WO2018234118A1 - Traitement de maladies inflammatoires par des inhibiteurs de l'activité de c5a - Google Patents

Traitement de maladies inflammatoires par des inhibiteurs de l'activité de c5a Download PDF

Info

Publication number
WO2018234118A1
WO2018234118A1 PCT/EP2018/065676 EP2018065676W WO2018234118A1 WO 2018234118 A1 WO2018234118 A1 WO 2018234118A1 EP 2018065676 W EP2018065676 W EP 2018065676W WO 2018234118 A1 WO2018234118 A1 WO 2018234118A1
Authority
WO
WIPO (PCT)
Prior art keywords
antibody
antigen
binding
binding fragment
compound
Prior art date
Application number
PCT/EP2018/065676
Other languages
English (en)
Inventor
Renfeng Guo
Niels R RIEDEMANN
Original Assignee
Inflarx Gmbh
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Inflarx Gmbh filed Critical Inflarx Gmbh
Priority to SG11201912882QA priority Critical patent/SG11201912882QA/en
Priority to KR1020197038211A priority patent/KR20200020727A/ko
Priority to AU2018286754A priority patent/AU2018286754A1/en
Priority to JP2019570986A priority patent/JP2020524696A/ja
Priority to CN201880041303.4A priority patent/CN111201241A/zh
Priority to EA201992673A priority patent/EA201992673A1/ru
Priority to EP18732016.3A priority patent/EP3642230A1/fr
Priority to CA3066689A priority patent/CA3066689C/fr
Publication of WO2018234118A1 publication Critical patent/WO2018234118A1/fr
Priority to IL271074A priority patent/IL271074A/en

Links

Classifications

    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/10Anti-acne agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/545Medicinal preparations containing antigens or antibodies characterised by the dose, timing or administration schedule
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding

Definitions

  • the present invention relates to inhibitors of C5a activity and their use in the treatment of cutaneous, neutrophilic, inflammatory diseases in a subject.
  • C5a is one of the most potent inflammatory peptides, with a broad spectrum of functions (Guo and Ward, 2005). C5a exerts its effects through the high-affinity C5a receptors (C5aR and C5L2) (Ward, 2009). C5aR belongs to the rhodopsin family of G-protein-coupled receptors with seven transmembrane segments; C5L2 has a similar structure but appears not to be G-protein-coupled. It is currently believed that C5a exerts its biological functions primarily through C5a-C5aR interaction, as few biological responses have been found for C5a-C5L2 interaction. However, latest reports demonstrate signaling also through C5L2 activation (Rittirsch and others, 2008).
  • C5aR is widely expressed on myeloid cells including neutrophils, eosinophils, basophils, and monocytes, and non-myeloid cells in many organs, especially in the lung and liver, indicative of the importance of C5a/C5aR signaling.
  • Widespread up-regulation of C5aR expression occurs during the onset of sepsis, and blockade of C5a/C5aR interaction by anti- C5a, or anti-C5aR antibodies, or C5aR antagonists renders highly protective effects in rodent models of sepsis (Czermak and others, 1999; Huber-Lang and others, 2001; Riedemann and others, 2002).
  • C5a has a variety of biological functions (Guo and Ward, 2005).
  • C5a is a strong chemoattractant for neutrophils and also has chemotactic activity for monocytes and macrophages.
  • C5a causes an oxidative burst (0 2 consumption) in neutrophils and enhances phagocytosis and release of granular enzymes.
  • C5a has also been found to be a vasodilator.
  • C5a has been shown to be involved in modulation of cytokine expression from various cell types and to enhance expression of adhesion molecule expression on neutrophils. High doses of C5a can lead to nonspecific chemotactic "desensitization" of neutrophils, thereby causing broad dysfunction.
  • C5a Many inflammatory diseases are attributable to the effects of C5a, including sepsis, acute lung injury, inflammatory bowel disease, rheumatoid arthritis and others.
  • C5a Many inflammatory diseases are attributable to the effects of C5a, including sepsis, acute lung injury, inflammatory bowel disease, rheumatoid arthritis and others.
  • exposure of neutrophils to C5a can lead to neutrophil dysfunction and paralysis of signaling pathways, leading to defective assembly of NADPH oxidase, paralysis of MAPK signaling cascades, a great depression of oxidative burst, phagocytosis and chemotaxis (Guo and others, 2006; Huber-Lang and others, 2002).
  • Thymocytes apoptosis and delayed neutrophil apoptosis are two important pathogenic events for sepsis development, which are dependent on the presence of C5a.
  • C5a up-regulates ⁇ 2- integrin expression on neutrophils to promote cell migration into organs, one of the major causes for multi-organ failure (MOF). It is also found that C5a is attributable to the activation of the coagulation pathway that occurs in experimental sepsis. C5a stimulates the synthesis and release from human leukocytes of pro-inflammatory cytokines such as TNF-a, IL- ⁇ ⁇ , IL-6, IL- 8, and macrophage migration inhibitory factor (MIF). Given that complement activation is an event occurring during the onset of acute inflammation, C5a may come into play before emergence of most of the inflammatory "cytokine storm”. It appears that C5a plays a key role in orchestrating and amplifying the performance of the cytokine network and the formation of systemic inflammatory response syndrome (SIRS).
  • SIRS systemic inflammatory response syndrome
  • C5a affects the crosstalk between dendritic cells (DC) and ⁇ cells, and this may result in a large production of inflammatory mediators such as IL-17 (Xu and others, 2010).
  • An essential role for C5a has been established and defined in the generation of pathogenic Thl7 responses in systemic lupus erythematosus (SLE) (Pawaria and others, 2014).
  • SLE systemic lupus erythematosus
  • C5a is a key regulator for Treg cells offering a powerful suppressive effect for Treg propagation and induction (Strainic and others, 2013). Given the fact that Treg and TH17 are the essential players in the autoimmune disease setting, inhibition of C5a signaling would be expected to significantly reduce overactive immune status in the autoimmune diseases.
  • IFX- 1 is a chimaeric monoclonal IgG4 antibody which specifically binds to the soluble human complement split product C5a.
  • IFX-1 is composed of 1328 amino acids and has an approximate molecular weight of 148,472 Daltons.
  • the CDR and FR sequences of IFX-1 are disclosed in in Table 3 below.
  • IFX- 1 is expressed in a mammalian CHO cell line as recombinant protein and finally formulated in a phosphate buffered saline solution for intravenous administration.
  • the binding of this antibody to human C5a facilitates a highly effective blockade of C5a-induced biological effects by disabling C5a binding to and reacting with its corresponding cell-bound receptors.
  • IFX-1 In vitro analysis of IFX-1 demonstrates a strong binding capacity to soluble human C5a as well as a high blocking activity of C5a-induced biological effects such as lysozyme release from human neutrophils or CD l ib up-regulation in neutrophils in human whole blood.
  • One IFX-1 antibody reaches the capability of neutralizing the effects of 2 molecules C5a with close to 100% efficiency in experimental in vitro settings.
  • Clinical trials with IFX-1 have been ongoing to test its clinical efficacy in several inflammatory diseases including septic organ dysfunction and complex cardiac surgery.
  • Neutrophils terminally differentiated cells with a short lifespan in circulation, are the most abundant leukocytes in the human body.
  • neutrophils are characterized by their ability to act as phagocytic cells, release lytic enzymes from their granules and produce reactive oxygen species upon stimulation.
  • other stimuli such as immune complex can also induce the respiratory burst in neutrophils, leading to enhanced inflammation and the recruitment of inflammatory cells (Kaplan, 2013).
  • neutrophils After infiltrating into inflamed tissues, neutrophils engage in many other cell types, such as macrophages, dendritic cells (DCs), natural killer cells, lymphocytes and mesenchymal stem cells, regulate innate and adaptive immune responses. For instance, neutrophils can modulate DC maturation and the proliferation and polarization of T cells, and they can also directly prime antigen-specific T-helper type 1 and T-helper type 17 cells (Abi Abdallah and others, 2011).
  • DCs dendritic cells
  • natural killer cells IL-1 and lymphocytes and mesenchymal stem cells
  • neutrophils can modulate DC maturation and the proliferation and polarization of T cells, and they can also directly prime antigen-specific T-helper type 1 and T-helper type 17 cells (Abi Abdallah and others, 2011).
  • HS Hidradenitis Suppurativa
  • HS ulcerative colitis .
  • Nodules appear in the affected areas, and they progressively become swollen and rupture with the release of pus. This process occurs repeatedly leading to sinus tract formation and scars (Jemec, 2004). This disease course creates a frustrating situation for the patients but also for physicians. The point prevalence is reported to range between 1% and 4% (Jemec and others, 1996).
  • HS Hidradenitis suppurativa
  • Activated neutrophils could be an important effector cell type causing tissue damage through direct harmful effect or indirect regulatory effect toward other effect cells such as active T cells and TH17 in this disease setting.
  • HS may appear as an auto-inflammatory disease based on a defect in the hair follicle innate immunity (Revuz, 2009), which is supported by the fact that pro-inflammatory cytokines such as interleukin (IL)-ip, and tumor necrosis factor-a (TNF-a) are markedly increased in lesional and perilesional skin (Wollina and others, 2013).
  • pro-inflammatory cytokines such as interleukin (IL)-ip, and tumor necrosis factor-a (TNF-a) are markedly increased in lesional and perilesional skin (Wollina and others, 2013).
  • ND neutrophilic dermatoses
  • SS Sweet syndrome
  • PG pyoderma gangrenosum
  • SPD subcorneal pustular dermatosis
  • HS Hidradenitis suppurativa
  • PG Pyoderma gangrenosum
  • HS hidradenitis suppurativa
  • BADAS bowel-associated dermatosis-arthritis syndrome
  • fever flu-like symptoms
  • arthritis inflammatory skin involvement
  • lesions recalling different neutrophilic dermatoses such as papules and plaques (Sweet's syndrome), pustules and ulcers (pyoderma gangrenosum) or nodules, abscesses or fistulae (hidradenitis suppurativa).
  • neutrophilic panniculitis can be associated. Patients usually experience a symmetrical, non-erosive polyarthritis that predominantly involves small joints (Cugno and others, 2018).
  • SAPHO syndrome synovitis, acne, pustulosis, hyperostosis and osteitis (SAPHO) syndrome was initially described in 1987.
  • SAPHO syndrome is a rare condition, possibly to misdiagnosis. Although its pathogenesis is still elusive, there is increasing understanding that SAPHO shares similarities with other autoinflammatory diseases (Cugno and others, 2018).
  • Autoimmune diseases are defined by defective differentiation of self and non-self molecules, leading to inappropriate recognition of self molecules and tissues as foreign structures, and concomitant immune attack against host organs.
  • the pathogenesis of autoimmune diseases can generally be divided into two phases, immunization phase and effector phase.
  • Immunization phase is characterized by the emergence of autoreactive T- lymphocytes. Those T-cells then trigger a secondary response leading to tissue damaging phase by activating various other cell types (B-cells, cytotoxic T-cells, NK-cells, neutrophils, macrophages, osteoclasts, fibroblasts, etc.).
  • B-cells cytotoxic T-cells
  • NK-cells cytotoxic T-cells
  • neutrophils neutrophils
  • macrophages osteoclasts
  • fibroblasts etc.
  • the activation of those effector cells by the autoreactive T cells can be considered as the effector phase which can be mediated by multiple levels including autoantibody production, cytokin
  • Neutrophils could participate in the multiple steps of the autoimmune disease process, including antigen presentation, regulation of the activity of other immune cell types, and direct tissue damage. Neutrophils can expose/release autoantigens when activated, or when dying by apoptosis, or during formation of neutrophil extracellular traps (NETs). They can also contribute to tissue deposition of autoantibodies or, as an effector cell type, they can induce tissue damage themselves.
  • NETs neutrophil extracellular traps
  • autoimmune diseases such as, rheumatoid arthritis (RA), systemic lupus erythematosus (SLE), bullous pemphigoid, epidermolysis bullosa acquisita, ANCA-associated vasculitis, familial Mediterranean fever, cryopyrin-associated periodic disorders (CAPS) and gout, etc.
  • RA rheumatoid arthritis
  • SLE systemic lupus erythematosus
  • CAS cryopyrin-associated periodic disorders
  • gout etc.
  • rheumatoid neutrophilic dermatosis is a rare cutaneous manifestation in patients with severe rheumatoid arthritis. It mainly affects patients with severe seropositive rheumatoid arthritis, predominantly women (ration 2: 1), but it has been observed also in seronegative rheumatoid arthritis (Cugno and others, 2018).
  • the present inventors have now surprisingly found that molecules inhibiting C5a signaling, e.g. an anti-C5a antibody, are exceptionally well-suited for the treatment of Hidradenitis suppurativa.
  • the present inventors have additionally studied the physiological mechanism leading to neutrophil activation and found out that C5a is the key driver of neutrophil activation.
  • the present invention relates to a compound for use in the treatment of a cutaneous, neutrophilic, inflammatory disease in a subject, wherein the compound is an inhibitor of C5a activity, and wherein the cutaneous, neutrophilic, inflammatory disease is selected from the group consisting of hidradenitis suppurativa (HS); Pyoderma gangrenosum (PG); PAPA (pyogenic arthritis, PG and acne); PASH (PG, acne and hidradenitis suppurativa); PAPASH (pyogenic arthritis, acne, PG and hidradenitis suppurativa); Sweet syndrome (SS); subcorneal pustular dermatosis (SPD); epidermolysis bullosa acquisita, erythema elevatum diutinum (EED); neutrophilic panniculitis; bowel-associated dermatosis-arthritis syndrome (BAD AS); SAPHO (synovitis, acne, pustulosis, hyperost
  • HS
  • the present invention relates to a method for the treatment of a cutaneous, neutrophilic, inflammatory disease in a subject, comprising the step of:
  • adenitis suppurativa HS
  • Pyoderma gangrenosum PG
  • PAPA pyogenic arthritis, PG and acne
  • PASH PG, acne and hidradenitis suppurativa
  • PAPASH pyogenic arthritis, acne, PG and hidradenitis suppurativa
  • Sweet syndrome SS
  • subcorneal pustular dermatosis SPD
  • the present invention relates to a use of a compound for the preparation of a pharmaceutical composition for the treatment of a cutaneous, neutrophilic, inflammatory disease, wherein the compound is an inhibitor of C5a activity, and wherein the cutaneous, neutrophilic, inflammatory disease is selected from the group consisting of hidradenitis suppurativa (HS); Pyoderma gangrenosum (PG); PAPA (pyogenic arthritis, PG and acne); PASH (PG, acne and hidradenitis suppurativa); PAPASH (pyogenic arthritis, acne, PG and hidradenitis suppurativa); Sweet syndrome (SS); subcorneal pustular dermatosis (SPD); epidermolysis bullosa acquisita, erythema elevatum diutinum (EED); neutrophilic panniculitis; bowel-associated dermato sis-arthritis syndrome (BADAS); SAPHO (synovitis, acne, pus
  • FIG. 1 Blocking activity of IFX-1 to recombinant human C5a (rhC5a)-induced CDllb upregulation on blood neutrophils.
  • IFX-1-004 and IFX-1-012 represent two different production batches. Human whole blood was incubated with buffer, antibody alone, rhC5a alone, or combinations of different antibody concentration and rhC5a. After incubation, cells were stained with anti-mouse CD1 lb:FITC and CD1 lb MFI was analysed by flow cytometry. Results are presented as mean + SD. The percentage of IFX-1 blocking activity of C5a- induced CDl lb expression is marked (arrow). Statistical differences were calculated by One-Way- ANOVA, p values of p ⁇ 0.05 were statistically significant.
  • FIG. 1 Blocking activity of IFX-1 on endogenous C5a (eC5a)-driven CDllb upregulation on neutrophils.
  • ZAP Zymosan-activated human plasma
  • Whole blood was incubated with buffer, IFX-1 alone, ZAP alone, or combinations of IFX-1 and ZAP. After incubation, cells were stained with anti-mouse CDl lb:FITC and analysed by flow cytometry. Results are presented as mean + SD. The percentage of IFX-1 blocking activity of eC5a-induced CDl lb expression is marked (arrow). Statistical differences were calculated by One-Way-ANOVA, p values of p ⁇ 0.05 were statistically significant.
  • FIG. 3 Activation of blood neutrophils by zymosan and IFX-1 blocking activity.
  • Whole blood was incubated with HBSS, rhC5a and zymosan A alone, or combinations of different IFX-1 concentrations and rhC5a or zymosan A.
  • cells were stained with anti-mouse CDl lb:FITC, and CDl lb MFI was analysed by flow cytometry. Results are presented as mean + SD. The percentage of IFX-1 blocking activity of C5a-induced CDl lb expression is marked (arrow). Statistical differences were calculated by One-Way-ANOVA, p values of p ⁇ 0.05 were statistically significant.
  • IFX-1 inhibits zymosan-induced generation of IL-8 in human whole blood.
  • IL-8 concentrations were obtained by ELISA after incubation of human whole blood with different concentrations of zymosan A (as indicated on the x-axis) in the presence (empty circles) or absence (filled circles) of IFX-1. Results were presented as mean + SD.
  • FIG. 6 Effect of HS plasma on blood neutrophil activation and the potential role of C5a.
  • HS plasma samples were incubated with human whole blood in the presence and absence of IFX-1, and CDl lb expression on blood neutrophils was determined by flow cytometric analysis.
  • C5a levels in the control and HS samples were labeled in the embedded table.
  • HiSCR response post-IFX-1 treatment in HS patients is defined as a > 50% reduction in inflammatory lesion count (abscesses + inflammatory nodules), and no increase in abscesses or draining fistulas when compared with baseline.
  • FIG. 8 Blockade of C5a-induced CDllb upregulation via different anti-C5aR antibodies.
  • Whole blood as source of neutrophils was incubated with (spiked-) plasma samples in the absence or presence of respective anti-C5aR antibodies.
  • the blocking activity of each inhibitor was indicated as percentage on the corresponding sample.
  • PMX-53 Whole blood as source of neutrophils was incubated with (spiked-) plasma samples in the absence or presence of low-concentrated (A) or over-concentrated (B) C5aR antagonist PMX-53. The blocking activities of PMX-53 were indicated as percentage on the corresponding samples.
  • Whole blood as source of neutrophils was incubated with (spiked-) plasma samples in the absence or presence of IFX-1. The blocking activities of IFX-1 were indicated as percentage on the corresponding samples.
  • Avacopan Whole blood as source of neutrophils was incubated with (spiked-) plasma samples in the absence or presence of Avacopan. The blocking activities of Avacopan were indicated as percentage on the corresponding samples.
  • the terms used herein are defined as described in "A multilingual glossary of biotechnological terms: (IUPAC Recommendations)", Leuenberger, H.G.W, Nagel, B. and Kolbl, H. eds. (1995), Helvetica Chimica Acta, CH-4010 Basel, Switzerland).
  • C5a particularly refers to human C5a.
  • Human C5a is a 74 amino acid peptide with the following amino acid sequence:
  • the amino acid sequence of human C5 can be found under the accession number UniProtKB P01031 (C05_HUMAN).
  • inhibitor of C5a activity refers to any compound that in any way reduces the activity of C5a. This activity reduction can be achieved by directly or indirectly lowering the concentration of C5a, or by reducing the activity of C5a, or by preventing C5a from exerting its effects on one or more of its receptors (e.g. on C5aR or C5L2), or by reducing the concentration or activity of one or more receptors of C5a.
  • C5a receptor refers to any potential C5a binding ligand on the cell surface, especially to any receptor protein to which C5a may bind and elicit a reaction on said receptor (e.g. activation or inhibition of the receptor).
  • C5a receptor particularly encompasses the two receptors C5aR and C5L2.
  • Alternative names for C5aR are C5aRl and CD88.
  • An alternative name for C5L2 is C5aR2.
  • Certain embodiments of the present invention refer to an inhibitor of C5a that interferes with a C5a receptor (e.g. by binding to a C5a receptor, or by blocking expression of a C5a receptor).
  • a C5a receptor can refer to (i) C5aR or to (ii) C5L2 or to (iii) both C5aR and C5L2. This means that some inhibitors of C5a interfere with only one of the C5a receptors (i.e. either C5aR or C5L2), while other inhibitors of C5a interfere with both C5a receptors (i.e. both C5aR and C5L2).
  • protein ligand refers to any molecule composed of amino acids linked by peptide bonds, irrespective of the total size of the molecule, and that is capable of specifically binding to another molecule. Accordingly, the expression “protein ligand” comprises oligopeptides ( ⁇ 100 amino acids) and polypeptides (> 100 amino acids). The expression “protein ligand” also comprises cyclic peptides, irrespective of their size. The expression “protein ligand” particularly encompasses antibodies, antigen-binding fragments of antibodies, antibody- like proteins, and peptidomimetics.
  • a first compound e.g. a protein ligand or nucleic acid aptamer
  • a second compound e.g. a target protein
  • Kd dissociation constant Kd to said second compound of 1 mM or less, preferably 100 ⁇ or less, preferably 50 ⁇ or less, preferably 30 ⁇ or less, preferably 20 ⁇ or less, preferably 10 ⁇ or less, preferably 5 ⁇ or less, more preferably 1 ⁇ or less, more preferably 900 nM or less, more preferably 800 nM or less, more preferably 700 nM or less, more preferably 600 nM or less, more preferably 500 nM or less, more preferably 400 nM or less, more preferably 300 nM or less, more preferably 200 nM or less, even more preferably 100 nM or less, even more preferably 90 nM or less, even more preferably 80 nM or less, even more preferably 70 nM or
