-27 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
US5545806A (en) 1990-08-29 1996-08-13 Genpharm International, Inc. Ransgenic non-human animals for producing heterologous antibodies
US5770429A (en) 1990-08-29 1998-06-23 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
US5698426A (en) 1990-09-28 1997-12-16 Ixsys, Incorporated Surface expression libraries of heteromeric receptors
GB9028123D0 (en) 1990-12-28 1991-02-13 Erba Carlo Spa Monoclonal antibodies against human tumor necrosis factor alpha
JP3672306B2 (en) 1991-04-10 2005-07-20 ザ スクリップス リサーチ インスティテュート Heterodimeric receptor library using phagemids
GB9113120D0 (en) 1991-06-18 1991-08-07 Kodak Ltd Photographic processing apparatus
IT1249708B (en) 1991-09-23 1995-03-09 Tecnogen Scpa METHOD FOR DETERMINING THE TUMORAL NECROSIS FACTOR (TNF) IN BIOLOGICALLY ACTIVE FORM.
ATE275198T1 (en) 1991-12-02 2004-09-15 Medical Res Council PRODUCTION OF ANTIBODIES ON PHAGE SURFACES BASED ON ANTIBODIES SEGMENT LIBRARIES.
WO1993014083A1 (en) 1992-01-09 1993-07-22 Janssen Pharmaceutica N.V. Pharmaceutically active substituted benzimidazole derivatives
US5733743A (en) 1992-03-24 1998-03-31 Cambridge Antibody Technology Limited Methods for producing members of specific binding pairs
EP0682705A1 (en) 1993-02-03 1995-11-22 N.V. Innogenetics S.A. Tnf-alpha muteins and a process for preparing them
CA2119089A1 (en) 1993-03-29 1994-09-30 David Banner Tumor necrosis factor muteins
WO1995015982A2 (en) 1993-12-08 1995-06-15 Genzyme Corporation Process for generating specific antibodies
WO1995020401A1 (en) 1994-01-31 1995-08-03 Trustees Of Boston University Polyclonal antibody libraries
US5516637A (en) 1994-06-10 1996-05-14 Dade International Inc. Method involving display of protein binding pairs on the surface of bacterial pili and bacteriophage
DE4440613C1 (en) 1994-11-14 1996-07-25 Leica Ag Device and method for the detection and demodulation of an intensity-modulated radiation field
TW453995B (en) 1995-12-15 2001-09-11 Novartis Ag Certain alpha-substituted arylsulfonamido acetohydroxamic acids
JP2978435B2 (en) 1996-01-24 1999-11-15 チッソ株式会社 Method for producing acryloxypropyl silane
CN1204230A (en) 1997-12-08 1999-01-06 李建安 Protective cover for hand-held telephone set and its making method
US20040009535A1 (en) 1998-11-27 2004-01-15 Celltech R&D, Inc. Compositions and methods for increasing bone mineralization
US7101974B2 (en) 2000-03-02 2006-09-05 Xencor TNF-αvariants
AU2001287041B2 (en) 2000-09-01 2006-06-08 Biogen Idec Ma Inc. Pyridine derivatives useful as CD40:CD154 binding interruptor compounds and use thereof to treat immunological complications
CA2446193C (en) 2001-06-05 2011-11-01 Boehringer Ingelheim Pharmaceuticals, Inc. 1,4-disubstituted benzo-fused cycloalkyl urea compounds
US6632810B2 (en) 2001-06-29 2003-10-14 Kowa Co., Ltd. Cyclic diamine compound with condensed-ring groups
JP2003040888A (en) 2001-07-30 2003-02-13 Sankyo Co Ltd Imidazole derivative
WO2004012673A2 (en) 2002-08-01 2004-02-12 Wyeth, Methods and reagents relating to inflammation and apoptosis
US7993864B2 (en) 2002-12-03 2011-08-09 Ucb Pharma S.A. Assay for identifying antibody producing cells
NZ543976A (en) 2003-05-09 2008-04-30 Pharmexa As Immunogenic human TNF alpha analogues with reduced cytotoxicity and methods of their preparation
GB0312481D0 (en) 2003-05-30 2003-07-09 Celltech R&D Ltd Antibodies
US7989594B2 (en) 2003-07-01 2011-08-02 Celltech R & D Limited Modified antibody fab fragments
GB0315457D0 (en) 2003-07-01 2003-08-06 Celltech R&D Ltd Biological products
GB0315450D0 (en) 2003-07-01 2003-08-06 Celltech R&D Ltd Biological products
EA013614B1 (en) 2003-07-15 2010-06-30 Амджен Инк. Human anti-ngf neutralizing antibodies as selective ngf pathway inhibitors
