WO2013005603A1 - Agent for prevention and/or treatment of allergic inflammation in conjunctiva - Google Patents

Agent for prevention and/or treatment of allergic inflammation in conjunctiva Download PDF

Info

Publication number
WO2013005603A1
WO2013005603A1 PCT/JP2012/066207 JP2012066207W WO2013005603A1 WO 2013005603 A1 WO2013005603 A1 WO 2013005603A1 JP 2012066207 W JP2012066207 W JP 2012066207W WO 2013005603 A1 WO2013005603 A1 WO 2013005603A1
Authority
WO
WIPO (PCT)
Prior art keywords
polypeptide
conjunctiva
allergic
sema3a
semaphorin
Prior art date
Application number
PCT/JP2012/066207
Other languages
French (fr)
Japanese (ja)
Other versions
WO2013005603A9 (en
Inventor
純美 田中
信久 水木
良郎 五嶋
Original Assignee
公立大学法人横浜市立大学
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 公立大学法人横浜市立大学 filed Critical 公立大学法人横浜市立大学
Publication of WO2013005603A1 publication Critical patent/WO2013005603A1/en
Publication of WO2013005603A9 publication Critical patent/WO2013005603A9/en

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • C07K14/4701Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
    • C07K14/4702Regulators; Modulating activity
    • C07K14/4703Inhibitors; Suppressors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/1703Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • A61K38/1709Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/18Growth factors; Growth regulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • A61P27/14Decongestants or antiallergics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents

Definitions

  • the present invention relates to a preventive and / or therapeutic agent for allergic inflammation in the conjunctiva.
  • Non-patent Document 1 Non-patent Document 1
  • steroid eye drops have strong effects, they have strong side effects such as increased intraocular pressure in the eye, cataract, retinopathy, induction and exacerbation of infection, vascular abnormalities, skin symptoms, and hirsutism. It is not possible. For this reason, non-steroidal eye drops are being used as alternative treatments, and eye drops that are more effective than steroids and avoid side effects such as keratoconjunctival peripheral nerve sensory disturbance and conjunctival vascular abnormalities have been developed.
  • Non-patent Document 3 A typical semaphorin molecule Sema3A ⁇ whose function in the immune system has been clarified has been energetically analyzed around the world for its function as a guidance factor in the nervous system (Non-patent Document 3), but immune cell migration The inhibitory effect has also been reported recently (Non-patent Document 4). Suppression of T cell receptor (TCR) signal pathway and MAP kinase signal pathway or inhibition of actin accumulation suppresses activation, and NP-1NP / pelexin-A4 complex is negative for Temacell immune response as a receptor for Sema3A (Non-patent Documents 5 and 6).
  • TCR T cell receptor
  • An object of the present invention is to provide eye drops and subconjunctival injections for intractable allergic conjunctival diseases including seasonal allergic conjunctivitis.
  • Sema3A semaphorin 3A
  • the gist of the present invention is as follows. (1) A preventive and / or therapeutic agent for allergic inflammation in the conjunctiva, containing semaphorin 3A as an active ingredient. (2) The preventive and / or therapeutic agent according to (1), wherein the allergic inflammation in the conjunctiva is seasonal allergic conjunctivitis. (3) The preventive and / or therapeutic agent according to (2), wherein the seasonal allergic conjunctivitis is hay fever. (4) A method for preventing and / or treating allergic inflammation in the conjunctiva, comprising administering to a subject a pharmaceutically effective amount of semaphorin 3A. (5) Use of semaphorin 3A for prevention and / or treatment of allergic inflammation in the conjunctiva. (6) Semaphorin 3A for use in a method for preventing and / or treating allergic inflammation in the conjunctiva.
  • the biologics of the present invention are greatly different from conventional antiallergic eye drops as follows. ⁇ It is an advantage that it is as safe and safe as other anti-allergic eye drops. ⁇ Maintain normal corneal tissue and corneal epithelium. ⁇ As a therapeutic agent, it does not cause side effects such as keratoconjunctival peripheral nerve sensory disturbance and conjunctival vascular abnormalities. ⁇ Maintains normal conjunctival epithelial and goblet cell function. -Prevention and / or treatment of eosinophilic conjunctival vasculitis. ⁇ Can be used with other antiallergic eye drops. ⁇ Prevents dryness. ⁇ There are almost no side effects and immediate effects can be expected.
  • Sema3A can be expected to be effective 2 times / day for 4 to 6 times / day of antiallergic eye drops. • Initial therapy is effective twice a day. ⁇ No abnormalities in the anterior chamber. No abnormalities in intraocular tissues such as cornea, ciliary body, and lens. Does not cause secondary ciliary dysfunction or secondary glaucoma. ⁇ Does not cause secondary cataract than steroids. -Less damage to intraocular tissues. ⁇ Because it contains no preservatives, there is little burden on the eyes even with frequent and relatively long-term instillation. ⁇ Can be used without worrying about side effects such as sleepiness. ⁇ It can be expected that the patient's QOL will be better than allergy medicine.
  • the biologic of the present invention has a pharmacological action that cannot be achieved with chemical substances.
  • the present invention provides a preventive and / or therapeutic agent for allergic inflammation in the conjunctiva, containing semaphorin 3A as an active ingredient.
  • the active ingredient semaphorin 3A may be a natural type or a mutant.
  • Specific examples of natural semaphorin 3A or semaphorin 3A mutant include a region consisting of any of the following polypeptides (a) to (c), and have an activity of preventing and / or treating allergic inflammation in the conjunctiva: The polypeptide which has can be mentioned.
  • preventive and / or therapeutic activity of allergic inflammation in the conjunctiva means an activity that improves or prevents the symptoms of the conjunctival lesion of allergic inflammation (eg, allergic conjunctivitis) in the conjunctiva. Means. Whether or not the polypeptide has the activity is evaluated by, for example, administering the polypeptide to the animal developing allergic conjunctivitis by instillation, subconjunctival administration, etc., and improving the symptoms of the lesion. Can be examined.
  • a model animal such as a mouse having an allergic conjunctivitis described in Examples below can be used.
  • This mouse was sensitized by administering ragweed pollen as an antigen, and caused allergic conjunctivitis by inducing antigen instillation. In addition to clinical symptoms such as tears, eyes, and hyperemia and edema, Eosinophils that have infiltrated into are observed.
  • the evaluation of the prevention and / or treatment activity of allergic inflammation in the conjunctiva using this mouse can be carried out by, for example, applying polymorphism for a certain period of time by instillation and subconjunctival administration to the conjunctival lesion of the mouse, as specifically described in the following examples. This can be done by administering the peptide and evaluating the presence or absence of improvement in the symptoms of the lesion.
  • the average score value of the conjunctivitis symptoms of the allergic conjunctivitis onset group administered the polypeptide for a certain period significantly decreased compared to the average score value of the conjunctivitis symptoms of the allergic conjunctivitis onset group of the untreated allergic conjunctivitis, the average score value of the conjunctivitis symptoms of the allergic conjunctivitis onset group administered the polypeptide for a certain period significantly decreased, Alternatively, for example, when the average score of the polypeptide administration group is about two-thirds or less, preferably about one-half or less of the average score of the untreated individuals, the polypeptide is allergenic. It can be judged that it has the activity of preventing and / or treating conjunctivitis.
  • the number of eosinophils infiltrated into the conjunctiva is determined by removing the conjunctival tissue from animals with allergic conjunctivitis, preparing a specimen, confirming eosinophil infiltration with an optical microscope, and calculating the number. It can be carried out. For example, the average value of the number of eosinophils in the allergic conjunctivitis-onset population administered with the polypeptide for a certain period was significantly lower than the average value of the number of eosinophils in the untreated allergic conjunctivitis-onset population.
  • the average eosinophil count of the polypeptide administration group is about 2/3 or less, preferably about 1/2 or less of the average eosinophil count of the untreated population It can be determined that the polypeptide has a prophylactic and / or therapeutic activity for allergic conjunctivitis.
  • polypeptide refers to a molecule formed by peptide bonding of a plurality of (two or more) amino acids, and includes not only high molecular weight molecules having a large number of amino acids but also the number of amino acids. Low molecular weight molecules (oligopeptides) and proteins are also included.
  • “having an amino acid sequence” means that amino acid residues are arranged in such an order.
  • a polypeptide having the amino acid sequence represented by SEQ ID NO: 2 means a Met77Gly Trp Leu ... (omitted) ... Pro Arg Ser Val amino acid sequence of 771 amino acid residues Means polypeptide.
  • Semaphorin 3A protein sequences human and chicken, respectively
  • GenBank accession numbers NM006080 and U02528, respectively.
  • Semaphorins are proteins that have been identified as molecules that guide nerve axon elongation. It is also called collapsin because it was discovered based on the activity of collapsing the growth cone at the tip of the neurite. Semaphorins constitute a secreted or transmembrane protein family, and more than 30 members have been identified to date from invertebrates such as nematodes to vertebrates such as humans.
  • Semaphorin 3A is a secreted protein member of semaphorin and is a protein molecule known as a repulsive nerve axon guidance molecule.
  • Secreted semaphorins such as Sema3A have three structural domains: a semaphorin (sema) domain of ⁇ 500aa amino terminus, a C-2 type immunoglobulin (Ig) domain, and a basic terminal domain. It is known that the region necessary for Sema3A to exert physiological activities such as nerve elongation-inhibiting action is a region of 70 amino acids corresponding to the 166th to 235th of the sema domain (Koppel et al., Neuron , Vol.
  • the 70 amino acid region (hereinafter referred to as “70aa region”) is a region of 166aa to 235aa in both SEQ ID NO: 2 and SEQ ID NO: 4.
  • Sema3A has been identified from various animals such as humans, chickens, mice and rats. The homology of these Sema3A is as high as about 85% when compared as a whole protein, and is even higher as about 95% or more when compared only with the above 70 amino acid region.
  • the sema domain is 61aa to 567aa
  • the Ig domain is 592aa to 655aa
  • the basic terminus is 726aa to 772aa (Feiner et al., Neuron, Vol.19, 539) -545, 1997).
  • human Sema3A having the amino acid sequence of SEQ ID NO: 2 the sema domain is 61aa to 567aa
  • the Ig domain is 591aa to 654aa
  • the basic terminal domain is 726aa to 771aa.
  • the polypeptide (a) is a polypeptide comprising a region containing at least the 70aa region of the chicken Sema3A protein having the amino acid sequence shown in SEQ ID NO: 4. Specifically, from the full length of SEQ ID NO: 4 Or a fragment thereof containing the 70aa region.
  • a polypeptide contained as an active ingredient in the preventive and / or therapeutic agent of the present invention includes a region comprising the polypeptide of (a) and has an activity of preventing and / or treating allergic inflammation in the conjunctiva. (Hereinafter referred to as “active ingredient polypeptide a” for convenience).
  • the active ingredient polypeptide a may be composed only of the polypeptide (a) as long as it has preventive and / or therapeutic activity for allergic inflammation in the conjunctiva, and may be one end of the polypeptide (a). Alternatively, an amino acid or polypeptide added at both ends may be used.
  • the active ingredient polypeptide a consisting only of the polypeptide (a) are not particularly limited, and examples thereof include a polypeptide consisting of the entire amino acid sequence shown in SEQ ID NO: 4 (ie, chicken Sema3A protein).
  • chicken Sema3A protein has preventive and / or therapeutic activity for allergic inflammation in the conjunctiva is as specifically described in the following examples.
  • a fragment lacking a part of the N-terminus or C-terminus of chicken Sema3A protein has the activity of preventing and / or treating allergic inflammation in the conjunctiva, it is within the scope of the present invention as an active ingredient polypeptide a. Is included.
  • the active ingredient polypeptide a in which a polypeptide or the like is added to the polypeptide of (a) are not particularly limited.
  • the 70aa region of SEQ ID NO: 4 corresponds to a secreted semaphorin other than Sema3A.
  • Examples include chimeric proteins constructed by replacing the regions, and polypeptides in which various tags such as His tags, FLAG tags, and myc tags are added to the polypeptide (a).
  • secretory semaphorins other than Sema3A include, but are not limited to, collapsin 2, collapsin 3, and collapsin 5.
  • collapsin 2 The amino acid sequences of collapsin 2, collapsin 3 and collapsin 5 are known as described in Feiner et al., Neuron, vol. 19, 539-545 (1997).
  • the chimeric protein constructed by replacing the region containing at least the 70aa region of Sema3A with the corresponding region of collapsin 2 retains the DRG retraction activity of Sema3A, and is similar to Sema3A. Demonstrate physiological activity. Therefore, such a chimeric protein also has the activity of preventing and / or treating allergic inflammation in the conjunctiva, and thus is included in the scope of the present invention as the active ingredient polypeptide a.
  • Sema3A lacking the Ig domain also has DRG regression activity (Koppel et al., 1997), but such a polypeptide is also present in the region of Sema3A containing 70aa at the N-terminal side of the Ig domain.
  • a polypeptide to which a basic domain is added is included in the active ingredient polypeptide a and is within the scope of the present invention.
  • the specific examples described above are merely examples, and any amino acid or polypeptide added to the polypeptide (a) can be used as long as it has preventive and / or therapeutic activity for allergic inflammation in the conjunctiva.
  • the active ingredient polypeptide a is included in the scope of the present invention.
  • the polypeptide of (b) above is a polypeptide comprising a region containing at least the 70aa region of the human Sema3A protein having the amino acid sequence shown in SEQ ID NO: 2, specifically, from the full length of SEQ ID NO: 2. Or a fragment thereof containing the 70aa region.
  • Another example of a polypeptide contained as an active ingredient in the preventive and / or therapeutic agent of the present invention includes a region consisting of the polypeptide (b), and prevents and / or treats allergic inflammation in the conjunctiva.
  • a polypeptide having activity hereinafter referred to as “active ingredient polypeptide b” for convenience).
  • chicken Sema3A (SEQ ID NO: 4) and human Sema3A (SEQ ID NO: 2) is 86% over the entire protein length, and is even higher at 95% when compared only with the 70aa region important for the physiological activity of Sema3A.
  • chicken Sema3A can treat or prevent allergic conjunctivitis in mice that are not chickens, so chicken sema3A can be used to treat allergic conjunctivitis other than chickens and mice such as humans. It is also possible to prevent.
  • the active ingredient polypeptide b such as human Sema3A protein is also useful for the preparation of a preventive and / or therapeutic agent for allergic inflammation in the conjunctiva such as allergic conjunctivitis, like the above active ingredient polypeptide a such as chicken Sema3A protein. It is.
  • the active ingredient polypeptide b may be composed only of the polypeptide of (b) as long as it has preventive and / or therapeutic activity for allergic inflammation in the conjunctiva.
  • an amino acid or polypeptide may be added to one or both ends of the polypeptide (b). Specific examples of such a polypeptide are the same as those of the active ingredient polypeptide a.
  • the polypeptide consisting of the full length of SEQ ID NO: 2 ie, human Sema3A protein
  • allergic inflammation in the conjunctiva is not particularly limited.
  • examples thereof include fragments thereof having prophylactic and / or therapeutic activity, chimeric proteins according to the above embodiment with secreted semaphorins other than Sema3A, polypeptides obtained by adding various tags to the polypeptide (b), and the like.
  • These specific examples are also merely examples, and any amino acid or polypeptide added to the polypeptide of (b) can be used as long as it has the activity of preventing and / or treating allergic inflammation in the conjunctiva.
  • the active ingredient polypeptide b is included in the scope of the present invention.
  • the polypeptide (c) is a polypeptide in which a small number (preferably 1 to several) amino acid residues of the polypeptide (a) or (b) are substituted, deleted and / or inserted.
  • Peptide which is a polypeptide having homology with the original sequence of 80% or more, preferably 90% or more, more preferably 95% or more, and still more preferably 98% or more.
  • a protein may have almost the same function as the original protein even when a small number of amino acid residues in the amino acid sequence of the protein are substituted, deleted, or inserted. Are widely known to those skilled in the art.
  • the region consisting of the polypeptide of (c) can also bring about the physiological activity of the Sema3A protein in the same manner as the 70aa region described above, it includes the region consisting of the polypeptide of (c) above, and is allergic in the conjunctiva.
  • a polypeptide having preventive and / or therapeutic activity for inflammation (hereinafter referred to as “active ingredient polypeptide c” for convenience) is also prevented and / or treated according to the present invention in the same manner as the active ingredient polypeptides a and b. Useful for the preparation of agents.
  • the “homology” of the amino acid sequences means that both amino acid sequences are aligned so that the amino acid residues of the two amino acid sequences to be compared match as much as possible, and the number of matched amino acid residues is defined as the total number of amino acid residues. It is the percentage divided by the number.
  • a gap is appropriately inserted in one or both of the two sequences to be compared as necessary.
  • sequence alignment can be performed using a known program such as BLAST, FASTA, CLUSTAL W, and the like.
  • the total number of amino acid residues is the number of residues obtained by counting one gap as one amino acid residue.
  • the homology is the total number of amino acid residues in the longer sequence, and the number of amino acid residues matched. Is calculated by dividing.
  • the 20 amino acids constituting the natural protein are neutral amino acids having low side chains (Gly, Ile, Val, Leu, Ala, Met, Pro), and neutral amino acids having hydrophilic side chains (Asn).
  • the polypeptide (c) is specifically a polypeptide having a sequence homology of 80% or more with a polypeptide consisting of the full length of the amino acid sequence represented by SEQ ID NO: 2 or SEQ ID NO: 4 (ie, Sema3A protein) Or a polypeptide having a sequence homology of 80% or more with a Sema3A protein fragment containing a 70aa region.
  • the active ingredient polypeptide c is a polypeptide consisting only of these polypeptides, or an amino acid or polypeptide added to one or both ends of the polypeptide (c), and the conjunctiva A polypeptide having an activity of preventing and / or treating allergic inflammation in Specific examples thereof are the same as those of the active ingredient polypeptides a and b, and are not particularly limited.
  • the polypeptide having the full length of SEQ ID NO: 2 or 4 ie, Sema3A protein
  • a polypeptide having 80% or more sequence homology with a Sema3A protein fragment having preventive and / or therapeutic activity for allergic inflammation in the conjunctiva 80% or more sequence homology with a region containing 70aa region