  • binding preferably relates to a specific binding.
  • Specific binding means that a compound (e.g. a protein ligand or nucleic acid aptamer) binds stronger to a target such as an epitope for which it is specific compared to the binding to another target.
  • a compound binds stronger to a first target compared to a second target, if it binds to the first target with a dissociation constant (Kd) which is lower than the dissociation constant for the second target.
  • Kd dissociation constant
  • the dissociation constant (Kd) for the target to which the compound binds specifically is more than 10-fold, preferably more than 20-fold, more preferably more than 50-fold, even more preferably more than 100-fold, 200-fold, 500-fold or 1000-fold lower than the dissociation constant (Kd) for the target to which the compound does not bind specifically.
  • Kd (usually measured in “mol/L”, sometimes abbreviated as
  • M is intended to refer to the dissociation equilibrium constant of the particular interaction between a compound (e.g. a protein ligand) and a target molecule.
  • Methods for determining binding affinities of compounds are known to a person of ordinary skill in the art and can be selected for instance from the following methods known in the art: Surface Plasmon Resonance (SPR) based technology, Bio-layer interferometry (BLI), enzyme-linked immunosorbent assay (ELISA), flow cytometry, isothermal titration calorimetry (ITC), analytical ultracentrifugation, radioimmunoassay (RIA or IRMA) and enhanced chemiluminescence (ECL).
  • SPR Surface Plasmon Resonance
  • BBI Bio-layer interferometry
  • ELISA enzyme-linked immunosorbent assay
  • flow cytometry isothermal titration calorimetry
  • ITC isothermal titration calorimetry
  • analytical ultracentrifugation RIA or IRMA
  • ECL enhanced chemiluminescence
  • the dissociation constant Kd is determined at 20°C, 25°C, 30°C, or 37°C. If
  • an “epitope”, also known as antigenic determinant, is the part of a macromolecule that is recognized by the immune system, specifically by antibodies, B cells, or T cells.
  • an “epitope” is the part of a macromolecule capable of binding to a compound (e.g. an antibody or antigen-binding fragment thereof) as described herein.
  • binding preferably relates to a specific binding.
  • Epitopes usually consist of chemically active surface groupings of molecules such as amino acids or sugar side chains and usually have specific three-dimensional structural characteristics, as well as specific charge characteristics. Conformational and non-conformational epitopes can be distinguished in that the binding to the former but not the latter is lost in the presence of denaturing solvents.
  • a “paratope” is the part of an antibody that binds to the epitope.
  • a “paratope” is the part of a compound (e.g. a protein ligand) as described herein that binds to the epitope.
  • antibody typically refers to a glycoprotein comprising at least two heavy (H) chains and two light (L) chains inter-connected by disulfide bonds, or an antigen-binding portion thereof.
  • antibody also includes all recombinant forms of antibodies, in particular of the antibodies described herein, e.g. antibodies expressed in prokaryotes, unglycosylated antibodies, antibodies expressed in eukaryotes (e.g. CHO cells), glycosylated antibodies, and any antigen-binding antibody fragments and derivatives as described below.
  • Each heavy chain is comprised of a heavy chain variable region (abbreviated herein as VH or V H ) and a heavy chain constant region.
  • Each light chain is comprised of a light chain variable region (abbreviated herein as VL or V L ) and a light chain constant region.
  • VL light chain variable region
  • the VH and VL regions can be further subdivided into regions of hypervariability, termed complementarity determining regions (CDR), interspersed with regions that are more conserved, termed framework regions (FR).
  • CDR complementarity determining regions
  • FR framework regions
  • Each VH and VL is composed of three CDRs and four FRs, arranged from amino-terminus to carboxy- terminus in the following order: FR1, CDR1, FR2, CDR2, FR3, CDR3, FR4.
  • the variable regions of the heavy and light chains contain a binding domain that interacts with an antigen.
  • the constant regions of the antibodies may mediate the binding of the immunoglobulin to host tissues or factors, including various cells of the immune system (e.g., effector cells) and the first component (Clq) of the classical complement system
  • antigen-binding fragment of an antibody (or simply “binding portion”), as used herein, refers to one or more fragments of an antibody that retain the ability to specifically bind to an antigen. It has been shown that the antigen-binding function of an antibody can be performed by fragments of a full-length antibody.
  • binding fragments encompassed within the term "antigen-binding portion" of an antibody include (i) Fab fragments, monovalent fragments consisting of the VL, VH, CL and CH domains; (ii) F(ab') 2 fragments, bivalent fragments comprising two Fab fragments linked by a disulfide bridge at the hinge region; (iii) Fd fragments consisting of the VH and CH domains; (iv) Fv fragments consisting of the VL and VH domains of a single arm of an antibody, (v) dAb fragments (Ward et al., (1989) Nature 341: 544-546), which consist of a VH domain; (vi) isolated complementarity determining regions (CDR), and (vii) combinations of two or more isolated CDRs which may optionally be joined by a synthetic linker.
  • Fab fragments monovalent fragments consisting of the VL, VH, CL and CH domains
  • F(ab') 2 fragments bivalent fragments compris
  • the two domains of the Fv fragment, VL and VH are coded for by separate genes, they can be joined, using recombinant methods, by a synthetic linker that enables them to be made as a single protein chain in which the VL and VH regions pair to form monovalent molecules (known as single chain Fv (scFv); see e.g., Bird et al. (1988) Science 242: 423-426; and Huston et al. (1988) Proc. Natl. Acad. Sci. USA 85: 5879- 5883).
  • single chain Fv single chain Fv
  • Such single chain antibodies are also intended to be encompassed within the term "antigen-binding fragment" of an antibody.
  • a further example is a binding-domain immunoglobulin fusion protein comprising (i) a binding domain polypeptide that is fused to an immunoglobulin hinge region polypeptide, (ii) an immunoglobulin heavy chain CH2 constant region fused to the hinge region, and (iii) an immunoglobulin heavy chain CH3 constant region fused to the CH2 constant region.
  • the binding domain polypeptide can be a heavy chain variable region or a light chain variable region.
  • the binding-domain immunoglobulin fusion proteins are further disclosed in US 2003/0118592 and US 2003/0133939. These antibody fragments are obtained using conventional techniques known to those skilled in the art, and the fragments are screened for utility in the same manner as are intact antibodies.
  • antigen-binding fragments are so-called microantibodies, which are derived from single CDRs.
  • microantibodies which are derived from single CDRs.
  • Heap et al., 2005 describe a 17 amino acid residue microantibody derived from the heavy chain CDR3 of an antibody directed against the gpl20 envelope glycoprotein of HIV-1 (Heap C.J. et al. (2005) Analysis of a 17 -amino acid residue, virus-neutralizing microantibody. J. Gen. Virol. 86: 1791-1800).
  • small antibody mimetics comprising two or more CDR regions that are fused to each other, preferably by cognate framework regions.
  • antibody or antigen-binding fragment thereof refers to immunoglobulin molecules and immunologically active portions of immunoglobulin molecules, i.e. molecules that contain an antigen-binding site that immuno specifically binds an antigen. Also comprised are immunoglobulin-like proteins that are selected through techniques including, for example, phage display to specifically bind to a target molecule or target epitope.
  • the immunoglobulin molecules of the invention can be of any type (e.g., IgG, IgE, IgM, IgD, IgA and IgY), class (e.g., IgGl, IgG2, preferably IgG2a and IgG2b, IgG3, IgG4, IgAl and IgA2) or subclass of immunoglobulin molecule.
  • type e.g., IgG, IgE, IgM, IgD, IgA and IgY
  • class e.g., IgGl, IgG2, preferably IgG2a and IgG2b, IgG3, IgG4, IgAl and IgA2
  • subclass of immunoglobulin molecule e.g., IgG, IgE, IgM, IgD, IgA and IgY
  • subclass of immunoglobulin molecule e.g., IgG, IgE, IgM, Ig
  • Antibodies and antigen-binding fragments thereof usable in the invention may be from any animal origin including birds and mammals.
  • the antibodies or fragments are from human, chimpanzee, rodent (e.g. mouse, rat, guinea pig, or rabbit), chicken, turkey, pig, sheep, goat, camel, cow, horse, donkey, cat, or dog origin. It is particularly preferred that the antibodies are of human or murine origin.
  • Antibodies of the invention also include chimeric molecules in which an antibody constant region derived from one species, preferably human, is combined with the antigen-binding site derived from another species, e.g. mouse.
  • antibodies of the invention include humanized molecules in which the antigen-binding sites of an antibody derived from a non-human species (e.g. from mouse) are combined with constant and framework regions of human origin.
  • antibodies of the invention can be obtained directly from hybridomas which express the antibody, or can be cloned and recombinantly expressed in a host cell (e.g., a CHO cell, or a lymphocytic cell).
  • a host cell e.g., a CHO cell, or a lymphocytic cell.
  • host cells are microorganisms, such as E. coli, and fungi, such as yeast.
  • they can be produced recombinantly in a transgenic non-human animal or plant.
  • chimeric antibody refers to those antibodies wherein one portion of each of the amino acid sequences of heavy and light chains is homologous to corresponding sequences in antibodies derived from a particular species or belonging to a particular class, while the remaining segment of the chain is homologous to corresponding sequences in another species or class.
  • the variable region of both light and heavy chains mimics the variable regions of antibodies derived from one species of mammals, while the constant portions are homologous to sequences of antibodies derived from another.
  • One clear advantage to such chimeric forms is that the variable region can conveniently be derived from presently known sources using readily available B-cells or hybridomas from non-human host organisms in combination with constant regions derived from, for example, human cell preparations.
  • variable region has the advantage of ease of preparation and the specificity is not affected by the source, the constant region being human is less likely to elicit an immune response from a human subject when the antibodies are injected than would the constant region from a non- human source.
  • the definition is not limited to this particular example.
  • humanized antibody refers to a molecule having an antigen-binding site that is substantially derived from an immunoglobulin from a non-human species, wherein the remaining immunoglobulin structure of the molecule is based upon the structure and/or sequence of a human immunoglobulin.
  • the antigen-binding site may either comprise complete variable domains fused onto constant domains or only the complementarity determining regions (CDR) grafted onto appropriate framework regions in the variable domains.
  • CDR complementarity determining regions
  • Antigen-binding sites may be wild-type or modified by one or more amino acid substitutions, e.g. modified to resemble human immunoglobulins more closely.
  • Some forms of humanized antibodies preserve all CDR sequences (for example a humanized mouse antibody which contains all six CDRs from the mouse antibody). Other forms have one or more CDRs which are altered with respect to the original antibody.
  • human antibodies include antibodies having variable and constant regions derived from human germline immunoglobulin sequences.
  • the human antibodies of the invention may include amino acid residues not encoded by human germline immunoglobulin sequences (e.g., mutations introduced by random or site-specific mutagenesis in vitro or by somatic mutation in vivo).
  • Human antibodies of the invention include antibodies isolated from human immunoglobulin libraries or from animals transgenic for one or more human immunoglobulin and that do not express endogenous immunoglobulins, as described for example in U.S. Patent No. 5,939,598 by Kucherlapati & Jakobovits.
  • the term "monoclonal antibody” as used herein refers to a preparation of antibody molecules of single molecular composition.
  • a monoclonal antibody displays a single binding specificity and affinity for a particular epitope.
  • the monoclonal antibodies are produced by a hybridoma which includes a B cell obtained from a non-human animal, e.g. mouse, fused to an immortalized cell.
  • recombinant antibody includes all antibodies that are prepared, expressed, created or isolated by recombinant means, such as (a) antibodies isolated from an animal (e.g., a mouse) that is transgenic or transchromosomal with respect to the immunoglobulin genes or a hybridoma prepared therefrom, (b) antibodies isolated from a host cell transformed to express the antibody, e.g. from a transfectoma, (c) antibodies isolated from a recombinant, combinatorial antibody library, and (d) antibodies prepared, expressed, created or isolated by any other means that involve splicing of immunoglobulin gene sequences to other DNA sequences.
  • transfectoma includes recombinant eukaryotic host cells expressing an antibody, such as CHO cells, NS/0 cells, HEK293 cells, HEK293T cells, plant cells, or fungi, including yeast cells.
  • a heterologous antibody is defined in relation to a transgenic organism producing such an antibody. This term refers to an antibody having an amino acid sequence or an encoding nucleic acid sequence corresponding to that found in an organism not consisting of the transgenic organism, and being generally derived from a species other than the transgenic organism.
  • heterohybrid antibody refers to an antibody having light and heavy chains of different organismal origins.
  • an antibody having a human heavy chain associated with a murine light chain is a heterohybrid antibody.
  • antibodies and antigen-binding fragments thereof suitable for use in the present invention include, but are not limited to, polyclonal, monoclonal, monovalent, bispecific, heteroconjugate, multispecific, recombinant, heterologous, heterohybrid, chimeric, humanized (in particular CDR-grafted), deimmunized, or human antibodies, Fab fragments, Fab' fragments, F(ab') 2 fragments, fragments produced by a Fab expression library, Fd, Fv, disulfide- linked Fvs (dsFv), single chain antibodies (e.g. scFv), diabodies or tetrabodies (Holliger P. et al. (1993) Proc. Natl.
  • nanobodies also known as single domain antibodies
  • anti-idiotypic antibodies including, e.g., anti-Id antibodies to antibodies described herein
  • epitope-binding fragments of any of the above include, e.g., anti-Id antibodies to antibodies described herein, and epitope-binding fragments of any of the above.
  • the antibodies described herein are preferably isolated.
  • An "isolated antibody” as used herein is intended to refer to an antibody which is substantially free of other antibodies having different antigenic specificities (e.g., an isolated antibody that specifically binds to C5a is substantially free of antibodies that specifically bind antigens other than C5a).
  • An isolated antibody that specifically binds to an epitope, isoform or variant of human C5a may, however, have cross-reactivity to other related antigens, e.g. from other species (e.g. C5a species homologs, such as rat C5a).
  • an isolated antibody may be substantially free of other cellular material and/or chemicals.
  • a combination of "isolated" monoclonal antibodies relates to antibodies having different specificities and being combined in a well-defined composition.
  • naturally occurring refers to the fact that an object can be found in nature.
  • a polypeptide or polynucleotide sequence that is present in an organism (including viruses) that can be isolated from a source in nature and which has not been intentionally modified by man in the laboratory is naturally occurring.
  • nucleic acid aptamer refers to a nucleic acid molecule that has been engineered through repeated rounds of in vitro selection or SELEX (systematic evolution of ligands by exponential enrichment) to bind to a target molecule (for a review see: Brody E.N. and Gold L. (2000), Aptamers as therapeutic and diagnostic agents. J. Biotechnol. 74(1):5-13).
  • the nucleic acid aptamer may be a DNA or RNA molecule.
  • the aptamers may contain modifications, e.g.
  • modified nucleotides such as 2'-fluorine-substituted pyrimidines, and/or may comprise one or more nucleotides with L-ribose units (or L-deoxyribose) instead of the standard D-ribose units (or D-deoxyribose units).
  • antibody-like protein refers to a protein that has been engineered (e.g. by mutagenesis of loops) to specifically bind to a target molecule.
  • an antibody-like protein comprises at least one variable peptide loop attached at both ends to a protein scaffold. This double structural constraint greatly increases the binding affinity of the antibody-like protein to levels comparable to that of an antibody.
  • the length of the variable peptide loop typically consists of 10 to 20 amino acids.
  • the scaffold protein may be any protein having good solubility properties.
  • the scaffold protein is a small globular protein.
  • Antibody-like proteins include without limitation affibodies, affilins, affimers, affitins, alphabodies, anticalins, avimers, DARPins (designed ankyrin repeat proteins), fynomers, Kunitz domain peptides, and monobodies (for review see: Binz H.K. et al. (2005) Engineering novel binding proteins from nonimmuno globulin domains. Nat. Biotechnol. 23(10): 1257-1268).
  • Antibody-like proteins can be derived from large libraries of mutants, e.g. be panned from large phage display libraries and can be isolated in analogy to regular antibodies. Also, antibody-like binding proteins can be obtained by combinatorial mutagenesis of surface-exposed residues in globular proteins. Antibody-like proteins are sometimes referred to as "peptide aptamers" or as "antibody mimetics”.
  • a "peptidomimetic” is a small protein-like chain designed to mimic a peptide.
  • Peptidomimetics typically arise from modification of an existing peptide in order to alter the molecule's properties. For example, they may arise from modifications to change the molecule's stability or biological activity. This can have a role in the development of drug-like compounds from existing peptides. These modifications involve changes to the peptide that will not occur naturally (such as altered backbones and the incorporation of non-natural amino acids).
  • the term "small molecule” refers to a molecule with a molecular weight of 2 kDa or less, preferably with a molecular weight of 1 kDa or less.
  • small molecule particularly refers to molecules that are neither oligopeptides nor oligonucleotides.
  • the general expression "wherein A competes with B for binding to C”, is used to define the binding properties of the compound listed in position A.
  • Said compound A binds to C and compound B also binds to C but compound A and compound B cannot bind to C at the same time; i.e. A and B bind to the same epitope (or at least to overlapping epitopes) on C.
  • Such competition in binding can be determined by competitive ELISA or by Surface Plasmon Resonance (SPR) based technology or by any of the other techniques listed above in the context of the determination of binding affinities. If not explicitly stated otherwise, the competing binding properties of a compound are determined by ELISA at 20°C using equimolar concentrations of the two competing compounds.
  • SPR Surface Plasmon Resonance
  • a "cutaneous, neutrophilic, inflammatory disease” refers to any disease that is associated with an inflammation of the skin and with a neutrophilic infiltrate into the skin (e.g. into the epidermis) of an individual afflicted by said disease.
  • HS hidradenitis suppurativa
  • PAPA pyogenic arthritis, PG and acne
  • PASH PG, acne and hidradenitis suppurativa
  • PAPASH pyogenic arthritis, acne, PG and hidradenitis suppurativa
  • Sweet syndrome SS
  • SPD subcorneal pustular dermatosis
  • EED neutrophilic panniculitis