US7268116B2 (en) 2003-10-02 2007-09-11 Genhunter Corp. Methods and compositions for producing secreted trimeric receptor analogs and biologically active fusion proteins
TW200526780A (en) 2004-01-06 2005-08-16 Hayashibara Biochem Lab TNF antagonists and TNF inhibitors comprising the same as the active ingredient
GB0411186D0 (en) 2004-05-19 2004-06-23 Celltech R&D Ltd Biological products
CN103254309B (en) 2005-05-18 2017-09-26 埃博灵克斯股份有限公司 For the improved nanometer body of tumor necrosis factor αTM
DE102005023617A1 (en) 2005-05-21 2006-11-23 Aspre Ag Method for mixing colors in a display
WO2007060411A1 (en) 2005-11-24 2007-05-31 Ucb Pharma S.A. Anti-tnf alpha antibodies which selectively inhibit tnf alpha signalling through the p55r
MX343879B (en) * 2007-05-21 2016-11-25 Alderbio Holdings Llc Novel rabbit antibody humanization methods and humanized rabbit antibodies.
WO2008144753A2 (en) 2007-05-21 2008-11-27 Alder Biopharmaceuticals, Inc. Antibodies to tnf alpha and use thereof
AR067786A1 (en) 2007-08-03 2009-10-21 Alcon Mfg Ltd INHIBITION RELATED TO ARNI (INTERFERENCE RNA) OF TNFALFA SIGNALS (TUMOR NECROSIS FACTOR) FOR THE TREATMENT OF GLAUCOMA
EP2535349A1 (en) 2007-09-26 2012-12-19 UCB Pharma S.A. Dual specificity antibody fusions
US9365644B2 (en) 2008-04-23 2016-06-14 Epitomics, Inc. Anti-TNFα antibody
HUE039692T2 (en) 2008-06-25 2019-01-28 Esbatech Alcon Biomed Res Unit Stable and soluble antibodies inhibiting tnf
PE20110708A1 (en) 2008-11-20 2011-10-23 Panacea Biotec Ltd INHIBITING PEPTIDES OF THE ALPHA TUMOR NECROSIS FACTOR
JP2010172307A (en) 2009-01-30 2010-08-12 Saitama Medical Univ METHOD AND APPARATUS FOR PREDICTING PHARMACOLOGICAL EFFICACY OF SOLUBLE TNFalpha/LTalpha RECEPTOR PREPARATION ON RHEUMATOID ARTHRITIS
WO2010118404A2 (en) 2009-04-09 2010-10-14 California Institute Of Technology Methods for creating or identifying compounds that bind tumor necrosis factor alpha
CA2758964A1 (en) 2009-04-16 2010-10-21 Abbott Biotherapeutics Corp. Anti-tnf-.alpha. antibodies and their uses
RU2012147249A (en) 2010-04-07 2014-05-20 Эббви Инк. TNF-α- BINDING PROTEINS
US8377441B2 (en) 2010-08-03 2013-02-19 National Cheng Kung University Treating breast cancer with anti-IL-19 antibody
CA2853974C (en) 2010-10-30 2020-11-10 Kindex Therapeutics, Llc Cis 3,4-dihydroxy-2-(3-methylbutanoyl)-5-(3-methylbutyl)-4-(4-methylpentanoyl)cyclopent-2-en-1-one derivatives, substantially enantiomerically pure compositions and methods
AR084210A1 (en) 2010-12-08 2013-05-02 Abbott Lab PROTEINS OF UNION TO TNF-a
EP2721066A1 (en) 2011-06-17 2014-04-23 Glaxo Group Limited Tumour necrosis factor receptor 1 antagonists
CN103930126A (en) 2011-08-12 2014-07-16 Bsrc亚历山大.弗莱明 TNF superfamily trimerization inhibitors
CN104428293B (en) * 2012-06-11 2018-06-08 Ucb生物制药私人有限公司 Adjust the benzimidazole of TNF α
EP2867674B1 (en) * 2012-06-28 2018-10-10 UCB Biopharma SPRL A method for identifying compounds of therapeutic interest
GB201212513D0 (en) 2012-07-13 2012-08-29 Ucb Pharma Sa Therapeutic agents
BR112015000675B1 (en) * 2012-07-13 2022-07-12 UCB Biopharma SRL Imidazopyridine derivatives as modulators of TNF activity
ES2825999T3 (en) 2012-09-10 2021-05-17 Xencor Inc A dominant negative inhibitor of TNF-alpha for use in the treatment of neurological disorders of the CNS
CN104955480A (en) 2013-01-25 2015-09-30 西蒙有限公司 Compositions for selective reduction of circulating bioactive soluble TNF and methods for treating TNF-mediated disease
JP2016517344A (en) 2013-03-12 2016-06-16 アネロテック・インコーポレイテッドAnellotech,Inc. Regeneration of catalytic rapid pyrolysis catalyst
US9908944B2 (en) 2013-07-15 2018-03-06 Novo Nordisk A/S Antibodies that bind urokinase plasminogen activator
GB201321739D0 (en) 2013-12-09 2014-01-22 Ucb Pharma Sa Therapeutic agents