  • Examples include chimeric proteins in which a region and other regions of secreted semaphorins other than Sema3A are fused, polypeptides in which various tags are added to the polypeptide of (c), and the like. These specific examples are also merely examples, and any amino acid or polypeptide added to the polypeptide of (c) may have any activity for preventing and / or treating allergic inflammation in the conjunctiva.
  • the active ingredient polypeptide c is included in the scope of the present invention.
  • the active ingredient polypeptide c is particularly preferably a polypeptide whose homology in the 70aa region is higher than the homology of the entire region comprising the polypeptide (c).
  • the active ingredient polypeptide c is a polypeptide having 80% homology with the polypeptide having the amino acid sequence of SEQ ID NO: 2, those having a homology in the 70aa region of more than 80% are preferable.
  • Polypeptides having homology in the region of 90% or more, more preferably 95% or more, further preferably 98% or more, and more preferably 70aa region are identical.
  • the active ingredient polypeptide c is a chimeric protein in which a region having 80% sequence homology with the sema domain of human Sema3A protein and a region other than the sema domain of secreted semaphorin other than Sema3A are fused.
  • the homology in the 70aa region in the sema domain is preferably more than 80%, in particular, the homology in the 70aa region is 90% or more, more preferably 95% or more, more preferably 98% or more, more preferably Chimeric proteins having the same sequence in the 70aa region are preferred.
  • the active ingredient polypeptides a to c described above in particular, 80% or more, preferably 90% or more, more preferably 95% or more, and further preferably, a polypeptide having the amino acid sequence represented by SEQ ID NO: 2 or SEQ ID NO: 4.
  • a polypeptide having a homology of 98% or more is preferable.
  • the polypeptide contained as an active ingredient in the preventive and / or therapeutic agent of the present invention is a polypeptide having the amino acid sequence represented by SEQ ID NO: 2 or SEQ ID NO: 4.
  • a sugar chain or a polyethylene glycol (PEG) chain is added to the polypeptide or at least a part of the amino acids constituting the polypeptide in order to increase the stability of the polypeptide in vivo.
  • PEG polyethylene glycol
  • a technique using a D-form amino acid is widely known and used. By adding sugar chains or PEG chains, or by using D-form amino acids as at least part of the amino acids that make up the polypeptide, it is less susceptible to degradation by peptidases in vivo, reducing the polypeptide in vivo by half. The period becomes longer.
  • polypeptide used in the present invention may be subjected to these known modifications for in vivo stabilization as long as it has preventive and / or therapeutic activity for allergic inflammation in the conjunctiva.
  • polypeptide in the claims and in the claims is used to include those modified for in vivo stabilization, unless the context clearly indicates otherwise.
  • Glycosylation to polypeptides is well known, for example, Sato M, Furuike T, Sadamoto R, Fujitani N, Nakahara T, Niikura K, Monde K, Kondo H, Nishimura S., "Glycoinsulins: dendritic sialylolulins displaying a prolonged blood-sugar-lowering activity. ", J Am Chem Soc. 2004 Nov 3; 126 (43): 14013-22, Sato M, Sadamoto R, Niikura K, Monde K, Kondo H, Nishimura S," Site- specific introduction of sialic acid into insulin. ", Angew Chem Int Ed Engl. 2004 Mar 12; 43 (12): 1516-20.
  • the sugar chain can be bound to the N-terminus, C-terminus, or an amino acid therebetween, but is preferably bound to the N-terminus or C-terminus so as not to inhibit the activity of the polypeptide.
  • the number of sugar chains to be added is preferably 1 or 2, and preferably 1.
  • the sugar chain is preferably monosaccharide to tetrasaccharide, more preferably disaccharide or trisaccharide.
  • the sugar chain can be bound to a free amino group or carboxyl group of the polypeptide directly or via a spacer structure such as a methylene chain having about 1 to 10 carbon atoms.
  • the PEG chain can be attached to the N-terminus, C-terminus, or the amino acid between them, and usually one or two PEG chains are attached to a free amino group or carboxyl group on the polypeptide.
  • the molecular weight of the PEG chain is not particularly limited, but is usually 3000 to 7000. Degrees, preferably those of about 5000 is used.
  • a method in which at least a part of amino acids constituting a polypeptide is D-form is also known, for example, Brenneman DE, Spong CY, Hauser JM, Abebe D, Pinhasov A, Golian T, Gozes I., "Protective peptides that are orally active and mechanistically nonchiral. ", J Pharmacol Exp Ther. 2004 Jun; 309 (3): 1190-7 and Wilkemeyer MF, Chen SY, Menkari CE, Sulik KK, Charness ME.,” Ethanol antagonist peptides: structural specificity without "J. Pharmacol. Exp. Ther. 2004. Jun; 309 (3): 1183-9.
  • a part of the amino acids constituting the polypeptide may be D-form, but since the activity of the polypeptide is not inhibited as much as possible, all of the amino acids constituting the polypeptide are D-form amino acids rather than only a part of the amino acids. It is preferable that
  • Semaphorin 3A used as an active ingredient in the present invention can be easily prepared by a conventional method using, for example, a commercially available peptide synthesizer. Moreover, it can prepare easily using a well-known genetic engineering method. For example, a Sema3A gene cDNA is prepared by RT-PCR from RNA extracted from a tissue expressing the Sema3A gene, and the full length or a desired part of the cDNA is incorporated into an expression vector and introduced into a host cell. The desired polypeptide can be obtained. RNA extraction, RT-PCR, cDNA integration into a vector, and introduction of a vector into a host cell can be performed by known methods.
  • vectors and host cells to be used are well known, and various types are commercially available.
  • a method for producing a chimeric protein is also well known in this field.
  • a chimeric protein can be produced by the method described in the above-mentioned paper by Koppel et al.
  • the stabilization modification can also be easily performed by a known method as described in each of the above documents.
  • Semaphorin 3A which is an active ingredient in the preventive and / or therapeutic agent of the present invention, is effective in treating and / or preventing allergic inflammation in the conjunctiva.
  • Allergic inflammation in the conjunctiva includes eosinophilic conjunctival disease, allergic conjunctivitis (including seasonal allergic conjunctivitis such as pollen conjunctivitis), allergic keratoconjunctivitis, allergic blepharitis, hay fever, atopic keratoconjunctivitis Semaphorin 3A is useful for preventive initial treatment (therapy) of these diseases, pre-seasonal administration and / or therapeutic agent conservative treatment, maintenance therapy / induction therapy. is there.
  • the subject of administration of the preventive and / or therapeutic agent of the present invention is a mammal, and examples thereof include humans, dogs, cats, rabbits, hamsters and the like.
  • the use of semaphorin 3A derived from the same species as the patient to be prevented or treated is considered to have a higher prevention or treatment effect, and is also desirable from the viewpoint of safety in clinical application. Therefore, for example, when the subject to which the preventive and / or therapeutic agent of the present invention is administered is a human, the prophylactic and / or therapeutic agent containing the active ingredient polypeptide b as an active ingredient is particularly preferable.
  • the preventive and / or therapeutic agent of the present invention may consist of semaphorin 3A alone, or a pharmaceutically acceptable excipient, stabilizer, preservative, buffer, suitable for each dosage form, Additives such as solubilizers, emulsifiers, diluents, tonicity agents and the like can be mixed as appropriate to prepare a preparation. Formulation methods and usable additives are well known in the field of pharmaceutical preparations, and any method and additive can be used.
  • the preventive and / or therapeutic agent of the present invention is used by administering to the conjunctival part to be prevented or the conjunctival lesion part to be treated.
  • the administration method include local administration (instillation on the conjunctiva, administration by subconjunctival injection, etc.) and the like.
  • the dosage form include eye ointments, injections, eye drops and the like. The dosage is appropriately selected according to symptoms, age, body weight, administration method and the like, and is not particularly limited.
  • the amount of active ingredient is usually about 6000 to 60000 U per day for the target animal, preferably In the case of subconjunctival injection amount, the amount of active ingredient is usually about 500-5000 U per day, preferably about 500-5000 U, and divided into 1 to several times. Administered. Preferably, depending on the degree of symptom improvement, it is administered once or several times daily for several days to several months, or regularly once or several times every other day.
  • “1 U” is a semaphorin titer that collapses a growth cone of 50% of NGF-sensitive DRG neurons with a stock solution of a purified polypeptide solution obtained by a conventional method. collapse assay) (Luoet al., Cell vol.75, 217-227, 1993).
  • the preventive and / or therapeutic agent of the present invention may be used alone or in combination with other prophylactic and / or therapeutic agents.
  • the prophylactic and / or therapeutic agent of the present invention can be used in combination with anti-histamine eye drops, non-steroid eye drops and steroid eye drops, which are conventional anti-allergic eye drops.
  • Example 1 (Materials and methods) Ragweed pollen (short ragweed pollen: RW) was used as an antigen and an emulsion with aluminum hydroxide (Alum) was prepared and administered intraperitoneally twice to BALB / c mice at 10-day intervals. Instillation of RW after sensitization induces allergic conjunctivitis (EC) in mice, and the immediate phase observes and evaluates clinical scores 30 minutes after instillation, collects conjunctiva 24 hours after instillation, and infiltrate eosinophils Numbers were counted. As a control, normal mice were instilled with PBS. The late phase was sampled one week after the start of instillation. The number of infiltrating eosinophils was counted.
  • EC allergic conjunctivitis
  • mice For animal experiments, BALB / cCrSlc female mice purchased from Japan SLC Co., Ltd. (Hamamatsu, Japan) at the age of 5 weeks were used. Mice are raised in a constant temperature and humidity chamber at room temperature 25 ⁇ 1 ° C and humidity 55 ⁇ 5% under light and dark conditions with a 12-hour cycle, and with standard mouse food (solid feed: 5LJ1 (PMI FEEDS) and sterile water) There were 6 mice per group, and a total of 120 mice were used.
  • 5LJ1 PMI FEEDS
  • Sema3A administration Semaphorin 3A was prepared as previously reported (Journal of Investigative Dermatology (2008) 128, 2842-2849). Briefly, a chicken semaphorin 3A stable expression strain was cultured, and semaphorin 3A protein produced in the culture supernatant was purified by a conventional method. The semaphorin 3A stable expression strain is obtained by incorporating a chicken semaphorin 3A gene (SEQ ID NO: 3) into HEK293 cells. The activity of the obtained semaphorin 3A protein was measured by a collapse assay using the dorsal root ganglion of the chicken. The regression assay was performed by the following method.
  • DRG Dorsal root ganglia
  • the extracted DRG was placed on the bottom of a glass containing NGF-containing medium, and cultured in a static place for 24 hours in a 37 ° C. incubator.
  • the semaphorin 3A solution obtained by purification as described above is diluted 1-fold (stock solution) to 100,000-fold and administered at 37 ° C for 30 minutes. Fixed after ground culture. Under the microscope, calculate the percentage (%) of all NGF-sensitive DRG neurons that have collapsed growth cones, and determine the semaphorin 3A concentration (X-fold dilution) at which 50% collapse is observed. U / ml.
  • the titer of semaphorin 3A obtained above was 30,000 ⁇ U / mL.
  • Semaphorin-3A was instilled twice daily on both eyelids at a concentration of 10,100,1000 units.
  • the antigen solution was made into a single eye drop of 10 ⁇ l ⁇ l.
  • 2 ⁇ l was administered subconjunctivally to both eyes of the animal at intervals of 1 day.
  • the spleen was extracted from normal BALB / c mice. Prepare spleen cells and seed 100 mL of spleen cells of appropriate concentration (6.25 ⁇ 10 5 cells / mL to 2.5 ⁇ 10 6 cells / mL) onto a T cell plate coated with anti-CD3 antibody (BD Pharmingen). Semaphorin-3A was administered at a concentration of 10,100,1000 units, and the control group was administered PBS.
  • the cells were cultured in a CO 2 incubator at 37 ° C. for 45 to 48 hours.
  • the OD at a measurement wavelength of 490 nm was measured with a microplate spectrophotometer at a control wavelength of 700 nm.
  • Sema3A was confirmed to suppress T cell proliferation (FIG. 4). It was revealed that T cells induce eosinophils. (Yuki Fukushima: Clinical study of allergies 28 (4), 2008).
  • the semaphorin molecule group has heretofore been known as a nerve guidance factor, but recently, it has become clear that various semaphorin molecules are also involved in the immune system. It has long been known that the nervous system and the immune system have a lot in common. In particular, semaphorin molecules involved in antigen-presenting cell-T cell interactions in the immune system have been discovered. It has been confirmed in vitro that a semaphorin 3A molecule is coated with a T cell receptor complex antibody to induce T cell activation and suppress T cell proliferation.
  • Neuropilin-1 was found in human dendritic cells and resting T cells, T cells bound to Neuropilin-1-expressing Cos7 cells, and Neuropilin-1 was CD3 at the interface between dendritic cells and naive T cells. And co-localize.
  • Anti-Neuropilin-1 antibody also suppresses T cell proliferation by dendritic cells.
  • the semaphorin 3A molecule which is a glycan in the nervous system, has the effect of suppressing the migration of immune cells, but its action cannot be inhibited by an antibody against Neuropilin. Therefore, Sema3A suppresses the proliferation of T cells in the immune system. It is suggested that it is carried by different receptors.
  • Sema3A ophthalmic solution is confirmed to be fast-acting
  • the effect on allergic conjunctivitis is Sema3A 1000 U / ml ophthalmic solution, histamine H1 receptor antagonist 0.1% olopatadine hydrochloride ophthalmic solution, 0.025% levocabastine hydrochloride ophthalmic solution, mediator release inhibition RW antigen solution was instilled 15 minutes after instillation of the drug 0.5% tranilast ophthalmic solution, 0.01% ibudilast ophthalmic solution, steroid drug 0.02% fluorometholone ophthalmic solution or PBS (control), and the conjunctivitis and conjunctivitis inhibitory effects were compared. Subsequent conjunctivitis symptoms due to antigen induction were observed at each measurement time point, and a conjunctivitis score was assigned.
  • Conjunctivitis symptoms such as hyperemia, edema, eyes, and tears were observed for 30 minutes after instillation, and evaluated according to the scores for conjunctivitis symptoms shown in Table 1.
  • the amount of instillation was 1 10 ⁇ , and the animals were instilled twice daily for 6 days in total.
  • Sema3A was ⁇ 0 for control 3+ (moderate), eye drops for hyperemia, edema, eyes and tears 1 to 2+, suggesting a therapeutic effect (upper figure 3).
  • the count section was the range including the pupil and optic disc from the center. Data were examined as mean ⁇ standard error of all mice, one-way analysis of variance and Tukey-Kramer multiple comparison test.
  • Mouse conjunctiva was frozen in liquid nitrogen embedded in OCT compound (VWR, Sawanee, GA, USA). This was prepared and fixed with methanol.
  • ⁇ Sema3A instillation and subconjunctival administration showed a clear therapeutic effect in the RW allergic conjunctivitis model, which suppressed the increase in conjunctivitis score and decreased the increase in eosinophils and T cells at the same site.
  • ⁇ Scores for allergic conjunctivitis symptoms (Table 1) were statistically calculated using a total of 5 items. Differences in clinical symptom suppression were confirmed over the 4-week observation period. There was no significant change from week 3 to week 4 (FIG. 1). ⁇ Significant difference in eosinophil infiltration score was observed in Sema3A concentration difference.
  • Neuropilin-1 is one of the receptors for VascularelEndothelial Growth Factor (VEGF), and Sema3A has been reported to antagonistically inhibit angiogenesis, but Sema3A 1000 units are instilled and conjunctival function by subconjunctival administration in this study. Insufficiency, corneal damage caused by it, and new blood vessels were not observed during the experiment, so suppression of corneal new blood vessels is expected.
  • VEGF VascularelEndothelial Growth Factor
  • Sema3A Although application of Sema3A has been reported, such as nerve guidance and atopic dermatitis, in fact, the incidence of eosinophilic inflammatory diseases in the mucous membrane is high in the United States and Europe, and the disease concept has been established, pathological analysis and treatment Efforts are being made to establish the law. Since the Sema3A molecule did not show a pathological change at the tissue level due to administration to the conjunctiva, a therapeutic effect is expected for the above-mentioned eosinophilic inflammatory mucosal disease.
  • Non-steroidal eye drops are not effective enough. Prolonged use causes refractory corneal epithelial disorder and dry eye. Steroid eye drops cause increased corneal lesions due to increased intraocular pressure, secondary glaucoma, secondary cataracts, and susceptibility to infection, leading to the risk of corneal transplantation and the final surgery.
  • the present invention can be used for the prevention and / or treatment of allergic inflammation in the conjunctiva.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Veterinary Medicine (AREA)
  • Organic Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Immunology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Zoology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Epidemiology (AREA)
  • Pulmonology (AREA)
  • Marine Sciences & Fisheries (AREA)
  • Toxicology (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Molecular Biology (AREA)
  • Ophthalmology & Optometry (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

Provided are eye drops, and a subconjunctival injection drug for refractory allergic conjunctival disease, foremost for seasonal allergic conjunctivitis. This agent for prevention and/or treatment of allergic inflammation in the conjunctiva contains semaphorin-3A as an active component.