  • BAD AS bowel-associated dermatosis- arthritis syndrome
  • SAPHO synovitis, acne, pustulosis, hyperostosis, and osteitis
  • rheumatoid neutrophilic dermatosis familial Mediterranean fever, cryopyrin- associated disorders, gout, and Schnitzler syndrome.
  • HS-related disease comprises without limitation Pyoderma gangrenosum (PG); PAPA (pyogenic arthritis, PG and acne); PASH (PG, acne and hidradenitis suppurativa); PAPASH (pyogenic arthritis, acne, PG and hidradenitis suppurativa); Sweet syndrome (SS); and subcorneal pustular dermatosis (SPD).
  • PG Pyoderma gangrenosum
  • PAPA pyogenic arthritis, PG and acne
  • PASH PG, acne and hidradenitis suppurativa
  • PAPASH pyogenic arthritis, acne, PG and hidradenitis suppurativa
  • SS Sweet syndrome
  • SPD subcorneal pustular dermatosis
  • IFX-1 (alternative name: CaCP29; InflaRx GmbH, Germany) is an antibody specifically binding to C5a.
  • the CDR sequences and FR sequences of IFX-1 are disclosed in WO 2015/140304 Al (Table 3), the content of which is hereby incorporated by reference in its entirety.
  • INab708 (InflaRx GmbH, Germany) is another antibody specifically binding to C5a.
  • the CDR sequences and FR sequences of INab708 are also disclosed in WO 2015/140304 Al (Table 3), the content of which is incorporated by reference in its entirety.
  • MED 1-7814 (Medlmmune) is a recombinant humanized anti-C5a antibody.
  • the crystal structure of the human C5a in complex with MEDI7814 is available in the RCSB Protein Data Bank under 4UU9 (DOI: 10.2210/pdb4uu9/pdb).
  • ALXN-1007 (Alexion) is a humanized anti-C5a antibody.
  • NOX-D21 (Noxxon) is a PEGylated mixed L-RNA/DNA-aptamer (SpiegelmerTM) with the sequence 40 kDaPEG-aminohexyl-GCG AUG (dU)GG UGG UGA AGG GUU GUU GGG (dU)GU CGA CGC A(dC)G C (SEQ ID NO: 34).
  • NOX-D21 targets C5a (Hyzewicz J, Tanihata J, Kuraoka M, Nitahara-Kasahara Y, Beylier T, Ruegg UT, Vater A, and Takeda S. 2017.
  • Eculizumab (Alternative names: SolirisTM, 5G 1 - 1 ; h5G 1.1; Alexion Pharmaceuticals) is a recombinant humanized monoclonal IgG2/4K antibody produced by murine myeloma cell culture and purified by standard bioprocess technology. Eculizumab specifically binds to human C5. Eculizumab contains human constant regions from human IgG2 sequences and human IgG4 sequences and murine complementarity-determining regions grafted onto the human framework light- and heavy-chain variable regions. Eculizumab is composed of two 448 amino acid heavy chains and two 214 amino acid light chains and has a molecular weight of approximately 148 kDa.
  • the heavy chain and light chain of eculizumab are disclosed, for example, in WO 2016/061066 Al as SEQ ID NO: 1 and SEQ ID NO: 34, respectively.
  • Nucleic acids that encode the heavy and light chains of eculizumab are disclosed, for example, in U.S. Patent No. 6,355,245.
  • ALXN1210 (Alternative name: BNJ441; Alexion Pharmaceuticals) is an anti-C5 antibody.
  • the heavy and light chains of ALXN1210 are disclosed in WO 2016/209956 Al as SEQ ID NOs: 14 and 11, respectively.
  • ALXN5500 (Alexion) is a humanized anti-C5 antibody. It is a next-generation eculizumab candidate.
  • LFG316 (Alternative name: Tesidolumab, NOV-4; Morphosys, Novartis) is an anti-C5 antibody.
  • CoversinTM (alternative names: EV 576; PAS-coversin; rEV 576; Tissue targeted CoversinTM - Akari; Akari Therapeutics, Evolutec) is a recombinant protein molecule (16.7 kDa) derived from a salivary molecule from the Ornithodros moubata tick where it assists the parasite to feed without provoking a host immunological response.
  • the amino acid sequence of the EV576 protein i.e. Coversin
  • CoversinTM binds to C5.
  • RA 101495 (Ra Pharma) is a macrocyclic synthetic peptide inhibitor of C5 (Ricardo A,
  • Zimura® (Alternative names: Anti-C5 aptamer; ARC- 187; ARC- 1905; Avacincaptad pegol sodium; OphthoTech Corporation, Archemix Corporation) is a pegylated RNA aptamer that inhibits complement factor C5.
  • the nucleotide sequence of ARC 1905 i.e. Zimura
  • SEQ ID NO: 67 SEQ ID NO: 67
  • AMY-201 (Amyndas Pharmaceuticals) is an engineered form of Factor H that directly links the regulatory and surface -recognition domains; thus, it is a sort of mini-FH molecule.
  • Mirococept (alternative names: APT070 and APT 070C; originator: Adprotech; developer: Inflazyme Pharmaceuticals) consists of the first three short consensus domains of human complement receptor 1, manufactured in recombinant bacteria and modified with a membrane-targeting amphiphilic peptide based on the naturally occurring membrane-bound myristoyl-electrostatic switch peptide (Souza DG, Esser D, Bradford R, Vieira AT, and Teixeira MM. 2005.
  • APT070 (Mirococept), a membrane-localised complement inhibitor, inhibits inflammatory responses that follow intestinal ischaemia and reperfusion injury. Br J Pharmacol 145(8): 1027- 1034).
  • BikacioMab (Novelmed) is an F(ab) 2 fragment of an anti-factor Bb antibody termed
  • Antibody NM001 is produced by hybridoma cell line 1D3 deposited under ATCC accession number PTA-8543.
  • Lampalizumab (alternative names: Anti-factor D Fab; FCFD4514S; RG7417; TNX- 234; originator: Tanox, Developer: Genentech) is a humanized anti-Factor D Fab fragment that inhibits Factor D and the alternative complement pathway, through binding to an exosite on factor D.
  • ALN-CC5 (Alnylam) is an RNAi therapeutic targeting human, primate and rodent C5. Exemplary iRNA compositions targeting the C5 gene are described in WO 2016/044419.
  • Avacopan also known by the name CCX168; Chemocentryx
  • CCX168 Chemocentryx
  • MW 581.66 g/mol MW 581.66 g/mol
  • avacopan is (2R,3S)-2-[4-(cyclopentylamino)phenyl]- l-(2-fluoro-6-methylbenzoyl)-N-[4-methyl-3-(trifluoromethyl)phenyl]piperidine-3- carboxamide.
  • Avacopan is a selective inhibitor of C5aR.
  • the term "avacopan" refers to the compound according to formula I as well as to physiologically tolerable salts thereof.
  • the inhibitor of C5a activity is a compound having the formula II
  • C 1 is selected from the group consisting of aryl and heteroaryl, wherein the heteroaryl group has from 1-3 heteroatoms as ring members selected from N, O and S; and wherein said aryl and heteroaryl groups are optionally substituted with from 1 to 3 R 1 substituents;
  • C 2 is selected from the group consisting of aryl and heteroaryl, wherein the heteroaryl group has from 1-3 heteroatoms as ring members selected from N, O and S; and wherein said aryl and heteroaryl groups are optionally substituted with from 1 to 3 R 2 substituents;
  • C 3 is selected from the group consisting of C 1-8 alkyl or heteroalkyl, C 3 - 8 cycloalkyl, C 3 - 8 cycloalkyl-Ci-4 alkyl, aryl, aryl-Ci-4 alkyl, heteroaryl, heteroaryl-Ci-4 alkyl, heterocyclo alkyl or heterocycloalkyl-Ci-4 alkyl, wherein the heterocycloalkyl group or portion has from 1-3 heteroatoms selected from N, O and S, and wherein the heteroaryl group has from 1-3 heteroatoms as ring members selected from N, O and S, and each C 3 is optionally substituted with from 1-3 R 3 substituents;
  • each R 1 is independently selected from the group consisting of halogen, -CN, -R c , -C0 2 R a , - CONR a R b , -C(0)R a , -OC(0)NR a R b , -NR b C(0)R a , -NR b C(0)2R c , -NR a -C(0)NR a R b , - NR a C(0)NR a R b , -NR a R b , -OR a , and -S(0) 2 NR a R b ; wherein each R a and R b is independently selected from hydrogen, C 1-8 alkyl, and C 1-8 haloalkyl, or when attached to the same nitrogen atom can be combined with the nitrogen atom to form a five or six- membered ring having from 0 to 2 additional heteroatoms as ring members selected from N, O or S, and is optionally substituted with one or
  • each R 2 is independently selected from the group consisting of halogen, -CN, -N0 2 , -R f , - C0 2 R d , -CONR d R e , -C(0)R d , -OC(0)NR d R e , -NR e C(0)R d , -NR e C(0) 2 R f , - NR d C(0)NR d R e , -NR d C(0)NR d R e , -NR d R e , -OR d , and -S(0) 2 NR d R e ; wherein each R d and R e is independently selected from hydrogen, C 1-8 alkyl, and C 1-8 haloalkyl, or when attached to the same nitrogen atom can be combined with the nitrogen atom to form a five or six-membered ring having from 0 to 2 additional heteroatoms as ring members selected from N, O or S, and is optionally
  • each R 3 is independently selected from the group consisting of halogen, -CN, -R 1 , -C0 2 R g , - CONR g R h , -C(0)R g , -C(0)R ⁇ -OC(0)NR g R h , -NR h C(0)R , -NR h C0 2 R ⁇ -
  • X is hydrogen or CH3.
  • the inhibitor of C5a activity is a compound of the following formula III:
  • R 1 is selected from the group consisting of H, -0-CH 2 -0-P(0)OROR b , -0-C(0)-Ci_ 6 alkylene- IAX 1 , 0-P(0)OROR b , and -0-C(0)-A 1 -(Ci- 3 alkylene) n -C 4 -7 heterocyclyl wherein the C 4 -7 heterocyclyl is optionally substituted with 1 to 6 R c groups;
  • a 1 is selected from the group consisting of Ce-io aryl, C3-10 cycloalkyl, C5-10 heteroaryl and C5- 10 heterocyclyl, each of which is optionally substituted with 1 to 5 R x which can be the same or different;
  • L 2 is independently selected from the group consisting of a bond, -0-C(0)-Ci-6 alkylene-, and
  • X 1 is independently selected from the group consisting of -NR e R f , -P(0)OROR b , -O-
  • R 2 is selected from the group consisting of H, -L 3 -Ci-6 alkylene-L 4 -X 2 , -L 3 - (C 1-6 alkylene) m -
  • L 3 is independently selected from the group consisting of -C(0)-0-, and -C(O)-;
  • L 4 is independently selected from the group consisting of a bond, -0-C(0)-C 2 -6 alkenylene-, -
  • NR d -C(0)-Ci-6 alkylene- and -0-C(0)-Ci-6 alkylene- is optionally substituted with
  • X 2 is independently selected from the group consisting of -NR ⁇ , -P(0)OROR b , -O-
  • a 2 is selected from the group consisting of Ce-io aryl, C3-10 cycloalkyl, C5-10 heteroaryl and C5- 10 heterocyclyl, each of which is optionally substituted with 1 to 5 R x which can be the same or different;
  • R 3 is H or -L 5 -P(0)OROR b wherein L 5 is independently selected from the group consisting of a bond and -CH 2 -0-;
  • each R x is independently selected from the group consisting of halogen, C 1-6 alkyl, C 1-6 haloalkyl, Ci-e heteroalkyl, CN, NR y R z , SR y and OR y ;
  • each R c is independently selected from the group consisting of halogen, C 1-6 alkyl, C 1-6 haloalkyl, Ci-e heteroalkyl, CN, NR y R z , SR y and OR y ;
  • each R a , R b , R d , R e , R f , R g , R k , R 1 , R y and R z is independently selected from the group consisting of H and C 1-6 alkyl;
  • each R h is independently selected from the group consisting of H and C 1-6 alkyl wherein the Ci- 6 alkyl is optionally substituted with 1 to 5 substituents independently selected from C0 2 H, NR'R J , C 6 -io aryl, C3-10 cycloalkyl, C5-10 heteroaryl and C5-10 heterocyclyl, wherein each R 1 and R j is independently H or C 1-6 alkyl;
  • R 1 , R 2 and R 3 are H, and one of R 1 , R 2 and R 3 is other than H.
  • PMX-53 is a potent antagonist of C5aR (CD88). It is a circular peptide composed of six amino acids, with the following sequence: Ac-Phe-cyclo(Orn-Pro-D-Cha-Trp-Arg) with a lactam bridge between Orn-2 and Arg-6. Since PMX-53 contains at least one D-amino acid (i.e.
  • PMX-53 is commercially available by bio-techne GmbH (Wiesbaden-Nordenstadt, Germany), Cat. No. 5473.
  • the inhibitor of C5a activity is a cyclic peptide or peptidomimetic compound of the formula IV
  • A is H, alkyl, aryl, NH 2 , NH-alkyl, N(alkyl) 2 , NH-aryl, NH-acyl, NH-benzoyl, NHS0 3 ,
  • NHS0 2 -alkyl NHS0 2 -aryl, OH, O-alkyl, or O-aryl;
  • B is an alkyl, aryl, phenyl, benzyl, naphthyl or indole group, or the side chain of a D- or L- amino acid, but is not the side chain of glycine, D-phenylalanine, L- homophenylalanine, L-tryptophan, L-homotryptophan, L- tyrosine, or L- homotyrosine;
  • C is the side chain of a D-, L- or homo-amino acid, but is not the side chain of isoleucine, phenylalanine, or cyclohexylalanine;
  • D is the side chain of a neutral D-amino acid, but is not the side chain of glycine or D-alanine, a bulky planar side chain, or a bulky charged side chain;
  • E is a bulky substituent, but is not the side chain of D-tryptophan, L-N-methyltryptophan, L- homophenylalanine, L-2-naphthyl L-tetrahydroisoquinoline, L-cyclohexylalanine, D- leucinc, L-fluorenylalanine, or L-histidine;
  • F is the side chain of L-arginine, L-homoarginine, L-citrulline, or L-canavanine, or a bioisostere thereof; and X 1 is -(CH 2 ) n NH- or (CH 2 ) n S-, where n is an integer of from 1 to 4; -(CH 2 ) 2 0-; -(CH 2 ) 3 0; - (CH 2 ) 3 -; -(CH 2 ) 4 -, -CH 2 -COCHRNH-: or -CH 2 -CHCOCHRNH-, where R is the side chain of any common or uncommon amino acid.
  • common amino acid refers to the twenty proteinogenic amino acids that are defined by the standard genetic code.
  • uncommon amino acid includes, but is not restricted to, D-amino acids, homo-amino acids, N-alkyl amino acids, dehydroamino acids, aromatic amino acids other than phenylalanine, tyrosine and tryptophan, ortho-, meta- or para-aminobenzoic acid, ornithine, citrulline, canavanine, norleucinc, ⁇ -glutamic acid, aminobutyric acid, L-fluorenylalanine, L-3-benzothienylalanine, and ⁇ , ⁇ -disubstituted amino acids.
  • C5aR CD88
  • Specific antagonists of C5aR (CD88) suitable for practicing the present invention include PMX95, PMX218, PMX200, PMX273, PMX205, and PMX201, as disclosed in WO 2008/009062 Al.
  • Clone S5/1 is a monoclonal antibody recognizing the human receptor for C5a (CD88). Clone S5/1 was raised against a synthetic peptide comprising the N-terminal domain of the C5aR (Metl-Asn31). The antibody has been shown to inhibit the binding of C5a to its receptor. It is commercially available via Hycult Biotech (Uden, The Netherlands), Cat. No. HM2094.
  • Clone 7H110 is a monoclonal mouse antibody recognizing the human receptor for C5a (CD88). It is commercially available via Biomol GmbH (Hamburg, Germany); Cat. No. C2439- 60N.
  • a "patient” means any mammal or bird who may benefit from a treatment with the compound described herein (i.e. with an inhibitor of C5a activity described herein).
  • a “patient” is selected from the group consisting of laboratory animals (e.g. mouse or rat), domestic animals (including e.g. guinea pig, rabbit, chicken, turkey, pig, sheep, goat, camel, cow, horse, donkey, cat, or dog), or primates including chimpanzees and human beings. It is particularly preferred that the "patient” is a human being.
  • treat means accomplishing one or more of the following: (a) reducing the severity and/or duration of the disorder; (b) limiting or preventing development of symptoms characteristic of the disorder(s) being treated; (c) inhibiting worsening of symptoms characteristic of the disorder(s) being treated; (d) limiting or preventing recurrence of the disorder(s) in patients that have previously had the disorder(s); and (e) limiting or preventing recurrence of symptoms in patients that were previously symptomatic for the disorder(s).
  • prevent means preventing that a disorder occurs in a subject.
  • an “effective amount” is an amount of a therapeutic agent sufficient to achieve the intended purpose.
  • the effective amount of a given therapeutic agent will vary with factors such as the nature of the agent, the route of administration, the size and species of the animal to receive the therapeutic agent, and the purpose of the administration.
  • the effective amount in each individual case may be determined empirically by a skilled artisan according to established methods in the art.
  • “Pharmaceutically acceptable” means approved by a regulatory agency of the Federal or a state government or listed in the U.S. Pharmacopeia or other generally recognized pharmacopeia for use in animals, and more particularly in humans.
  • the present invention is directed to a compound for use in the treatment of a cutaneous, neutrophilic, inflammatory disease in a subject, wherein the compound is an inhibitor of C5a activity, and wherein the cutaneous, neutrophilic, inflammatory disease is selected from the group consisting of hidradenitis suppurativa (HS); Pyoderma gangrenosum (PG); PAPA (pyogenic arthritis, PG and acne); PASH (PG, acne and hidradenitis suppurativa); PAPASH (pyogenic arthritis, acne, PG and hidradenitis suppurativa); Sweet syndrome (SS); subcorneal pustular dermatosis (SPD); epidermolysis bullosa acquisita, erythema elevatum diutinum (EED); neutrophilic panniculitis; bowel-associated dermatosis-arthritis syndrome (BAD AS); SAPHO (synovitis, acne, pustulosis, hyperost
  • HS
  • the present invention is directed to a method for the treatment of a cutaneous, neutrophilic, inflammatory disease in a subject, comprising the step of:
  • adenitis suppurativa HS
  • Pyoderma gangrenosum PG
  • PAPA pyogenic arthritis, PG and acne
  • PASH PG, acne and hidradenitis suppurativa
  • PAPASH pyogenic arthritis, acne, PG and hidradenitis suppurativa
  • Sweet syndrome SS
  • subcorneal pustular dermatosis SPD
  • the present invention is directed to a use of a compound for the preparation of a pharmaceutical composition for the treatment of a cutaneous, neutrophilic, inflammatory disease, wherein the compound is an inhibitor of C5a activity, and wherein the cutaneous, neutrophilic, inflammatory disease is selected from the group consisting of hidradenitis suppurativa (HS); Pyoderma gangrenosum (PG); PAPA (pyogenic arthritis, PG and acne); PASH (PG, acne and hidradenitis suppurativa); PAPASH (pyogenic arthritis, acne, PG and hidradenitis suppurativa); Sweet syndrome (SS); subcorneal pustular dermatosis (SPD); epidermolysis bullosa acquisita, erythema elevatum diutinum (EED); neutrophilic panniculitis; bowel-associated dermato sis-arthritis syndrome (BADAS); SAPHO (synovitis, acne, pus
  • the inhibitor of C5a activity is:
  • C5 lowers the concentration of C5 (for example, by inhibiting formation and/or activity of C3 convertase; by inhibiting formation and/or activity of C5 convertase; by inhibiting the transcription of the C5 gene; by blocking translation of the C5 mRNA; by increasing degradation of the C5 mRNA; by increasing degradation of the C5 protein; or by prevention secretion of C5 from the liver);
  • C5a concentration of C5a (for example, by increasing degradation of the C5a protein); inhibits the binding between C5a and a C5a receptor (for example by binding to C5a or by binding to a C5a receptor);
  • C5a receptor lowers the concentration of a C5a receptor (for example, by inhibiting transcription of a C5a receptor gene; by blocking translation of a C5a receptor mRNA; by increasing degradation of a C5a receptor mRNA; by increasing degradation of a C5a receptor protein); and/or inhibits the activity of a C5a receptor.
  • the inhibitor of C5a activity is selected from the group consisting of a protein ligand (as defined above); an oligonucleotide; and a small molecule (as defined above). Oligonucleotides acting as inhibitors of C5a activity can achieve their inhibitory effect for example by binding to nucleic acid molecules (thereby inhibiting transcription and/or translation) or by binding to proteins (e.g. when the oligonucleotides are nucleic acid aptamers).
  • the inhibitor of C5a activity is a protein ligand that specifically binds to C5 protein, or to C5a protein, or to a C5a receptor protein.
  • the protein ligand is selected from the group consisting of
  • antibodies e.g. anti-C5 antibodies, anti-C5a antibodies, anti-C5aR antibodies, or anti-
  • proteins acting on the complement pathway e.g. Coversin
  • peptides e.g. RA101495 (Ra Pharma, Cambridge, MA); PMX-53 (bio-techne GmbH
  • the inhibitor of C5a activity is a protein ligand or an oligonucleotide, preferably a protein ligand, that specifically binds to a conformational epitope formed by amino acid sequences NDETCEQRA (SEQ ID NO: 2) and SHKDMQL (SEQ ID NO: 3) of human C5a. Binding to the conformational formed by the amino acid sequences according to SEQ ID NOs: 2 and 3 means that the protein ligand or oligonucleotide binds to at least one amino acid within the amino acid sequence according to SEQ ID NO: 2 and to at least one amino acid within the amino acid sequence according to SEQ ID NO: 3.
  • SEQ ID NO: 2 corresponds to amino acids 30-38 of human C5a.
  • SEQ ID NO: 3 corresponds to amino acids 66-72 of human C5a.
  • the protein ligand or oligonucleotide preferably the protein ligand, binds to at least one amino acid within the amino acid sequence according to DETCEQR (SEQ ID NO: 4).
  • SEQ ID NO: 4 corresponds to amino acids 31-37 of human C5a.
  • the protein ligand or oligonucleotide binds to at least one amino acid within the amino acid sequence according to HKDMQ (SEQ ID NO: 5), more preferably to at least one amino acid within the amino acid sequence KDM.
  • SEQ ID NO: 5 corresponds to amino acids 67-71 of human C5a; the sequence KDM corresponds to amino acids 68-70 of human C5a.