GB201321732D0 (en) 2013-12-09 2014-01-22 Ucb Pharma Sa Therapeutic agents
GB201321741D0 (en) 2013-12-09 2014-01-22 Ucb Pharma Sa Therapeutic agents
GB201321730D0 (en) 2013-12-09 2014-01-22 Ucb Pharma Sa Therapeutic agents
GB201321738D0 (en) 2013-12-09 2014-01-22 Ucb Pharma Sa Therapeutic Agents
GB201321745D0 (en) 2013-12-09 2014-01-22 Ucb Pharma Sa Therapeutic agents
GB201321735D0 (en) 2013-12-09 2014-01-22 Ucb Pharma Sa Therapeutic Agents
GB201321729D0 (en) 2013-12-09 2014-01-22 Ucb Pharma Sa Therapeutic agents
GB201321746D0 (en) 2013-12-09 2014-01-22 Ucb Pharma Sa Therapeutic agents
GB201321736D0 (en) 2013-12-09 2014-01-22 Ucb Pharma Sa Therapeutic agents
GB201321737D0 (en) 2013-12-09 2014-01-22 Ucb Pharma Sa Therapeutic Agents
GB201321743D0 (en) 2013-12-09 2014-01-22 Ucb Pharma Sa Therapeutic agents
BR112016013018A2 (en) 2013-12-09 2017-08-08 Ucb Biopharma Sprl FUSED BICYCLIC HETEROAROMATIC DERIVATIVES AS MODULATORS OF TNF ACTIVITY
EP3080112B1 (en) 2013-12-09 2020-06-03 UCB Biopharma SRL Imidazopyridine derivatives as modulators of tnf activity
GB201321740D0 (en) 2013-12-09 2014-01-22 Ucb Pharma Sa Therapeutic agents
GB201321734D0 (en) 2013-12-09 2014-01-22 Ucb Pharma Sa Therapeutic Agents
GB201321733D0 (en) 2013-12-09 2014-01-22 Ucb Pharma Sa Therapeutic agents
GB201321742D0 (en) 2013-12-09 2014-01-22 Ucb Pharma Sa Therapeutic agents
GB201321728D0 (en) 2013-12-09 2014-01-22 Ucb Pharma Sa Therapeutic agents
GB201321731D0 (en) 2013-12-09 2014-01-22 Ucb Pharma Sa Therapeutic agents
GB201321748D0 (en) 2013-12-09 2014-01-22 Ucb Pharma Sa Therapeutic agents
GB201321744D0 (en) 2013-12-09 2014-01-22 Ucb Pharma Sa Therapeutic agents
GB201321749D0 (en) 2013-12-09 2014-01-22 Ucb Pharma Sa Therapeutic agents
US10202405B2 (en) 2014-10-03 2019-02-12 Ucb Biopharma Sprl Fused pentacyclic imidazole derivatives
BR112017015661A2 (en) 2015-01-30 2018-03-20 Saitama Medical University anti-alk2 antibody
US10112944B2 (en) 2015-03-18 2018-10-30 Bristol-Myers Squibb Company Heterocyclic compounds useful as inhibitors of TNF
AU2016233289A1 (en) 2015-03-18 2017-11-09 Bristol-Myers Squibb Company Tricyclic heterocyclic compounds useful as inhibitors of TNF
AU2016233288A1 (en) 2015-03-18 2017-11-09 Bristol-Myers Squibb Company Substituted tricyclic heterocyclic compounds
UY36629A (en) 2015-04-17 2016-11-30 Abbvie Inc INDAZOLONAS AS MODULATORS OF THE TNF SIGNALING
UY36630A (en) 2015-04-17 2016-11-30 Abbvie Inc TRICYCLIC MODULATORS OF TNF SIGNALING
EP3286195A1 (en) 2015-04-17 2018-02-28 AbbVie Inc. Indazolones as modulators of tnf signaling
GB201509893D0 (en) 2015-06-08 2015-07-22 Ucb Biopharma Sprl Therapeutic agents
GB201509888D0 (en) 2015-06-08 2015-07-22 Ucb Biopharma Sprl Therapeutic agents
GB201509885D0 (en) 2015-06-08 2015-07-22 Ucb Biopharma Sprl Therapeutic agents
GB201510758D0 (en) 2015-06-18 2015-08-05 Ucb Biopharma Sprl Novel TNFa structure for use in therapy
JP6955482B2 (en) 2015-08-03 2021-10-27 ブリストル−マイヤーズ スクイブ カンパニーBristol−Myers Squibb Company Heterocyclic compounds useful as modifiers of TNF alpha
WO2017023902A1 (en) 2015-08-03 2017-02-09 Bristol-Myers Squibb Company Cyclic compounds useful as modulators of tnf alpha
EA201892142A1 (en) 2016-04-01 2019-04-30 Юсб Байофарма Спрл CONDENSED PENTACYCLIC DERIVATIVES OF IMIDAZOLE AS TNF ACTIVITY MODULATORS
JP6968092B2 (en) 2016-04-01 2021-11-17 ユーシービー バイオファルマ エスアールエル Condensed pentacyclic imidazole derivative as a modulator of TNF activity
CN109219609B (en) 2016-04-01 2022-02-01 Ucb生物制药私人有限公司 Fused hexacyclic imidazole derivatives as modulators of TNF activity
WO2017167996A1 (en) 2016-04-01 2017-10-05 Ucb Biopharma Sprl Fused pentacyclic imidazole derivatives as modulators of tnf activity
GB201621907D0 (en) 2016-12-21 2017-02-01 Ucb Biopharma Sprl And Sanofi Antibody epitope

