CN111249277A - Application of carboxyamidotriazole compound in preparing medicine for treating or preventing autoinflammatory diseases - Google Patents

Application of carboxyamidotriazole compound in preparing medicine for treating or preventing autoinflammatory diseases Download PDF

Info

Publication number
CN111249277A
CN111249277A CN201811467680.2A CN201811467680A CN111249277A CN 111249277 A CN111249277 A CN 111249277A CN 201811467680 A CN201811467680 A CN 201811467680A CN 111249277 A CN111249277 A CN 111249277A
Authority
CN
China
Prior art keywords
syndrome
disease
inflammatory
deficiency
cai
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
CN201811467680.2A
Other languages
Chinese (zh)
Inventor
朱蕾
叶菜英
段梦园
李娟�
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Guangdong Yinzhu Medical Technology Co.,Ltd.
Original Assignee
Institute of Basic Medical Sciences of CAMS
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 Institute of Basic Medical Sciences of CAMS filed Critical Institute of Basic Medical Sciences of CAMS
Priority to CN201811467680.2A priority Critical patent/CN111249277A/en
Publication of CN111249277A publication Critical patent/CN111249277A/en
Pending legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41921,2,3-Triazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]

Abstract

The invention belongs to the field of medicines, and particularly relates to an application of a carboxyamidotriazole compound or a pharmaceutically acceptable salt thereof in preparing a medicine for treating or preventing autoinflammatory diseases.