Description

結膜におけるアレルギー性炎症の予防及び/又は治療剤Preventive and / or therapeutic agent for allergic inflammation in conjunctiva
 本発明は、結膜におけるアレルギー性炎症の予防及び/又は治療剤に関する。 The present invention relates to a preventive and / or therapeutic agent for allergic inflammation in the conjunctiva.
 近年、花粉症に代表されるアレルギー性疾患が増加しており、多臓器に関わる、国民の約30%を占める、大きな社会問題となっている。アレルギー性結膜疾患の患者の約85%は花粉性と推定されており、1%以上の割合で重症例が存在する(非特許文献1)。これらアレルギー性疾患の増加の要因として、大気汚染物質との関連が指摘されている。 In recent years, allergic diseases represented by hay fever have increased, and this has become a major social problem involving about 30% of the people involved in multiple organs. About 85% of patients with allergic conjunctival diseases are estimated to be pollen, and severe cases exist at a rate of 1% or more (Non-patent Document 1). It has been pointed out that it is related to air pollutants as a factor in the increase of these allergic diseases.
 現在、処方可能な抗アレルギー点眼薬は9種類あり、抗ヒスタミン点眼薬が主流である。しかしながら、非ステロイド点眼薬は効果が十分とはいえない。アレルギー性結膜疾患はI型アレルギー反応が関与する結膜疾患の総称であり、抗アレルギー点眼薬のみでは種々の病変を抑制することは不可能であり、ステロイド点眼薬が処方されてきた(非特許文献2)。 Currently, there are nine types of antiallergic eye drops that can be prescribed, and antihistamine eye drops are the mainstream. However, non-steroidal eye drops are not effective enough. Allergic conjunctival disease is a general term for conjunctival diseases involving type I allergic reactions. Anti-allergic eye drops alone cannot suppress various lesions, and steroid eye drops have been prescribed (non-patent literature). 2).
 また一方で、ステロイド点眼薬は、効果は強いものの、眼局所の眼圧上昇、白内障、網膜症、感染症の誘発と増悪、血管異常、皮膚症状、多毛などの副作用も強く、安易に継続することはできない。そのため、非ステロイド点眼薬が代わりの治療剤として、ステロイド薬より角結膜末梢神経知覚障害、結膜血管異常などの副作用を避ける、効果の強い点眼薬の開発が行われている。 On the other hand, although steroid eye drops have strong effects, they have strong side effects such as increased intraocular pressure in the eye, cataract, retinopathy, induction and exacerbation of infection, vascular abnormalities, skin symptoms, and hirsutism. It is not possible. For this reason, non-steroidal eye drops are being used as alternative treatments, and eye drops that are more effective than steroids and avoid side effects such as keratoconjunctival peripheral nerve sensory disturbance and conjunctival vascular abnormalities have been developed.
 免疫系での機能が明らかになっている代表的なセマフォリン分子 Sema3A は、神経系におけるガイダンス因子としての機能が世界中で精力的に解析されていた(非特許文献3)が、免疫細胞遊走抑制作用も近年報告もされている(非特許文献4)。T cell レセプター(TCR)シグナル経路やMAPキナーゼシグナル経路の阻止、あるいはアクチンの集積阻害より活性化を抑制する、また、NP-1 /pelexin-A4複合体がSema3AのレセプターとしてT cell免疫応答の負の制御に関与すると報告された(非特許文献5及び6)。 A typical semaphorin molecule Sema3A 機能 whose function in the immune system has been clarified has been energetically analyzed around the world for its function as a guidance factor in the nervous system (Non-patent Document 3), but immune cell migration The inhibitory effect has also been reported recently (Non-patent Document 4). Suppression of T cell receptor (TCR) signal pathway and MAP kinase signal pathway or inhibition of actin accumulation suppresses activation, and NP-1NP / pelexin-A4 complex is negative for Temacell immune response as a receptor for Sema3A (Non-patent Documents 5 and 6).
 しかし、セマフォリン分子 Sema3Aがアレルギー性結膜疾患に有効であるという報告はない。 However, there is no report that the semaphorin molecule Sema3A is effective for allergic conjunctival diseases.
 本発明は、季節性アレルギー性結膜炎をはじめとする難治性アレルギー性結膜疾患の点眼薬、結膜下注射薬を提供することを目的とする。 An object of the present invention is to provide eye drops and subconjunctival injections for intractable allergic conjunctival diseases including seasonal allergic conjunctivitis.
 本発明者らは、セマフォリン3A (Sema3A)分子が、これまでにない眼科領域での薬理作用により、季節性アレルギー性結膜炎をはじめとする重症、難治性アレルギー性結膜疾患の新規治療薬として有効であることを見出し、本発明を完成させるに至った。Sema3Aは、結膜におけるアレルギー性炎症の予防及び/又は治療に有効であると考えられる。 The present inventors have demonstrated that semaphorin 3A (Sema3A) molecule is effective as a novel therapeutic agent for severe and refractory allergic conjunctival diseases including seasonal allergic conjunctivitis due to its unprecedented pharmacological action in the ophthalmic field. As a result, the present invention has been completed. Sema3A is thought to be effective in preventing and / or treating allergic inflammation in the conjunctiva.
 本発明の要旨は、以下の通りである。
(1)セマフォリン3Aを有効成分として含有する、結膜におけるアレルギー性炎症の予防及び/又は治療剤。
(2)結膜におけるアレルギー性炎症が季節性アレルギー性結膜炎である(1)記載の予防及び/又は治療剤。
(3)季節性アレルギー性結膜炎が花粉症である(2)記載の予防及び/又は治療剤。
(4)セマフォリン3Aの医薬的に有効な量を被験者に投与することを含む、結膜におけるアレルギー性炎症を予防及び/又は治療する方法。
(5)結膜におけるアレルギー性炎症の予防及び/又は治療のためのセマフォリン3Aの使用。
(6)結膜におけるアレルギー性炎症を予防及び/又は治療する方法に使用するためのセマフォリン3A。
The gist of the present invention is as follows.
(1) A preventive and / or therapeutic agent for allergic inflammation in the conjunctiva, containing semaphorin 3A as an active ingredient.
(2) The preventive and / or therapeutic agent according to (1), wherein the allergic inflammation in the conjunctiva is seasonal allergic conjunctivitis.
(3) The preventive and / or therapeutic agent according to (2), wherein the seasonal allergic conjunctivitis is hay fever.
(4) A method for preventing and / or treating allergic inflammation in the conjunctiva, comprising administering to a subject a pharmaceutically effective amount of semaphorin 3A.
(5) Use of semaphorin 3A for prevention and / or treatment of allergic inflammation in the conjunctiva.
(6) Semaphorin 3A for use in a method for preventing and / or treating allergic inflammation in the conjunctiva.
 本発明の生物製剤が従来の抗アレルギ-点眼薬と大きく異なることは下記のような点である。
・他の抗アレルギ-点眼薬と同等及び同等以上の有効性が認められ、安全性が高いことが利点である。
・角膜組織、角膜上皮を正常に保つ。
・代わり治療剤として、ステロイド薬より角結膜末梢神経知覚障害、結膜血管異常などの副作用を起さない。
・結膜上皮、杯状細胞機能を正常に保つ。
・好酸球性結膜血管炎の予防及び/又は治療可能。
・他の抗アレルギー点眼薬と併用可能。
・ドライの発生防止。
・副作用はほとんどない上に,即効性が期待できる。
・重症アレルギー性結膜疾患にSema3A単剤で効果的であることが期待できる。
・抗アレルギー点眼薬の4回~6回/日の点眼に対し、Sema3Aは2回/日の点眼で効果が期待できる。
・初期療法は2回/日で有効である。
・前房内異常所見なし。角膜、毛様体、水晶体など眼内組織異常所見なし。続発毛様体機能不全、続発緑内障を起さない。
・ステロイド薬より続発白内障を起さない。
・眼内組織にダメージが少ない。
・防腐剤が入っていないので、頻回点眼、比較的長期点眼でも、目に負担が少ない。
・眠気など副作用を気にせずに使用できる。
・今までのアレルギー医療よりも、患者のQOLが優れることが期待できる。
The biologics of the present invention are greatly different from conventional antiallergic eye drops as follows.
・ It is an advantage that it is as safe and safe as other anti-allergic eye drops.
・ Maintain normal corneal tissue and corneal epithelium.
・ As a therapeutic agent, it does not cause side effects such as keratoconjunctival peripheral nerve sensory disturbance and conjunctival vascular abnormalities.
・ Maintains normal conjunctival epithelial and goblet cell function.
-Prevention and / or treatment of eosinophilic conjunctival vasculitis.
・ Can be used with other antiallergic eye drops.
・ Prevents dryness.
・ There are almost no side effects and immediate effects can be expected.
-Expected to be effective with Sema3A alone for severe allergic conjunctival disease.
・ Sema3A can be expected to be effective 2 times / day for 4 to 6 times / day of antiallergic eye drops.
• Initial therapy is effective twice a day.
・ No abnormalities in the anterior chamber. No abnormalities in intraocular tissues such as cornea, ciliary body, and lens. Does not cause secondary ciliary dysfunction or secondary glaucoma.
・ Does not cause secondary cataract than steroids.
-Less damage to intraocular tissues.
・ Because it contains no preservatives, there is little burden on the eyes even with frequent and relatively long-term instillation.
・ Can be used without worrying about side effects such as sleepiness.
・ It can be expected that the patient's QOL will be better than allergy medicine.
 本発明の生物製剤は、化学物質では達成できない薬理作用を持つ。 The biologic of the present invention has a pharmacological action that cannot be achieved with chemical substances.
 本明細書は、本願の優先権の基礎である日本国特許出願、特願2011‐147038の明細書および/または図面に記載される内容を包含する。 This specification includes the contents described in the specification and / or drawings of the Japanese patent application, Japanese Patent Application No. 2011-147038, which is the basis of the priority of the present application.
臨床所見合計スコアの変化量。Change in total clinical findings score. 好酸球浸潤スコアの変化量。Change in eosinophil infiltration score. 即時相スコア(上)と遅発相スコア(下)。点眼薬a:0.025%レボカバスチン塩酸塩点眼液、点眼薬b:0.02%フルオロメトロン点眼液、点眼薬c:0.5%トラニラスト点眼液、点眼薬d:0.1%オロパタジン塩酸塩点眼液、点眼薬e:0.01%イブジラスト点眼液、点眼薬f:セマフォリン3A(Semaphorin-3A) 1000U/ml点眼液。Immediate phase score (above) and late phase score (below). Eye drops a: 0.025% levocabastine hydrochloride ophthalmic solution, eye drops b: 0.02% fluorometholone ophthalmic solution, eye drops c: 0.5% tranilast ophthalmic solution, eye drops d: 0.1% olopatadine hydrochloride ophthalmic solution, eye drops e: 0.01 % Ibudilast ophthalmic solution, ophthalmic solution f: Semaphorin-3A 1000 U / ml ophthalmic solution. T cell増殖アッセイの結果。Results of T cell proliferation assay.
 以下、本発明を詳細に説明する。 Hereinafter, the present invention will be described in detail.
 本発明は、セマフォリン3Aを有効成分として含有する、結膜におけるアレルギー性炎症の予防及び/又は治療剤を提供する。 The present invention provides a preventive and / or therapeutic agent for allergic inflammation in the conjunctiva, containing semaphorin 3A as an active ingredient.
 有効成分であるセマフォリン3Aは、天然型のものであっても、変異体であってもよい。天然型セマフォリン3A又はセマフォリン3A変異体の具体例としては、下記(a)~(c)のいずれかのポリペプチドからなる領域を含み、結膜におけるアレルギー性炎症の予防及び/又は治療活性を有するポリペプチドを挙げることができる。
 (a) 配列表の配列番号4に示されるアミノ酸配列中の連続する70個以上のアミノ酸から成り、配列番号4中の166aa~235aa(「第166番アミノ酸~第235番アミノ酸」を意味する)の領域を含むポリペプチド。
 (b) 配列表の配列番号2に示されるアミノ酸配列中の連続する70個以上のアミノ酸から成り、配列番号2中の166aa~235aaの領域を含むポリペプチド。
 (c) (a)又は(b)のポリペプチドと80%以上の配列相同性を有するポリペプチド。
The active ingredient semaphorin 3A may be a natural type or a mutant. Specific examples of natural semaphorin 3A or semaphorin 3A mutant include a region consisting of any of the following polypeptides (a) to (c), and have an activity of preventing and / or treating allergic inflammation in the conjunctiva: The polypeptide which has can be mentioned.
(a) Consists of 70 or more consecutive amino acids in the amino acid sequence shown in SEQ ID NO: 4 in the sequence listing, and 166aa to 235aa in SEQ ID NO: 4 (meaning "166th amino acid to 235th amino acid") A polypeptide comprising the region of
(b) A polypeptide comprising a region of 166aa to 235aa in SEQ ID NO: 2 consisting of 70 or more consecutive amino acids in the amino acid sequence shown in SEQ ID NO: 2 in the sequence listing.
(c) A polypeptide having 80% or more sequence homology with the polypeptide of (a) or (b).
 ここで、「結膜におけるアレルギー性炎症の予防及び/又は治療活性」とは、結膜におけるアレルギー性炎症(例えば、アレルギー性結膜炎等)の結膜病変部の症状を改善又は該症状の増悪を防止する活性を意味する。ポリペプチドが該活性を有するか否かは、例えば、アレルギー性結膜炎を発症している動物に点眼、結膜下投与等によりポリペプチドを投与し、病変部の症状が改善するか否かを評価することによって調べることができる。アレルギー性結膜炎発症動物としては、例えば、下記実施例に記載されるアレルギー性結膜炎発症マウスのようなモデル動物を用いることができる。このマウスは、ブタクサ花粉などを抗原として投与することにより感作し、抗原点眼誘発により、アレルギー性結膜炎を発症させたもので、涙、目やに、充血や浮腫などの臨床症状に加えて、結膜中に浸潤した好酸球が観察される。このマウスを用いた結膜におけるアレルギー性炎症の予防及び/又は治療活性の評価は、下記実施例に具体的に記載するように、例えば、マウスの結膜病変部に点眼と結膜下投与により一定期間ポリペプチドを投与し、病変部の症状の改善の有無を評価することにより行なうことができる。結膜病辺部の症状の改善の有無は、例えば、下記実施例に記載されるように、充血や浮腫などの臨床症状、結膜中に浸潤した好酸球数などを結膜炎の指標として評価することができる。例えば、無処置のアレルギー性結膜炎発症個体群の結膜炎症状のスコア平均値と比較して、一定期間ポリペプチドを投与したアレルギー性結膜炎発症個体群の結膜炎症状のスコア平均値が有意に低下した場合、又は、例えば、ポリペプチド投与群のスコア平均値が無処置個体群のスコア平均値の約3分の2以下、好ましくは約2分の1以下となった場合には、該ポリペプチドはアレルギー性結膜炎の予防及び/又は治療活性を有すると判断できる。 Here, “preventive and / or therapeutic activity of allergic inflammation in the conjunctiva” means an activity that improves or prevents the symptoms of the conjunctival lesion of allergic inflammation (eg, allergic conjunctivitis) in the conjunctiva. Means. Whether or not the polypeptide has the activity is evaluated by, for example, administering the polypeptide to the animal developing allergic conjunctivitis by instillation, subconjunctival administration, etc., and improving the symptoms of the lesion. Can be examined. As an animal having an allergic conjunctivitis, for example, a model animal such as a mouse having an allergic conjunctivitis described in Examples below can be used. This mouse was sensitized by administering ragweed pollen as an antigen, and caused allergic conjunctivitis by inducing antigen instillation. In addition to clinical symptoms such as tears, eyes, and hyperemia and edema, Eosinophils that have infiltrated into are observed. The evaluation of the prevention and / or treatment activity of allergic inflammation in the conjunctiva using this mouse can be carried out by, for example, applying polymorphism for a certain period of time by instillation and subconjunctival administration to the conjunctival lesion of the mouse, as specifically described in the following examples. This can be done by administering the peptide and evaluating the presence or absence of improvement in the symptoms of the lesion. The presence or absence of improvement of symptoms in the conjunctival disease area, for example, as described in the following examples, evaluate clinical symptoms such as hyperemia and edema, the number of eosinophils infiltrated in the conjunctiva as an index of conjunctivitis Can do. For example, compared to the average score value of the conjunctivitis symptoms of the allergic conjunctivitis onset group of the untreated allergic conjunctivitis, the average score value of the conjunctivitis symptoms of the allergic conjunctivitis onset group administered the polypeptide for a certain period significantly decreased, Alternatively, for example, when the average score of the polypeptide administration group is about two-thirds or less, preferably about one-half or less of the average score of the untreated individuals, the polypeptide is allergenic. It can be judged that it has the activity of preventing and / or treating conjunctivitis.
 また、結膜中に浸潤した好酸球数の測定は、アレルギー性結膜炎発症動物から結膜組織を摘出し、標本を作製し、光学顕微鏡で好酸球浸潤を確認して、数を計算することにより行うことができる。例えば、無処置のアレルギー性結膜炎発症個体群の好酸球数の平均値と比較して、一定期間ポリペプチドを投与したアレルギー性結膜炎発症個体群の好酸球数の平均値が有意に低下した場合、又は、例えば、ポリペプチド投与群の好酸球数平均値が無処置個体群の好酸球数平均値の約3分の2以下、好ましくは約2分の1以下となった場合には、該ポリペプチドはアレルギー性結膜炎の予防及び/又は治療活性を有すると判断できる。 The number of eosinophils infiltrated into the conjunctiva is determined by removing the conjunctival tissue from animals with allergic conjunctivitis, preparing a specimen, confirming eosinophil infiltration with an optical microscope, and calculating the number. It can be carried out. For example, the average value of the number of eosinophils in the allergic conjunctivitis-onset population administered with the polypeptide for a certain period was significantly lower than the average value of the number of eosinophils in the untreated allergic conjunctivitis-onset population. Or, for example, when the average eosinophil count of the polypeptide administration group is about 2/3 or less, preferably about 1/2 or less of the average eosinophil count of the untreated population It can be determined that the polypeptide has a prophylactic and / or therapeutic activity for allergic conjunctivitis.
 なお、本発明において、「ポリペプチド」とは、複数(2以上)のアミノ酸がペプチド結合することによって形成される分子をいい、構成するアミノ酸数が多い高分子量の分子のみならず、アミノ酸数が少ない低分子量の分子(オリゴペプチド)や、タンパク質も包含される。 In the present invention, “polypeptide” refers to a molecule formed by peptide bonding of a plurality of (two or more) amino acids, and includes not only high molecular weight molecules having a large number of amino acids but also the number of amino acids. Low molecular weight molecules (oligopeptides) and proteins are also included.
 また、本発明において、「アミノ酸配列を有する」とは、アミノ酸残基がそのような順序で配列しているという意味である。従って、例えば、「配列番号2で示されるアミノ酸配列を有するポリペプチド」とは、Met Gly Trp Leu ・・・(中略)・・・Pro Arg Ser Valのアミノ酸配列を持つ771アミノ酸残基のサイズのポリペプチドを意味する。 In the present invention, “having an amino acid sequence” means that amino acid residues are arranged in such an order. Thus, for example, “a polypeptide having the amino acid sequence represented by SEQ ID NO: 2” means a Met77Gly Trp Leu ... (omitted) ... Pro Arg Ser Val amino acid sequence of 771 amino acid residues Means polypeptide.
 配列番号2及び4に示されるアミノ酸配列は、セマフォリン3Aタンパク質(それぞれヒト及びニワトリ)の配列であり、それぞれGenBankにアクセッション番号NM006080及びU02528として登録されている。セマフォリンとは、神経軸索の伸長をガイドする分子として同定されたタンパク質である。神経突起の先端にある成長円錐を退縮(collapse)させる活性をもとに発見されたことから、コラプシンと呼ばれることもある。セマフォリンは、分泌型又は膜貫通型のタンパク質ファミリーを構成し、現在までに線虫などの無脊椎動物からヒトなどの脊椎動物までで30種類を超えるメンバーが同定されている。セマフォリン3A(Sema3A)は、セマフォリンの分泌型タンパク質メンバーの1つであり、反発性神経軸索ガイダンス分子として知られるタンパク質分子である。Sema3A等の分泌型セマフォリンは、~500aaアミノ末端のセマフォリン(sema)ドメイン、C-2型免疫グロブリン(Ig)ドメイン、及び塩基性末端ドメインの3つの構造ドメインを有する。Sema3Aが神経伸長抑制作用等の生理活性を発揮するのに必要な領域は、semaドメインのうち、特に166~235番目にあたる70アミノ酸の領域であることが知られている(Koppel et al., Neuron, Vol.19, 531-537, 1997)。該70アミノ酸の領域(以下「70aa領域」という)は、配列番号2及び配列番号4のいずれにおいても166aa~235aaの領域である。Sema3Aは、ヒト、ニワトリ、マウス、ラット等の種々の動物から同定されている。これらのSema3Aの相同性は、タンパク質全体として比較するとおおよそ85%前後と高く、上記した70アミノ酸の領域のみで比較すると、95%程度以上とさらに高い。なお、配列番号4のアミノ酸配列を有するニワトリSema3Aでは、semaドメインは61aa~567aa、Igドメインは592aa~655aa、塩基性末端は726aa~772aaである(Feiner et al., Neuron, Vol.19, 539-545, 1997参照)。配列番号2のアミノ酸配列を有するヒトSema3Aでは、semaドメインは61aa~567aa、Igドメインは591aa~654aa、塩基性末端ドメインは726aa~771aaである。 The amino acid sequences shown in SEQ ID NOs: 2 and 4 are semaphorin 3A protein sequences (human and chicken, respectively), and are registered in GenBank as accession numbers NM006080 and U02528, respectively. Semaphorins are proteins that have been identified as molecules that guide nerve axon elongation. It is also called collapsin because it was discovered based on the activity of collapsing the growth cone at the tip of the neurite. Semaphorins constitute a secreted or transmembrane protein family, and more than 30 members have been identified to date from invertebrates such as nematodes to vertebrates such as humans. Semaphorin 3A (Sema3A) is a secreted protein member of semaphorin and is a protein molecule known as a repulsive nerve axon guidance molecule. Secreted semaphorins such as Sema3A have three structural domains: a semaphorin (sema) domain of ~ 500aa amino terminus, a C-2 type immunoglobulin (Ig) domain, and a basic terminal domain. It is known that the region necessary for Sema3A to exert physiological activities such as nerve elongation-inhibiting action is a region of 70 amino acids corresponding to the 166th to 235th of the sema domain (Koppel et al., Neuron , Vol. 19, 531-537, 1997). The 70 amino acid region (hereinafter referred to as “70aa region”) is a region of 166aa to 235aa in both SEQ ID NO: 2 and SEQ ID NO: 4. Sema3A has been identified from various animals such as humans, chickens, mice and rats. The homology of these Sema3A is as high as about 85% when compared as a whole protein, and is even higher as about 95% or more when compared only with the above 70 amino acid region. In the chicken Sema3A having the amino acid sequence of SEQ ID NO: 4, the sema domain is 61aa to 567aa, the Ig domain is 592aa to 655aa, and the basic terminus is 726aa to 772aa (Feiner et al., Neuron, Vol.19, 539) -545, 1997). In human Sema3A having the amino acid sequence of SEQ ID NO: 2, the sema domain is 61aa to 567aa, the Ig domain is 591aa to 654aa, and the basic terminal domain is 726aa to 771aa.