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11448655B2 (en) 2015-06-18 2022-09-20 UCB Biopharma SRL Method for identifying a modulator of the TNFα or CD40L interaction with their cognate receptors
US11674967B2 (en) 2015-06-18 2023-06-13 UCB Biopharma SRL Method of identifying potential inhibitors of APO TNFα trimers
US11174311B2 (en) 2016-12-21 2021-11-16 UCB Biopharma SRL Antibody against trimeric TNFα complex

Also Published As

Publication number Publication date
US10883996B2 (en) 2021-01-05
US20180231562A1 (en) 2018-08-16
CA2987823C (en) 2023-08-22
CN108055865B (en) 2021-06-15
EP3988936A1 (en) 2022-04-27
JP2023052250A (en) 2023-04-11
PT3311169T (en) 2021-11-03
JP6948954B2 (en) 2021-10-13
IL256099B2 (en) 2024-06-01
KR20180012863A (en) 2018-02-06
JP2018525333A (en) 2018-09-06
US10705094B2 (en) 2020-07-07
CN111574517B (en) 2022-11-18
EP3311167B1 (en) 2020-09-23
CN111574517A (en) 2020-08-25
CN107771286A (en) 2018-03-06
EP3311168A1 (en) 2018-04-25
JP7159420B2 (en) 2022-10-24
PL3311171T3 (en) 2022-03-28
US20180203016A1 (en) 2018-07-19
ES2895486T3 (en) 2022-02-21
ES2836769T3 (en) 2021-06-28
JP2022023060A (en) 2022-02-07
GB201510758D0 (en) 2015-08-05
JP2018522230A (en) 2018-08-09
CN108139407B (en) 2020-10-30
AU2015398984A1 (en) 2017-12-21
US11448655B2 (en) 2022-09-20
WO2016202411A1 (en) 2016-12-22
JP2018520668A (en) 2018-08-02
US20220074947A1 (en) 2022-03-10
IL256099B1 (en) 2024-02-01
CN107810419A (en) 2018-03-16
EP3960759A1 (en) 2022-03-02
CA2988723A1 (en) 2016-12-22
CL2017003246A1 (en) 2018-04-27
EP3311167A1 (en) 2018-04-25
CN108290945A (en) 2018-07-17
PT3311171T (en) 2022-01-20
US20210088530A1 (en) 2021-03-25
US20180172701A1 (en) 2018-06-21
KR102599907B1 (en) 2023-11-07
EP3311171B1 (en) 2021-12-01
WO2016202414A1 (en) 2016-12-22
US20200400678A1 (en) 2020-12-24
CA2987827C (en) 2023-08-29
CA2987827A1 (en) 2016-12-22
CA2987698A1 (en) 2016-12-22
US20180171008A1 (en) 2018-06-21
EP3311170B1 (en) 2021-10-13
IL256097A (en) 2018-02-28
JP2019218382A (en) 2019-12-26
JP6659030B2 (en) 2020-03-04
WO2016202415A1 (en) 2016-12-22
CA2988516A1 (en) 2016-12-22
US20180172702A1 (en) 2018-06-21
KR20180048571A (en) 2018-05-10
US20230194541A1 (en) 2023-06-22
MX2017015757A (en) 2018-04-24
IL256099A (en) 2018-02-28
EA201890082A1 (en) 2018-07-31
PT3311170T (en) 2021-11-03
WO2016202413A1 (en) 2016-12-22
JP6752344B2 (en) 2020-09-09
AU2015398984B2 (en) 2022-05-26
CL2017003189A1 (en) 2018-07-06
US11674967B2 (en) 2023-06-13
ES2902415T3 (en) 2022-03-28