  • the protein ligand or oligonucleotide binds to at least one amino acid within the amino acid sequence DETCEQR (SEQ ID NO: 4) and to at least one amino acid within the amino acid sequence HKDMQ (SEQ ID NO: 5).
  • the protein ligand or oligonucleotide preferably the protein ligand, binds to at least one amino acid within the amino acid sequence DETCEQR (SEQ ID NO: 4) and to at least one amino acid within the amino acid sequence KDM.
  • the two sequences forming the conformational epitope of C5a are separated by 1-50 contiguous amino acids that do not participate in binding to the binding moiety of the invention.
  • amino acids that do not participate in binding to the binding moiety of the invention will be referred to as "non-binding amino acids”.
  • the two sequences forming the conformational epitope are preferably separated by 6-45 contiguous non-binding amino acids, more preferably by 12-40 contiguous non-binding amino acids, more preferably by 18-35 contiguous non- binding amino acids, more preferably by 24-30 contiguous non-binding amino acids, more preferably by 25-29 contiguous non-binding amino acids, even more preferably by 26-28 contiguous non-binding amino acids, and most preferably by 27 contiguous non-binding amino acids.
  • the protein ligand or oligonucleotide, preferably the protein ligand, specifically binding to a conformational epitope of C5a has a binding constant to human C5a with a Kd value of 10 nM or less, preferably 9 nM or less, more preferably 8 nM or less, more preferably 7 nM or less, more preferably 6 nM or less, more preferably 5 nM or less, more preferably 4 nM or less, more preferably 3 nM or less, more preferably 2 nM or less, and even more preferably 1 nM or less.
  • the dissociation constant Kd between the binding moiety and human C5a is between 1 pM (picomolar) and 5 nM (nanomolar), more preferably between 2 pM and 4 nM, more preferably between 5 pM and 3 nM, more preferably between 10 pM and 2 nM, more preferably between 50 pM and 1 nM, more preferably between 100 pM and 900 pM, more preferably between 200 pM and 800 pM, more preferably between 300 pM and 700 pM, and even more preferably between 400 pM and 600 pM.
  • the protein ligand or oligonucleotide, preferably the protein ligand, specifically binding to C5a exhibits at least 75% blocking activity, preferably at least 80% blocking activity, more preferably at least 85% blocking activity, more preferably at least 90% blocking activity, more preferably at least 95% blocking activity for biological effects induced by one molecule C5a, particularly human C5a.
  • These particular blocking activities refer to those embodiments, wherein the binding moiety comprises a single paratope binding to C5a, preferably human C5a.
  • the binding moiety comprises two or more C5a-specific paratopes, said blocking activities of at least 75%, preferably at least 80%, more preferably at least 85%, etc.
  • the binding moiety exhibits at least 75% blocking activity, preferably at least 80% blocking activity, more preferably at least 85% blocking activity, more preferably at least 90% blocking activity, and more preferably at least 95% blocking activity for biological effects induced by C5a.
  • a preferred biological effect to be blocked is C5a-induced lysozyme release from human whole blood cells. Assays for determining this C5a-induced lysozyme release and its blocking are described, for example, in WO 2011/063980 Al and in the corresponding US national stage application US 2012/0231008 Al.
  • the protein ligand is an antibody or an antigen-binding fragment thereof, wherein said antibody or antigen-binding fragment thereof comprises
  • heavy chain CDR3 sequence optionally comprises 1, 2, or 3 amino acid exchanges, preferably conservative amino acid exchanges, 1, 2, or 3 amino acid deletions, and/or 1, 2, or 3 amino acid additions.
  • the protein ligand is an antibody or an antigen-binding fragment thereof, wherein said antibody or antigen-binding fragment thereof comprises
  • a light chain CDR3 sequence as set forth in SEQ ID NO: 9; wherein the light chain CDR3 sequence optionally comprises 1, 2, or 3 amino acid exchanges, preferably conservative amino acid exchanges, 1, 2, or 3 amino acid deletions, and/or 1, 2, or 3 amino acid additions.
  • the protein ligand is an antibody or an antigen-binding fragment thereof, wherein said antibody or antigen-binding fragment thereof comprises
  • heavy chain CDR3 sequence optionally comprises 1, 2, or 3 amino acid exchanges, preferably conservative amino acid exchanges, 1, 2, or 3 amino acid deletions, and/or 1, 2, or 3 amino acid additions;
  • the light chain CDR3 sequence optionally comprises 1, 2, or 3 amino acid exchanges, preferably conservative amino acid exchanges, 1, 2, or 3 amino acid deletions, and/or 1, 2, or 3 amino acid additions.
  • the protein ligand is an antibody or an antigen-binding fragment thereof, wherein said antibody or antigen-binding fragment thereof comprises at least one of the following sequences:
  • heavy chain CDR2 sequence optionally comprises 1, 2, or 3 amino acid exchanges, preferably conservative amino acid exchanges, 1, 2, or 3 amino acid deletions, and/or 1, 2, or 3 amino acid additions;
  • the light chain CDR2 sequence optionally comprises 1, 2, or 3 amino acid exchanges, preferably conservative amino acid exchanges, 1, 2, or 3 amino acid deletions, and/or 1, 2, or 3 amino acid additions; wherein the heavy chain CDR1 sequence optionally comprises 1, 2 or 3 amino acid exchanges, preferably conservative amino acid exchanges, 1, 2, or 3 amino acid deletions, and/or 1, 2, or 3 amino acid additions; and
  • the light chain CDR1 sequence optionally comprises 1, 2, or 3 amino acid exchanges, preferably conservative amino acid exchanges, 1, 2, or 3 amino acid deletions, and/or 1, 2, or 3 amino acid additions.
  • the total number of these optional changes recited above in each one of the amino acid sequences according to SEQ ID NO: 6, SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, and SEQ ID NO: 17, i.e. the total number of exchanges, deletions and additions in each sequence, is 1 or 2.
  • the total number of exchanges, deletions, and additions added up for all CDRs present in an antibody or antigen-binding fragment thereof is between 1 and 5 (e.g. 1, 2, 3, 4, or 5).
  • the protein ligand is an antibody or an antigen-binding fragment thereof, comprises one of the sets A to H of heavy chain CDR3, heavy chain CDR2, and heavy chain CDR1 sequences as listed below in Table 1, wherein each heavy chain CDR3 sequence optionally comprises 1, 2, or 3 amino acid exchanges, preferably conservative amino acid exchanges, 1, 2, or 3 amino acid deletions, and/or 1, 2, or 3 amino acid additions;
  • each heavy chain CDR2 sequence optionally comprises 1, 2, or 3 amino acid exchanges, preferably conservative amino acid exchanges, 1, 2, or 3 amino acid deletions, and/or 1, 2, or 3 amino acid additions;
  • each heavy chain CDR1 sequence optionally comprises 1, 2, or 3 amino acid exchanges, preferably conservative amino acid exchanges, 1, 2, or 3 amino acid deletions, and/or 1, 2, or 3 amino acid additions:
  • Table 1 Sets of heavy chain CDR sequences suitable for use in the antibodies or fragments thereof of the present invention
  • the protein ligand is an antibody or an antigen-binding fragment thereof, comprises one of the following sets I to IV of light chain CDR3, light chain CDR2, and light chain CDR1 sequences as listed in Table 2, wherein each light chain CDR3 sequence optionally comprises 1, 2, or 3 amino acid exchanges, preferably conservative amino acid exchanges, 1, 2, or 3 amino acid deletions, and/or 1, 2, or 3 amino acid additions;
  • each light chain CDR2 sequence optionally comprises 1, 2, or 3 amino acid exchanges, preferably conservative amino acid exchanges, 1, 2, or 3 amino acid deletions, and/or 1, 2, or 3 amino acid additions;
  • each light chain CDR1 sequence optionally comprises 1, 2, or 3 amino acid exchanges, preferably conservative amino acid exchanges, 1, 2, or 3 amino acid deletions, and/or 1, 2, or 3 amino acid additions.
  • Table 2 Sets of light chain CDR sequences suitable for use in the antibodies or fragments thereof of the present invention
  • the protein ligand is an antibody or an antigen-binding fragment thereof, comprises one of the heavy CDR sets A-H listed above in Table 1 and one of the light chain CDR sets I- IV listed above in Table 2, i.e.
  • each heavy chain CDR3 sequence optionally comprises 1, 2, or 3 amino acid exchanges, preferably conservative amino acid exchanges, 1, 2, or 3 amino acid deletions, and/or 1, 2, or 3 amino acid additions;
  • each heavy chain CDR2 sequence optionally comprises 1, 2, or 3 amino acid exchanges, preferably conservative amino acid exchanges, 1, 2, or 3 amino acid deletions, and/or 1, 2, or 3 amino acid additions;
  • each heavy chain CDR1 sequence optionally comprises 1, 2, or 3 amino acid exchanges, preferably conservative amino acid exchanges, 1, 2, or 3 amino acid deletions and/or 1, 2, or 3 amino acid additions;
  • each light chain CDR3 sequence optionally comprises 1, 2, or 3 amino acid exchanges, in particular conservative amino acid exchanges, 1, 2, or 3 amino acid deletions, and/or 1, 2, or 3 amino acid additions;
  • each light chain CDR2 sequence optionally comprises 1, 2, or 3 amino acid exchanges, preferably conservative amino acid exchanges, 1, 2, or 3 amino acid deletions, and/or 1, 2, or 3 amino acid additions;
  • each light chain CDR1 sequence optionally comprises 1, 2, or 3 amino acid exchanges, preferably conservative amino acid exchanges, 1, 2, or 3 amino acid deletions and/or 1, 2, or 3 amino acid additions.
  • the protein ligand is an antibody or an antigen-binding fragment thereof, comprises a VH domain that comprises, essentially consists of or consists of (i) the VH domain of IFX-1 or (ii) the VH domain of INab708.
  • FR1, CDR1, FR2, CDR2, FR3, CDR3, and FR4 sequences defining the VH domains of IFX-1 and INab708 are shown below in Table 3.
  • the protein ligand is an antibody or an antigen-binding fragment thereof, comprises a VL domain that comprises, essentially consists of or consists of (i) the VL domain of IFX-1 or (ii) the VL domain of INab708.
  • Heavy Chain Heavy Chain:
  • FR2 WVKQRPGQGLEWIGR FR2: WVKQRPGHGLEWIGE
  • CDR3 CARGNDGYYGFAY
  • CDR3 CTRRGLYDGSSYFAY
  • FR4 WGQGTLVTVSS
  • FR4 WGQGTLVTVSA
  • Light Chain Light Chain:
  • FR1 DIVLTQSPASLAVSLGQRATIS
  • FR1 DIVLTQSPASLAVSLGQRATIS (SEQ ID NO: 22) (SEQ ID NO: 30)
  • FR2 WYQQKPGQPPKLL FR2: WYQQKPGQPPKLL
  • CDR3 CQQSNEDPYT
  • CDR3 CQQNNEDPLT
  • the inhibitor of C5a activity is an oligonucleotide that specifically binds to C5, or to C5a, or to a C5a receptor.
  • the oligonucleotide is a nucleic acid aptamer.
  • the nucleic acid aptamer may be selected from the group consisting of DNA-aptamers, D-RNA aptamers, and L-RNA aptamers (e.g., SpiegelmersTM).
  • the inhibitor of C5a activity reduces expression of C5 protein or a C5a receptor protein.
  • said inhibitor of C5a activity that reduces expression of C5 protein or a C5a receptor protein is an oligonucleotide selected from the group consisting of antisense DNA, antisense RNA, siRNA, and miRNA.
  • the C5a receptor is C5aR and/or C5L2. In preferred embodiments of any aspect of the present invention, the C5a receptor is C5aR (also known as CD88 or C5aRl).
  • the inhibitor of C5a activity is selected from the group consisting of:
  • the cutaneous, neutrophilic, inflammatory disease is a cutaneous, neutrophilic, inflammatory disease.
  • an auto-inflammatory disease (more precisely: a cutaneous, neutrophilic, auto- inflammatory disease); or
  • an autoimmune disease with cutaneous inflammation (more precisely: an autoimmune disease with cutaneous, neutrophilic inflammation).
  • the cutaneous, neutrophilic, inflammatory disease is an auto-inflammatory disease selected from the group consisting of hidradenitis suppurativa (HS); Pyoderma gangrenosum (PG); PAPA (pyogenic arthritis, PG and acne); PASH (PG, acne and hidradenitis suppurativa); PAPASH (pyogenic arthritis, acne, PG and hidradenitis suppurativa); Sweet syndrome (SS); subcorneal pustular dermatosis (SPD); epidermolysis bullosa acquisita, erythema elevatum diutinum (EED); neutrophilic panniculitis; bowel-associated dermatosis-arthritis syndrome (BADAS); and SAPHO (synovitis, acne, pustulosis, hyperostosis, and osteitis) syndrome.
  • HS hidradenitis suppurativa
  • PG Pyoderma gangrenosum
  • PAPA
  • the cutaneous, neutrophilic, inflammatory disease is HS or a HS -related disease selected from the group consisting of Pyoderma gangrenosum (PG); PAPA (pyogenic arthritis, PG and acne); PASH (PG, acne and hidradenitis suppurativa); PAPASH (pyogenic arthritis, acne, PG and hidradenitis suppurativa); Sweet syndrome (SS); and subcorneal pustular dermatosis (SPD).
  • PG Pyoderma gangrenosum
  • PAPA pyogenic arthritis, PG and acne
  • PASH PG, acne and hidradenitis suppurativa
  • PAPASH pyogenic arthritis, acne, PG and hidradenitis suppurativa
  • SS Sweet syndrome
  • SPD subcorneal pustular dermatosis
  • the cutaneous, neutrophilic, inflammatory disease is an autoimmune disease with cutaneous inflammation selected from the group consisting of rheumatoid neutrophilic dermatosis; familial Mediterranean fever, cry opyrin- associated disorders, gout, and Schnitzler syndrome.
  • the compound is to be administered at a dose of 800 mg once per week or at a dose of 800 mg twice per week. In further embodiments of the first or third aspect,
  • the inhibitor of C5a activity is a compound specifically binding to C5a (preferably selected from the group consisting of IFX-1, INab708, MEDI-7814, ALXN-1007, NOX-D21, and an antigen-binding fragment thereof; more preferably the inhibitor of C5a activity is selected from the group consisting of IFX-1, INab708, MEDI-7814, ALXN-1007 and an antigen-binding fragment thereof; even more preferably, the inhibitor of C5a activity is selected from the group consisting of IFX-1 and an antigen-binding fragment thereof; most preferably the inhibitor of C5a activity is IFX-1); and
  • the cutaneous, neutrophilic, inflammatory disease is hidradenitis suppurativa (HS); and the compound is to be administered at a dose of 800 mg once per week or at a dose of 800 mg twice per week.
  • HS hidradenitis suppurativa
  • the inhibitor of C5a activity is to be administered intravenously. In further embodiments of the first or third aspect, the inhibitor of C5a activity is to be administered twice per week at a dose of 800 mg in the first week of treatment and once per week at a dose of 800 mg in the second and subsequent weeks of treatment. In further embodiments of the first or third aspect, the total duration of treatment is between 5 and 12 weeks (e.g. 5 weeks, 6, weeks, 7 weeks, 8 weeks, 9 weeks, 10 weeks, 11 weeks, or 12 weeks).
  • the compound is administered at a dose of 800 mg once per week or at a dose of 800 mg twice per week. In further embodiments of the second aspect,
  • the inhibitor of C5a activity is a compound specifically binding to C5a (preferably selected from the group consisting of IFX-1, INab708, MEDI-7814, ALXN-1007, NOX-D21, and an antigen-binding fragment thereof; more preferably the inhibitor of C5a activity is selected from the group consisting of IFX-1, INab708, MEDI-7814, ALXN-1007 and an antigen-binding fragment thereof; even more preferably, the inhibitor of C5a activity is selected from the group consisting of IFX-1 and an antigen-binding fragment thereof; most preferably the inhibitor of C5a activity is IFX-1); and
  • the cutaneous, neutrophilic, inflammatory disease is hidradenitis suppurativa (HS); and the compound is administered at a dose of 800 mg once per week or at a dose of 800 mg twice per week.
  • HS hidradenitis suppurativa
  • the inhibitor of C5a activity is administered intravenously.
  • the compound is administered twice per week at a dose of 800 mg in the first week of treatment and once per week at a dose of 800 mg in the second and subsequent weeks of treatment.
  • the total duration of treatment is between 5 and 12 weeks (e.g. 5 weeks, 6, weeks, 7 weeks, 8 weeks, 9 weeks, 10 weeks, 11 weeks, or 12 weeks).
  • a compound e.g. an inhibitor of C5a activity described herein
  • a pharmaceutical composition comprising the compound
  • administration may be parenterally, transmucosally, e.g., orally, nasally, rectally, intravaginally, sublingually, submuco sally, transdermally, or by inhalation.
  • administration is parenteral, e.g., via intravenous or intraperitoneal injection, and also including, but is not limited to, intra-arterial, intramuscular, intradermal and subcutaneous administration.
  • the compound described herein e.g. an inhibitor of C5a activity described herein
  • a pharmaceutical composition comprising the compound is administered locally, it can be injected directly into the organ or tissue to be treated.
  • compositions adapted for oral administration may be provided as capsules or tablets; as powders or granules; as solutions, syrups or suspensions (in aqueous or non-aqueous liquids); as edible foams or whips; or as emulsions.
  • Tablets or hard gelatine capsules may comprise lactose, starch or derivatives thereof, magnesium stearate, sodium saccharine, cellulose, magnesium carbonate, stearic acid or salts thereof.
  • Soft gelatine capsules may comprise vegetable oils, waxes, fats, semi-solid, or liquid polyols etc. Solutions and syrups may comprise water, polyols and sugars.
  • An active agent intended for oral administration may be coated with or admixed with a material that delays disintegration and/or absorption of the active agent in the gastrointestinal tract (e.g., glyceryl monostearate or glyceryl distearate may be used).
  • a material that delays disintegration and/or absorption of the active agent in the gastrointestinal tract e.g., glyceryl monostearate or glyceryl distearate may be used.
  • a material that delays disintegration and/or absorption of the active agent in the gastrointestinal tract e.g., glyceryl monostearate or glyceryl distearate may be used.
  • glyceryl monostearate or glyceryl distearate may be used.
  • compositions adapted for transdermal administration may be provided as discrete patches intended to remain in intimate contact with the epidermis of the recipient for a prolonged period of time.
  • Pharmaceutical compositions adapted for topical administration may be provided as ointments, creams, suspensions, lotions, powders, solutions, pastes, gels, sprays, aerosols or oils.
  • a topical ointment or cream is preferably used.
  • the active ingredient may be employed with either a paraffinic or a water-miscible ointment base.
  • the active ingredient may be formulated in a cream with an oil-in-water base or a water-in-oil base.
  • compositions adapted for topical administration to the eye include eye drops.
  • the active ingredient can be dissolved or suspended in a suitable carrier, e.g., in an aqueous solvent.
  • Pharmaceutical compositions adapted for topical administration in the mouth include lozenges, pastilles and mouthwashes.
  • Pharmaceutical compositions adapted for nasal administration may comprise solid carriers such as powders (preferably having a particle size in the range of 20 to 500 microns). Powders can be administered in the manner in which snuff is taken, i.e., by rapid inhalation through the nose from a container of powder held close to the nose.
  • compositions adopted for nasal administration may comprise liquid carriers, e.g., nasal sprays or nasal drops.
  • compositions may comprise aqueous or oil solutions of the active ingredient.
  • Compositions for administration by inhalation may be supplied in specially adapted devices including, but not limited to, pressurized aerosols, nebulizers or insufflators, which can be constructed so as to provide predetermined dosages of the active ingredient.
  • Pharmaceutical compositions may also be administered via the nasal cavity to the lungs.
  • compositions adapted for rectal administration may be provided as suppositories or enemas.
  • Pharmaceutical compositions adapted for vaginal administration may be provided as pessaries, tampons, creams, gels, pastes, foams or spray formulations.
  • compositions adapted for parenteral administration include aqueous and non-aqueous sterile injectable solutions or suspensions, which may contain antioxidants, buffers, bacteriostats and solutes that render the compositions substantially isotonic with the blood of an intended recipient.
  • Other components that may be present in such compositions include water, alcohols, polyols, glycerine and vegetable oils, for example.