 上記(a)のポリペプチドとは、配列番号4に示されるアミノ酸配列を有するニワトリSema3Aタンパク質のうちの少なくとも70aa領域を含む領域から成るポリペプチドであり、具体的には、配列番号4の全長から成るポリペプチド又は70aa領域を含むその断片である。本発明の予防及び/又は治療剤に有効成分として含有されるポリペプチドの1つの例は、(a)のポリペプチドから成る領域を含み、かつ、結膜におけるアレルギー性炎症の予防及び/又は治療活性を有するポリペプチドである(以下、便宜的に「有効成分ポリペプチドa」と呼ぶ)。有効成分ポリペプチドaとしては、結膜におけるアレルギー性炎症の予防及び/又は治療活性を有する限り、(a)のポリペプチドのみから成るものであってもよく、また、(a)のポリペプチドの一端又は両端にアミノ酸又はポリペプチドが付加されたものであってもよい。 The polypeptide (a) is a polypeptide comprising a region containing at least the 70aa region of the chicken Sema3A protein having the amino acid sequence shown in SEQ ID NO: 4. Specifically, from the full length of SEQ ID NO: 4 Or a fragment thereof containing the 70aa region. One example of a polypeptide contained as an active ingredient in the preventive and / or therapeutic agent of the present invention includes a region comprising the polypeptide of (a) and has an activity of preventing and / or treating allergic inflammation in the conjunctiva. (Hereinafter referred to as “active ingredient polypeptide a” for convenience). The active ingredient polypeptide a may be composed only of the polypeptide (a) as long as it has preventive and / or therapeutic activity for allergic inflammation in the conjunctiva, and may be one end of the polypeptide (a). Alternatively, an amino acid or polypeptide added at both ends may be used.
 (a)のポリペプチドのみから成る有効成分ポリペプチドaの具体例としては、特に限定されないが、例えば、配列番号4に示されるアミノ酸配列の全長から成るポリペプチド(すなわちニワトリSema3Aタンパク質)が挙げられる。ニワトリSema3Aタンパク質が結膜におけるアレルギー性炎症の予防及び/又は治療活性を有することは、下記実施例に具体的に記載される通りである。また、ニワトリSema3Aタンパク質のN末端又はC末端の一部を欠失した断片であっても、結膜におけるアレルギー性炎症の予防及び/又は治療活性を有する限り、有効成分ポリペプチドaとして本発明の範囲に包含される。 Specific examples of the active ingredient polypeptide a consisting only of the polypeptide (a) are not particularly limited, and examples thereof include a polypeptide consisting of the entire amino acid sequence shown in SEQ ID NO: 4 (ie, chicken Sema3A protein). . The fact that chicken Sema3A protein has preventive and / or therapeutic activity for allergic inflammation in the conjunctiva is as specifically described in the following examples. In addition, even if a fragment lacking a part of the N-terminus or C-terminus of chicken Sema3A protein has the activity of preventing and / or treating allergic inflammation in the conjunctiva, it is within the scope of the present invention as an active ingredient polypeptide a. Is included.
 また、(a)のポリペプチドにポリペプチド等が付加された有効成分ポリペプチドaの具体例としては、特に限定されないが、例えば、配列番号4の70aa領域をSema3A以外の分泌型セマフォリンの対応領域に入れ替えて構築したキメラタンパク質、及び(a)のポリペプチドにHisタグ、FLAGタグ、mycタグ等の各種タグ等が付加されたポリペプチド等が挙げられる。上記Sema3A以外の分泌型セマフォリンとしては、例えばコラプシン2、コラプシン3、コラプシン5等が挙げられるが、これらに限定されない。なお、コラプシン2、コラプシン3及びコラプシン5のアミノ酸配列は、Feiner et al., Neuron, vol.19, 539-545 (1997)に記載される通り公知である。Koppelらの上記論文に開示される通り、Sema3Aの少なくとも70aa領域を含む領域をコラプシン2の対応領域に入れ替えて構築したキメラタンパク質は、Sema3Aが有するDRG退縮活性を保持しており、Sema3Aと同様の生理活性を発揮する。従って、このようなキメラタンパク質も、結膜におけるアレルギー性炎症の予防及び/又は治療活性を有するので、有効成分ポリペプチドaとして本発明の範囲に包含される。さらに、Igドメインを欠失したSema3AもDRG退縮活性を有するが(Koppel et al., 1997)、このようなポリペプチドも、Sema3AのうちIgドメインよりもN末端側の70aaを含む領域にSema3Aの塩基性ドメインを付加したポリペプチドとして、有効成分ポリペプチドaに包含され、本発明の範囲に含まれる。なお、上記した具体例は単なる例示であり、(a)のポリペプチドにいかなるアミノ酸又はポリペプチドが付加されたものであっても、結膜におけるアレルギー性炎症の予防及び/又は治療活性を有する限り、有効成分ポリペプチドaとして本発明の範囲に包含される。 In addition, specific examples of the active ingredient polypeptide a in which a polypeptide or the like is added to the polypeptide of (a) are not particularly limited. For example, the 70aa region of SEQ ID NO: 4 corresponds to a secreted semaphorin other than Sema3A. Examples include chimeric proteins constructed by replacing the regions, and polypeptides in which various tags such as His tags, FLAG tags, and myc tags are added to the polypeptide (a). Examples of secretory semaphorins other than Sema3A include, but are not limited to, collapsin 2, collapsin 3, and collapsin 5. The amino acid sequences of collapsin 2, collapsin 3 and collapsin 5 are known as described in Feiner et al., Neuron, vol. 19, 539-545 (1997). As disclosed in the above paper by Koppel et al., The chimeric protein constructed by replacing the region containing at least the 70aa region of Sema3A with the corresponding region of collapsin 2 retains the DRG retraction activity of Sema3A, and is similar to Sema3A. Demonstrate physiological activity. Therefore, such a chimeric protein also has the activity of preventing and / or treating allergic inflammation in the conjunctiva, and thus is included in the scope of the present invention as the active ingredient polypeptide a. Furthermore, Sema3A lacking the Ig domain also has DRG regression activity (Koppel et al., 1997), but such a polypeptide is also present in the region of Sema3A containing 70aa at the N-terminal side of the Ig domain. A polypeptide to which a basic domain is added is included in the active ingredient polypeptide a and is within the scope of the present invention. The specific examples described above are merely examples, and any amino acid or polypeptide added to the polypeptide (a) can be used as long as it has preventive and / or therapeutic activity for allergic inflammation in the conjunctiva. The active ingredient polypeptide a is included in the scope of the present invention.
 上記(b)のポリペプチドとは、配列番号2に示されるアミノ酸配列を有するヒトSema3Aタンパク質のうちの少なくとも70aa領域を含む領域から成るポリペプチドであり、具体的には、配列番号2の全長から成るポリペプチド又は70aa領域を含むその断片である。本発明の予防及び/又は治療剤に有効成分として含有されるポリペプチドのもう1つの例は、(b)のポリペプチドから成る領域を含み、かつ、結膜におけるアレルギー性炎症の予防及び/又は治療活性を有するポリペプチドである(以下、便宜的に「有効成分ポリペプチドb」と呼ぶ)。ニワトリのSema3A(配列番号4)とヒトのSema3A(配列番号2)との相同性は、タンパク質全長では86%であり、Sema3Aの生理活性に重要な70aa領域のみで比較すると95%と更に高い。下記実施例に記載される通り、ニワトリのSema3Aがニワトリではないマウスのアレルギー性結膜炎を治療又は予防できるのであるから、ニワトリのSema3Aを用いてヒト等のニワトリ及びマウス以外のアレルギー性結膜炎を治療又は予防することも可能である。従って、ヒトSema3Aタンパク質等の有効成分ポリペプチドbも、ニワトリSema3Aタンパク質等の上記有効成分ポリペプチドaと同様に、アレルギー性結膜炎等の結膜におけるアレルギー性炎症の予防及び/又は治療剤の調製に有用である。有効成分ポリペプチドbも、上記した有効成分ポリペプチドaと同様に、結膜におけるアレルギー性炎症の予防及び/又は治療活性を有する限り、(b)のポリペプチドのみから成るものであってもよく、また、(b)のポリペプチドの一端又は両端にアミノ酸又はポリペプチドが付加されたものであってもよい。このようなポリペプチドの具体例は、上記有効成分ポリペプチドaと同様であり、例えば、特に限定されないが、配列番号2の全長から成るポリペプチド(すなわちヒトSema3Aタンパク質)、結膜におけるアレルギー性炎症の予防及び/又は治療活性を有するその断片、Sema3A以外の分泌型セマフォリンとの上記態様によるキメラタンパク質、(b)のポリペプチドに各種タグ等が付加されたポリペプチド等が挙げられる。これらの具体例も単なる例示であり、(b)のポリペプチドにいかなるアミノ酸又はポリペプチドが付加された態様のものであっても、結膜におけるアレルギー性炎症の予防及び/又は治療活性を有する限り、有効成分ポリペプチドbとして本発明の範囲に包含される。 The polypeptide of (b) above is a polypeptide comprising a region containing at least the 70aa region of the human Sema3A protein having the amino acid sequence shown in SEQ ID NO: 2, specifically, from the full length of SEQ ID NO: 2. Or a fragment thereof containing the 70aa region. Another example of a polypeptide contained as an active ingredient in the preventive and / or therapeutic agent of the present invention includes a region consisting of the polypeptide (b), and prevents and / or treats allergic inflammation in the conjunctiva. A polypeptide having activity (hereinafter referred to as “active ingredient polypeptide b” for convenience). The homology between chicken Sema3A (SEQ ID NO: 4) and human Sema3A (SEQ ID NO: 2) is 86% over the entire protein length, and is even higher at 95% when compared only with the 70aa region important for the physiological activity of Sema3A. As described in the examples below, chicken Sema3A can treat or prevent allergic conjunctivitis in mice that are not chickens, so chicken sema3A can be used to treat allergic conjunctivitis other than chickens and mice such as humans. It is also possible to prevent. Therefore, the active ingredient polypeptide b such as human Sema3A protein is also useful for the preparation of a preventive and / or therapeutic agent for allergic inflammation in the conjunctiva such as allergic conjunctivitis, like the above active ingredient polypeptide a such as chicken Sema3A protein. It is. Similarly to the active ingredient polypeptide a described above, the active ingredient polypeptide b may be composed only of the polypeptide of (b) as long as it has preventive and / or therapeutic activity for allergic inflammation in the conjunctiva. In addition, an amino acid or polypeptide may be added to one or both ends of the polypeptide (b). Specific examples of such a polypeptide are the same as those of the active ingredient polypeptide a. For example, the polypeptide consisting of the full length of SEQ ID NO: 2 (ie, human Sema3A protein), allergic inflammation in the conjunctiva, is not particularly limited. Examples thereof include fragments thereof having prophylactic and / or therapeutic activity, chimeric proteins according to the above embodiment with secreted semaphorins other than Sema3A, polypeptides obtained by adding various tags to the polypeptide (b), and the like. These specific examples are also merely examples, and any amino acid or polypeptide added to the polypeptide of (b) can be used as long as it has the activity of preventing and / or treating allergic inflammation in the conjunctiva. The active ingredient polypeptide b is included in the scope of the present invention.
 上記(c)のポリペプチドは、上記(a)又は(b)のポリペプチドのうちの少数(好ましくは1ないし数個)のアミノ酸残基が置換し、欠失し及び/又は挿入されたポリペプチドであって、元の配列と80%以上、好ましくは90%以上、より好ましくは95%以上、さらに好ましくは98%以上の相同性を有するポリペプチドである。一般に、タンパク質において、該タンパク質のアミノ酸配列のうち少数のアミノ酸残基が置換され、欠失され又は挿入された場合であっても、元のタンパク質とほぼ同じ機能を有している場合があることは、当業者において広く知られている。従って、上記(c)のポリペプチドから成る領域も、上記した70aa領域と同様にSema3Aタンパク質の生理活性をもたらし得るため、上記(c)のポリペプチドから成る領域を含み、かつ、結膜におけるアレルギー性炎症の予防及び/又は治療活性を有するポリペプチド(以下、便宜的に「有効成分ポリペプチドc」と呼ぶ)も、上記有効成分ポリペプチドa及びbと同様に、本発明の予防及び/又は治療剤の調製に有用である。 The polypeptide (c) is a polypeptide in which a small number (preferably 1 to several) amino acid residues of the polypeptide (a) or (b) are substituted, deleted and / or inserted. Peptide, which is a polypeptide having homology with the original sequence of 80% or more, preferably 90% or more, more preferably 95% or more, and still more preferably 98% or more. In general, a protein may have almost the same function as the original protein even when a small number of amino acid residues in the amino acid sequence of the protein are substituted, deleted, or inserted. Are widely known to those skilled in the art. Therefore, since the region consisting of the polypeptide of (c) can also bring about the physiological activity of the Sema3A protein in the same manner as the 70aa region described above, it includes the region consisting of the polypeptide of (c) above, and is allergic in the conjunctiva. A polypeptide having preventive and / or therapeutic activity for inflammation (hereinafter referred to as “active ingredient polypeptide c” for convenience) is also prevented and / or treated according to the present invention in the same manner as the active ingredient polypeptides a and b. Useful for the preparation of agents.
 ここで、アミノ酸配列の「相同性」とは、比較すべき2つのアミノ酸配列のアミノ酸残基ができるだけ多く一致するように両アミノ酸配列を整列させ、一致したアミノ酸残基数を、全アミノ酸残基数で除したものを百分率で表したものである。上記整列の際には、必要に応じ、比較する2つの配列の一方又は双方に適宜ギャップを挿入する。このような配列の整列化は、例えばBLAST、FASTA、CLUSTAL W等の周知のプログラムを用いて行なうことができる。ギャップが挿入される場合、上記全アミノ酸残基数は、1つのギャップを1つのアミノ酸残基として数えた残基数となる。このようにして数えた全アミノ酸残基数が、比較する2つの配列間で異なる場合には、相同性(%)は、長い方の配列の全アミノ酸残基数で、一致したアミノ酸残基数を除して算出される。なお、天然のタンパク質を構成する20種類のアミノ酸は、低極性側鎖を有する中性アミノ酸(Gly, Ile, Val, Leu, Ala, Met, Pro)、親水性側鎖を有する中性アミノ酸(Asn, Gln, Thr, Ser, Tyr Cys)、酸性アミノ酸(Asp, Glu)、塩基性アミノ酸(Arg, Lys, His)、芳香族アミノ酸(Phe, Tyr, Trp)のように類似の性質を有するものにグループ分けでき、これらの間での置換であればポリペプチドの性質が変化しないことが多いことが知られている。従って、上記(a)又は(b)のポリペプチド中のアミノ酸残基を置換する場合には、これらの各グループの間で置換することにより、有効成分として用いるポリペプチドの結膜におけるアレルギー性炎症の予防及び/又は治療活性を維持できる可能性が高くなる。 Here, the “homology” of the amino acid sequences means that both amino acid sequences are aligned so that the amino acid residues of the two amino acid sequences to be compared match as much as possible, and the number of matched amino acid residues is defined as the total number of amino acid residues. It is the percentage divided by the number. In the above alignment, a gap is appropriately inserted in one or both of the two sequences to be compared as necessary. Such sequence alignment can be performed using a known program such as BLAST, FASTA, CLUSTAL W, and the like. When gaps are inserted, the total number of amino acid residues is the number of residues obtained by counting one gap as one amino acid residue. When the total number of amino acid residues counted in this way is different between the two sequences to be compared, the homology (%) is the total number of amino acid residues in the longer sequence, and the number of amino acid residues matched. Is calculated by dividing. The 20 amino acids constituting the natural protein are neutral amino acids having low side chains (Gly, Ile, Val, Leu, Ala, Met, Pro), and neutral amino acids having hydrophilic side chains (Asn). , Gln, Thr, Ser, Tyr Cys), acidic amino acids (Asp, Glu), basic amino acids (Arg, Lys, His), aromatic amino acids (Phe, Tyr, Trp) It is known that the properties of a polypeptide often do not change as long as it can be grouped and the substitution between them. Therefore, when substituting amino acid residues in the polypeptide of (a) or (b) above, by substituting between these groups, allergic inflammation in the conjunctiva of the polypeptide used as the active ingredient The possibility of maintaining preventive and / or therapeutic activity is increased.
 上記(c)のポリペプチドとは、具体的には、配列番号2若しくは配列番号4で示されるアミノ酸配列の全長から成るポリペプチド(すなわちSema3Aタンパク質)と80%以上の配列相同性を有するポリペプチド、又は、70aa領域を含むSema3Aタンパク質断片と80%以上の配列相同性を有するポリペプチドである。従って、上記有効成分ポリペプチドcとは、これらのポリペプチドのみから成るポリペプチド、又は、(c)のポリペプチドの一端又は両端にアミノ酸又はポリペプチドが付加されたものであって、かつ、結膜におけるアレルギー性炎症の予防及び/又は治療活性を有するポリペプチドである。これらの具体例は、上記有効成分ポリペプチドa及びbと同様であり、特に限定されないが、例えば、配列番号2又は4の全長から成るポリペプチド(すなわちSema3Aタンパク質)と80%以上の配列相同性を有するポリペプチド、結膜におけるアレルギー性炎症の予防及び/又は治療活性を有するSema3Aタンパク質断片と80%以上の配列相同性を有するポリペプチド、70aa領域を含む領域と80%以上の配列相同性を有する領域と、Sema3A以外の分泌型セマフォリンのその他の領域とを融合させたキメラタンパク質、(c)のポリペプチドに各種タグ等が付加されたポリペプチド等が挙げられる。これらの具体例も単なる例示であり、(c)のポリペプチドにいかなるアミノ酸又はポリペプチドが付加された態様のものであっても、結膜におけるアレルギー性炎症の予防及び/又は治療活性を有する限り、有効成分ポリペプチドcとして本発明の範囲に包含される。 The polypeptide (c) is specifically a polypeptide having a sequence homology of 80% or more with a polypeptide consisting of the full length of the amino acid sequence represented by SEQ ID NO: 2 or SEQ ID NO: 4 (ie, Sema3A protein) Or a polypeptide having a sequence homology of 80% or more with a Sema3A protein fragment containing a 70aa region. Therefore, the active ingredient polypeptide c is a polypeptide consisting only of these polypeptides, or an amino acid or polypeptide added to one or both ends of the polypeptide (c), and the conjunctiva A polypeptide having an activity of preventing and / or treating allergic inflammation in Specific examples thereof are the same as those of the active ingredient polypeptides a and b, and are not particularly limited. For example, the polypeptide having the full length of SEQ ID NO: 2 or 4 (ie, Sema3A protein) has a sequence homology of 80% or more. A polypeptide having 80% or more sequence homology with a Sema3A protein fragment having preventive and / or therapeutic activity for allergic inflammation in the conjunctiva, 80% or more sequence homology with a region containing 70aa region Examples include chimeric proteins in which a region and other regions of secreted semaphorins other than Sema3A are fused, polypeptides in which various tags are added to the polypeptide of (c), and the like. These specific examples are also merely examples, and any amino acid or polypeptide added to the polypeptide of (c) may have any activity for preventing and / or treating allergic inflammation in the conjunctiva. The active ingredient polypeptide c is included in the scope of the present invention.