Description

Application of carboxyamidotriazole compound in preparing medicine for treating or preventing autoinflammatory diseases
Technical Field
The invention belongs to the field of medicines, and particularly relates to an application of a carboxyamidotriazole compound or a pharmaceutically acceptable salt thereof in preparing a medicine for treating or preventing autoinflammatory diseases.
Background
Autoinflammatory diseases (AUID) are a group of immune system diseases mediated by innate immunity (innate immunity) that have been increasingly recognized in recent years with obvious host predisposition and whose clinical manifestations are characterized by abnormally elevated inflammatory responses. AUID is mostly a rare disease, and has been proposed for the first time since 1999 (non-patent document 1), and has been developed to a group of widely covered diseases, from monogenic diseases such as Familial Mediterranean Fever (FMF), TNF receptor-related periodic syndrome (TRAPS), Mevalonate Kinase Deficiency (MKD), NLRP12 autoinflamation disease (the nuclear-binding oligomeric kinase-like protein (NLRP)12 autoinflamation disease (NLRP12AD)), and the like to diseases such as adult Stull inflammatory disease (AOAOS), Crohn's disease (CD's), and the like.
The clinical manifestations of AUID are recurrent febrile symptoms, acute arthritic symptoms and chronic systemic inflammatory response symptoms caused by systemic inflammatory response. The monogenic AUID mostly occurs in the early stage of a newborn or an infant, the polygenic AUID is late in onset and can reach the adolescence or even an adult, most of the patients have fever which repeatedly lasts for several days to several weeks, and the patients have nonspecific manifestations of body mass reduction, hypodynamia, general malaise, flu-like symptoms, lymphadenopathy, splenomegaly and the like and usually disappear along with the recovery of body temperature; there may also be skin, muscle, joint, eye, ear, blood system, gastrointestinal tract, respiratory tract, nervous system and cardiovascular system involvement, which can lead to patient disability and even death.
Since AUID is a genetic disease and the disease persists for life, the main goal of treatment is to quickly alleviate disease activity and normalize inflammatory responses in the body, thereby reducing tissue damage and complications (including amyloidosis), returning patients to normal life and improving their quality of life. Currently, first line treatment drugs for AUID include colchicine, non-steroidal anti-inflammatory drugs, and glucocorticoids. However, the above drugs have many and serious adverse reactions after long-term use, such as colchicine, which has an early common gastrointestinal reaction rate of 80%, and severe cases can cause dehydration, electrolyte disorder and the like, and long-term use can cause severe hemorrhagic gastroenteritis, nervous system toxicity, hematopoietic dysfunction, renal function impairment and the like. Furthermore, AUID is a disease mediated by an innate immune imbalance that is not a specific therapeutic agent and some patients appear ineffective against such agents.
Recently, as the understanding of the mechanism of the disease is advanced, targeting biological agents against inflammatory factors activated in the inflammatory response pathway have brought about the clinical treatment of AUID with the finding that IL-1 inhibitors have become the first-line drugs for the treatment of IL-1 receptor antagonist Deficiency (DIRA) and cold-inflammatory-associated periodic thermal syndrome (CAPS), and are also recognized as the main choice for other AUID patients who are ineffective against first-line drugs such as colchicine, and in addition, TNF- α inhibitors and IL-6 inhibitors have been used (non-patent documents 2 to 5).
The carboxyamidotriazole is a novel antitumor drug, has the chemical name of 5-amino-1- { [3, 5-dichloro-4- (4-chlorobenzoyl) phenyl ] methyl } -1H-1,2, 3-triazole-4-formamide, and has the chemical structural formula:
Figure BDA0001890221040000021
patent document 1 discloses that the compound has an antitumor effect. Further, patent document 2 discloses that carboxyamidotriazole or a pharmaceutically acceptable salt thereof has an effect of preventing and/or treating silicosis.
However, there has been no report on the effect of carboxyamidotriazole compounds in treating autoinflammatory diseases.
Documents of the prior art
Patent document
Patent document 1: european patent EP0644880
Patent document 2: chinese patent CN101919843
Non-patent document
Non-patent document 1: cell.1999,97:133-144
Non-patent document 2: clin Rev Allergy immunol.2013, 45; 2:227-235
Non-patent document 3: ann Rheum dis.2015, 74; 9:1636-1644
Non-patent document 4: front pharmacol.2017, 22; 8:278
Non-patent document 5: clin Immunol.2018, pii: S1521-6616(18):30432-30437
Disclosure of Invention
Problems to be solved by the invention
As described above, in the treatment of AUID, including first-line treatment drugs such as colchicine, non-steroidal anti-inflammatory drugs and glucocorticoids, there are serious adverse reactions, and the problem of recurrence of disease often occurs after withdrawal, and some patients show no effect on the drugs, in recent years, biological agents having a targeted action, such as IL-1 inhibitors, TNF- α inhibitors and IL-6 inhibitors, are protein preparations, which can be administered only by injection, are liable to cause reactions at the site of infusion and local injection, and can themselves become new antigens to stimulate the body to produce antibodies, thereby rendering the drugs ineffective, and the drugs have an immunosuppressive action, so long-term administration of drugs can cause fatal adverse reactions such as severe infection, and further, the price is very high.
Accordingly, an object of the present invention is to provide a novel method for treating or preventing an autoinflammatory disease, and more specifically, to provide a prophylactic/therapeutic agent effective for an autoinflammatory disease.
Means for solving the problems
As a result of intensive studies to solve the above problems, the present inventors have found that a carboxyamidotriazole-based compound or a pharmaceutically acceptable salt thereof is effective for the prevention/treatment of auto-inflammatory diseases, and have completed the present invention. That is, the present invention relates to the following (1) to (5).
(1) The application of the carboxyamidotriazole compound shown in the formula (A) or the pharmaceutically acceptable salt thereof in preparing the medicine for treating or preventing the autoinflammatory diseases,
Figure BDA0001890221040000041
wherein X represents CH2S, O or C ═ O; r4Represents Cl, CF3Br or CH3;R5Represents Cl, Br or NO2
(2) The use according to (1), wherein the carboxyamidotriazole compound is 5-amino-1- { [3, 5-dichloro-4- (4-chlorobenzoyl) phenyl ] methyl } -1H-1,2, 3-triazole-4-carboxamide.
(3) The use according to (1), wherein the pharmaceutically acceptable salt is selected from any one or more of the group consisting of hydrochloride, sulfate, orotate and acetate.
(4) The use according to any one of (1) to (3), wherein the autoinflammatory disease is a STING-related vascular disease selected from familial mediterranean fever, TNF receptor-related periodic syndrome, mevalonate kinase deficiency, NLRP12autoinflammatory disease, coldtin-related periodic heat syndrome, familial cold autoinflammatory syndrome, Muckle-Wells syndrome, neonatal multisystem inflammatory disease/chronic infantile neurocutaneous joint syndrome, Blau syndrome, chronic relapsing multifocal osteomyelitis, pyogenic arthritis-pyoderma-acne syndrome, Majeed syndrome, IL-1 receptor antagonist deficiency, IL-36receptor antagonist deficiency, PFAPA syndrome, nucleotide oligomerization domain 2-related autoinflammatory disease, Duchenne syndrome, STING-related vascular disease, baby-onset STING-related vascular disease, adenosine deaminase 2 deficiency, proteasome-related autoinflammatory syndrome, Guillain-West village syndrome, joint contracture-muscular atrophy-microcytoanaemia-lipodystrophy-related syndrome, chronic atypical neutrophilic dermatosis with fever and lipoatrophy, lipodystrophy-induced juvenile lipodystrophy syndrome, Schnitzler's syndrome, interferon-mediated autoinflammatory diseases, Aicardi-Gouti's syndrome, interferon-stimulated response gene 15deficiency, dysregulation of spinal cartilage dysplasia, phospholipase C gamma 2-related antibody deficiency and immune dysregulation, CARD 14-mediated psoriasis, defects of linear ubiquitin chain assembly complexes, sideroblasts anemia, immunodeficiency, fever and developmental elongation syndrome, familial macrojawbiosis, early onset sarcoidosis, recurrent grapevine type 1, HOIL-1deficiency, HOIP deficiency, generalized impetigo, palmoplantar pustulosis, subcortical pustulosis, tardive pressure urticaria, inflammatory acrodermatitis, neutrophilic panniculitis, PASH syndrome, SAPHO syndrome, pediatric SAPHO syndrome, autoinflammatory bone disease, chronic aseptic osteomyelitis, disseminated superficial actinic porokeratosis, familial chronic lichenification keratosis, H syndrome, Pyrin-associated autoinflammatory disease with neutrophilic dermatosis, macrophage activation syndrome, Singleton-Merten syndrome, X-linked reticuloendosis, TORCH syndrome, OTULIN-associated autoinflammatory disease, a20 under-haplotype dose, adult Still disease, pseudogout, systemic juvenile idiopathic arthritis, crohn's disease, inflammatory bowel disease, severe infant inflammatory bowel disease, Sweet syndrome, familial hemophagocytosis, inflammatory body disease, macrophage activation syndrome associated with NLRC4, and autoinflammatory disease associated with NLRP1 with arthritis and dyskeratosis.
(5) The use according to (4), wherein the autoinflammatory disease is a pyretic-related autoinflammatory disease selected from familial mediterranean fever, chronic relapsing multifocal osteomyelitis, mevalonate kinase deficiency, NLRP12autoinflammatory disease, cold-related periodic heat syndrome, familial cold autoinflammatory syndrome, Muckle-Wells syndrome, neonatal multisystem inflammatory disease/chronic infantile neurocutaneous joint syndrome, suppurative arthritis-pyoderma-acne syndrome, Majeed syndrome, IL-1 receptor antagonist deficiency, IL-36receptor antagonist deficiency, Schnitzler syndrome, recurrent hydatidiform type 1, SAPHO syndrome, childhood SAPHO syndrome, autoinflammatory bone disease, chronic aseptic osteomyelitis, familial chronic lichenification disease, Pyrrin-related autoinflammatory disease accompanied by neutrophilic skin disease, a20 single dose deficiency, adult Still disease, pseudogout, systemic juvenile idiopathic arthritis, severe infant inflammatory bowel disease, Sweet syndrome, NLRC 4-associated macrophage activation syndrome, NLRP 1-associated autoinflammatory diseases with arthritis and dyskeratosis, phospholipase C γ 2-associated antibody deficiency and immune dysregulation, Blau syndrome, nucleotide oligomerization domain 2-associated autoinflammatory diseases, minor syndrome, CARD 14-mediated psoriasis, linear ubiquitin chain assembly complex deficiency, early onset sarcoidosis, HOIL-1deficiency, HOIP deficiency, generalized pustulosis, inflammatory limb dermatitis, OTULIN-associated autoinflammatory diseases, tumor necrosis factor receptor-associated periodic syndrome, pfstratum corneum apa syndrome, impetigo dermatosis, disseminated superficial actinic porokeratosis, X-linked reticulum abnormality, and H syndrome.
Effects of the invention
According to the present invention, a novel prophylactic/therapeutic agent effective for auto-inflammatory diseases can be provided, which comprises a carboxyamidotriazole-based compound or a pharmaceutically acceptable salt thereof as an active ingredient. Further, according to the present invention, a novel method for treating or preventing an auto-inflammatory disease can be provided.
Drawings
FIG. 1 shows the effect of CAI (carboxyamiditazole, carboxyamidotriazole) on the levels of IL-1 β, IL-6 and TNF- α secretion by Peripheral Blood Mononuclear Cells (PBMC) in patients with Blau Syndrome (BS).*P<0.05,**P<0.01。
FIG. 2 shows the effect of CAI on nuclear translocation of NF-. kappa. B p65 in PBMCs of BS patients.
FIG. 3 shows the effect of CAI on I κ B and p-p65 expression in PBMCs of BS patients.
FIG. 4 shows the effect of CAI on the level of IL-1 β, IL-6 and TNF- α secretion by PBMCs of Familial Mediterranean Fever (FMF) patients.*P<0.05,**P<0.01,***P<0.001。
FIG. 5 shows the effect of CAI on the levels of IL-1 β and IL-6 secretion by PBMCs of patients with tumor necrosis factor receptor-related periodic syndrome (TRAPS).*P<0.05。
FIG. 6 shows the effect of CAI on the level of IL-1 β and IL-6 secretion by PBMCs of patients with NLRP12 auto-inflammatory disease (NLRP12 AD).*P<0.05。
FIG. 7 shows the effect of CAI on the level of IL-1 β and IL-6 secretion by PBMCs of patients with Duchenne syndrome (YAO syndrome).**P<0.01,***P<0.001。
FIG. 8 shows CAI versus cold inflammatory associated cycle feverEffect of IL-1 β secretion levels in PBMCs of patients with syndrome (CAPS) -Mulkle-Wells syndrome (MWS).*P<0.05,**P<0.01,***P<0.001。
FIG. 9 shows the effect of CAI on the levels of IL-1 β, IL-6 and TNF- α secretion by PBMCs of adult Still disease (AOSD) patients.*P<0.05。
Detailed Description
The carboxyamidotriazole compound in the invention comprises carboxyamidotriazole and analogues and derivatives thereof. Specifically, the carboxyamidotriazole compound provided by the invention comprises a compound with a structure shown in the following formula (I):
Figure BDA0001890221040000071
wherein R is1Has the structure of formula (II):
Figure BDA0001890221040000072
wherein p is an integer of 0 to 2; m is an integer of 0 to 4; n is an integer of 0 to 5; x can be O, S, SO2、C=O、CHCN、CH2Or C ═ NR6
Wherein R is6Is hydrogen, (C1-C6) alkanyl, hydroxy, (C1-C6) alkyloxy, amino, (C1-C6) alkanamino, dialkylamino or cyano;
R4and R5Respectively halogen, cyano, trifluoromethyl, (C1-C6) alkanoyl, nitro, (C1-C6) alkanyl, (C1-C6) alkyloxy, carbonyl, carbalkoxy, trifluoromethoxy, acetylamino, (C1-C6) alkylthio, (C1-C6) alkansulfonyl, trichloroethylene, trifluoromethylthio, trifluoromethylsulfinyl or trifluoromethylsulfonyl;
R2is amino, (C1-C6) alkylamino, dialkylamino, acetamido, acetamimide, ureidoacyl, carboxamido, carboximide or guanidino;
R3is carbamoyl, cyano, carbamoyl, imineOr N-hydroxycarbamoyl.
More specifically, the carboxyamidotriazole compound shown in the formula (I) is shown in the formula (II), n and m are respectively 0, 1 or 2; p is 1; x is O, S, C ═ O or CH2;R4Is fluorine, chlorine, bromine, methyl, trifluoromethyl, cyano, methoxycarbonyl, trifluoromethoxy, trifluoromethylthio, nitro or trichloroethylene; r5Is chlorine, bromine, fluorine, methyl, trifluoromethyl, cyano, carbalkoxy, trifluorovinyl or nitro.
More specifically, the carboxyamidotriazole compound shown in the formula (I) has a structural formula shown as the following formula (A):
Figure BDA0001890221040000081
in the formula, X is CH2S, O or C ═ O; r4Is Cl, CF3Br or CH3;R5Is Cl, Br or NO2
More specifically, the carboxyamidotriazole compound represented by the formula (I) is 5-amino-1- { [3, 5-dichloro-4- (4-chlorobenzoyl) phenyl ] methyl } -1H-1,2, 3-triazole-4-carboxamide represented by the following structural formula, that is, carboxyamidotriazole (hereinafter, may be abbreviated as CAI).
Figure BDA0001890221040000091
As pharmaceutically acceptable salts of the carboxyamidotriazole compounds, hydrochloride, sulfate, orotate or acetate salts may be mentioned.
The invention comprises the application of the carboxyamidotriazole compound or the pharmaceutically acceptable salt thereof in preparing a medicament for treating or preventing autoinflammatory diseases (AUID).
AUID is caused by congenital abnormalities in the immune system, including the dysregulation of neutrophils, macrophages, NK cells and related molecules of innate immunity that protect the body from external pathogens. Unlike autoimmune diseases (e.g., systemic lupus erythematosus), monocytes are primarily macrophages, rather than T, B lymphocytes, in AUID, which are involved in the inflammatory response impairment process, and autoantibodies or antigen-specific T cells are not present in patients with AUID.
AUID has now developed into a group of diseases covering a wide range, including in particular the following: familial Mediterranean Fever (FMF), tumor necrosis factor receptor-associated periodic syndrome (TNFactor-associated periodic syndrome, TRAPS), mevalonate kinase deficiency (mevalonate kinase deficiency, MKD, high IgD have been considered as serum markers for diagnosing this disease, so also called high IgD syndrome (HIDS), NLRP12 auto-inflammatory disease (the same as NLRP 12-associated systemic fever syndrome, NLRP 36-auto-inflammatory disease), NLRP 3632-auto-thermal syndrome (NLRP-associated cold-related syndrome, NLRP 36-auto-thermal syndrome, NLRP 32-auto-thermal syndrome, NLRP 2-auto-thermal syndrome, MRP 2-auto-thermal syndrome, cryoprin-associated cutaneous syndrome, CAPS, Familial Cold Autoimmune Syndrome (FCAS), Multisystemic inflammatory disease of newborn (neonatal-infant neurocutaneous joint syndrome/chronic infant neurocutaneous joint syndrome, MWS), Nomid/CINCA, Blau Syndrome (BS), chronic recurrent multifocal osteomyelitis (chronic multifocal cutaneous syndrome, CRMO), pyogenic arthritis-pyoderma-acne syndrome (pathologic, metabolic syndrome of deficiency of P-adrenocortical receptor), Pacific inflammatory syndrome of pyogenic arthritis, pyogenic skin-acne syndrome (PAS-associated systemic inflammatory syndrome, DIRAS), multiple sclerosis-systemic inflammatory syndrome (MWS), multiple sclerosis-systemic inflammatory disease of newborn (respiratory syndrome/chronic infectious disease of newborn), multiple sclerosis of pyogenic skin syndrome (PAS-associated inflammatory disease of deficiency of P-receptor of P-interstitial receptor (PAS-36), multiple sclerosis-associated with multiple sclerosis, multiple sclerosis of pyogenic arthritis-acne syndrome (chronic inflammatory disease of deficiency of P-inflammatory disease of PAS-receptor (PAS-related inflammatory disease of PAS-receptor of Pacific disease of the human, mP-epidermal syndrome (PAS-associated with deficiency of P-receptor of P-inhibitor of P-receptor (DIR-associated with IL-receptor of P-antagonist of P-receptor of P-mediated multiple infection of the P-receptor (PAS, DIR-mediated multiple infection of the same, S, P-associated with the receptor of the same, P-associated with the receptor of the same, P-associated with the, DITRA), PFAPA syndrome (PFAPA syndrome, also known as Marshall syndrome or periodic fever, aphthous stomatitis, pharyngitis and lymphadenitis syndrome), nucleotide-oligomerization domain 2-associated autoinflammatory disease (NAID, also known as NOD2-associated autoinflammatory disease), Yaosdrome syndrome (NAID), STING-associated vascular disease (STING-associated vascular disorder (NAID)), STING-associated vascular disease (STING-associated vascular disease, also known as juvenile inflammatory disease, acute inflammatory disease-associated vascular disease (SAAD-associated vascular disease), Denumerous-related inflammatory disease (ADA-associated Adenosine kinase, Adeno syndrome, Lipase-related inflammatory disease, Adeno-associated Adenosine-2-associated hormone, Adeno-related hormone, Adeno-related hormone-vascular disease, Adeno-associated hormone-kinase (NAID-related hormone-2-associated Adenosine kinase, Adeno-related hormone-2-associated with Adenosine kinase, Adeno-related hormone-related disorder (ADA-related hormone-related syndrome, Adeno-related hormone-related to Adenosine-related hormone-related disorder (ADA, Adeno-related hormone-related syndrome, Adeno-related hormone-related to NID, Adeno-related hormone-related disorder, NNS, also known as Nakajo-Nishimura syndrome, also known as Japanese autoimmune syndrome with lipoatrophy (JASL)), joint contracture-muscular atrophy-microcytoanaemia-lipodystrophy-associated lipodystrophy syndrome (JMP), Chronic atypical neutrophilic dermatosis with fever and lipoatrophy (Chronic lymphocytic leukemia with lipodystrophy and interstitial dystrophy syndrome), juvenile adipocyte dysbolism (Chronic inflammatory-induced dysadiposis syndrome, also known as autoimmune-interferon syndrome), autoimmune response syndrome (interferon-induced dysadiposis syndrome), acute adiposis syndrome (Chronic inflammatory-induced dysadiposis syndrome, also known as autoimmune response syndrome), ISG)15 deficiency (ISG15deficiency), spinal cartilage dysplasia with immunoregulatory abnormalities (SPENCDI), Phospholipase C gamma 2(PLC gamma 2) associated antibody deficiencies and immune dysregulation (Phospholipase-C gamma 2-associated systemic diabetes and immune dysregulation, PLAD), CARD14mediated psoriasis (CARD14mediated psoriasis, CAMPS), linear ubiquitin chain assembly complex (linear ubiquitin chain assembly complex, LUBAC) deficiency, sideroblastic cellular anemia, immunodeficiency, fever and prolonged development syndrome (sideroblastic anemia, febrile without toxicity syndrome, febrile disease and prolonged development syndrome (siderobial leukemia, febrile disease and hypernociceptive disease, hypereosinophilic dyscraving disease, hypereosinophilic dysfunction, hypereosinophilic disease, hypereosinophilic, keratoderma cuticle (SCPD), urticaria tardive, inflammatory acrodermatitis, neutrophilic panniculitis, PASH syndrome (PASHSynrome, i.e., pyoderma gangrenosum, acne, hidradenitis suppurativa syndrome, pyoderma gangrenosum, acid, and epithelial high sensitivity syndrome), SAPHO syndrome (SAPHO syndrome, i.e., synovitis, acne, impetigo, hyperosteogeny and osteomyelitis syndrome, syndrome-acid-pustulosis-hyperositis syndrome), SAPHO syndrome in children, auto inflammatory bone disease (autoimmune bone disease), chronic osteomyelitis (chronic non inflammatory disease, CNO), diffuse superficial actinic porosis (systemic sclerosis, chronic inflammatory dermatosis), scleroderma gangrene-keratosis syndrome (DSpoliosis), chronic inflammatory dermatosis (inflammatory dermatosis, acute keratosis, scleroderma gangrenosum, scleroderma gangrene-keratoderma, scleroderma gangreniformis syndrome (acute keratoderma purpuria-associated with acute keratosis, scleroderma gangreniformis syndrome, scleroderma, pyemia-keratoderma syndrome (DSA 3, chronic keratoderma, scleroderma keratoderma keratosis, scleroderma keratoderma keratitis, scleroderma keratitis syndrome, PAAND), Macrophage Activation Syndrome (MAS), Singleton-Merten Syndrome (Singleton-Merten Syndrome, SMS), X-linked reticuloendochrome abnormality (X-linked reticuloendotheral Syndrome, XLPDR), TORCH Syndrome (TORCH Syndrome), OTULIN-related auto-inflammatory disease (The OTULIN-related auto-inflammatory Syndrome, ORAS), A20 underdose (A20haploinsufficiency, HA20), adult Stilli disease (adult acute lung disease, AOSD), gout (gout), pseudogout, juvenile idiopathic arthritis (BD) systemic inflammatory bowel disease (acute bowel disease, Behcet's disease), Crohn's disease (Crohn 'disease), Crohn's disease, FHL), an inflammasome disease (inflamasocata), NLRC 4-related macrophage activation syndrome (NLRC4-MAS, also called syndrome of enterocoliticas and inflammatory associated with inflammation with circulation in NLRC4(SCAN4)), an NLRP 1-related autoinflammatory disease accompanied by arthritis and dyskeratosis, and the like.
According to the pathogenesis, AUID can be divided into inflammatory body diseases (inflammation related disorders of IL-1 β activation diseases (IL-1 β activation disorders), NF-kappa B activation diseases (NF-kappa B activation disorders), protein folding diseases (protein folding disorders), complement related diseases (complement related disorders), macrophage activation related diseases (macrophage activation disorders), mitochondrial dysfunction related diseases (mitotic sepsis functional associated diseases), cytokine signal transduction disorder related diseases (cytokine dysfunction related diseases) and other etiological inflammatory diseases including autoimmune reticuloendotheliosis, macrophage deficiency related diseases including acute lymphokinesis syndrome, chronic inflammatory bowel disease, coronary heart disease.
Furthermore, the present invention includes a method of treating a patient having AUID, the method comprising the step of administering to the patient an effective amount of a carboxyamidotriazole compound or a pharmaceutically acceptable salt thereof.
The dosage employed will, of course, depend on the particular disease being treated, as well as other factors including age, weight, health, severity of symptoms, route of administration, frequency of treatment, and whether other drugs are concomitantly used during the treatment period. The dosage of the active compound to be used can be readily determined by conventional methods known to those of ordinary skill in the art. For the carboxyamidotriazole compounds of the invention or pharmaceutically acceptable salts thereof, the daily adult dose is generally in the range of about 10mg to 300mg, preferably in the range of about 50mg to 200 mg. For example, a total daily dose of between 50mg and 200mg of the carboxyamidotriazole compound of the invention may be administered orally to a human suffering from AUID. Alternatively, a total daily dose of between 50mg and 200mg of the carboxyamidotriazole compound of the invention may be administered rectally to a human suffering from AUID.
In addition, the invention also comprises a pharmaceutical composition containing the carboxyamidotriazole compound or the pharmaceutically acceptable salt thereof. More specifically, the carboxyamidotriazole compound or a pharmaceutically acceptable salt thereof may be formulated into a pharmaceutical composition using standard pharmaceutically acceptable carriers, fillers, solubilizers, stabilizers, and the like well known to those skilled in the art.
The pharmaceutical composition comprising the carboxyamidotriazole compound or a pharmaceutically acceptable salt thereof is administered to an individual in need of such a medicament by any of a variety of routes including, but not limited to, topical, oral, intravenous, intramuscular, intraarterial, intramedullary, intrathecal, intraventricular, transdermal, subcutaneous, intraperitoneal, intranasal, enteral, topical, sublingual or rectal means.
The oral preparation may be a solid preparation such as a tablet, a capsule, a powder, a granule, or a liquid preparation such as a solution, a suspension, etc. Formulations suitable for oral administration may contain a pharmaceutically acceptable carrier or excipient. Pharmaceutically acceptable carriers or excipients suitable for solid formulations (such as tablets or capsules) may be: for example, binders (e.g., acacia, gelatin, dextrin, hydroxypropyl cellulose, methyl cellulose, polyvinylpyrrolidone), diluents (e.g., lactose, sucrose, mannitol, corn starch, potato starch, calcium phosphate, calcium citrate, crystalline cellulose), lubricants (e.g., magnesium stearate, calcium stearate, stearic acid, talc, anhydrous silica gel), disintegrants (e.g., corn starch, potato starch, carboxymethyl cellulose calcium, alginic acid), and wetting agents (e.g., sodium lauryl sulfate). Pharmaceutically acceptable carriers or excipients suitable for liquid formulations (e.g. solutions or suspensions) may be: for example, aqueous lytic enzymes (e.g., water), suspending agents (e.g., acacia, gelatin, methylcellulose, sodium carboxymethylcellulose, hydroxymethylcellulose, aluminum stearate gel), surfactants (e.g., lecithin, sorbitan monooleate, glycerol monostearate), and non-aqueous lytic enzymes (e.g., polyethylene glycol 400, glycerol, propylene glycol, vegetable oils). In addition, the liquid formulation may contain preservatives (e.g., methyl paraben, propyl paraben), flavoring agents and/or coloring agents.
The enteral administration (enema) can be in the form of an aqueous solution or suspension prepared using the above-mentioned water-soluble enzyme or suspending agent. If necessary, the enema can be a sol or gel preparation prepared using a thickening agent such as polyacrylic acid, gelatin, etc.
Suppositories can be prepared by conventional methods by mixing carboxyamidotriazole or a pharmaceutically acceptable salt, analogue, derivative thereof with a commercially available oily base such as Witepsole or the like or a water-soluble base such as polyethylene glycol, glycerol, gelatin or the like. The suppository can be capsule suppository, tablet suppository or ointment suppository.
The external preparation may include external powders, ointments, creams, etc.
Examples
The present invention will be further illustrated by the following examples, but the present invention is not limited to the following examples.
AUID covers a wide range of but has a uniform characteristic, and is expressed as systemic inflammatory response due to the increase of proinflammatory factors IL-1 β, TNF- α and IL-6, including recurrent febrile symptoms, acute arthritis symptoms and systemic chronic inflammatory response symptoms, while the decrease of cytokine levels directly reflects the remission of diseases.recently, inhibitors against these three cytokines are also increasingly used for AUID treatment and show good therapeutic effects (see, for example, Recommendations for the management of autoimmune diseases. Ann Rheum Dis.2015,74(9):1636 and 1644. scientists, in evaluating the therapeutic effects of drugs on AUID, often adopt a method of evaluating the therapeutic effects of drugs on patients' Peripheral Blood Mononuclear Cells (PBMC) secreted by detecting the levels of PBMC of the patients after administration to evaluate the therapeutic effects of drugs on the drugs (see, for example, culture of International drugs for PBMC-1436, culture for evaluation of IL-14383, 1436, and so on the results of various antigens).
Example 1 Effect of CAI on secretion of proinflammatory cytokines by PBMCs of patients with Blau Syndrome (BS)
PBMC of BS patients were routinely isolated and cell suspensions were prepared in 1X 10 DMEM high-glucose medium containing 10% fetal bovine serum6Individual cells/well were seeded in 24-well plates. The experimental groups were as follows:
(1) BS group: adding only culture medium;
(2) CAI treatment group: adding CAI with final concentration of 40 μ M;
(3) LPS + Muramyl Dipeptide (MDP) group: LPS (final concentration 10ng/ml) and MDP (final concentration 10. mu.g/ml) were added simultaneously;
(4) LPS + MDP + CAI group: LPS (final concentration 10ng/ml), MDP (final concentration 10. mu.g/ml) and CAI (final concentration 40. mu.M) were added simultaneously.
Placing at 37 ℃ and 5% CO2After 22h of culture in an incubator, cell supernatants were collected and frozen at-20 ℃ and the levels of IL-1 β, IL-6 and TNF- α in the cell supernatants were determined by enzyme-linked immunosorbent assay (ELISA).
As shown in FIG. 1, PBMC of BS patients secreted high levels of IL-1 β, IL-6 and TNF- α, and administration of 40 μ M CAI treatment significantly reduced the levels of the above cytokines in the supernatant, with inhibition rates of 25.66% (P <0.05), 87.84% (P <0.05) and 90.75% (P <0.01), respectively, after 22h of LPS and MDP stimulation of PBMC cells of BS patients, the levels of IL-1 β, IL-6 and TNF- α secretion were further significantly increased, while 40 μ M CAI had significant inhibition effects on LPS and MDP-induced increases in the levels of IL-1 β, IL-6 and TNF- α secretion, with inhibition rates of 66.90% (P <0.05), 54.93% (P <0.05) and 44.66% (P <0.05), respectively, suggesting that CAI may have a good therapeutic effect on BS.
Example 2 Effect of CAI on Nuclear translocation of NF- κ B p65 in PBMCs of BS patients
NF-. kappa.B is a nuclear transcription factor having important transcriptional regulation effects on cytokines such as IL-1 β, IL-6 and TNF- α. the NF-. kappa.B family includes p50/p105, p52/p100, Rel (p65), RelB, c-Rel, which are usually present in the form of homo-or heterodimers, most commonly as heterodimers consisting of p50/p 65. in the resting state, p50/p65 binds to its repressor IkB and exists in the cytoplasm as inactive complexes.
PBMC of BS patients were routinely isolated and cell suspensions were prepared in 1X 10 DMEM high-glucose medium containing 10% fetal bovine serum6Individual cells/well were seeded in 24-well plates previously plated with sterile slides and the experimental groups were the same as in example 1. Placing at 37 ℃ and 5% CO2Culturing in an incubator for 22h, then removing culture medium in the hole, and observing nuclear translocation of NF-kappa B p65 by adopting immunofluorescence staining combined with a laser confocal microscope. NF- κ B p65 antibody was purchased from Cell Signaling, and anti-rabbit Alexa Fluor488 was purchased from Biyuntian Biotechnology.
As shown in FIG. 2, NF-. kappa. B p65 was expressed at a high level in PBMCs of the BS group and localized in the nuclei, and the nuclear region NF-. kappa. B p65 showed significantly decreased green fluorescence expression and fluorescence intensity after 40. mu.MCAI treatment. After LPS and MDP stimulation is carried out on PBMC of a BS patient, the fluorescence intensity of NF-kappa B p65 is further enhanced, the expression level is obviously increased, 40 mu M CAI can obviously weaken the fluorescence intensity enhancement of NF-kappa B p65 induced by LPS and MDP, and part of fluorescence is distributed in cytoplasm. This result suggests that CAI has a significant inhibitory effect on the enhanced nuclear translocation of NF-. kappa. B p65 in PBMCs of BS patients, and that CAI may inhibit the activation of NF-. kappa.B in PBMCs of BS patients.
Example 3 Effect of CAI on I κ B and p-p65 expression in PBMCs of BS patients
In addition, phosphorylation of p65 increases its ability to bind to DNA, increasing its transcriptional activation capacity, thus, the level of phosphorylation of I κ B α and I κ B α in PBMCs from BS patients was examined.
PBMC of BS patients were routinely isolated and cell suspensions were prepared in 1X 10 DMEM high-glucose medium containing 10% fetal bovine serum6Individual cells/well were seeded in 24-well plates. The experimental groups were as follows:
(1) BS group: adding only culture medium;
(2) CAI 10 μ M group: adding CAI to a final concentration of 10. mu.M
(3) CAI 20 μ M group: adding CAI to a final concentration of 20. mu.M
(4) CAI 40 μ M group: adding CAI to a final concentration of 40. mu.M
Placing at 37 ℃ and 5% CO2After culturing in an incubator for 22h, extracting total protein of cells in each hole, freezing and storing at-80 ℃, detecting the expression levels of I kappa B and p-p65 by adopting a western blot method, and using β -actin as an internal reference.
As shown in FIG. 3, the expression level of I κ B in the CAI (10, 20, 40 μ M) group was significantly increased and the expression level of p-p65 was significantly decreased compared to the BS group, the results suggest that CAI may exert therapeutic effects on BS by reducing I κ B degradation and p65 phosphorylation in PBMC of BS patients, inhibiting NF- κ B p65 nuclear translocation, and thus reducing IL-1 β, IL-6, and TNF- α levels.
Example 4 Effect of CAI on secretion of proinflammatory cytokines from PBMCs of patients with Familial Mediateranean (FMF)
PBMC of FMF patients were isolated routinely, and cell suspensions were prepared in 1X 10 DMEM high-glucose medium containing 10% fetal bovine serum6Individual cells/well were seeded in 24-well plates. The experimental groups were as follows:
(1) FMF group: adding only culture medium;
(2) CAI 10 μ M group: adding CAI with final concentration of 10 μ M;
(3) CAI 20 μ M group: adding CAI with a final concentration of 20 mu M;
(4) CAI 40 μ M group: adding CAI with final concentration of 40 μ M;
placing at 37 ℃ and 5% CO2After 24h of culture in an incubator, cell supernatants were collected and frozen at-20 ℃ and the levels of IL-1 β, IL-6 and TNF- α in the cell supernatants were determined by ELISA.
As shown in FIG. 4, CAI significantly reduced the higher levels of IL-1 β, IL-6 and TNF- α secreted by PBMCs of FMF patients, where CAI (10, 20, 40. mu.M) inhibited IL-1 β secretion by 26.71% (P <0.05), 38.44% (P <0.01) and 49.87% (P <0.001), 40. mu.M CAI inhibited IL-6 secretion by 51.26% (P <0.001) and 40. mu.MCAI inhibited TNF- α secretion by 22.07% (P <0.05), respectively.
Example 5 Effect of CAI on the secretion of proinflammatory cytokines from PBMCs of patients with TNF receptor-assisted periodic syndrome (TRAPS)
PBMC of TRAPS patients are routinely isolated and cell suspensions are prepared in DMEM high-sugar medium containing 10% fetal bovine serum at 2X 106Individual cells/well were seeded in 24-well plates. The experimental groups were as follows:
(1) TRAPS group: adding only culture medium;
(2) CAI treatment group: adding CAI with final concentration of 40 μ M;
(3) LPS group: LPS (final concentration 1. mu.g/ml) was added;
(4) LPS + CAI 40 μ M group: LPS (final concentration 1. mu.g/ml) and CAI (final concentration 40. mu.M) were added simultaneously;
placing at 37 ℃ and 5% CO2After 3h of culture in an incubator, cell supernatants were collected and frozen at-20 ℃ and the levels of IL-1 β and IL-6 in the cell supernatants were determined by ELISA.
As shown in FIG. 5, 40 μ M CAI reduced the higher IL-1 β and IL-6 levels in the PBMC culture supernatants of TRAPS patients, with inhibition rates of 17.49% (P <0.05) and 61.11% (P <0.05), respectively.
Example 6 Effect of CAI on secretion of proinflammatory cytokines from PBMCs of NLRP12auto inflammatory disease (NLRP12auto inflammatory disease, NLRP12AD) patients
PBMC of NLRP12AD patient are isolated conventionally, and cell suspension is prepared in DMEM high-sugar medium containing 10% fetal bovine serum at 2X 106Individual cells/well were seeded in 24-well plates. The experimental groups were as follows:
(1) NLRP12AD group: adding only culture medium;
(2) CAI 10 μ M group: adding CAI with final concentration of 10 μ M;
(3) CAI 20 μ M group: adding CAI with a final concentration of 20 mu M;
(4) CAI 40 μ M group: adding CAI with final concentration of 40 μ M;
placing at 37 ℃ and 5% CO2After 3h of culture in an incubator, cell supernatants were collected and frozen at-20 ℃ and the levels of IL-1 β and IL-6 in the cell supernatants were determined by ELISA.
As shown in FIG. 6, CAI significantly reduced the higher IL-1 β and IL-6 levels in the PBMC culture supernatants of NLRP12AD patients, where the inhibition of IL-1 β by CAI (20, 40. mu.M) was 75.79% (P <0.05) and 55.98% (P <0.05), respectively, and the inhibition of IL-6 by CAI (10, 20, 40. mu.M) was 62.22% (P <0.05), 72.56% (P <0.05) and 69.51% (P <0.05), respectively, suggesting that CAI may have a good therapeutic effect on NLRP12 AD.
Example 7 Effect of CAI on secretion of proinflammatory cytokines from PBMCs of patients with Duchenne syndrome (Yao syndrome, formerly known as NOD 2-associatedauntomatic disease (NAID))
PBMC of patients with Securian syndrome are separated conventionally, and a cell suspension is prepared by using a DMEM high-sugar medium containing 10% fetal bovine serumAt 1X 106Individual cells/well were seeded in 24-well plates. The experimental groups were as follows:
(1) group of syndromes of wanted: adding only culture medium;
(2) CAI 10 μ M group: adding CAI with final concentration of 10 μ M;
(3) CAI 20 μ M group: adding CAI with a final concentration of 20 mu M;
(4) CAI 40 μ M group: adding CAI with final concentration of 40 μ M;
placing at 37 ℃ and 5% CO2After 22h of culture in an incubator, cell supernatants were collected and frozen at-20 ℃ and the levels of IL-1 β and IL-6 in the cell supernatants were determined by ELISA.
As shown in FIG. 7, CAI significantly reduced the levels of IL-1 β and IL-6 in the supernatant of PBMC cultures from patients with Duchenne syndrome, in which CAI (20, 40. mu.M) inhibited IL-1 β secretion by 55.53% (P <0.01) and 56.98% (P <0.01), respectively, and CAI (10, 20, 40. mu.M) inhibited IL-6 secretion by 31.43% (P <0.01), 30.69% (P <0.01) and 52.72% (P <0.001), respectively.
Example 8 Effect of CAI on secretion of proinflammatory cytokines by PBMCs of CAPS patients
CAPS is caused by continuous activation of NLRP3 inflammasome due to NLRP3 gene mutation and is characterized by IL-1 β increase, leading to periodic attacks of inflammation, and is classified into Familial Cold Autoinflammatory Syndrome (FCAS), Muckle-Wells syndrome (MWS) and chronic infant neurocutaneous joint syndrome (NOMID/CINCA) according to clinical manifestations from mild to severe.
PBMC of CAPS-MWS patients are separated conventionally, and cell suspension is prepared by DMEM high-sugar medium containing 10% fetal bovine serum and is used for 2X 106Individual cells/well were seeded in 24-well plates. The experimental groups were as follows:
(1) normal control group (CON): normal PBMC, only adding culture medium, and not performing any treatment;
(2) CAPS group: CAPS was added to the medium only;
(3) CAI treatment group: adding CAI with final concentration of 40 μ M;
(4) LPS group: LPS (final concentration 1. mu.g/ml) was added;
(5) LPS + CAI 10 μ M group: LPS (final concentration 1. mu.g/ml) and CAI (final concentration 10. mu.M) were added simultaneously;
(6) LPS + CAI 20 μ M group: LPS (final concentration 1. mu.g/ml) and CAI (final concentration 20. mu.M) were added simultaneously;
(7) LPS + CAI 40 μ M group: LPS (final concentration 1. mu.g/ml) and CAI (final concentration 40. mu.M) were added simultaneously;
placing at 37 ℃ and 5% CO2After 3h of culture in an incubator, cell supernatants were collected and frozen at-20 ℃ and the level of IL-1 β in the cell supernatants was determined by ELISA.
As shown in FIG. 8, compared with normal PBMC, the level of IL-1 β secreted by PBMC of CAPS-MWS patients was significantly increased (P <0.05), the level of IL-1 β in the supernatant was significantly decreased by 40 μ M CAI administration, the inhibition rate was 85.95%, which is significantly different (P < 0.01). the level of secreted IL-1 β was significantly increased (P <0.01) after LPS stimulation of PBMC cells of CAPS-MWS patients, and the inhibition rates of IL-1 β secretion induced by LPS were significantly increased (10, 20, 40 μ M) and reached 74.15% (P <0.001), 73.11% (P <0.01) and 93.42% (P <0.001), respectively.
Example 9 Effect of CAI on secretion of proinflammatory cytokines from PBMCs of adult still disease (AOSD) patients
PBMC of AOSD patients are separated conventionally, and cell suspension is prepared by DMEM high-sugar medium containing 10% fetal bovine serum and is used for 2X 106Individual cells/well were seeded in 24-well plates. The experimental groups were as follows:
(1) AOSD group: adding only culture medium;
(2) CAI treatment group: adding CAI with final concentration of 40 μ M;
placing at 37 ℃ and 5% CO2After 3h of culture in an incubator, cell supernatants were collected and frozen at-20 ℃ and the levels of IL-1 β, IL-6 and TNF- α in the cell supernatants were determined by ELISA.
As shown in FIG. 9, administration of 40 μ M CAI treatment significantly reduced the levels of IL-1 β, IL-6, and TNF- α in the supernatant with inhibition of 10.25% (P <0.05), 70.20% (P <0.05), and 48.01% (P <0.05), respectively.
From the results of examples 1-9, CAI is known to be effective in treating autoinflammatory diseases including Blau syndrome, familial mediterranean fever, TNF receptor-related periodic syndrome, NLRP12autoinflammatory disease, Dutch's syndrome, cryoinflamin-related periodic fever syndrome, and adult still's disease.
While the present invention has been described in detail and with reference to specific embodiments thereof, it will be apparent to one skilled in the art that various changes and modifications can be made therein without departing from the spirit and scope thereof.

Claims (5)

1. The application of the carboxyamidotriazole compound shown in the formula (A) or the pharmaceutically acceptable salt thereof in preparing the medicine for treating or preventing the autoinflammatory diseases,
Figure FDA0001890221030000011
wherein X represents CH2S, O or C ═ O; r4Represents Cl, CF3Br or CH3;R5Represents Cl, Br or NO2
2. Use according to claim 1, wherein the carboxyamidotriazole compound is 5-amino-1- { [3, 5-dichloro-4- (4-chlorobenzoyl) phenyl ] methyl } -1H-1,2, 3-triazole-4-carboxamide.
3. The use according to claim 1, wherein the pharmaceutically acceptable salt is selected from any one or more of the group consisting of hydrochloride, sulfate, orotate and acetate.
4. The use according to any one of claims 1 to 3, wherein the autoinflammatory disease is a STING-related vascular disease selected from familial mediterranean fever, TNF receptor-related periodic syndrome, mevalonate kinase deficiency, NLRP12autoinflammatory disease, coldtin-related periodic heat syndrome, familial cold autoinflammatory syndrome, Muckle-Wells syndrome, neonatal multisystem inflammatory disease/chronic infantile neurocutaneous joint syndrome, Blau syndrome, chronic relapsing multifocal osteomyelitis, pyogenic arthritis-pyoderma gangrenosum-acne syndrome, Majeed syndrome, IL-1 receptor antagonist deficiency, IL-36receptor antagonist deficiency, PFAPA syndrome, nucleotide oligomerization domain 2-related autoinflammatory disease, Duchenne syndrome, STING-related vascular disease, baby-onset STING-related vascular disease, adenosine deaminase 2 deficiency, proteasome-related autoinflammatory syndrome, Guillain-West village syndrome, joint contracture-muscular atrophy-microcytoanaemia-lipodystrophy-related syndrome, chronic atypical neutrophilic dermatosis with fever and lipoatrophy, lipodystrophy-induced juvenile lipodystrophy syndrome, Schnitzler's syndrome, interferon-mediated autoinflammatory diseases, Aicardi-Gouti's syndrome, interferon-stimulated response gene 15deficiency, dysregulation of spinal cartilage dysplasia, phospholipase C gamma 2-related antibody deficiency and immune dysregulation, CARD 14-mediated psoriasis, defects of linear ubiquitin chain assembly complexes, sideroblasts anemia, immunodeficiency, fever and developmental elongation syndrome, familial macrojawbiosis, early onset sarcoidosis, recurrent grapevine type 1, HOIL-1deficiency, HOIP deficiency, generalized impetigo, palmoplantar pustulosis, subcortical pustular dermatosis, delayed-onset urticaria with pressure, inflammatory acrodermatitis, neutrophilic panniculitis, PASH syndrome, SAPHO syndrome, childhood SAPHO syndrome, auto-inflammatory bone disease, chronic aseptic osteomyelitis, disseminated superficial actinic keratosis, familial lichenification, H syndrome, Pyrin-related auto-inflammatory disease with neutrophilic dermatosis, macrophage activation syndrome, Singleton-Merten syndrome, X-linked reticuloendosis, TORCH syndrome, OTULIN-related auto-inflammatory disease, A20 under-single dose, adult Still disease, pseudogout, systemic juvenile idiopathic arthritis, severe infant inflammatory bowel disease, Sweet syndrome, familial hemophagocytosis, inflammatory corpuscular disease, NLRC 4-associated macrophage activation syndrome and NLRP 1-associated autoinflammatory diseases accompanied by arthritis and dyskeratosis.
5. The use according to claim 4, wherein the auto-inflammatory disease is a disease selected from familial mediterranean fever, chronic recurrent multifocal osteomyelitis, mevalonate kinase deficiency, NLRP12 auto-inflammatory disease, cold inflammatory hormone-related periodic heat syndrome, familial cold auto-inflammatory syndrome, Muckle-Wells syndrome, neonatal multisystem inflammatory disease/chronic infantile neurocutaneous joint syndrome, pyogenic arthritis-pyoderma gangrenosum-acne syndrome, Majeed syndrome, IL-1 receptor antagonist deficiency, IL-36receptor antagonist deficiency, Schnitzler syndrome, recurrent hydatidiform type 1, SAPHO syndrome, childhood SAPHO syndrome, auto-inflammatory bone disease, chronic aseptic osteomyelitis, familial chronic lichenification keratosis, Pyrin-related auto-inflammatory disease with neutrophilic skin disease, a20 single dose deficiency, adult Still disease, pseudogout, systemic juvenile idiopathic arthritis, severe infant inflammatory bowel disease, Sweet syndrome, NLRC 4-associated macrophage activation syndrome, NLRP 1-associated autoinflammatory diseases with arthritis and dyskeratosis, phospholipase C γ 2-associated antibody deficiency and immune dysregulation, Blau syndrome, nucleotide oligomerization domain 2-associated autoinflammatory diseases, minor syndrome, CARD 14-mediated psoriasis, linear ubiquitin chain assembly complex deficiency, early onset sarcoidosis, HOIL-1deficiency, HOIP deficiency, generalized pustulosis, inflammatory limb dermatitis, OTULIN-associated autoinflammatory diseases, tumor necrosis factor receptor-associated periodic syndrome, pfstratum corneum apa syndrome, impetigo dermatosis, disseminated superficial actinic porokeratosis, X-linked reticulum abnormality, and H syndrome.
CN201811467680.2A 2018-12-03 2018-12-03 Application of carboxyamidotriazole compound in preparing medicine for treating or preventing autoinflammatory diseases Pending CN111249277A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN201811467680.2A CN111249277A (en) 2018-12-03 2018-12-03 Application of carboxyamidotriazole compound in preparing medicine for treating or preventing autoinflammatory diseases

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN201811467680.2A CN111249277A (en) 2018-12-03 2018-12-03 Application of carboxyamidotriazole compound in preparing medicine for treating or preventing autoinflammatory diseases

Publications (1)

Publication Number Publication Date
CN111249277A true CN111249277A (en) 2020-06-09

Family

ID=70948791

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201811467680.2A Pending CN111249277A (en) 2018-12-03 2018-12-03 Application of carboxyamidotriazole compound in preparing medicine for treating or preventing autoinflammatory diseases

Country Status (1)

Country Link
CN (1) CN111249277A (en)

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006037106A2 (en) * 2004-09-24 2006-04-06 Rfe Pharma Llc Carboxy-amido-triazoles for the localized treatment of ocular diseases
CN101164538A (en) * 2006-10-16 2008-04-23 中国医学科学院基础医学研究所 Application of carboxyamido-triazoles compound and its salt in preparing medicine for treating painful diseases and/or inflammatory diseases
CN101618036A (en) * 2008-06-30 2010-01-06 中国医学科学院基础医学研究所 Novel medicinal application of carboxyl aminotriazole and acceptable salts of the carboxyl aminotriazole
CN101919843A (en) * 2009-06-10 2010-12-22 中国医学科学院基础医学研究所 Novel application of carboxyamidotriazole compound and salt thereof in preparation of medicaments for preventing and treating interstitial pneumonia or pulmonary fibrosis
CN103251601A (en) * 2006-12-06 2013-08-21 巧妙疗法股份有限公司 Use of carboxyamidotriazole (cai) orotate in macular degeneration

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006037106A2 (en) * 2004-09-24 2006-04-06 Rfe Pharma Llc Carboxy-amido-triazoles for the localized treatment of ocular diseases
CN101164538A (en) * 2006-10-16 2008-04-23 中国医学科学院基础医学研究所 Application of carboxyamido-triazoles compound and its salt in preparing medicine for treating painful diseases and/or inflammatory diseases
CN103251601A (en) * 2006-12-06 2013-08-21 巧妙疗法股份有限公司 Use of carboxyamidotriazole (cai) orotate in macular degeneration
CN101618036A (en) * 2008-06-30 2010-01-06 中国医学科学院基础医学研究所 Novel medicinal application of carboxyl aminotriazole and acceptable salts of the carboxyl aminotriazole
CN101919843A (en) * 2009-06-10 2010-12-22 中国医学科学院基础医学研究所 Novel application of carboxyamidotriazole compound and salt thereof in preparation of medicaments for preventing and treating interstitial pneumonia or pulmonary fibrosis

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
LEE GOLDMAN,MD等: "《西氏内科学 下册》", 31 May 2015, 世界图书出版西安公司 *
XIAOWAN DU等: "Therapeutic efficacy of carboxyamidotriazole on 2,4,6-trinitrobenzene sulfonic acid-induced colitis model is associated with the inhibition of NLRP3 inflammasome and NF-κB activation", 《INTERNATIONAL IMMUNOPHARMACOLOGY》 *
吴永贵等: "《当代内科学进展》", 31 January 2016, 安徽科学技术出版社 *

Similar Documents

Publication Publication Date Title
US20220339169A1 (en) Methods of treating or selecting a treatment for a subject resistant to tnf inhibitor using a nlrp3 antagonist
Schreiber et al. Safety and efficacy of recombinant human interleukin 10 in chronic active Crohn's disease
KR101524165B1 (en) Methods for improving the pharmacokinetics of hiv integrase inhibitors
AU2011380510B2 (en) Methods for treating gout flares
CA2945263A1 (en) Use of gsk-3 inhibitors or activators which modulate pd-1 or t-bet expression to modulate t cell immunity
EP2059234B1 (en) Method for reducing or alleviating inflammation in the digestive tract
JP7125353B2 (en) Pharmaceutical composition for use in treating HTLV-1-associated myelopathy
JP2011190199A (en) Medicament for rheumatoid arthritis
US20180055803A1 (en) Methods for Treating Gout Flares
US20230346719A1 (en) Multiple myeloma treatment
JP2021106625A (en) Il-34 antisense oligonucleotide and method of using the same
TW202310841A (en) Lou064 for treating multiple sclerosis
CN111249277A (en) Application of carboxyamidotriazole compound in preparing medicine for treating or preventing autoinflammatory diseases
EP2575799A1 (en) Methods for treating inflammatory autoimmune disorders
WO2021002887A1 (en) Gut-targeted nlrp3 antagonists and their use in therapy
JP6192142B2 (en) How to treat gout redness
Semo-Oz et al. The Role of Anti–IL-1 Medications in Autoinflammatory Disease
EP3664786B1 (en) Method for treating schnitzler&#39;s syndrome
US20170136089A1 (en) Compositions and methods of regulating bone resorption
US7329643B2 (en) Inhibition of HMGB1 release by fetuin
WO2024023696A1 (en) Dosing regimen for a nlrp3 inhibitor
KR20240055038A (en) LOU064 for the treatment of multiple sclerosis
WO2023078807A1 (en) A class iia hdac inhibitor and decitabine to treat myopathy
Baum et al. THU0128 Evaluation of Safety, Pharmacokinetics and Pharmacodynamics of BI 638683, A Novel CCR1 Antagonist
Bakshi et al. Periodic fevers and autoinflammatory conditions

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
TA01 Transfer of patent application right
TA01 Transfer of patent application right

Effective date of registration: 20201210

Address after: Room 726, 7th floor, building a, Central Europe center, No.2, South Lingnan Avenue, Lecong Town, Shunde District, Foshan City, Guangdong Province 528300

Applicant after: Guangdong Yinzhu Medical Technology Co.,Ltd.

Address before: 100005 No. three, 5 Dongdan, Beijing, Dongcheng District

Applicant before: INSTITUTE OF BASIC MEDICAL SCIENCES, CHINESE ACADEMY OF MEDICAL SCIENCES