  • Compositions adapted for parenteral administration may be presented in unit-dose or multi-dose containers, for example sealed ampules and vials, and may be stored in a freeze-dried (lyophilized) condition requiring only the addition of a sterile liquid carrier, e.g., sterile saline solution for injections, immediately prior to use.
  • a sterile liquid carrier e.g., sterile saline solution for injections, immediately prior to use.
  • Extemporaneous injection solutions and suspensions may be prepared from sterile powders, granules and tablets.
  • a compound described herein e.g. an inhibitor of C5a activity described herein
  • a pharmaceutical composition adapted for intravenous administration to human beings.
  • compositions for intravenous administration are solutions in sterile isotonic aqueous buffer.
  • the composition may also include a solubilizing agent and a local anesthetic such as lidocaine to ease pain at the site of the injection.
  • the ingredients are supplied either separately or mixed together in unit dosage form, for example, as a dry lyophilized powder or water-free concentrate in a hermetically- sealed container such as an ampule or sachette indicating the quantity of active agent.
  • composition is to be administered by infusion, it can be dispensed with an infusion bottle containing sterile pharmaceutical grade water or saline.
  • an ampule of sterile saline can be provided so that the ingredients may be mixed prior to administration.
  • a compound e.g. an inhibitor of C5a activity described herein
  • a pharmaceutical composition comprising the compound
  • the compound may be administered using intravenous infusion, an implantable osmotic pump, a transdermal patch, liposomes, or other modes of administration.
  • a pump may be used (see Sefton (1987) CRC Crit. Ref. Biomed. Eng. 14: 201; Buchwald et al. (1980) Surgery 88:507; Saudek et al. (1989) N. Eng. J. Med. 321: 574).
  • the compound in another embodiment, can be delivered in a vesicle, in particular a liposome (see Langer (1990) Science 249: 1527-1533; Treat et al. (1989) in Liposomes in the Therapy of Infectious Disease and Cancer, Lopez-Berestein and Fidler (eds.), Liss, N.Y., 353-365; WO 91/04014; U.S. 4,704,355).
  • a liposome see Langer (1990) Science 249: 1527-1533; Treat et al. (1989) in Liposomes in the Therapy of Infectious Disease and Cancer, Lopez-Berestein and Fidler (eds.), Liss, N.Y., 353-365; WO 91/04014; U.S. 4,704,355).
  • polymeric materials can be used (see Medical Applications of Controlled Release (1974) Langer and Wise (eds.), CRC Press: Boca Raton, Fla.; Controlled Drug Bioavailability, Drug Product Design and Performance, (1984) Smolen and Ball (eds.), Wiley: N.Y.; Ranger and Peppas (1953) J. Macromol. Sci. Rev. Macromol. Chem. 23: 61; see also Levy et al. (1985) Science 228: 190; During et al. (1989) Am. Neurol. 25: 351; Howard et al. (1989) J. Neurosurg. 71: 105).
  • a controlled release system can be placed in proximity of the therapeutic target, i.e., the target cells, tissue or organ, thus requiring only a fraction of the systemic dose (see, e.g., Goodson (1984) 115-138 in Medical Applications of Controlled Release, vol. 2).
  • Other controlled release systems are discussed in the review by Langer (1990, Science 249: 1527-1533).
  • a compound described herein e.g. an inhibitor of C5a activity described herein
  • a pharmaceutical composition comprising the compound locally to the area in need of treatment.
  • This may be achieved by, for example, and not by way of limitation, local infusion during surgery, topical application, e.g., in conjunction with a wound dressing after surgery, by injection, by means of a catheter, by means of a suppository, or by means of an implant, said implant being of a porous, non-porous, or gelatinous material, including membranes, such as silastic membranes, or fibers.
  • the preferred effective dose will be determined by a skilled artisan based upon considering several factors which will be known to one of ordinary skill in the art. Such factors include the particular form of the pharmaceutical composition, e.g. polypeptide or vector, and its pharmacokinetic parameters such as bioavailability, metabolism, half-life, etc., which will have been established during the usual development procedures typically employed in obtaining regulatory approval for a pharmaceutical compound. Further factors in considering the dose include the condition or disease to be prevented and or treated or the benefit to be achieved in a normal individual, the body mass of the patient, the route of administration, whether administration is acute or chronic, concomitant medications, and other factors well known to affect the efficacy of administered pharmaceutical agents. Thus the precise dosage should be decided according to the judgment of the practitioner and each patient's circumstances, e.g., depending upon the condition and the immune status of the individual patient, according to standard clinical techniques. EXAMPLES
  • Zymosan A was dissolved to 2 mg/ml in 50 ml sterile saline and boiled for 1 h at 100°C. After centrifugation, supernatant was discarded and the pellet was resuspended in 50 ml sterile saline. After a second centrifugation step, pellet was resuspended in 5 ml sterile saline to obtain a 20 mg/ml stock solution. Stock solution was aliquoted and stored at -20°C until use. To activate the plasma, Zymosan A stock solution and 100 ⁇ plasma were mixed and incubated at 37°C for 30 min. After incubation, tubes were centrifuged and the supernatant was aliquoted and stored at -20°C until use. 1.2 CD1 lb assay using rhC5a or ZAP as stimulants
  • Human whole blood was stimulated with rhC5a or ZAP.
  • the antibodies were diluted to final Ab/Ag molar ratios of 1: 1 and 0.5: 1.
  • IFX-1 was diluted to reach final Ab/Ag molar ratios of approximately 4: 1/3: 1/2: 1/1: 1/0.5: 1.
  • Blood only with buffer served as a non- stimulation control to assess the baseline CD l ib expression.
  • Blood with antibody alone was used to determine the effects on CD l ib expression of the antibody under non- stimulated condition.
  • the complete mixture (Ab/Ag/blood) was incubated at 37°C for 20 min to evaluate C5a- induced up-regulation of CD l ib.
  • After addition of anti-mouse CDl lb:FITC samples were incubated for 30 min on ice to minimize background staining.
  • Granulocytes were gated and mean fluorescence intensity (MFI) of FITC labeled (CD l ib expressing) granulocytes was examined by flow cytometer.
  • MFI mean fluorescence intensity
  • Human IL-8 ELISA was performed as recommended in the instruction manual under section 'Assay procedure" (eBioscience Inc., San Diego, CA). Briefly, coating was performed overnight at 4°C using 100 ⁇ lx capture antibody. Plates were blocked using 200 ⁇ lx assay diluents at RT for 1 h. Standard stock solutions were diluted with lx assay diluents to the desired concentration, followed by 6 serial 1:2 dilutions. Sample supernatants were diluted as required in lx assay diluents.
  • Purified anti-human C5a monoclonal antibody (InflaRx GmbH, Jena, Germany) was coated overnight with a final concentration of 0.5 ⁇ g/mL on the ELISA plate. After blocking with the assay diluent (lx PBS with 0.05% Tween 20 and 2% heat-inactivated FBS), calibration samples (recombinant human C5a, Sigma, Taufkirchen, Germany) and samples diluted in assay diluent were incubated for 90 minutes at room temperature.
  • assay diluent lx PBS with 0.05% Tween 20 and 2% heat-inactivated FBS
  • Mouse anti-human C5/C5a antibody clone 561 (Hycult Biotech, Uden, The Netherlands) diluted to 2 ⁇ g/mL in assay diluent was applied as the primary detection antibody for a 60-minute incubation at room temperature, followed by a 30-minute incubation with the secondary horseradish peroxidase labeled antibody (goat anti-mouse IgG2a polyclonal antibody, SouthernBiotech, Birmingham, USA) diluted to 0.05 ⁇ g/mL in assay diluent. Color development was performed with tetramethylbenzidine substrate solution (TMB, Biozol, Eching, Germany) and was stopped with 3.7 N sulfuric acid.
  • TMB tetramethylbenzidine substrate solution
  • the OD was read as the absorbance of 450 nm by Tecan Infinite® 200 reader with Tecan MagellanTM (Tecan Group, Maennedorf, Switzerland).
  • Tecan MagellanTM Tecan MagellanTM (Tecan Group, Maennedorf, Switzerland).
  • the in-house developed C5a ELISA was validated according to the EMA guideline on bioanalytical method validation.
  • Intra-assay and inter-assay precision tested with five different concentrations showed a coefficient of variance (CV) of 0.65 % to 4.96 % and 1.50 % to 4.88 % for six and 18 repetitions, respectively.
  • CV coefficient of variance
  • Recovery analysis of the spiked recombinant human C5a in buffer resulted in recoveries of 86.98 + 1.20 % (mean + SD) at the lower limit of quantification and 91.50 + 3.29 % at the upper limit of quantification.
  • No cross-reactivity for C3, C3a and C4 and cross- reactivity of ⁇ 0.01% for C5b-6 was detected.
  • Human IgG4 antibodies did not interfere with the assay.
  • the mean C5a level in citrate plasma from 20 human volunteers is 17.08 ng/mL + 6.96 ng/mL with a range from 7.52 ng/mL to 30.17 ng/mL.
  • C3a ELISA (BD OptEIATM Human C3a ELISA Kit, BD Bioscience, Germany) was conducted according to the manufacturer instruction.
  • C5b-9 concentration was determined using the C5b-9 ELISA validated by InflaRx based on the BD OptEIATM Human C5b-9 ELISA Set (BD Bioscience).
  • C5a concentration was measured using the C5a ELISA established and validated by InflaRx described above.
  • CD1 lb up-regulation is a sensitive hallmark for neutrophil activation
  • CD1 lb levels on neutrophils were employed to evaluate the neutrophil activation.
  • the human whole blood model was used to assess the blocking activity of IFX-1 to recombinant human C5a (rhC5a) in this study. Human whole blood was incubated with buffer, antibody alone, rhC5a alone, or combinations of different antibody concentration and rhC5a. After incubation, cells were stained with anti- mouse CDl lb:FITC and CD l ib MFI was analysed by flow cytometry checking for the activation levels of blood neutrophils.
  • zymosan-activated plasma As a source for endogenous C5a (eC5a), zymosan-activated plasma (ZAP) was used to stimulate the blood neutrophils. The amount of eC5a in ZAP was measured using a commercial C5a ELISA Kit. The data presented here ( Figure 2) point out that eC5a in ZAP induced comparable levels of CD l ib up-regulation to rhC5a. The presence of IFX-1 significantly decreased the CD l ib expression on human neutrophils, even at an Ab:Ag molar ratio of 0.5: 1. The overall blocking activity of IFX-1 to ZAP-induced CD l ib up-regulation ranged from 100% to 82% depending on the Ab:Ag ratio.
  • IFX-1 could specifically block CD l ib upregulation up to 100%. It can therefore be concluded that eC5a is the sole driver for neutrophil activation upon ZAP stimulation, and IFX-1 can completely block it.
  • Zymosan A as an active fungus cell wall component, can induce strong inflammatory responses in human whole blood as characterized by the activation of neutrophils with elevated cytokines and chemokines levels.
  • human whole blood was spiked with zymosan A in the presence or absence of IFX-1, and CD l ib expression on blood neutrophils was measured by flow cytometric analysis.
  • CD1 lb on blood neutrophils was strongly upregulated at the presence of zymosan in human whole blood.
  • the zymosan- stimulated CDl lb upregulation can be suppressed by 79% - 93% depending on the concentration of IFX-1 added.
  • the CDl lb up-regulation stimulated by rhC5a was 100% blocked by IFX-1. Therefore, it is affirmative that the CDl lb up-regulation on blood neutrophils upon zymosan A stimulation is caused primarily by eC5a. In addition, it can be concluded that eC5a, once generated in the whole blood by zymosan A, binds to IFX-1 first, thereby blocking its access to its natural receptors.
  • IL-8 levels were measured and used to assess the inflammatory response.
  • IL-8 concentrations after various doses of zymosan A stimulation ranged from 458 pg/ml to 3218 pg/ml in the absence of IFX-1.
  • the presence of IFX-1 significantly reduced IL-8 generation upon stimulation with various concentrations of zymosan A and the reduction rate up to 54% was observed.
  • zymosan-induced inflammatory responses are largely dependent on the presence of C5a.
  • Circulating concentrations of complement factors C3a and C5a as well as membrane attack complex sC5b-9 were determined in the plasma of 54 patients and of 14 healthy controls as well as in the pus of seven patients. As shown in Figure 5, circulating C5a was significantly greater in patient plasma than in control plasma (P ⁇ 0.01), and the differences of C3a and C5b- 9 between patients and controls were of similar significance. Therefore, it can be concluded that systemic complement activation occurs in HS. Given the essential role of complement activation in the innate and adaptive immunity, the inventors assumed that targeting of complement activation could be a new therapeutic strategy for the treatment of HS.
  • the HS plasma samples with high levels of C5a were chosen and assessed by employing the human whole blood model. As shown in Figure 6, in contrast to the control plasma samples with low C5a levels (Ctrl 008 and Ctrl 012), HS plasma samples (Pat. 088 and Pat. 092) with high levels of C5a strongly upregulated CDl lb expression on blood neutrophils. Recombinant human C5a was used as the positive control, while the plasma from healthy volunteers was chosen as the negative control.
  • CDl lb upregulation induced by HS plasma can be 100% suppressed by IFX-1, indicating that C5a is the most important activator in the HS plasma to initiate neutrophil activation. From these novel results the inventors concluded that blockade of C5a in HS patients is sufficient to achieve a strong suppression on neutrophil activation.
  • IFX-1 Primary objective of the trial was to explore the safety and tolerability of IFX-1 administered over 8 weeks. Secondary objectives of the trial were to assess the pharmacokinetics and pharmacodynamics of IFX- 1 as well as to generate preliminary data on the efficacy of IFX-1 on clinical endpoints (e.g., HiSCR, DLQI, VAS for disease status, VAS for pain, HS-PGA, modified Sartorius Score) to generate further hypotheses.
  • clinical endpoints e.g., HiSCR, DLQI, VAS for disease status, VAS for pain, HS-PGA, modified Sartorius Score
  • a primary failure is defined as an at least 12 week treatment with a biological compound without effect and a secondary failure as achieving an initial response after at least 12 week treatment with a biological compound followed by a relapse.
  • corticosteroids defined as daily intake of prednisone or equivalent more than 1 mg/kg for the last three weeks; 9. Intake of immunosuppressive drugs within the past 30 days (e.g., cyclosporine, tacrolimus)
  • IFX-1 is well tolerated by HS patients. There were no drug-related serious adverse events reported over the treatment period.
  • HiSCR Hidradenitis Suppurativa Clinical Response
  • HiSCR is defined by the status of three types of lesions (defining criteria): abscesses (fluctuant, with or without drainage, tender or painful), inflammatory nodules (tender, erythematous, pyogenic granuloma lesion) and draining fistulas (sinus tracts, with communications to skin surface, draining purulent fluid).
  • the proposed definition of responders to treatment is: (i) at least a 50% reduction in ANs, (ii) no increase in the number of abscesses, and (iii) no increase in the number of draining fistulas from baseline.
  • HiSCR has been validated recently as a responsive and clinically meaningful endpoint of the inflammatory manifestation of HS (Kimball and others, 2014).
  • Humira EMA assessment report :
  • Photographic documentation of the affected areas confirmed these findings by a highly reduced inflammation on the skin, as evidenced by the visual reduction of inflammatory swollenness and redness post treatment.
  • anti-C5a represents a powerful anti-inflammatory agent in the disease setting of HS.
  • This clinical finding demonstrates that blockade of C5a is highly effective to reduce the activation of neutrophils thereby effectively alleviating cutaneous neutrophilic inflammatory disorders. 4. BLOCKING THE CDIIB UPREGULATION INDUCED BY ACTIVATED COMPLEMENT FACTOR IN HLDRADENITIS SUPPURATIVA PATIENTS VIA C5A-C5AR AXIS INHIBITION
  • CD l ib also known as integrin alpha M
  • integrin alpha M integrin alpha M
  • Enhanced expression of CDl lb/CD18 therefore reflects an inflammatory triggering event.
  • the human CD l ib assay is conducted using flow cytometry to detect FITC-conjugated anti-CD l ib antibody on the surface of neutrophils.
  • Activated complement products, especially elevated endogenous C5a (eC5a) in HS patient plasma samples can strongly upregulate CD l ib expression through the binding of C5a to its receptor C5aR (CD88) on neutrophils. Consequently, blockade of the C5a-C5aR axis is expected to abolish or attenuate CD1 lb upregulation on the surface of neutrophils.
  • C5a exerts its effects through interacting with the high-affinity C5a receptors (C5aR and C5L2) (Guo and Ward, 2005).
  • C5aR belongs to the rhodopsin family of G-protein-coupled receptors with seven transmembrane segments, while C5L2 is not G-protein-coupled. It is generally understood that C5a-C5aR signaling is very important in the pathogenesis of proinflammatory outcomes (Ward, 2009). Therefore, targeting the C5aR is another strategy for inhibiting complement-dependent inflammatory diseases.
  • a series of small molecules derived from the C-terminus of C5a were developed as C5aR antagonists.
  • the lead compound cyclic hexapeptide PMX-53 (AcF-[OP(D-Cha)WR]) (Finch et al., 1999) was shown to attenuate injury in numerous animal models of inflammation following intravenous, subcutaneous, intraperitoneal, and oral administration (Proctor et al. 2006). With their structural similarity to C5a, such antagonists compete with C5a for the C5a receptors on neutrophils (March et al., 2004). In addition, anti-C5aR antibodies could block the binding of C5a to C5aR, thereby reducing the accumulation and activation of myeloid-derived suppressor cells and neutrophils (Markiewski et al., 2008).
  • Avacopan (CCX168) is an orally-administered small molecule drug candidate that selectively inhibits the complement C5a receptor (C5aR), and is being developed for inflammatory and autoimmune diseases (Bekker et al., 2008; Jayne et al., 2017)
  • Pat. 092 plasma 70.01 8801.00 276.12
  • o Staining buffer 1% heat-inactivated FBS + 0.1% sodium azide in lx PBS solution ⁇ FBS, Thermo Fisher Scientific (Darmstadt, Germany), Cat. No. 10099133
  • Red blood cells were then lysed with lx FACS Lysing solution at room temperature for 10 min. 2 mL Staining buffer was used to wash the remaining cells twice. After centrifugation at 2500 rpm for 3 min, cells were resuspended in 0.5 mL Staining buffer and ready for FACS analysis. A gate was set on the FSC vs. SSC plot to allow analysis only of cells with the size of neutrophils.
  • the percentage of blocking activity (BA) of the C5a-C5aR axis blockade test material was calculated using the formula below. Where MFI is the mean fluorescence intensity emitted from the CDl lb-bound FITC on neutrophils.
  • BA (%) (MFI Patient plasma -MFI Test material spiked patient plasma) ⁇ (MFI Patient plasma MFI huPp) X 100 b) Statistical analysis
  • CDl lb expression on neutrophils was evaluated with plasma samples from two healthy blood donors (Ctrl 009 and Ctrl 010) and two diagnosed HS patients (Pat. 088 and Pat. 092).
  • the mean fluorescence intensity (MFI) of the Ctrl-samples was 2156.9+114.3, which falls within the non- stimulated CDl lb baseline expression range (MFI ⁇ 3500).
  • MFI mean fluorescence intensity
  • rhC5a 20 nM recombinant human C5a
  • Keratinocytes and neutrophils are important sources of proinflammatory molecules in hidradenitis suppurativa. Br J Dermatol 174(3):514-521.
  • Complement C5a regulates IL-17 by affecting the crosstalk between DC and gammadelta T cells in CLP-induced sepsis. Eur J Immunol 40(4): 1079- 1088.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Public Health (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Veterinary Medicine (AREA)
  • General Chemical & Material Sciences (AREA)
  • Immunology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Dermatology (AREA)
  • Molecular Biology (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Rheumatology (AREA)
  • Pain & Pain Management (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Compounds Of Unknown Constitution (AREA)

Abstract

La présente invention concerne des inhibiteurs de l'activité de C5a et leur utilisation dans le traitement de maladies cutanées, neutrophiles, inflammatoires chez un sujet.
PCT/EP2018/065676 2017-06-23 2018-06-13 Traitement de maladies inflammatoires par des inhibiteurs de l'activité de c5a WO2018234118A1 (fr)

Priority Applications (9)

Application Number Priority Date Filing Date Title
SG11201912882QA SG11201912882QA (en) 2017-06-23 2018-06-13 Treatment of inflammatory diseases with inhibitors of c5a activity
KR1020197038211A KR20200020727A (ko) 2017-06-23 2018-06-13 C5a 활성의 억제제를 사용한 염증 질병의 치료
AU2018286754A AU2018286754A1 (en) 2017-06-23 2018-06-13 Treatment of inflammatory diseases with inhibitors of C5A activity
JP2019570986A JP2020524696A (ja) 2017-06-23 2018-06-13 C5a活性のインヒビターでの炎症性疾患の処置
CN201880041303.4A CN111201241A (zh) 2017-06-23 2018-06-13 用C5a活性抑制剂治疗炎性疾病
EA201992673A EA201992673A1 (ru) 2017-09-07 2018-06-13 ЛЕЧЕНИЕ ВОСПАЛИТЕЛЬНЫХ ЗАБОЛЕВАНИЙ С ПОМОЩЬЮ ИНГИБИТОРОВ АКТИВНОСТИ C5a
EP18732016.3A EP3642230A1 (fr) 2017-06-23 2018-06-13 Traitement de maladies inflammatoires par des inhibiteurs de l'activité de c5a
CA3066689A CA3066689C (fr) 2017-06-23 2018-06-13 Traitement de maladies inflammatoires par des inhibiteurs de l'activite de c5a
IL271074A IL271074A (en) 2017-06-23 2019-12-01 Treatment of inflammatory diseases with c5a activity inhibitors

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
EP17177657 2017-06-23
EP17177657.8 2017-06-23
EP17189938.8 2017-09-07
EP17189938 2017-09-07

Publications (1)

Publication Number Publication Date
WO2018234118A1 true WO2018234118A1 (fr) 2018-12-27

Family

ID=62636193

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2018/065676 WO2018234118A1 (fr) 2017-06-23 2018-06-13 Traitement de maladies inflammatoires par des inhibiteurs de l'activité de c5a

Country Status (10)

Country Link
EP (1) EP3642230A1 (fr)
JP (1) JP2020524696A (fr)
KR (1) KR20200020727A (fr)
CN (1) CN111201241A (fr)
AU (1) AU2018286754A1 (fr)
CA (1) CA3066689C (fr)
IL (1) IL271074A (fr)
SG (1) SG11201912882QA (fr)
TW (1) TWI786132B (fr)
WO (1) WO2018234118A1 (fr)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10633434B2 (en) 2016-06-14 2020-04-28 Regeneron Pharmaceuticals, Inc. Anti-C5 antibodies
WO2020182974A1 (fr) * 2019-03-14 2020-09-17 Morphosys Ag Anticorps ciblant c5ar
WO2021146320A1 (fr) * 2020-01-13 2021-07-22 Visterra, Inc. Molécules d'anticorps anti-c5ar1 et leurs utilisations
WO2021259160A1 (fr) * 2020-06-24 2021-12-30 Staidson (Beijing) Biopharmaceuticals Co., Ltd. Anticorps reconnaissant de manière spécifique c5a et leurs utilisations
US11365265B2 (en) 2017-12-13 2022-06-21 Regeneron Pharmaceuticals, Inc. Anti-C5 antibody combinations and uses thereof
US11912781B2 (en) 2021-01-13 2024-02-27 Visterra, Inc. Humanized complement 5A receptor 1 antibodies and methods of use thereof

Citations (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4704355A (en) 1985-03-27 1987-11-03 New Horizons Diagnostics Corporation Assay utilizing ATP encapsulated within liposome particles
WO1991004014A1 (fr) 1989-09-21 1991-04-04 Synergen, Inc. Procede de transport de compositions a travers la barriere hemato-encephalique
WO1999000406A1 (fr) 1997-06-25 1999-01-07 The University Of Queensland AGONISTES ET ANTAGONISTES CYCLIQUES DE RECEPTEURS DE C5a ET DE RECEPTEURS COUPLES A LA PROTEINE G
US5939598A (en) 1990-01-12 1999-08-17 Abgenix, Inc. Method of making transgenic mice lacking endogenous heavy chains
WO2003033528A1 (fr) 2001-10-17 2003-04-24 University Of Queensland Peptides cycliques comme antagonistes du recepteur couple a la proteine g
US20030118592A1 (en) 2001-01-17 2003-06-26 Genecraft, Inc. Binding domain-immunoglobulin fusion proteins
US20030133939A1 (en) 2001-01-17 2003-07-17 Genecraft, Inc. Binding domain-immunoglobulin fusion proteins
WO2005079363A2 (fr) 2004-02-12 2005-09-01 Archemix Corporation Agents therapeutiques a base d'aptameres utiles dans le traitement de troubles lies a un complement
WO2008009062A1 (fr) 2006-07-21 2008-01-24 Promics Pty Ltd Traitement de l'hyperplasie intimale et d'affections associées
WO2008029167A1 (fr) 2006-09-08 2008-03-13 Varleigh Limited Procédé de traitement de troubles du nerf périphérique
WO2010075257A1 (fr) 2008-12-22 2010-07-01 Chemocentryx, Inc. Antagonistes de c5ar
WO2011063980A1 (fr) 2009-11-26 2011-06-03 Inflarx Gmbh Fractions de liaison anti-c5a à activité bloquante élevée
WO2011163640A1 (fr) 2010-06-24 2011-12-29 Chemocentryx, Inc. Antagonistes de c5ar
WO2015140304A1 (fr) 2014-03-20 2015-09-24 Inflarx Gmbh Inhibiteurs de c5a pour le traitement de la pneumonie virale
WO2016044419A1 (fr) 2014-09-16 2016-03-24 Alnylam Pharmaceuticals, Inc. Compositions d'arni de composant du complément c5 et leurs procédés d'utilisation
WO2016061066A1 (fr) 2014-10-15 2016-04-21 Alexion Pharmaceuticals, Inc. Procedes de replication de culture cellulaire de production d'eculizumab a grande echelle
WO2016102877A1 (fr) * 2014-12-23 2016-06-30 Galderma Research & Development Nouveaux composés antagonistes des récepteurs cxcr1 et cxcr2 aux chimiokines, et leur utilisation dans le traitement de pathologies médiées par des chimiokines
WO2016209956A1 (fr) 2015-06-26 2016-12-29 Alexion Pharmaceuticals, Inc. Procédé de traitement d'un patient soumis à une vaccination avec l'éculizumab ou un variant de l'éculizumab
WO2017176620A2 (fr) 2016-04-04 2017-10-12 Chemocentryx, Inc. Antagonistes de c5ar solubles

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU2005226792A1 (en) * 2004-03-26 2005-10-06 Promics Pty Limited Treatment of neurological conditions using complement C5a receptor modulators
GB0905790D0 (en) * 2009-04-03 2009-05-20 Alligator Bioscience Ab Novel polypeptides and use thereof
SG11201507400SA (en) * 2013-03-14 2015-10-29 Alnylam Pharmaceuticals Inc Complement component c5 irna compositions and methods of use thereof
DK2994488T3 (da) * 2013-05-08 2022-10-03 Novo Nordisk As Anvendelse af c5ar-antagonister
WO2018184739A1 (fr) * 2017-04-03 2018-10-11 Inflarx Gmbh Traitement de maladies inflammatoires par des inhibiteurs de l'activité de c5a

Patent Citations (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4704355A (en) 1985-03-27 1987-11-03 New Horizons Diagnostics Corporation Assay utilizing ATP encapsulated within liposome particles
WO1991004014A1 (fr) 1989-09-21 1991-04-04 Synergen, Inc. Procede de transport de compositions a travers la barriere hemato-encephalique
US5939598A (en) 1990-01-12 1999-08-17 Abgenix, Inc. Method of making transgenic mice lacking endogenous heavy chains
WO1999000406A1 (fr) 1997-06-25 1999-01-07 The University Of Queensland AGONISTES ET ANTAGONISTES CYCLIQUES DE RECEPTEURS DE C5a ET DE RECEPTEURS COUPLES A LA PROTEINE G
US20030118592A1 (en) 2001-01-17 2003-06-26 Genecraft, Inc. Binding domain-immunoglobulin fusion proteins
US20030133939A1 (en) 2001-01-17 2003-07-17 Genecraft, Inc. Binding domain-immunoglobulin fusion proteins
WO2003033528A1 (fr) 2001-10-17 2003-04-24 University Of Queensland Peptides cycliques comme antagonistes du recepteur couple a la proteine g
WO2005079363A2 (fr) 2004-02-12 2005-09-01 Archemix Corporation Agents therapeutiques a base d'aptameres utiles dans le traitement de troubles lies a un complement
WO2008009062A1 (fr) 2006-07-21 2008-01-24 Promics Pty Ltd Traitement de l'hyperplasie intimale et d'affections associées
WO2008029167A1 (fr) 2006-09-08 2008-03-13 Varleigh Limited Procédé de traitement de troubles du nerf périphérique
WO2010075257A1 (fr) 2008-12-22 2010-07-01 Chemocentryx, Inc. Antagonistes de c5ar
WO2011063980A1 (fr) 2009-11-26 2011-06-03 Inflarx Gmbh Fractions de liaison anti-c5a à activité bloquante élevée
US20120231008A1 (en) 2009-11-26 2012-09-13 Renfeng Guo Anti-C5A Binding Moieties with High Blocking Activity
WO2011163640A1 (fr) 2010-06-24 2011-12-29 Chemocentryx, Inc. Antagonistes de c5ar
WO2015140304A1 (fr) 2014-03-20 2015-09-24 Inflarx Gmbh Inhibiteurs de c5a pour le traitement de la pneumonie virale
WO2016044419A1 (fr) 2014-09-16 2016-03-24 Alnylam Pharmaceuticals, Inc. Compositions d'arni de composant du complément c5 et leurs procédés d'utilisation
WO2016061066A1 (fr) 2014-10-15 2016-04-21 Alexion Pharmaceuticals, Inc. Procedes de replication de culture cellulaire de production d'eculizumab a grande echelle
WO2016102877A1 (fr) * 2014-12-23 2016-06-30 Galderma Research & Development Nouveaux composés antagonistes des récepteurs cxcr1 et cxcr2 aux chimiokines, et leur utilisation dans le traitement de pathologies médiées par des chimiokines
WO2016209956A1 (fr) 2015-06-26 2016-12-29 Alexion Pharmaceuticals, Inc. Procédé de traitement d'un patient soumis à une vaccination avec l'éculizumab ou un variant de l'éculizumab
WO2017176620A2 (fr) 2016-04-04 2017-10-12 Chemocentryx, Inc. Antagonistes de c5ar solubles

Non-Patent Citations (82)

* Cited by examiner, † Cited by third party
Title
"Drug Product Design and Performance", 1984, WILEY, article "Controlled Drug Bioavailability"
"Helvetica Chimica Acta", 1995, article "A multilingual glossary of biotechnological terms: (IUPAC Recommendations"
"Medical Applications of Controlled Release", 1974, CRC PRESS
"UniProtKB", Database accession no. P01031
ABI ABDALLAH DS; EGAN CE; BUTCHER BA; DENKERS EY: "Mouse neutrophils are professional antigen-presenting cells programmed to instruct Th1 and Thl7 T-cell differentiation", INT IMMUNOL, vol. 23, no. 5, 2011, pages 317 - 326
ALMAGRO; FRANSSON, FRONTIERS IN BIOSCIENCE, vol. 13, 2008, pages 1619 - 1633
ARUN CUMPELIK ET AL: "Neutrophil microvesicles resolve gout by inhibiting C5a-mediated priming of the inflammasome", ANNALS OF THE RHEUMATIC DISEASES, vol. 75, no. 6, 5 August 2015 (2015-08-05), GB, pages 1236 - 1245, XP055454249, ISSN: 0003-4967, DOI: 10.1136/annrheumdis-2015-207338 *
BEKKER, P. ET AL.: "Characterization of Pharmacologic and Pharmacokinetic Properties of CCX168, a Potent and Selective Orally Administered Complement 5a Receptor Inhibitor, Based on Preclinical Evaluation and Randomized Phase 1 Clinical Study", PLOS ONE, vol. 11, 2016, pages e0164646
BINZ H.K. ET AL.: "Engineering novel binding proteins from nonimmunoglobulin domains", NAT. BIOTECHNOL., vol. 23, no. 10, 2005, pages 1257 - 1268, XP002381839, DOI: doi:10.1038/nbt1127
BIRD ET AL., SCIENCE, vol. 242, 1988, pages 423 - 426
BRAUN-FALCO M; KOVNERYSTYY O; LOHSE P; RUZICKA T: "Pyoderma gangrenosum, acne, and suppurative hidradenitis (PASH)--a new autoinflammatory syndrome distinct from PAPA syndrome", J AM ACAD DERMATOL, vol. 66, no. 3, 2012, pages 409 - 415, XP055059087, DOI: doi:10.1016/j.jaad.2010.12.025
BRODY E.N.; GOLD L.: "Aptamers as therapeutic and diagnostic agents", J. BIOTECHNOL., vol. 74, no. 1, 2000, pages 5 - 13, XP009031105
BUCHWALD ET AL., SURGERY, vol. 88, 1980, pages 507
CARLOS, T. M.; HARLAN, J. M.: "Membrane proteins involved in phagocyte adherence to endothelium", IMMUNOL REV, vol. 114, 1990, pages 5 - 28, XP001008136, DOI: doi:10.1111/j.1600-065X.1990.tb00559.x
CLINICALTRIALS.GOV: "Archive History for NCT03001622: Studying Complement Inhibition in Patients With Moderate to Severe Hidradenitis Suppurativa", 20 March 2017 (2017-03-20), XP055489573, Retrieved from the Internet <URL:https://clinicaltrials.gov/ct2/history/NCT03001622?V_3=View#StudyPageTop> [retrieved on 20180702] *
CUGNO M; BORGHI A; MARZANO AV: "PAPA, PASH and PAPASH Syndromes: Pathophysiology, Presentation and Treatment", AM J CLIN DERMATOL., 2017
CUGNO M; GUALTIEROTTI R; MERONI PL; MARZANO AV: "Inflammatory Joint Disorders and Neutrophilic Dermatoses: a Comprehensive Review", CLINIC REV ALLERG IMMUNOL, vol. 54, 2018, pages 269 - 281, XP036468720, DOI: doi:10.1007/s12016-017-8629-0
CUGNO MASSIMO ET AL: "PAPA, PASH and PAPASH Syndromes: Pathophysiology, Presentation and Treatment", AMERICAN JOURNAL OF CLINICAL DERMATOLOGYNEW ZEALANDAPR 2017, ADIS INTERNATIONAL LTD, NZ, vol. 18, no. 4, 25 February 2017 (2017-02-25), pages 555 - 562, XP009503738, ISSN: 1179-1888, DOI: 10.1007/S40257-017-0265-1 *
CZERMAK BJ; SARMA V; PIERSON CL; WARNER RL; HUBER-LANG M; BLESS NM; SCHMAL H; FRIEDL HP; WARD PA: "Protective effects of C5a blockade in sepsis", NAT MED, vol. 5, no. 7, 1999, pages 788 - 792
DURING ET AL., ANN. NEUROL., vol. 25, 1989, pages 351
EQS GROUP AG ET AL: "InflaRx initiates exploratory phase II trial with IFX-1, a first-in-class anti-complement C5a antibody, in patients with Hidradenitis Suppurativa - dgap.de", 4 January 2017 (2017-01-04), XP055425518, Retrieved from the Internet <URL:http://www.dgap.de/dgap/News/corporate/inflarx-initiates-exploratory-phase-trial-with-ifx-firstinclass-anticomplement-antibody-patients-with-hidradenitis-suppurativa/?newsID=980687> [retrieved on 20171115] *
FINCH, A. M. ET AL.: "Low-molecular-weight peptidic and cyclic antagonists of the receptor for the complement factor C5a", J MED CHEM, vol. 42, 1999, pages 1965 - 1974, XP002137173, DOI: doi:10.1021/jm9806594
GOODSON, MEDICAL APPLICATIONS OF CONTROLLED RELEASE, vol. 2, 1984, pages 115 - 138
GRAILLE J ET AL: "PAPA, PASH, PAPASH, PsAPASH, PASS... des syndromes auto-inflammatoires PAS si simples", REVUE DE MEDECINE INTERNE, vol. 36, December 2015 (2015-12-01), XP029315654, ISSN: 0248-8663, DOI: 10.1016/J.REVMED.2015.10.218 *
GUO RF; RIEDEMANN NC; SUN L; GAO H; SHI KX; REUBEN JS; SARMA VJ; ZETOUNE FS; WARD PA: "Divergent signaling pathways in phagocytic cells during sepsis", J IMMUNOL, vol. 177, no. 2, 2006, pages 1306 - 1313
GUO RF; WARD PA: "Role of C5a in inflammatory responses", ANNU REV IMMUNOL, vol. 23, 2005, pages 821 - 852
HARDING J: "Eculizumab", DRUGS OF THE FU, PROUS SCIENCE, ES, vol. 29, no. 7, 1 July 2004 (2004-07-01), pages 673 - 676, XP008113000, ISSN: 0377-8282, DOI: 10.1358/DOF.2004.029.07.819330 *
HAVVA OZGE KESEROGLU ET AL: "A Case of Subcorneal Pustular Dermatosis Successfully Treated with Acitretin", ARCHIVES OF INFLAMMATION, vol. 1, no. 2, 27 October 2016 (2016-10-27), France, pages 1 - 3, XP055489858 *
HEAP C.J. ET AL.: "Analysis of a 17-amino acid residue, virus-neutralizing microantibody", J. GEN. VIROL., vol. 86, 2005, pages 1791 - 1800, XP002506464, DOI: doi:10.1099/VIR.080812-0
HOLLIGER P. ET AL., PROC. NATL. ACAD. SCI. U.S.A., vol. 90, no. 14, 1993, pages 6444 - 6448
HOWARD ET AL., J. NEUROSURG., vol. 71, 1989, pages 105
HUBER-LANG MS; SARMA JV; MCGUIRE SR; LU KT; GUO RF; PADGAONKAR VA; YOUNKIN EM; LAUDES U; RIEDEMANN NC; YOUNGER JG: "Protective effects of anti-C5a peptide antibodies in experimental sepsis", FASEB J, vol. 15, no. 3, 2001, pages 568 - 570
HUBER-LANG MS; YOUNKIN EM; SARMA JV; MCGUIRE SR; LU KT; GUO RF; PADGAONKAR VA; CURNUTTE JT; ERICKSON R; WARD PA: "Complement-induced impairment of innate immunity during sepsis", J IMMUNOL, vol. 169, no. 6, 2002, pages 3223 - 3231
HUSTON ET AL., PROC. NATL. ACAD. SCI. USA, vol. 85, 1988, pages 5879 - 5883
HYZEWICZ J; TANIHATA J; KURAOKA M; NITAHARA-KASAHARA Y; BEYLIER T; RUEGG UT; VATER A; TAKEDA S: "Low-Intensity Training and the C5a Complement Antagonist NOX-D21 Rescue the mdx Phenotype through Modulation of Inflammation", AM. J. PATHOL., vol. 187, no. 5, 2017, pages 1147 - 1161
JAYNE, D. R. W. ET AL.: "Randomized Trial of C5a Receptor Inhibitor Avacopan in ANCA-Associated Vasculitis", J AM SOC NEPHROL, vol. 28, 2017, pages 2756 - 2767
JEMEC GB: "Medical treatment of hidradenitis suppurativa", EXPERT OPIN PHARMACOTHER, vol. 5, no. 8, 2004, pages 1767 - 1770
JEMEC GB; HEIDENHEIM M; NIELSEN NH: "The prevalence of hidradenitis suppurativa and its potential precursor lesions", J AM ACAD DERMATOL, vol. 35, no. 2, 1996, pages 191 - 194
JORIZZO J L ET AL: "Low-dose weekly methotrexate for unusual neutrophilic vascular reactions: Cutaneous polyarteritis nodosa and Behcets disease", JOURNAL OF THE AMERICAN ACADEMY OF DERMATOLOGY, MOSBY, INC, US, vol. 24, no. 6, 1 June 1991 (1991-06-01), pages 973 - 978, XP023658556, ISSN: 0190-9622, [retrieved on 19910601], DOI: 10.1016/0190-9622(91)70156-V *
KAPLAN M: "ECULIZUMAB", CURRENT OPINION IN INVESTIGATIONAL D, PHARMAPRESS, US, vol. 3, no. 7, 1 July 2002 (2002-07-01), pages 1017 - 1023, XP009045356, ISSN: 1472-4472 *
KAPLAN MJ: "Role of neutrophils in systemic autoimmune diseases", ARTHRITIS RES THER, vol. 15, no. 5, 2013, pages 219
KIMBALL AB; JEMEC GB; YANG M; KAGELEIRY A; SIGNOROVITCH JE; OKUN MM; GU Y; WANG K; MULANI P; SUNDARAM M: "Assessing the validity, responsiveness and meaningfulness of the Hidradenitis Suppurativa Clinical Response (HiSCR) as the clinical endpoint for hidradenitis suppurativa treatment", BR J DERMATOL, vol. 171, no. 6, 2014, pages 1434 - 1442
KLOS, A. ET AL.: "The role of the anaphylatoxins in health and disease", MOL IMMUNOL, vol. 46, 2009, pages 2753 - 2766, XP026438375
KURZEN H; KUROKAWA I; JEMEC GB; EMTESTAM L; SELLHEYER K; GIAMARELLOS-BOURBOULIS EJ; NAGY I; BECHARA FG; SARTORIUS K; LAPINS J: "What causes hidradenitis suppurativa?", EXP DERMATOL, vol. 17, no. 5, 2008, pages 455 - 456
LANGER, SCIENCE, vol. 249, 1990, pages 1527 - 1533
LARSON, R. S.; SPRINGER, T. A.: "Structure and function of leukocyte integrins", IMMUNOL REV, vol. 114, 1990, pages 181 - 217
LEVY ET AL., SCIENCE, vol. 228, 1985, pages 190
LIMA AL; KARL I; GINER T; POPPE H; SCHMIDT M; PRESSER D; GOEBELER M; BAUER B: "Keratinocytes and neutrophils are important sources of proinflammatory molecules in hidradenitis suppurativa", BR J DERMATOL, vol. 174, no. 3, 2016, pages 514 - 521
LIN DANG ET AL: "Role of the complement anaphylatoxin C5a-receptor pathway in atopic dermatitis in mice", MOLECULAR MEDICINE REPORTS, vol. 11, no. 6, 4 February 2015 (2015-02-04), GR, pages 4183 - 4189, XP055426905, ISSN: 1791-2997, DOI: 10.3892/mmr.2015.3301 *
MARCH, D. R. ET AL.: "Potent cyclic antagonists of the complement C5a receptor on human polymorphonuclear leukocytes. Relationships between structures and activity", MOL PHARMACOL, vol. 65, 2004, pages 868 - 879, XP001205138, DOI: doi:10.1124/mol.65.4.868
MARIA ARGYROPOULOU ET AL: "AN OPEN-LABEL TRIAL TO ASSESS THE SAFETY OF IFX-1 IN PATIENTS WITH HIDRADENITIS SUPPURATIVA NOT ELIGIBLE FOR ADALIMUMAB", 4 August 2017 (2017-08-04), XP055425520, Retrieved from the Internet <URL:http://www.inflarx.de/dam/jcr:8dab6fc6-e244-4a70-a473-b982f9e59b59/Argyropoulou_2017_poster.pdf> [retrieved on 20171115] *
MARKIEWSKI, M. M. ET AL.: "Modulation of the antitumor immune response by complement", NAT IMMUNOL, vol. 9, 2008, pages 1225 - 1235
MARZANO AV: "Hidradenitis suppurativa, neutrophilic dermatoses and autoinflammation: what's the link?", BR J DERMATOL, vol. 174, no. 3, 2016, pages 482 - 483
MARZANO AV; CECCHERINI I; GATTORNO M; FANONI D; CAROLI F; RUSMINI M; GROSSI A; DE SIMONE C; BORGHI OM; MERONI PL: "Association of pyoderma gangrenosum, acne, and suppurative hidradenitis (PASH) shares genetic and cytokine profiles with other autoinflammatory diseases", MEDICINE (BALTIMORE, vol. 93, no. 27, 2014, pages e187
NAVARINI ALEXANDER A ET AL: "Neutrophilic dermatoses and autoinflammatory diseases with skin involvement-innate immune disorders", SEMINARS IN IMMUNOPATHOLOGY, SPRINGER BERLIN / HEIDELBERG, DE, vol. 38, no. 1, 30 November 2015 (2015-11-30), pages 45 - 56, XP035873042, ISSN: 1863-2297, [retrieved on 20151130], DOI: 10.1007/S00281-015-0549-6 *
NEMETH T; MOCSAI A: "The role of neutrophils in autoimmune diseases", IMMUNOL LETT, vol. 143, no. 1, 2012, pages 9 - 19, XP028475856, DOI: doi:10.1016/j.imlet.2012.01.013
NEMETH T; MOCSAI A; LOWELL CA: "Neutrophils in animal models of autoimmune disease", SEMIN IMMUNOL, vol. 28, no. 2, 2016, pages 174 - 186, XP029532877, DOI: doi:10.1016/j.smim.2016.04.001
NIKHIL DHINGRA ET AL: "Attenuated neutrophil axis in atopic dermatitis compared to psoriasis reflects TH17 pathway differences between these diseases", JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY, vol. 132, no. 2, 1 August 2013 (2013-08-01), AMSTERDAM, NL, pages 498 - 501.e3, XP055489537, ISSN: 0091-6749, DOI: 10.1016/j.jaci.2013.04.043 *
PAWARIA S; RAMANI K; MAERS K; LIU Y; KANE LP; LEVESQUE MC; BISWAS PS: "Complement component C5a permits the coexistence of pathogenic Thl7 cells and type I IFN in lupus", J IMMUNOL, vol. 193, no. 7, 2014, pages 3288 - 3295
PRAT L; BOUAZIZ JD; WALLACH D; VIGNON-PENNAMEN MD; BAGOT M: "Neutrophilic dermatoses as systemic diseases", CLIN DERMATOL, vol. 32, no. 3, 2014, pages 376 - 388
PROCTOR, L. M.; WOODRUFF, T. M.; SHARMA, P.; SHIELS, I. A.; TAYLOR, S. M.: "Transdermal pharmacology of small molecule cyclic C5a antagonists", ADV EXP MED BIOL, vol. 586, 2006, pages 329 - 345
QIU X.-Q. ET AL.: "Small antibody mimetics comprising two complementary-determining regions and a framework region for tumor targeting", NATURE BIOTECHNOLOGY, vol. 25, no. 8, 2007, pages 921 - 929, XP002714946, DOI: doi:10.1038/nbt1320
RANGER; PEPPAS, J. MACROMOL. SCI. REV. MACROMOL. CHEM., vol. 23, 1953, pages 61
RENFENG GUO ET AL: "IFX-1 blocking the anaphylatoxin C5a IFX-1 blocking the anaphylatoxin C5a IFX-1 blocking the anaphylatoxin C5a - an anti-inflammatory effect in patients with hidradenitis suppurativa", 29 August 2017 (2017-08-29), XP055425517, Retrieved from the Internet <URL:http://www.inflarx.de/dam/jcr:ec982d1e-f82b-4992-a1b0-ff6e69a04b6b/Guo_2017_poster.pdf> [retrieved on 20171115] *
REVUZ J.: "Hidradenitis suppurativa", J EUR ACAD DERMATOL VENEREOL, vol. 23, no. 9, 2009, pages 985 - 998
RICARDO A; ARATA M; DEMARCO S; DHAMNASKAR K; HAMMER R; FRIDKIS-HARELI M; RAJAGOPAL V; SEYB K; TANG G-Q; TOBE S: "Preclinical Evaluation of RA101495, a Potent Cyclic Peptide Inhibitor of C5 for the Treatment of Paroxysmal Nocturnal Hemoglobinuria", BLOOD, vol. 126, 2015, pages 939
RIEDEMANN NC; GUO RF; NEFF TA; LAUDES IJ; KELLER KA; SARMA VJ; MARKIEWSKI MM; MASTELLOS D; STREY CW; PIERSON CL: "Increased C5a receptor expression in sepsis", J CLIN INVEST, vol. 110, no. 1, 2002, pages 101 - 108
RIEDEMANN, N. C. ET AL.: "Controlling the anaphylatoxin C5a in diseases requires a specifically targeted inhibition", CLIN IMMUNOL, vol. 180, 2017, pages 25 - 32, XP055454362, DOI: doi:10.1016/j.clim.2017.03.012
RITTIRSCH D; FLIERL MA; NADEAU BA; DAY DE; HUBER-LANG M; MACKAY CR; ZETOUNE FS; GERARD NP; CIANFLONE K; KOHL J: "Functional roles for C5a receptors in sepsis", NAT MED, vol. 14, no. 5, 2008, pages 551 - 557
SAUDEK ET AL., N. ENG. J. MED., vol. 321, 1989, pages 574
SEFTON, RC CRIT. REF. BIOMED. ENG., vol. 14, 1987, pages 201
SLADE DE; POWELL BW; MORTIMER PS: "Hidradenitis suppurativa: pathogenesis and management", BR J PLAST SURG, vol. 56, no. 5, 2003, pages 451 - 461
SMITH, C. W.; MARLIN, S. D.; ROTHLEIN, R.; TOMAN, C.; ANDERSON, D. C.: "Cooperative interactions of LFA-1 and Mac-1 with intercellular adhesion molecule-1 in facilitating adherence and transendothelial migration of human neutrophils in vitro", J CLIN INVEST, vol. 83, 1989, pages 2008 - 2017
SOUZA DG; ESSER D; BRADFORD R; VIEIRA AT; TEIXEIRA MM: "APT070 (Mirococept), a membrane-localised complement inhibitor, inhibits inflammatory responses that follow intestinal ischaemia and reperfusion injury", BR J PHARMACOL, vol. 145, no. 8, 2005, pages 1027 - 1034
STRAINIC MG; SHEVACH EM; AN F; LIN F; MEDOF ME: "Absence of signaling into CD4(+) cells via C3aR and C5aR enables autoinductive TGF-betal signaling and induction of Foxp3(+) regulatory T cells", NAT IMMUNOL, vol. 14, no. 2, 2013, pages 162 - 171
T TERNOWITZ ET AL: "Methotrexate inhibits the human C5a-induced skin response in patients with psoriasis.", J INVEST DERMATOL., vol. 89, no. 2, 1 August 1987 (1987-08-01), United States, pages 192 - 196, XP055455258 *
TREAT ET AL.: "Liposomes in the Therapy of Infectious Disease and Cancer", 1989, LISS, pages: 353 - 365
TZANETAKOU V; KANNI T; GIATRAKOU S; KATOULIS A; PAPADAVID E; NETEA MG; DINARELLO CA; VAN DER MEER JW; RIGOPOULOS D; GIAMARELLOS-BO: "Safety and Efficacy of Anakinra in Severe Hidradenitis Suppurativa: A Randomized Clinical Trial", JAMA DERMATOL, vol. 152, no. 1, 2016, pages 52 - 59
WARD ET AL., NATURE, vol. 341, 1989, pages 544 - 546
WARD PA.: "Functions of C5a receptors", J MOL MED (BERL, vol. 87, no. 4, 2009, pages 375 - 378, XP019681211
WOLLINA U; KOCH A; HEINIG B; KITTNER T; NOWAK A: "Acne inversa (Hidradenitis suppurativa): A review with a focus on pathogenesis and treatment", INDIAN DERMATOL ONLINE J, vol. 4, no. 1, 2013, pages 2 - 11
XU R; WANG R; HAN G; WANG J; CHEN G; WANG L; LI X; GUO R; SHEN B; LI Y: "Complement C5a regulates IL-17 by affecting the crosstalk between DC and gammadelta T cells in CLP-induced sepsis", EUR J IMMUNOL, vol. 40, no. 4, 2010, pages 1079 - 1088

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10633434B2 (en) 2016-06-14 2020-04-28 Regeneron Pharmaceuticals, Inc. Anti-C5 antibodies
US11479602B2 (en) 2016-06-14 2022-10-25 Regeneren Pharmaceuticals, Inc. Methods of treating C5-associated diseases comprising administering anti-C5 antibodies
US11492392B2 (en) 2016-06-14 2022-11-08 Regeneran Pharmaceuticals, Inc. Polynucleotides encoding anti-C5 antibodies
US11365265B2 (en) 2017-12-13 2022-06-21 Regeneron Pharmaceuticals, Inc. Anti-C5 antibody combinations and uses thereof
WO2020182974A1 (fr) * 2019-03-14 2020-09-17 Morphosys Ag Anticorps ciblant c5ar
WO2021146320A1 (fr) * 2020-01-13 2021-07-22 Visterra, Inc. Molécules d'anticorps anti-c5ar1 et leurs utilisations
US11773179B2 (en) 2020-01-13 2023-10-03 Visterra, Inc. Antibody molecules to C5aR1 and uses thereof
WO2021259160A1 (fr) * 2020-06-24 2021-12-30 Staidson (Beijing) Biopharmaceuticals Co., Ltd. Anticorps reconnaissant de manière spécifique c5a et leurs utilisations
US11912781B2 (en) 2021-01-13 2024-02-27 Visterra, Inc. Humanized complement 5A receptor 1 antibodies and methods of use thereof

Also Published As

Publication number Publication date
SG11201912882QA (en) 2020-01-30
CN111201241A (zh) 2020-05-26
CA3066689A1 (fr) 2018-12-27
AU2018286754A1 (en) 2019-12-19
TWI786132B (zh) 2022-12-11
KR20200020727A (ko) 2020-02-26
CA3066689C (fr) 2024-01-16
JP2020524696A (ja) 2020-08-20
IL271074A (en) 2020-01-30
TW201904611A (zh) 2019-02-01
EP3642230A1 (fr) 2020-04-29

Similar Documents

Publication Publication Date Title
US11273225B2 (en) Treatment of inflammatory diseases with inhibitors of C5a activity
US11890349B2 (en) Treatment of inflammatory diseases with inhibitors of C5A activity
CA3066689C (fr) Traitement de maladies inflammatoires par des inhibiteurs de l&#39;activite de c5a
KR102513989B1 (ko) 간 질환을 치료하거나 예방하는 방법
ES2711882T3 (es) Método de tratamiento de la nefropatía diabética
US20230158060A1 (en) Inhibitors of C5a for the Treatment of Corona Virus Infection
EP3220952B1 (fr) Méthode de traitement ou de prévention d&#39;un accident vasculaire cérébral
EA043076B1 (ru) Лечение воспалительных заболеваний с помощью ингибиторов активности c5a
US20240158483A1 (en) Anti-c1s antibodies and uses thereof

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 18732016

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 3066689

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 2018286754

Country of ref document: AU

Date of ref document: 20180613

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2019570986

Country of ref document: JP

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 20197038211

Country of ref document: KR

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2018732016

Country of ref document: EP

Effective date: 20200123