 有効成分ポリペプチドcとしては、特に、70aa領域における相同性が、(c)のポリペプチドから成る領域全体の相同性よりも高いポリペプチドが好ましい。例えば、有効成分ポリペプチドcが、配列番号2のアミノ酸配列を有するポリペプチドと80%の相同性を有するポリペプチドである場合、70aa領域における相同性が80%を超えるものが好ましく、特に、70aa領域における相同性が90%以上、より好ましくは95%以上、さらに好ましくは98%以上、さらに好ましくは70aa領域における配列が同一であるポリペプチドが好ましい。また、例えば、有効成分ポリペプチドcが、ヒトSema3Aタンパク質のsemaドメインと80%の配列相同性を有する領域と、Sema3A以外の分泌型セマフォリンのsemaドメイン以外の領域とを融合させたキメラタンパク質である場合、semaドメイン中の70aa領域における相同性が80%を超えるものが好ましく、特に、70aa領域における相同性が90%以上、より好ましくは95%以上、さらに好ましくは98%以上、さらに好ましくは70aa領域における配列が同一であるキメラタンパク質が好ましい。 The active ingredient polypeptide c is particularly preferably a polypeptide whose homology in the 70aa region is higher than the homology of the entire region comprising the polypeptide (c). For example, when the active ingredient polypeptide c is a polypeptide having 80% homology with the polypeptide having the amino acid sequence of SEQ ID NO: 2, those having a homology in the 70aa region of more than 80% are preferable. Polypeptides having homology in the region of 90% or more, more preferably 95% or more, further preferably 98% or more, and more preferably 70aa region are identical. Further, for example, the active ingredient polypeptide c is a chimeric protein in which a region having 80% sequence homology with the sema domain of human Sema3A protein and a region other than the sema domain of secreted semaphorin other than Sema3A are fused. In some cases, the homology in the 70aa region in the sema domain is preferably more than 80%, in particular, the homology in the 70aa region is 90% or more, more preferably 95% or more, more preferably 98% or more, more preferably Chimeric proteins having the same sequence in the 70aa region are preferred.
 上記した有効成分ポリペプチドaないしcの中でも、特に、配列番号2又は配列番号4で示されるアミノ酸配列を有するポリペプチドと80%以上、好ましくは90%以上、より好ましくは95%以上、さらに好ましくは98%以上の相同性を有するポリペプチドが好ましく、この中でも、上記したように、70aa領域における相同性がポリペプチド全体としての相同性よりも高いポリペプチドが好ましい。最も好ましくは、本発明の予防及び/又は治療剤に有効成分として含まれるポリペプチドは、配列番号2又は配列番号4で示されるアミノ酸配列を有するポリペプチドである。 Among the active ingredient polypeptides a to c described above, in particular, 80% or more, preferably 90% or more, more preferably 95% or more, and further preferably, a polypeptide having the amino acid sequence represented by SEQ ID NO: 2 or SEQ ID NO: 4. Is preferably a polypeptide having a homology of 98% or more. Among them, as described above, a polypeptide having a homology in the 70aa region higher than the homology of the entire polypeptide is preferable. Most preferably, the polypeptide contained as an active ingredient in the preventive and / or therapeutic agent of the present invention is a polypeptide having the amino acid sequence represented by SEQ ID NO: 2 or SEQ ID NO: 4.
 一般に、ポリペプチドから成る医薬において、生体内でのポリペプチドの安定性を高めるために、ポリペプチドに糖鎖やポリエチレングリコール(PEG)鎖を付加したり、ポリペプチドを構成するアミノ酸の少なくとも一部としてD体アミノ酸を用いたりする技術が広く知られており、用いられている。糖鎖やPEG鎖を付加したり、ポリペプチドを構成するアミノ酸の少なくとも一部としてD体アミノ酸を用いたりすることにより、生体内でのペプチダーゼによる分解を受けにくくなり、生体内におけるポリペプチドの半減期が長くなる。本発明に用いられるポリペプチドは、結膜におけるアレルギー性炎症の予防及び/又は治療活性を有する限り、生体内安定化のためのこれらの公知の修飾を施したものであってもよく、本明細書及び特許請求の範囲における「ポリペプチド」という語は、文脈上そうでないことが明らかな場合を除き、生体内安定化のための修飾を施したものも包含する意味で用いている。 In general, in a pharmaceutical comprising a polypeptide, a sugar chain or a polyethylene glycol (PEG) chain is added to the polypeptide or at least a part of the amino acids constituting the polypeptide in order to increase the stability of the polypeptide in vivo. A technique using a D-form amino acid is widely known and used. By adding sugar chains or PEG chains, or by using D-form amino acids as at least part of the amino acids that make up the polypeptide, it is less susceptible to degradation by peptidases in vivo, reducing the polypeptide in vivo by half. The period becomes longer. The polypeptide used in the present invention may be subjected to these known modifications for in vivo stabilization as long as it has preventive and / or therapeutic activity for allergic inflammation in the conjunctiva. The term “polypeptide” in the claims and in the claims is used to include those modified for in vivo stabilization, unless the context clearly indicates otherwise.
 ポリペプチドに対する糖鎖付加は周知であり、例えば、Sato M, Furuike T, Sadamoto R, Fujitani N, Nakahara T, Niikura K, Monde K, Kondo H, Nishimura S., "Glycoinsulins: dendritic sialyloligosaccharide-displaying insulins showing a prolonged blood-sugar-lowering activity.",J Am Chem Soc. 2004 Nov 3;126(43):14013-22やSato M, Sadamoto R, Niikura K, Monde K, Kondo H, Nishimura S,"Site-specific introduction of sialic acid into insulin.", Angew Chem Int Ed Engl. 2004 Mar 12;43(12):1516-20等に記載されている。糖鎖は、N末端、C末端又はそれらの間のアミノ酸に結合可能であるが、ポリペプチドの活性を阻害しないためにN末端又はC末端に結合することが好ましい。また、付加する糖鎖の個数は、1個又は2個が好ましく、1個が好ましい。糖鎖は、単糖から4糖が好ましく、さらには2糖又は3糖が好ましい。糖鎖は、ポリペプチドの遊離のアミノ基又はカルボキシル基に直接又は例えば炭素数1~10程度のメチレン鎖等のスペーサー構造を介して結合することができる。 Glycosylation to polypeptides is well known, for example, Sato M, Furuike T, Sadamoto R, Fujitani N, Nakahara T, Niikura K, Monde K, Kondo H, Nishimura S., "Glycoinsulins: dendritic sialylolulins displaying a prolonged blood-sugar-lowering activity. ", J Am Chem Soc. 2004 Nov 3; 126 (43): 14013-22, Sato M, Sadamoto R, Niikura K, Monde K, Kondo H, Nishimura S," Site- specific introduction of sialic acid into insulin. ", Angew Chem Int Ed Engl. 2004 Mar 12; 43 (12): 1516-20. The sugar chain can be bound to the N-terminus, C-terminus, or an amino acid therebetween, but is preferably bound to the N-terminus or C-terminus so as not to inhibit the activity of the polypeptide. The number of sugar chains to be added is preferably 1 or 2, and preferably 1. The sugar chain is preferably monosaccharide to tetrasaccharide, more preferably disaccharide or trisaccharide. The sugar chain can be bound to a free amino group or carboxyl group of the polypeptide directly or via a spacer structure such as a methylene chain having about 1 to 10 carbon atoms.
 ポリペプチドに対するPEG鎖の付加も周知であり、例えば、Ulbricht K, Bucha E, Poschel KA, Stein G, Wolf G, Nowak G., "The use of PEG-Hirudin in chronic hemodialysis monitored by the Ecarin Clotting Time: influence on clotting of the extracorporeal system and hemostatic parameters.", Clin Nephrol. 2006 Mar;65(3):180-90.やDharap SS, Wang Y, Chandna P, Khandare JJ, Qiu B, Gunaseelan S, Sinko PJ, Stein S, Farmanfarmaian A, Minko T., "Tumor-specific targeting of an anticancer drug delivery system by LHRH peptide.", Proc Natl Acad Sci USA. 2005 Sep 6;102(36):12962-7."等に記載されている。PEG鎖は、N末端、C末端又はそれらの間のアミノ酸に結合可能であり、通常、1個又は2個のPEG鎖が、ポリペプチド上の遊離のアミノ基やカルボキシル基に結合される。PEG鎖の分子量は、特に限定されないが、通常3000~7000程度、好ましくは5000程度のものが用いられる。 Addition of PEG chains to polypeptides is also well known, e.g. Ullbricht K, Bucha E, Poschel KA, Stein G, Wolf G, Nowak G., '' The use of PEG-Hirudin in chronic hemodialysis monitored by the Ecarin Clotting influence on clotting of the extracorporeal system and hemostatic parameters. ", Clin Nephrol. 2006 Mar; 65 (3): 180-90., Dharap SS, Wang Y, Chandna P, Khandare JJ, Qiu B, Gunaseelan S, Sinko Stein S, Farmanfarmaian A, Minko T., "Tumor-specific targeting of an anticancer drug delivery system by LHRH peptide.", Proc Natl Acad Sci USA. 2005 Sep 6; 102 (36): 12962-7. " The PEG chain can be attached to the N-terminus, C-terminus, or the amino acid between them, and usually one or two PEG chains are attached to a free amino group or carboxyl group on the polypeptide. The molecular weight of the PEG chain is not particularly limited, but is usually 3000 to 7000. Degrees, preferably those of about 5000 is used.
 ポリペプチドを構成するアミノ酸の少なくとも一部をD体とする方法も周知であり、例えば、Brenneman DE, Spong CY, Hauser JM, Abebe D, Pinhasov A, Golian T, Gozes I., "Protective peptides that are orally active and mechanistically nonchiral.", J Pharmacol Exp Ther. 2004 Jun;309(3):1190-7やWilkemeyer MF, Chen SY, Menkari CE, Sulik KK, Charness ME., "Ethanol antagonist peptides: structural specificity without stereospecificity.", J Pharmacol Exp Ther. 2004 Jun;309(3):1183-9.等に記載されている。ポリペプチドを構成するアミノ酸の一部をD体としてもよいが、ポリペプチドの活性をできるだけ阻害しないため、一部のみをD体にするよりは、ポリペプチドを構成するアミノ酸の全てをD体アミノ酸とすることが好ましい。 A method in which at least a part of amino acids constituting a polypeptide is D-form is also known, for example, Brenneman DE, Spong CY, Hauser JM, Abebe D, Pinhasov A, Golian T, Gozes I., "Protective peptides that are orally active and mechanistically nonchiral. ", J Pharmacol Exp Ther. 2004 Jun; 309 (3): 1190-7 and Wilkemeyer MF, Chen SY, Menkari CE, Sulik KK, Charness ME.," Ethanol antagonist peptides: structural specificity without "J. Pharmacol. Exp. Ther. 2004. Jun; 309 (3): 1183-9. A part of the amino acids constituting the polypeptide may be D-form, but since the activity of the polypeptide is not inhibited as much as possible, all of the amino acids constituting the polypeptide are D-form amino acids rather than only a part of the amino acids. It is preferable that
 本発明で有効成分として用いられるセマフォリン3Aは、例えば市販のペプチド合成機を用いて常法により容易に調製することができる。また、公知の遺伝子工学的手法を用いて容易に調製することができる。例えば、Sema3A遺伝子を発現している組織から抽出したRNAから、Sema3A遺伝子のcDNAをRT-PCRにより調製し、該cDNAの全長又は所望の一部を発現ベクターに組み込んで、宿主細胞中に導入し、目的とするポリペプチドを得ることができる。RNAの抽出、RT-PCR、ベクターへのcDNAの組み込み、ベクターの宿主細胞への導入は周知の方法により行なうことができる。また、用いるベクターや宿主細胞も周知であり、種々のものが市販されている。キメラタンパク質の作製方法もこの分野で周知であり、例えば上記Koppelらの論文に記載される方法でキメラタンパク質を作製することができる。また、上記安定化修飾も、上記各文献に記載されているような周知の方法により容易に行なうことができる。 Semaphorin 3A used as an active ingredient in the present invention can be easily prepared by a conventional method using, for example, a commercially available peptide synthesizer. Moreover, it can prepare easily using a well-known genetic engineering method. For example, a Sema3A gene cDNA is prepared by RT-PCR from RNA extracted from a tissue expressing the Sema3A gene, and the full length or a desired part of the cDNA is incorporated into an expression vector and introduced into a host cell. The desired polypeptide can be obtained. RNA extraction, RT-PCR, cDNA integration into a vector, and introduction of a vector into a host cell can be performed by known methods. Further, vectors and host cells to be used are well known, and various types are commercially available. A method for producing a chimeric protein is also well known in this field. For example, a chimeric protein can be produced by the method described in the above-mentioned paper by Koppel et al. The stabilization modification can also be easily performed by a known method as described in each of the above documents.
 本発明の予防及び/又は治療剤における有効成分であるセマフォリン3Aは、結膜におけるアレルギー性炎症の治療及び/又は予防に有効である。結膜におけるアレルギー性炎症としては、好酸球性結膜疾患、アレルギー性結膜炎(花粉性結膜炎などの季節性アレルギー性結膜炎を含む)、アレルギー性角結膜炎、アレルギー性眼瞼炎、花粉症、アトピー性角結膜炎、アレルギー性瞼皮膚炎などを挙げることができ、セマフォリン3Aは、これらの疾患の予防初期治療(療法)、季節前投与及び/又は治療剤保存的治療剤、維持療法・導入療法に有用である。 Semaphorin 3A, which is an active ingredient in the preventive and / or therapeutic agent of the present invention, is effective in treating and / or preventing allergic inflammation in the conjunctiva. Allergic inflammation in the conjunctiva includes eosinophilic conjunctival disease, allergic conjunctivitis (including seasonal allergic conjunctivitis such as pollen conjunctivitis), allergic keratoconjunctivitis, allergic blepharitis, hay fever, atopic keratoconjunctivitis Semaphorin 3A is useful for preventive initial treatment (therapy) of these diseases, pre-seasonal administration and / or therapeutic agent conservative treatment, maintenance therapy / induction therapy. is there.
 本発明の予防及び/又は治療剤の投与対象は哺乳動物であり、例えばヒト、イヌ、ネコ、ウサギ、ハムスター等が挙げられる。なお、予防又は治療対象の患者と同一の生物種由来のセマフォリン3Aを用いた方が、予防又は治療効果も高いと考えられるし、臨床応用上の安全性の観点からも望ましい。従って、例えば、本発明の予防及び/又は治療剤の投与対象がヒトの場合には、上記した有効成分ポリペプチドbを有効成分とする予防及び/又は治療剤が特に好ましい。 The subject of administration of the preventive and / or therapeutic agent of the present invention is a mammal, and examples thereof include humans, dogs, cats, rabbits, hamsters and the like. The use of semaphorin 3A derived from the same species as the patient to be prevented or treated is considered to have a higher prevention or treatment effect, and is also desirable from the viewpoint of safety in clinical application. Therefore, for example, when the subject to which the preventive and / or therapeutic agent of the present invention is administered is a human, the prophylactic and / or therapeutic agent containing the active ingredient polypeptide b as an active ingredient is particularly preferable.
 本発明の予防及び/又は治療剤は、セマフォリン3Aのみから成っていてもよいし、各投与形態に適した薬理学的に許容される賦形剤、安定化剤、保存剤、緩衝剤、溶解補助剤、乳化剤、希釈剤、等張化剤などの添加剤を適宜混合させて製剤とすることもできる。製剤化方法及び使用可能な添加剤は、医薬製剤の分野において周知であり、いずれの方法及び添加剤をも用いることができる。 The preventive and / or therapeutic agent of the present invention may consist of semaphorin 3A alone, or a pharmaceutically acceptable excipient, stabilizer, preservative, buffer, suitable for each dosage form, Additives such as solubilizers, emulsifiers, diluents, tonicity agents and the like can be mixed as appropriate to prepare a preparation. Formulation methods and usable additives are well known in the field of pharmaceutical preparations, and any method and additive can be used.
 本発明の予防及び/又は治療剤は、予防すべき結膜部又は治療すべき結膜病変部に投与して用いられる。投与方法としては、局所投与(結膜上への点眼投与、結膜下注射投与など)などが挙げられる。剤型としては、眼軟膏剤、注射剤、点眼剤などが挙げられる。投与量は、症状、年齢、体重、投与方法等に応じて適宜選択され、特に限定されないが、点眼量の場合、通常、対象動物に対し有効成分量として1日6000~60000 U程度、好ましくは4000~40000 U程度であり、結膜下注射量の場合、通常、対象動物に対し有効成分量として1日500~5000 U程度、好ましくは500~5000 U程度であり、1回ないし数回に分けて投与される。好ましくは、症状の改善の程度に応じ、数日ないし数ヶ月間にわたり、毎日1回若しくは数回、ないしは数日おきに1日若しくは数回、定期的に投与がなされる。ここで、「1 U」とは、常法により得られる精製ポリペプチド溶液の原液でNGF感受性DRGニューロンの50%の成長円錐を退縮(collapse)させるセマフォリン力価のことであり、退縮アッセイ(collapse assay)(Luoet al., Cell vol.75, 217-227, 1993)により調べることができる。 The preventive and / or therapeutic agent of the present invention is used by administering to the conjunctival part to be prevented or the conjunctival lesion part to be treated. Examples of the administration method include local administration (instillation on the conjunctiva, administration by subconjunctival injection, etc.) and the like. Examples of the dosage form include eye ointments, injections, eye drops and the like. The dosage is appropriately selected according to symptoms, age, body weight, administration method and the like, and is not particularly limited. In the case of eye drops, the amount of active ingredient is usually about 6000 to 60000 U per day for the target animal, preferably In the case of subconjunctival injection amount, the amount of active ingredient is usually about 500-5000 U per day, preferably about 500-5000 U, and divided into 1 to several times. Administered. Preferably, depending on the degree of symptom improvement, it is administered once or several times daily for several days to several months, or regularly once or several times every other day. Here, “1 U” is a semaphorin titer that collapses a growth cone of 50% of NGF-sensitive DRG neurons with a stock solution of a purified polypeptide solution obtained by a conventional method. collapse assay) (Luoet al., Cell vol.75, 217-227, 1993).
 本発明の予防及び/又は治療剤は、単独で用いてもよいし、他の予防及び/又は治療剤等と併用することもできる。本発明の予防及び/又は治療剤は、従来の抗アレルギー点眼薬である抗ヒスタミン点眼薬、非ステロイド点眼薬やステロイド点眼薬とは作用機序が異なるため、これらと併用することができる。 The preventive and / or therapeutic agent of the present invention may be used alone or in combination with other prophylactic and / or therapeutic agents. The prophylactic and / or therapeutic agent of the present invention can be used in combination with anti-histamine eye drops, non-steroid eye drops and steroid eye drops, which are conventional anti-allergic eye drops.
 以下、実施例に基づいて本発明を詳細に説明するが、本発明はこれらの実施例に限定されるものではない。 Hereinafter, the present invention will be described in detail based on examples, but the present invention is not limited to these examples.
〔実施例1〕
(材料と方法)
 ブタクサ花粉(short ragweed pollen: RW)を抗原とし、水酸化アルミニウム(Alum)とのエマルジョンを作成し,BALB/cマウスに10日間隔で計2回腹腔内投与した。感作後RWを点眼することによりマウスにアレルギー性結膜炎(EC)を誘導し,即発相は点眼30分後に臨床スコアを観察し、評価し、点眼24時間後に結膜を採取し、浸潤好酸球数を計数した。対照として正常マウスにPBSを点眼した。遅発相は点眼開始から1週間後にサンプルをとった。浸潤好酸球数を計数した。
[Example 1]
(Materials and methods)
Ragweed pollen (short ragweed pollen: RW) was used as an antigen and an emulsion with aluminum hydroxide (Alum) was prepared and administered intraperitoneally twice to BALB / c mice at 10-day intervals. Instillation of RW after sensitization induces allergic conjunctivitis (EC) in mice, and the immediate phase observes and evaluates clinical scores 30 minutes after instillation, collects conjunctiva 24 hours after instillation, and infiltrate eosinophils Numbers were counted. As a control, normal mice were instilled with PBS. The late phase was sampled one week after the start of instillation. The number of infiltrating eosinophils was counted.
1.動物
 実験には、5週齢で日本SLC 株式会社(浜松、日本)より購入した BALB/cCrSlc雌マウスを用いた。マウスは室温 25±1℃、湿度55±5%の恒温恒湿室で 12 時間サイクルの明暗条件下で、また標準的なマウス餌(固形飼料:5LJ1(PMI FEEDS) および滅菌水を与えて飼育した。マウスは1群6匹とし、合計120匹を用いた。 
1. For animal experiments, BALB / cCrSlc female mice purchased from Japan SLC Co., Ltd. (Hamamatsu, Japan) at the age of 5 weeks were used. Mice are raised in a constant temperature and humidity chamber at room temperature 25 ± 1 ° C and humidity 55 ± 5% under light and dark conditions with a 12-hour cycle, and with standard mouse food (solid feed: 5LJ1 (PMI FEEDS) and sterile water) There were 6 mice per group, and a total of 120 mice were used.
2.アレルギー性結膜炎の誘発
 能動免疫によるアレルギー性結膜炎(EC)の誘発。ブタクサ花粉(RW)を腹腔に10日間隔で計2 回投与による能動感作 および抗原液を点眼チャレンジすることにより誘発した。 
・アレルギー性結膜疾患誘発方法: RWを抗原とし、Alumとエマルジョンを作成した。RWはPolysciences(ウォリントン、ペンシルバニア州、アメリカ合衆国)から購入した。水酸化アルミニウム(みょうばん)はシグマ(セントルイス、ミズーリ州、アメリカ合衆国)から購入した。
・ブタクサ花粉(RW)を腹腔内注射することにより全身感作を行った。腹腔に10日間隔で計2 回投与による能動感作、全身感作10日後に抗原RWをPBSに溶かして、点眼チャレンジし、アレルギー性結膜炎を誘発し、即時相は、結膜炎症状を点眼後30分間観察し、24時後組織サンプルをとった。 遅発相は、チャレンジ1週間後にサンプルをとった。充血、浮腫、涙、眼やに等の結膜炎症状を点眼後30分間観察し、評価した。


































2. Induction of allergic conjunctivitis Induction of allergic conjunctivitis (EC) by active immunity. Ragweed pollen (RW) was induced in the abdominal cavity by active sensitization by administration twice in total at 10-day intervals and by instillation challenge with an antigen solution.
・ Allergic conjunctival disease induction method: Alum and emulsion were prepared using RW as an antigen. RW was purchased from Polysciences (Warrington, Pennsylvania, USA). Aluminum hydroxide was purchased from Sigma (St. Louis, Missouri, USA).
-Whole body sensitization was performed by intraperitoneal injection of ragweed pollen (RW). Active sensitization by administration twice at a 10-day interval in the abdominal cavity, antigen RW was dissolved in PBS 10 days after systemic sensitization and challenged by instillation to induce allergic conjunctivitis. The tissue sample was taken after 24 hours. The late phase was sampled one week after challenge. Conjunctival inflammation such as hyperemia, edema, tears, and eyes was observed for 30 minutes after instillation and evaluated.


































Figure JPOXMLDOC01-appb-T000001
参考
(*細川 友和 ら スギ花粉誘発によるアレルギー性結膜炎に及ぼすディーゼル排気曝露の影響http://www.env.go.jp/chemi/report/h16-13/02_2_2.pdf)
(*高村ら アレルギー・免疫Vol.15,No3,2008)
Figure JPOXMLDOC01-appb-T000001
Reference (* Tomokazu Hosokawa et al. Effects of diesel exhaust exposure on allergic conjunctivitis induced by cedar pollen http://www.env.go.jp/chemi/report/h16-13/02_2_2.pdf)
(* Takamura et al. Allergy / Immunology Vol.15, No3,2008)
3.Sema3A投与 
 セマフォリン3Aは、既報のように調製した(Journal of Investigative Dermatology (2008) 128, 2842-2849)。簡単に説明すると、ニワトリセマフォリン3A安定発現株を培養し、常法により、培養上清に生産されたセマフォリン3Aタンパク質を精製した。なお、該セマフォリン3A安定発現株は、ニワトリのセマフォリン3A遺伝子(配列番号3)をHEK293細胞に組み込んだものである。 得られたセマフォリン3Aタンパク質の活性は、ニワトリの後根神経節を用いた退縮アッセイ(collapse assay)により測定した。退縮アッセイは、以下の方法により行なった。
3. Sema3A administration
Semaphorin 3A was prepared as previously reported (Journal of Investigative Dermatology (2008) 128, 2842-2849). Briefly, a chicken semaphorin 3A stable expression strain was cultured, and semaphorin 3A protein produced in the culture supernatant was purified by a conventional method. The semaphorin 3A stable expression strain is obtained by incorporating a chicken semaphorin 3A gene (SEQ ID NO: 3) into HEK293 cells. The activity of the obtained semaphorin 3A protein was measured by a collapse assay using the dorsal root ganglion of the chicken. The regression assay was performed by the following method.
 胚性7日目のニワトリ卵胚から後根神経節(DRG)を摘出した。摘出したDRGをNGF含有メディウム入りのガラス底面にのせ、37℃のインキュベーター内で24時間静地培養した。NGF感受性DRGニューロンが伸長しているのを確認後、上記の通り精製して得たセマフォリン3Aの溶液を1倍(原液)~10万倍に希釈して投与し、37℃で30分静地培養後固定した。顕微鏡下にて、全NGF感受性DRGニューロンのうち、成長円錐が退縮(collapse)しているニューロンの割合(%)を算出し、50%のcollapseを認めるセマフォリン3A濃度(X倍希釈)をX U/mlとした。 Dorsal root ganglia (DRG) were extracted from embryonic day 7 chick egg embryos. The extracted DRG was placed on the bottom of a glass containing NGF-containing medium, and cultured in a static place for 24 hours in a 37 ° C. incubator. After confirming that NGF-sensitive DRG neurons are growing, the semaphorin 3A solution obtained by purification as described above is diluted 1-fold (stock solution) to 100,000-fold and administered at 37 ° C for 30 minutes. Fixed after ground culture. Under the microscope, calculate the percentage (%) of all NGF-sensitive DRG neurons that have collapsed growth cones, and determine the semaphorin 3A concentration (X-fold dilution) at which 50% collapse is observed. U / ml.
 退縮アッセイの結果、上記で得られたセマフォリン3Aの力価は3万 U/mLであった。 As a result of the regression assay, the titer of semaphorin 3A obtained above was 30,000 μU / mL.
 セマフォリン3A (semaphorin-3A) 10,100,1000ユニットの濃度において、 動物の両眼 に毎日2回点眼した。抗原溶液は、1 回点眼量 10 μlとした。結膜下投与は、動物の両眼に1日間隔で2μlを結膜下に投与した。 Semaphorin-3A was instilled twice daily on both eyelids at a concentration of 10,100,1000 units. The antigen solution was made into a single eye drop of 10 μl μl. For subconjunctival administration, 2 μl was administered subconjunctivally to both eyes of the animal at intervals of 1 day.
4.対照群は同様の使用量でPBSを投与した。臨床スコア群は4週間にわたって観察した。  4). In the control group, PBS was administered at the same use amount. The clinical score group was observed over 4 weeks.
5.サンプリングはPBS、点眼、結膜下投与群で、それぞれ6、10、14日、計3回実施した。 5. Sampling was performed in PBS, eye drops, and subconjunctival administration groups for 6, 10, and 14 days, for a total of 3 times.
6.免疫T cell増殖の抑制確認
 正常BALB / cマウスから脾臓を摘出した。脾細胞を調製し適当な濃度(6.25×105個/mLから2.5×106個/mL)の脾細胞100 mLを抗CD3抗体(BD ファーミンジェン)でコーティングされたT細胞プレートに播種し、Semaphorin-3A 10,100,1000ユニットの濃度において投与し、対照群はPBSを投与した。
6). Confirmation of suppression of immune T cell proliferation The spleen was extracted from normal BALB / c mice. Prepare spleen cells and seed 100 mL of spleen cells of appropriate concentration (6.25 × 10 5 cells / mL to 2.5 × 10 6 cells / mL) onto a T cell plate coated with anti-CD3 antibody (BD Pharmingen). Semaphorin-3A was administered at a concentration of 10,100,1000 units, and the control group was administered PBS.
 CO2インキュベータに入れて37℃で45~48時間培養した。(プレート(抗マウスCD3)にBD バイオコートTM T細胞活性化プレート(抗マウスCD3)使用)
 マイクロプレート分光光度計で、対照波長700 nmで測定波長490 nmのODを測定した。in vitroでSema3AはT cell増殖の抑制が確認された(図4)。T細胞が好酸球を誘導することが明らかとなった。(福島敦樹:アレルギーの臨床28(4),2008)。
The cells were cultured in a CO 2 incubator at 37 ° C. for 45 to 48 hours. (Use of BD Biocoat TM T cell activation plate (anti-mouse CD3) on plate (anti-mouse CD3))
The OD at a measurement wavelength of 490 nm was measured with a microplate spectrophotometer at a control wavelength of 700 nm. In vitro, Sema3A was confirmed to suppress T cell proliferation (FIG. 4). It was revealed that T cells induce eosinophils. (Yuki Fukushima: Clinical study of allergies 28 (4), 2008).
 セマフォリン分子群は、従来、神経ガイダンス因子として知られてきたが、近年、免疫系においても種々のセマフォリン分子が関与していることが明らかになってきている。神経系と免疫系は、多くの共通性があることが以前より知られている。とくに免疫系における抗原提示細胞-T細胞間相互作用に関与するセマフォリン分子が発見された。in vitroでセマフォリン3A分子がT細胞レセプター複合体抗体をコーティングしたプレートで、T細胞活性化誘導、T細胞の増殖を抑制することが確認されている。 The semaphorin molecule group has heretofore been known as a nerve guidance factor, but recently, it has become clear that various semaphorin molecules are also involved in the immune system. It has long been known that the nervous system and the immune system have a lot in common. In particular, semaphorin molecules involved in antigen-presenting cell-T cell interactions in the immune system have been discovered. It has been confirmed in vitro that a semaphorin 3A molecule is coated with a T cell receptor complex antibody to induce T cell activation and suppress T cell proliferation.
 TordjmanらはNeuropilin-1はヒトの樹状細胞と休止期T細胞に発見し、T細胞はNeuropilin-1発現Cos7細胞に結合し、樹状細胞とナイーブT細胞の接触面においてNeuropilin-1はCD3とco-localizeする。また、抗Neuropilin-1抗体は樹状細胞によるT細胞の増殖を抑制する。神経系におけるリカンドであるセマフォリン3A分子には、免疫細胞の遊走を抑制する作用があるが、Neuropilinに対する抗体でその作用が阻害できないから、Sema3AはT細胞増殖の抑制は免疫系ではNeuropilin-1と異なる受容体により担われていることが示唆される。 Tordjman et al. Discovered that Neuropilin-1 was found in human dendritic cells and resting T cells, T cells bound to Neuropilin-1-expressing Cos7 cells, and Neuropilin-1 was CD3 at the interface between dendritic cells and naive T cells. And co-localize. Anti-Neuropilin-1 antibody also suppresses T cell proliferation by dendritic cells. The semaphorin 3A molecule, which is a glycan in the nervous system, has the effect of suppressing the migration of immune cells, but its action cannot be inhibited by an antibody against Neuropilin. Therefore, Sema3A suppresses the proliferation of T cells in the immune system. It is suggested that it is carried by different receptors.
7.Sema3A点眼薬として速効性の確認
点眼薬投与
 アレルギー性結膜炎に対する効果は、Sema3A 1000U/ml点眼液、ヒスタミンH1受容体拮抗薬0.1%オロパタジン塩酸塩点眼液、0.025%レボカバスチン塩酸塩点眼液、メディエーター遊離抑制薬0.5%トラニラスト点眼液、0.01%イブジラスト点眼液、ステロイド薬0.02%フルオロメトロン点眼液又はPBS(コントロール)を点眼後15分にRW抗原液を点眼し、結膜炎及び結膜炎抑制作用をそれぞれ比較した。その後の抗原惹起による結膜炎症状を各測定時点で観察し、結膜炎スコアを付け、そのスコアを指標として結膜炎抑制作用を評価した。
7). Sema3A ophthalmic solution is confirmed to be fast-acting The effect on allergic conjunctivitis is Sema3A 1000 U / ml ophthalmic solution, histamine H1 receptor antagonist 0.1% olopatadine hydrochloride ophthalmic solution, 0.025% levocabastine hydrochloride ophthalmic solution, mediator release inhibition RW antigen solution was instilled 15 minutes after instillation of the drug 0.5% tranilast ophthalmic solution, 0.01% ibudilast ophthalmic solution, steroid drug 0.02% fluorometholone ophthalmic solution or PBS (control), and the conjunctivitis and conjunctivitis inhibitory effects were compared. Subsequent conjunctivitis symptoms due to antigen induction were observed at each measurement time point, and a conjunctivitis score was assigned.
 充血、浮腫、目やに、涙等の結膜炎症状を点眼後30分間観察し、表1に示した結膜炎の症状に対するスコアにより評価した。1 回点眼量 10 μl とし、動物の両眼に毎日2回で計6 日間点眼した。 Conjunctivitis symptoms such as hyperemia, edema, eyes, and tears were observed for 30 minutes after instillation, and evaluated according to the scores for conjunctivitis symptoms shown in Table 1. The amount of instillation was 1 10 μ, and the animals were instilled twice daily for 6 days in total.
 即時相スコアでは、コントロール3+(中等度)、点眼薬は充血、浮腫、目やに、涙1~2+に対し、Sema3Aは±0であり、治療効果が示唆された(図3上)。 In the immediate phase score, Sema3A was ± 0 for control 3+ (moderate), eye drops for hyperemia, edema, eyes and tears 1 to 2+, suggesting a therapeutic effect (upper figure 3).
 遅発相スコアでは、コントロール4+(重度)、点眼薬は充血、浮腫、目やに、涙2~3+に対し、Sema3Aは2+であり、0.02%フルオロメトロンと対応する治療効果が示唆された(図3下)。 In the late phase score, control 4+ (severe), eye drops were congested, edema, eyes and tears 2 to 3+, Sema3A was 2+, suggesting a therapeutic effect corresponding to 0.02% fluorometholone (Fig. 3) under).
8.結膜組織の摘出と病理組織学的解析
 結膜を含む目について、眼瞼の周囲の毛を刈り落とし、眼瞼周囲を幅約 5 mm に切断してから眼瞼の先を約 2 mm 残して眼瞼周囲を切り落とした。全体の眼瞼と球結膜固有層を10%の緩衝ホルマリンで固定した。
8). Removal of the conjunctival tissue and histopathological analysis For eyes containing the conjunctiva, the hair around the eyelid is shaved off, the area around the eyelid is cut to a width of about 5 mm, and the tip of the eyelid is left behind to leave about 2 mm. It was. The entire eyelid and bulbar conjunctival lamina were fixed with 10% buffered formalin.
ギムザ染色
 粘膜中の好酸球浸潤を確認し、セクションは盲検のサンプルを与えられた2つのオブザーバでカウントされた。
Giemsa staining Confirmed infiltration of eosinophils in the mucosa, sections were counted in two observers given blinded samples.
 カウントのセクションは、中央部から瞳孔と視神経乳頭が含まれる範囲とした。データはすべてのマウスの平均値±標準誤差、一元配置分散分析及びTukey-Kramerの多重比較検定として検討した。 The count section was the range including the pupil and optic disc from the center. Data were examined as mean ± standard error of all mice, one-way analysis of variance and Tukey-Kramer multiple comparison test.
蛍光染色
Sema3Aの組織貯留確認。
Fluorescent staining
Sema3A tissue storage confirmation.
 マウス結膜は、OCT化合物(VWR、サワニー、ジョージア州、米国)に埋め込まれていた液体窒素中で凍結した。これを調製し、メタノールで固定した。 Mouse conjunctiva was frozen in liquid nitrogen embedded in OCT compound (VWR, Sawanee, GA, USA). This was prepared and fixed with methanol.
一次抗体
(ウサギ由来ポリクロナール抗体)Anti-Semaphorin 3A
メーカー Abcam, Cambridge
二次抗体
(チキン由来)ウサギ抗体 Alexa 488蛍光標識(緑色)
メーカー Molecular Probes-Invitrogen
核染色
To-PRO-3 iodide(青色)
メーカー Molecular Probes-Invitrogen
細胞骨格(アクチン標識)
Alexa Fluor 546 phalloidin(赤色)
メーカー Molecular Probes-Invitrogen
Primary antibody (rabbit-derived polyclonal antibody) Anti-Semaphorin 3A
Manufacturer Abcam, Cambridge
Secondary antibody (derived from chicken) Rabbit antibody Alexa 488 fluorescent label (green)
Manufacturer Molecular Probes-Invitrogen
Nuclear staining
To-PRO-3 iodide (blue)
Manufacturer Molecular Probes-Invitrogen
Cytoskeleton (actin labeling)
Alexa Fluor 546 phalloidin (red)
Manufacturer Molecular Probes-Invitrogen
9.統計処理
 実験結果は平均±標準誤差(S.E.) で表した。1群はすべて6匹とした。統計学的処理は、Dunnett 法による多重検定を行った。
9. Statistical processing The experimental results were expressed as mean ± standard error (SE). All groups consisted of 6 animals. For statistical processing, multiple tests by Dunnett's method were performed.
(結果)
 マウス実験的アレルギー性結膜炎モデルに対する効果 
・以上の実験によって、眼結膜の炎症組織像は好酸球を主体とした炎症性細胞浸潤、 出血傾向およびリンパ管の拡張が認められた。アレルギー反応に基づく炎症性細胞浸潤を主体とした結膜炎スコアの上昇、並びに結膜における好酸球及び T 細胞の増加が確認された。モデルはヒトのアレルギー性結膜炎と類似していることが認められた。 
・Sema3Aの点眼と結膜下投与は、RWアレルギー性結膜炎モデルにおいて、結膜炎スコアの上昇を抑制した、同部位の好酸球及び T 細胞の増加を減らし、明らかな治療効果が示された。 
・アレルギー性結膜炎の症状に対するスコア(表1)は、5 項目の合計で統計した。4週間の観察期間において臨床症状抑制の差が確認された。3週目から4週目においては有意な変化がなかった(図1)。
・Sema3Aの濃度差において、好酸球浸潤スコアに有意な差が認められた。(図2) 
さらに、好酸球浸潤スコアの差により、点眼群と結膜下投与群の有意差が確認された。 
・蛍光染色によりSema3Aは結膜組織移行性がよいことが証明された。 
・抗アレルギー点眼液ヒスタミンH1受容体拮抗薬とメディエーター遊離抑制薬、ステロイド点眼薬、Sema3Aの投与によって、Sema3Aの即効性が確認された(図3)。
・in vitroで、Sema3AのT細胞増殖抑制が確認された(図4)。
 Sema3Aの点眼と結膜下投与は、RWアレルギー性結膜炎モデルにおいて、明らかな治療効果を示した。 
・アレルギー性結膜炎の症状に対するスコア(表1)は5 項目の合計で統計し、4週間の観察期間において臨床症状の改善が確認された(図1)。 
(result)
Effect on mouse experimental allergic conjunctivitis model
-As a result of the above experiments, the inflammatory tissue image of the eye conjunctiva was found to have inflammatory cell infiltration, hemorrhagic tendency and lymphatic vessel dilation mainly consisting of eosinophils. An increase in the conjunctivitis score mainly due to inflammatory cell infiltration based on the allergic reaction and an increase in eosinophils and T cells in the conjunctiva were confirmed. The model was found to be similar to human allergic conjunctivitis.
・ Sema3A instillation and subconjunctival administration showed a clear therapeutic effect in the RW allergic conjunctivitis model, which suppressed the increase in conjunctivitis score and decreased the increase in eosinophils and T cells at the same site.
・ Scores for allergic conjunctivitis symptoms (Table 1) were statistically calculated using a total of 5 items. Differences in clinical symptom suppression were confirmed over the 4-week observation period. There was no significant change from week 3 to week 4 (FIG. 1).
・ Significant difference in eosinophil infiltration score was observed in Sema3A concentration difference. (Figure 2)
Furthermore, a significant difference between the ophthalmic group and the subconjunctival administration group was confirmed by the difference in the eosinophil infiltration score.
・ Fluorescence staining demonstrated that Sema3A has good conjunctival tissue migration.
-The immediate effect of Sema3A was confirmed by administration of antiallergic ophthalmic solution histamine H1 receptor antagonist and mediator release inhibitor, steroid eyedrops, and Sema3A (FIG. 3).
-In vitro, suppression of T cell proliferation by Sema3A was confirmed (Fig. 4).
Instillation and subconjunctival administration of Sema3A showed a clear therapeutic effect in the RW allergic conjunctivitis model.
・ Score for allergic conjunctivitis symptom (Table 1) was statistically made up of a total of 5 items, and improvement of clinical symptoms was confirmed in the observation period of 4 weeks (Fig. 1).
1)セマフォリン分子のレセプターとしてはNeuropilinやplexinファミリーに属する分子群が知られている。Neuropilin-1はVascular Endothelial Growth Factor (VEGF)のレセプターの一つでもあり、Sema3Aが拮抗的に血管新生を阻害するとの報告もあるが、Sema3A 1000ユニットを本試験で点眼および結膜下投与による結膜機能不全、それを原因とする角膜の障害、新生血管が実験中に認められなかったので、角膜新生血管の抑制が予想される。 1) As receptors for semaphorin molecules, molecular groups belonging to the Neuropilin and plexin families are known. Neuropilin-1 is one of the receptors for VascularelEndothelial Growth Factor (VEGF), and Sema3A has been reported to antagonistically inhibit angiogenesis, but Sema3A 1000 units are instilled and conjunctival function by subconjunctival administration in this study. Insufficiency, corneal damage caused by it, and new blood vessels were not observed during the experiment, so suppression of corneal new blood vessels is expected.
2)またSema3Aの適用は、神経ガイダンス、アトピー性皮膚炎など報告されているが、実際粘膜での好酸球性炎性疾患は欧米では発症率高く、その疾患概念が確立し病態解析と治療法の確立に向けた取り組みが進んている。Sema3A分子は結膜への投与による組織レベルでの病理的変化は認められなかったので、以上のような好酸球性炎性粘膜疾患にも治療効果が期待される。 2) Although application of Sema3A has been reported, such as nerve guidance and atopic dermatitis, in fact, the incidence of eosinophilic inflammatory diseases in the mucous membrane is high in the United States and Europe, and the disease concept has been established, pathological analysis and treatment Efforts are being made to establish the law. Since the Sema3A molecule did not show a pathological change at the tissue level due to administration to the conjunctiva, a therapeutic effect is expected for the above-mentioned eosinophilic inflammatory mucosal disease.
(考察)
 現在処方可能な抗アレルギー点眼薬はヒスタミンH1受容体拮抗薬(3種類)とメディエーター遊離抑制薬(6種類)、ステロイド点眼や内服薬、免疫調整剤点眼など、臨床で大体3種類が必要だが、薬の組み換えもしばしば必要とする。 
 非ステロイド点眼薬は効果が十分とはいえない。長期連用により難治性の角膜上皮障害、ドライアイを生じる。ステロイド点眼薬では眼圧上昇、続発緑内障、続発白内障、易感染性により、角膜の病変を悪化させ、角膜移植など、最終手術までに至る危険性がある。 
 炎症を繰り返す慢性疾患で、アレルギー疾患の自覚症状の中でも最も高い高率の90%以上に、また他のアレルギー疾患(鼻、気管)との合併が多いため、多大な費用と時間を費やすなど、生活に大きな支障を来して、患者のQOLが低下する。
(Discussion)
Currently, there are three types of anti-allergic eye drops that can be prescribed, including histamine H1 receptor antagonists (3 types), mediator release inhibitors (6 types), steroid drops, oral drugs, and immunomodulator drops. Often recombination is also required.
Non-steroidal eye drops are not effective enough. Prolonged use causes refractory corneal epithelial disorder and dry eye. Steroid eye drops cause increased corneal lesions due to increased intraocular pressure, secondary glaucoma, secondary cataracts, and susceptibility to infection, leading to the risk of corneal transplantation and the final surgery.
It is a chronic disease that repeats inflammation, the highest rate of all symptoms of allergic disease, more than 90%, and because there are many complications with other allergic diseases (nose, trachea), etc. It causes a major hindrance to life and lowers the patient's QOL.
 以上のリスクを克服するため、生物製剤は化学物質では達成できない薬理作用を持ち、眼内組織にダメージの少ないことが推定できる貴重な存在で、速効性のよい点眼薬になる可能性が期待される。
 本明細書で引用した全ての刊行物、特許および特許出願をそのまま参考として本明細書にとり入れるものとする。
In order to overcome the above risks, biologics have a pharmacological action that cannot be achieved with chemical substances, and are expected to become eye drops with a fast-acting effect because they can be presumed to have little damage to intraocular tissues. The
All publications, patents and patent applications cited herein are incorporated herein by reference in their entirety.
 本発明は、結膜におけるアレルギー性炎症の予防及び/又は治療に利用できる。 The present invention can be used for the prevention and / or treatment of allergic inflammation in the conjunctiva.
<配列番号1>
配列番号1は、ヒトのセマフォリン3A遺伝子のDNA配列を示す。
<配列番号2>
配列番号2は、ヒトのセマフォリン3Aのアミノ酸配列を示す。
<配列番号3>
配列番号3は、ニワトリのセマフォリン3A遺伝子のDNA配列を示す。
<配列番号4>
配列番号4は、ニワトリのセマフォリン3Aのアミノ酸配列を示す。
<SEQ ID NO: 1>
SEQ ID NO: 1 shows the DNA sequence of the human semaphorin 3A gene.
<SEQ ID NO: 2>
SEQ ID NO: 2 shows the amino acid sequence of human semaphorin 3A.
<SEQ ID NO: 3>
SEQ ID NO: 3 shows the DNA sequence of the chicken semaphorin 3A gene.
<SEQ ID NO: 4>
SEQ ID NO: 4 shows the amino acid sequence of chicken semaphorin 3A.

Claims (6)

  1. セマフォリン3Aを有効成分として含有する、結膜におけるアレルギー性炎症の予防及び/又は治療剤。 A prophylactic and / or therapeutic agent for allergic inflammation in the conjunctiva, comprising semaphorin 3A as an active ingredient.
  2. 結膜におけるアレルギー性炎症が季節性アレルギー結膜炎である請求項1記載の予防及び/又は治療剤。 The preventive and / or therapeutic agent according to claim 1, wherein the allergic inflammation in the conjunctiva is seasonal allergic conjunctivitis.
  3. 季節性アレルギー性結膜炎が花粉症である請求項2記載の予防及び/又は治療剤。 The preventive and / or therapeutic agent according to claim 2, wherein the seasonal allergic conjunctivitis is hay fever.
  4. セマフォリン3Aの医薬的に有効な量を被験者に投与することを含む、結膜におけるアレルギー性炎症を予防及び/又は治療する方法。 A method for preventing and / or treating allergic inflammation in the conjunctiva, comprising administering to a subject a pharmaceutically effective amount of semaphorin 3A.
  5. 結膜におけるアレルギー性炎症の予防及び/又は治療のためのセマフォリン3Aの使用。 Use of semaphorin 3A for the prevention and / or treatment of allergic inflammation in the conjunctiva.
  6. 結膜におけるアレルギー性炎症を予防及び/又は治療する方法に使用するためのセマフォリン3A。 Semaphorin 3A for use in a method for preventing and / or treating allergic inflammation in the conjunctiva.
PCT/JP2012/066207 2011-07-01 2012-06-26 Agent for prevention and/or treatment of allergic inflammation in conjunctiva WO2013005603A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
JP2011147038 2011-07-01
JP2011-147038 2011-07-01

Publications (2)

Publication Number Publication Date
WO2013005603A1 true WO2013005603A1 (en) 2013-01-10
WO2013005603A9 WO2013005603A9 (en) 2013-04-04

Family

ID=47436958

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2012/066207 WO2013005603A1 (en) 2011-07-01 2012-06-26 Agent for prevention and/or treatment of allergic inflammation in conjunctiva

Country Status (1)

Country Link
WO (1) WO2013005603A1 (en)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014178427A1 (en) * 2013-05-02 2014-11-06 学校法人順天堂 Method for controlling expression of semaphorin 3a
WO2015133459A1 (en) * 2014-03-03 2015-09-11 国立大学法人東京大学 Agent for preventing, suppressing or treating corneal disease or corneal damage, cell sheet, cell culture aid, and cell culture method
WO2016128966A1 (en) * 2015-02-10 2016-08-18 Medical Research & Development Fund for Health Services Bnai Zion Medical Center Semaphorin 3a for treatment and assessment of severity of inflammatory bowel disease
WO2020225400A1 (en) * 2019-05-09 2020-11-12 Boehringer Ingelheim International Gmbh Anti-sema3a antibodies and their uses for treating eye or ocular diseases
WO2023032994A1 (en) * 2021-08-31 2023-03-09 富士フイルム株式会社 Compound and labeled biomaterial using same

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2008107275A (en) * 2006-10-27 2008-05-08 Juntendo Allergic dermatitis diagnostic method
JP2008297243A (en) * 2007-05-31 2008-12-11 Yokohama City Univ Therapeutic or prophylactic agent for pruritic dermatosis

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2008107275A (en) * 2006-10-27 2008-05-08 Juntendo Allergic dermatitis diagnostic method
JP2008297243A (en) * 2007-05-31 2008-12-11 Yokohama City Univ Therapeutic or prophylactic agent for pruritic dermatosis

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
HARUNA SAWASHIRO ET AL.: "Semaphorin3A Toyo ga OVA Yuhatsusei Allergy-sei Bien ni Oyobosu Eikyo no Kento", THE JAPANESE PHARMACOLOGICAL SOCIETY KANTO BUKAI, vol. 123, 2010, pages 56 *
JUNKO YAMAGUCHI ET AL.: "Semaphorin3A wa Atopy-sei Hifuen no Hishin o Kaizen suru", JAPANESE JOURNAL OF ALLERGOLOGY, vol. 56, no. 8/9, 2007, pages 1052 *
MIN ZHOU ET AL.: "Preventive Treatment with Ibudilast Eyedrops : Effect on Allergic Conjunctivitis in Guinea Pigs", JOURNAL OF THE EYE, vol. 21, no. 12, 2004, pages 1679 - 1682 *
NAOKI KOMATSU ET AL.: "The therapeutic efficacy of CCR3 antagonist against murine allergic conjunctivitis", THE JOURNAL OF THE YONAGO MEDICAL ASSOCIATION, vol. 57, no. 2, 2006, pages 39 - 48 *

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014178427A1 (en) * 2013-05-02 2014-11-06 学校法人順天堂 Method for controlling expression of semaphorin 3a
JPWO2014178427A1 (en) * 2013-05-02 2017-02-23 学校法人順天堂 Semaphorin 3A expression regulation method
WO2015133459A1 (en) * 2014-03-03 2015-09-11 国立大学法人東京大学 Agent for preventing, suppressing or treating corneal disease or corneal damage, cell sheet, cell culture aid, and cell culture method
JP2015166323A (en) * 2014-03-03 2015-09-24 国立大学法人 東京大学 Agent for preventing, suppressing or treating corneal disease or corneal damage, cell sheet, cell culture aid, and cell culture method
WO2016128966A1 (en) * 2015-02-10 2016-08-18 Medical Research & Development Fund for Health Services Bnai Zion Medical Center Semaphorin 3a for treatment and assessment of severity of inflammatory bowel disease
US10736939B2 (en) 2015-02-10 2020-08-11 Medical Research & Development Fund for Health Services Bnai Zion Medical Center Semaphorin 3A for treatment and assessment of severity of inflammatory bowel disease
WO2020225400A1 (en) * 2019-05-09 2020-11-12 Boehringer Ingelheim International Gmbh Anti-sema3a antibodies and their uses for treating eye or ocular diseases
US11267880B2 (en) 2019-05-09 2022-03-08 Boehringer Ingelheim International Gmbh Anti-Sema3A antibodies and their uses for treating eye or ocular diseases
WO2023032994A1 (en) * 2021-08-31 2023-03-09 富士フイルム株式会社 Compound and labeled biomaterial using same

Also Published As

Publication number Publication date
WO2013005603A9 (en) 2013-04-04

Similar Documents

Publication Publication Date Title
US11738064B2 (en) Pharmaceutical composition for preventing and treating eye diseases, containing as active ingredient, fusion protein in which tissue-penetrating peptide and anti-vascular endothelial growth factor preparation are fused
EP2968471B1 (en) Peptides for use in the topical treatment of retinal neurodegenerative diseases, in particular in early stages of diabetic retinopathy and other retinal diseases in which neurodegeneration plays an essential role
US20130231464A1 (en) Methods of use of soluble cd24 for therapy of rheumatoid arthritis
US9233140B2 (en) Treatment methods for hypersensitive disorders
WO2013005603A1 (en) Agent for prevention and/or treatment of allergic inflammation in conjunctiva
JP2007529418A (en) Method of treating corneal transplant rejection by use of a VEGF antagonist
US20190002517A1 (en) Neuropeptide y-derived peptides
JP6440107B2 (en) Cell sheet production method, composition, cell culture aid, and cell culture method
JP6033433B2 (en) Method for treating hyperglycemia disorder using apolipoprotein AIV
Castro et al. Low-dose melittin is safe for intravitreal administration and ameliorates inflammation in an experimental model of uveitis
US20140038887A1 (en) Glial cell line derived neurotrophic factor, obesity, and obesity-related diseases and conditions
CN114945387A (en) Pharmaceutical composition for preventing or treating retinal neurodegenerative disease comprising Prox1 inhibitor as active ingredient
US20160272689A1 (en) Protamine in treatment of neuronal injuries
US10052362B2 (en) Glial cell line derived neurotrophic factor, obesity, and obesity-related diseases and conditions
Wilke et al. Complement regulation in the eye: implications for age-related macular degeneration
US20220211810A1 (en) Method of Treatment
KR20220000357A (en) Pharmaceutical composition for retinal neurodegenerative disease comprising inhibitor of Prox1 as an active ingredient
Jones The Role of Fibrillin-1 in Eye Development and Disease
Lee et al. https://www. jbc. org/cgi/doi/10.1074/jbc. RA120. 016053 The latest version is at
US20160331807A1 (en) Therapeutic use of vegfr-3 ligands
Class et al. Patent application title: NEUROPEPTIDE Y-DERIVED PEPTIDES Inventors: David Paul D. Woldbye (CopenhagenØ, DK) Casper Rene Gøtzsche (Copenhagen Ø, DK) Kristian Klemp (Charlottenlund, DK) Vladimir Berezin (Copenhagen N, DK) Vladimir Berezin (Copenhagen N, DK) Elisabeth Bock (Charlottenlund, DK)
BR112015020143B1 (en) TOPICAL PHARMACEUTICAL COMPOSITION IN TOPICAL EYE TREATMENT AND/OR IN THE PREVENTION OF NEURODEGENERATIVE DISEASES OF THE RETINA

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: 12807502

Country of ref document: EP

Kind code of ref document: A1

DPE1 Request for preliminary examination filed after expiration of 19th month from priority date (pct application filed from 20040101)
NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 12807502

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: JP