JP2018524577A (en) 2018-08-30
EP3311170A1 (en) 2018-04-25
EA201890078A1 (en) 2018-06-29
EA201890081A1 (en) 2018-06-29
PL3311169T3 (en) 2022-01-31
CA2987698C (en) 2023-07-25
US10969393B2 (en) 2021-04-06
JP2021109880A (en) 2021-08-02
EP3311169B1 (en) 2021-09-29
PT3311168T (en) 2023-07-11
CN108055865A (en) 2018-05-18
CN108139407A (en) 2018-06-08
CA2987823A1 (en) 2016-12-22
MX2017015481A (en) 2018-02-19
EP3311171A1 (en) 2018-04-25
ES2948915T3 (en) 2023-09-21
ES2895552T3 (en) 2022-02-21
JP2023100010A (en) 2023-07-14
EA201890080A1 (en) 2018-07-31
WO2016202412A1 (en) 2016-12-22
US10775385B2 (en) 2020-09-15
US20210132079A1 (en) 2021-05-06
CN107771286B (en) 2021-07-30
CN108290945B (en) 2022-09-30
CN107810419B (en) 2021-04-30
EP3311168B1 (en) 2023-05-10
US11022614B2 (en) 2021-06-01
JP2021153592A (en) 2021-10-07
PL3311168T3 (en) 2023-09-18
EP3311169A1 (en) 2018-04-25
JP6951256B2 (en) 2021-10-20
PL3311170T3 (en) 2022-02-14
JP2021038222A (en) 2021-03-11
JP6781718B2 (en) 2020-11-04
AU2015398985A1 (en) 2017-12-21
JP2018524311A (en) 2018-08-30
EP3995831A1 (en) 2022-05-11
JP7336178B2 (en) 2023-09-11

Similar Documents

Publication Publication Date Title
US20210140972A1 (en) Mechanism of action
US20220025033A1 (en) Antibody epitope
EA042528B1 (en) MECHANISM OF ACTION

Legal Events

Date Code Title Description
AS Assignment

Owner name: CELLTECH R&D LIMITED, UNITED KINGDOM

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:O'CONNELL, JAMES PHILIP;PORTER, JOHN ROBERT;LAWSON, ALASTAIR;AND OTHERS;SIGNING DATES FROM 20180329 TO 20180518;REEL/FRAME:054100/0788

Owner name: UCB BIOPHARMA SPRL, BELGIUM

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:UCB PHARMA S.A.;REEL/FRAME:054100/0809

Effective date: 20180607

Owner name: UCB PHARMA S.A., BELGIUM

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:UCB S.A.;REEL/FRAME:054100/0796

Effective date: 20180607

Owner name: UCB S.A., BELGIUM

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:CELLTECH R&D LIMITED;REEL/FRAME:054100/0785

Effective date: 20180607

Owner name: UCB BIOPHARMA SRL, BELGIUM

Free format text: CHANGE OF NAME;ASSIGNOR:UCB BIOPHARMA SPRL;REEL/FRAME:054126/0938

Effective date: 20191202

STPP Information on status: patent application and granting procedure in general

Free format text: APPLICATION DISPATCHED FROM PREEXAM, NOT YET DOCKETED

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: FINAL REJECTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED