TW201010988A - 5-(4-methanesulfonyl-phenyl)-thiazole derivatives for the treatment of acute and chronic inflammatory diseases - Google Patents

5-(4-methanesulfonyl-phenyl)-thiazole derivatives for the treatment of acute and chronic inflammatory diseases Download PDF

Info

Publication number
TW201010988A
TW201010988A TW098118927A TW98118927A TW201010988A TW 201010988 A TW201010988 A TW 201010988A TW 098118927 A TW098118927 A TW 098118927A TW 98118927 A TW98118927 A TW 98118927A TW 201010988 A TW201010988 A TW 201010988A
Authority
TW
Taiwan
Prior art keywords
compound
soil
tnf
disease
substituted
Prior art date
Application number
TW098118927A
Other languages
Chinese (zh)
Other versions
TWI403506B (en
Inventor
Royo Victor Rubio
La Hera Martinez Antonio De
De Mon Soto Melchor Alvarez
Munoz Ana Munoz
Original Assignee
Faes Farma Sa
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
Priority claimed from EP08380177A external-priority patent/EP2135864A1/en
Priority claimed from US12/139,661 external-priority patent/US7781594B2/en
Application filed by Faes Farma Sa filed Critical Faes Farma Sa
Publication of TW201010988A publication Critical patent/TW201010988A/en
Application granted granted Critical
Publication of TWI403506B publication Critical patent/TWI403506B/en

Links

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
    • A61K31/425Thiazoles
    • A61K31/4261,3-Thiazoles
    • 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
    • A61K31/425Thiazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/02Nasal agents, e.g. decongestants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/08Drugs for disorders of the urinary system of the prostate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/10Drugs for disorders of the urinary system of the bladder
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/04Antipruritics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/04Drugs for skeletal disorders for non-specific disorders of the connective tissue
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/02Drugs for disorders of the nervous system for peripheral neuropathies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D277/00Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings
    • C07D277/02Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings not condensed with other rings
    • C07D277/20Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members
    • C07D277/22Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members with only hydrogen atoms, hydrocarbon or substituted hydrocarbon radicals, directly attached to ring carbon atoms
    • C07D277/24Radicals substituted by oxygen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D277/00Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings
    • C07D277/02Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings not condensed with other rings
    • C07D277/20Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members
    • C07D277/32Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D277/34Oxygen atoms

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Immunology (AREA)
  • Pulmonology (AREA)
  • Diabetes (AREA)
  • Urology & Nephrology (AREA)
  • Hematology (AREA)
  • Dermatology (AREA)
  • Biomedical Technology (AREA)
  • Rheumatology (AREA)
  • Neurology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Epidemiology (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Cardiology (AREA)
  • Obesity (AREA)
  • Neurosurgery (AREA)
  • Communicable Diseases (AREA)
  • Vascular Medicine (AREA)
  • Pain & Pain Management (AREA)
  • Endocrinology (AREA)
  • Transplantation (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Emergency Medicine (AREA)
  • Oncology (AREA)
  • Otolaryngology (AREA)
  • Gastroenterology & Hepatology (AREA)

Abstract

The invention relates to the use of a compound of formula (I): or a pharmaceutically acceptable salt, prodrug and/or solvate thereof, in the preparation of a medicament for the treatment of an acute or chronic inflammatory disease, by inhibiting the production of at least one pro-inflammatory cytokine selected from TNF-alpha and IFN-gamma, or by inmunomodulating the IL-8 chemokine and/or the IL-10 regulatory cytokine. The invention also relates to novel compounds of formula (I'): or a pharmaceutically acceptable salt, prodrug and/or solvate thereof, as well as a pharmaceutical composition comprising said compound.

Description

201010988 Λ 六、發明說明: 【發明所屬之技術領域】 本發明關於衍生自5-(4-甲磺醯基-苯基)-噻唑之化合 物類於製備用於治療急性或慢性發炎疾病或狀況(諸如類 風濕性關節炎)之醫藥產品上之用途。 【先前技術】 〇 免疫學是區別自體與非自體之間之科學硏究。對自體 耐受性之崩解係自體免疫性疾病之起源。另外,其他狀況 諸如移植、動脈粥樣硬化、敗血性及非敗血性急性或慢性 發炎病理學和許多其他到目前爲止不被視爲自體免疫之疾 病亦展現由免疫細胞所媒介之病理機制。大多數作者認爲 免疫發炎效應物反應之活化係基於二個信號: -信號1包含刺激該細胞系抗原之受器(TCR-CD3 T細 胞受器複合物),以辨識該包埋在該主要組織相容性複合 〇 物(MHC)分子中之同源抗原。在B細胞中,細胞外可溶性 或膜結合性抗原與該免疫球蛋白CD79(Ig/CD79)細胞系受 器複合物交聯以遞送該信號1至該抗體分泌性淋巴細胞系 〇 -除抗原遞送之信號1之外,需要信號2以避免因反 應低落(anergy)或細胞系刪除所導致之耐受性。信號2係 由共刺激物遞送,諸如表現在專業抗原表現細胞(APC)(如 人單核細胞及彼等之分化系統後代)之表面上之CD86。 在先天性免疫發炎反應中,危險信號會被刺激模式辨 201010988 識受器(PRR)之微生物及自身調節性抗原或分裂原所提升 。PRR傳信途徑諸如TLR4(革蘭氏陰性細菌細胞壁脂多醣 或LPS之受器)遞送高量共刺激表面分子(也就是CD80或 CD86)之表現。接下來,共刺激配位體 CD80-CD86和T 幫助細胞表面之CD28交聯。信號1(也就是抗CD3抗體 交聯)與信號2(也就是CD28被特定抗體交聯)之組合實驗 性地模擬有效發炎效應物免疫反應之天然條件。値得注意 的是,經核准之免疫抑制藥物在高量共刺激之條件下對於 抑制前發炎細胞介素級聯之活化上有若干困難。後者發生 於自體免疫疾病和許多其他嚴重的急性或慢性發炎狀況中 〇 APC不只是後天免疫系統中決定免疫反應種類之關鍵 ,因爲它們深切地影響該挑戰細胞系所媒介的是效應物( 也就是T細胞幫助(Th)、細胞性細胞毒性(Th)、抗體產生 (B細胞))反應或是耐受性反應(反應低落或細胞凋亡)。 APC本身亦可遞送效應物直接免疫反應。一證據充分之實 例係單核細胞在癌症病患中產製及釋放高量之腫瘤壞死因 子a (TNF- α )。在該等病患中,TNF- α促進放大反應及血 管內皮細胞之活化,伴隨介白素8(IL-8)和其他前發炎細 胞介素之分泌以及前凝血劑活性。同時,TNF- α促進血 栓形成及缺血性腫瘤壞死,此作用導致惡病質素 (cachectin)或TNF- α之原始定義。然而,TNF- α係一涉 及許多其他疾病狀況之多向性細胞介素。經年累月的證據 顯示,LPS可刺激單核細胞分泌大量TNF- α,這強烈導 201010988 致敗血性休克之發生。最近,高量TNF- ct之不當產 分泌被認爲是自體免疫發炎疾病(諸如類風濕性關節 脊椎關節病、克隆氏病、葡萄膜炎及牛皮癖)之治療 。因此,目前已經廣泛地認爲使用抗TNF- α係治療 疾病之最新策略,其中降低可獲得之生物活性TNF-量可能導致改善該病患之狀況。 就這方面來說,可能得益於TNF- α拮抗劑治療 φ 病數目正在快速增加中,包括動脈粥樣硬化、代謝性 群、腦炎、病毒性肝炎、腎小球性腎炎、對腫瘤之不 炎反應及敗血性休克等。 除了該些由若干體細胞產製者之外,TNF- α有 主要的產製來源,單核細胞與Τ淋巴細胞。在獲得性 反應之過程中,令APC在前發炎指導情況下展示至 信號刺激IL-12 ρ70(ρ35及ρ40之雜二聚體)分泌,後 使該經活化之Th細胞之細胞介素分泌特性極化成爲 〇 型之共刺激分子。Thl細胞展現特徵化之細胞介素特 其中干擾素r (IFN- r )分泌係真正的足跡。該等細胞 泌高量之TNF - α。 由Thl分泌之IFN-τ促進許多對發炎疾病之了 可能重要之效果。一方面,其進一步活化單核細胞且 給定刺激所引起之 TNF- α分泌量。另一方面,其 TNF- α受器下游更強烈之信號途徑。總而言之,IFN-了其本身之直接效果諸如抗病毒活性之外,還能 MHC-II之表現且藉由增力D TNF- α仍然強烈之效果以 製及 炎、 目標 該些 α之 之疾 徵候 當發 二個 免疫 危險 者係 Th-1 性, 亦分 解上 增加 促使 r除 增加 造成 201010988 發炎及病灶。 前發炎級聯係由單核細胞之TNF-α或Thl細胞之 IFN-r及TNF- 〇:產製所起動,該級聯透過其他前發炎細 胞介素途徑(諸如IL-8)擴大。IL-8(不管其原始命名)被描 述爲一種由巨噬細胞和其他細胞種類(諸如上皮細胞)所產 製之趨化激素,其亦可由內皮細胞合成,因此也被稱爲 CXCL8。雖然嗜中性顆粒細胞係IL-8之主要標靶細胞, 但有相當廣泛種類之細胞(內皮細胞、巨噬細胞、肥大細 _ 胞、角質化細胞)對此趨化激素亦有反應。IL-8之主要功 能係誘導其標靶細胞(例如嗜中性顆粒細胞)之趨化性。在 嗜中性細胞中,移動及其主要功能吞噬作用所需之細胞生 理系列反應亦被引發,諸如細胞內Ca2 +增加、胞外分泌( 例如組織胺釋放)及呼吸爆發。IL-8可由任何具有TLR之 細胞分泌,TLR係與先天性免疫反應有關。IL-8之主要功 能係募集嗜中性細胞以吞噬該引起抗原模式TLR之抗原 。藉由傳送IL-8標靶細胞至內皮及其他標靶組織,因此 Q IL-8除了在身體防禦上之作用以外,還與許多TNF- α之 病理性作用之擴大及執行有關。 白血球之移動及回歸(homing)不僅由趨化激素和它們 的受器所調節,亦由若干黏附分子調節。在這些黏附分子 當中,選擇素CD62L被認爲是防止白血球移動至淋巴結 之治療目標,因此被當作排列非類固醇抗發炎藥物 (NSAID)之活體外效果之評估參數。 許多免疫抑制劑及抗TNF-α分子會影響正常之免疫 -8- 201010988 \ · 防禦機制,因爲它們增進對免疫細胞之細胞毒性作用或抑 制在成功效應物免疫反應之前之細胞系擴增底下之增生機 制。 考慮到細胞外空間分泌之TNF- α (不論是細胞質膜插 入或分泌之可溶性形式)之重要性,許多硏究已致力於設 計可阻斷細胞外TNF-α與TNF-受器I及/或TNF-受器II 之交互作用之治療劑。最適切之方法爲使用可溶性誘餌 φ TNF-受器以捕捉TNF-α,且由於解離需要長時間而防止 前發炎配位體與該細胞受器之交互作用。 第二種策略係以習知方式產製人化抗人TNF-α抗體 或產製也以給定疾病相關之其他分子爲標的之雙特異性單 鏈分子(例如在類風濕性關節炎中之抗VEGF/抗TNF-a) 。雖然上述之分子係TNF- α拮抗劑,它們限制它們的作 用機制以阻斷胞外分泌之TNF-α。 能驅使TNF- α產製及分泌之詳盡但不完全之標的清 φ 單可能爲:(a)驅使轉錄表現TNF- a之分子;(b)驅使 TNF- a -RNA從細胞核運送至細胞質及RNA剪切之分子; (c)指引TNF-α轉譯之分子;(d)調節TNF-cz -mRNA安定 性之分子;(e)指引高基小胞至前TNF- α表面形式錨定處 之細胞膜之分子;(f)與分泌釋放TNF-α有關之分子諸如 TNF-α轉換酶(TACE)及(g)調節前TNF-α之表面形式之內 化及其信號之分子。所有這些皆關於TNF-α產製及分泌 之細胞標的。 雖然有以不同方式設計之阻斷TNF-o:之產製之治療 201010988 劑,若能發現選擇性阻斷不只是TNF-α產製還有其他關 鍵前發炎細胞介素(如IFN-r)產製之新穎藥物將受到高度 重視。 【發明內容】 發明摘要 本發明之作者意外發現式(I)化合物顯示一些高度有 趣之免疫調節效果,它們可能適用於控制急性和慢性發炎 疾病之病理機制,因此具有潛在的臨床應用價値。具體地 說,本發明所使用之化合物已能抑制來自慢性發炎疾病( 諸如類風溼性關節炎)病患之週邊血液單核細胞(PBMC)之 TNF- α產製,亦能抑制該些細胞經T細胞刺激後之IFN-Τ分泌。此外,式(I)化合物已能調控分泌趨化激素(即 IL-8)和調節性細胞介素(即IL-10)之生物反應特性,且在 健康及疾病狀態下以不同之方式調控。式(I)化合物之整 體免疫調控效果不會對來自週邊血液之單核細胞造成任何 毒性效應。另外,在細胞分裂刺激後之活化及增生反應並 不受這些化合物之調節甚或增加。 在這一個小分子中就同時具有對數種前發炎細胞介素 如TNF-α及IFN-r之抑制效果與調節IL-8趨化激素及/ 或IL-1 0調節性細胞介素(所有均在系統性及器官特異性 自體免疫異常、移植、急性和慢性發炎疾病、某些代謝性 及退化性疾病與動脈粥樣硬化之病理生理學中具有關鍵重 要性)之潛能,使式(I)化合物得以歸類於新穎之免疫調節 201010988 \ 劑類別,並以具有臨床及治療重要性之各種可能之再程序 化層級之前發炎/調節性細胞介素及趨化激素之級聯爲標 的。 顯而易見的是,免疫細胞係無柄且進入器官以巡邏在 發炎狀況中被浸潤之身體組織,其中免疫細胞聚集於根據 疾病活性、目標器官及損害程度而分佈之病灶中。由於本 發明所使用之化合物被預期可調節該病理生理過程之若干 Ο 形態,因此可利用顯影試驗以特徵化受器數量、結合效率 、受器佔用率及藥物探針濃度。由於白血球具有回歸特性 ,偵測該治療性目標也能得到有關該標靶細胞及該病灶部 位之位置之資訊。 因此,式(I)化合物亦展現一種在藥物發展、臨床試 驗及個體化醫藥上作爲顯影生物標記之用途,這能提供的 不只是藥物動力學、分佈及投藥之資訊,還包含在臨床前 及臨床試驗中有關個體化反應模式之重要資料。後者可能 Φ 導致定義經確效、可靠、個體化之臨床終點之替代生物標 記,以供投予至需要該預後性及個體化評估式(I)化合物 或其醫藥組成物之有效量之病患,以最佳化該領域之技藝 人士所知之明確生物顯影技術。 根據第一態樣,本發明關於式(I)化合物: -11 - 201010988201010988 Λ VI. DESCRIPTION OF THE INVENTION: TECHNICAL FIELD OF THE INVENTION The present invention relates to compounds derived from 5-(4-methylsulfonyl-phenyl)-thiazole for use in the treatment of acute or chronic inflammatory diseases or conditions ( Use in pharmaceutical products such as rheumatoid arthritis. [Prior Art] 〇 Immunology is a scientific study that distinguishes between self and non-self. The disintegration of autologous tolerance is the origin of autoimmune diseases. In addition, other conditions such as transplantation, atherosclerosis, septic and non-septic acute or chronic inflammatory pathology and many other diseases that have not been considered autoimmune so far also exhibit pathological mechanisms mediated by immune cells. Most authors believe that the activation of the immune inflammatory effector response is based on two signals: - Signal 1 contains a receptor that stimulates the cell line antigen (TCR-CD3 T cell receptor complex) to identify the embedding in the primary A homologous antigen in a histocompatibility complex mites (MHC) molecule. In B cells, an extracellular soluble or membrane-bound antigen is cross-linked with the immunoglobulin CD79 (Ig/CD79) cell line receptor complex to deliver the signal 1 to the antibody secreting lymphocyte lineage - in addition to antigen delivery In addition to signal 1, signal 2 is required to avoid tolerance due to anergy or cell line deletion. Signal 2 is delivered by a costimulator, such as CD86 on the surface of specialized antigen-expressing cells (APCs), such as human monocytes and their descendants of the differentiation system. In the congenital immune inflammatory response, the risk signal is enhanced by the stimulating pattern of the microorganisms of the 201010988 recognizer (PRR) and autoregulatory antigens or mitogens. PRR signaling pathways such as TLR4 (the receptor for Gram-negative bacterial cell wall lipopolysaccharide or LPS) deliver high levels of costimulatory surface molecules (i.e., CD80 or CD86). Next, the co-stimulatory ligands CD80-CD86 and T help CD28 cross-linking on the cell surface. The combination of signal 1 (i.e., anti-CD3 antibody cross-linking) and signal 2 (i.e., CD28 is cross-linked by a particular antibody) experimentally mimics the natural conditions of an effective inflammatory effector immune response. It is noted that approved immunosuppressive drugs have several difficulties in inhibiting the activation of the proinflammatory cytokine cascade under conditions of high co-stimulation. The latter occurs in autoimmune diseases and many other serious acute or chronic inflammatory conditions. APC is not only the key to determining the type of immune response in the acquired immune system, because they profoundly affect the cell lineage that is the effector (also It is the T cell help (Th), cellular cytotoxic (Th), antibody production (B cell) response or tolerogenic response (low response or apoptosis). APC itself can also deliver an effector direct immune response. A well-documented example is that monocytes produce and release high amounts of tumor necrosis factor a (TNF-α) in cancer patients. In these patients, TNF-α promotes amplification and activation of vascular endothelial cells, accompanied by secretion of interleukin-8 (IL-8) and other proinflammatory cytokines, as well as procoagulant activity. At the same time, TNF-α promotes thrombosis and ischemic tumor necrosis, which results in the original definition of cachectin or TNF-α. However, TNF-α is a polytropic interleukin that is involved in many other disease states. Over the years, evidence shows that LPS can stimulate the secretion of large amounts of TNF-α by monocytes, which strongly leads to the occurrence of septic shock in 201010988. Recently, high levels of TNF-ct inappropriate secretion are considered to be treatments for autoimmune inflammatory diseases such as rheumatoid arthritis, Crohn's disease, uveitis, and psoriasis. Therefore, the latest strategy for the treatment of diseases using anti-TNF-α is now widely recognized, wherein reducing the amount of biologically active TNF-enable may lead to an improvement in the condition of the patient. In this regard, it may be possible to benefit from the rapid increase in the number of TNF-α antagonists, including atherosclerosis, metabolic group, encephalitis, viral hepatitis, glomerulonephritis, and tumors. Non-inflammatory reaction and septic shock. In addition to those produced by several somatic cells, TNF-α has a major source of production, monocytes and sputum lymphocytes. During the process of obtaining the response, APC is displayed under the guidance of pre-inflammatory to signal stimulation of IL-12 ρ70 (heterodimer of ρ35 and ρ40), and then the intercellular secretion characteristics of the activated Th cells. Polarization becomes a costimulatory molecule of the scorpion type. Thl cells exhibit a characteristic pathway of interleukin, in which the interferon r (IFN-r) secretion is a true footprint. These cells secrete high amounts of TNF-α. IFN-τ secreted by Th1 promotes many potentially important effects on inflammatory diseases. In one aspect, it further activates monocytes and gives the amount of TNF-[alpha] secretion caused by the stimulation. On the other hand, its TNF-α receptor is more strongly signaled downstream. In summary, IFN- has its own direct effects such as antiviral activity, but also the performance of MHC-II and by the force of D TNF-α still strong effect to make inflammation, target the symptoms of these alpha When two immune-impaired patients are Th-1, the increase in decomposition also causes the increase in r to cause inflammation and lesions in 201010988. The pre-inflammatory grade is initiated by IFN-r and TNF- 〇: production of TNF-[alpha] or Th1 cells of monocytes, which is expanded by other pro-inflammatory interleukin pathways, such as IL-8. IL-8 (regardless of its original nomenclature) is described as a chemokine produced by macrophages and other cell types, such as epithelial cells, which can also be synthesized by endothelial cells and is therefore also referred to as CXCL8. Although the neutrophil granulosa cell line is the primary target cell for IL-8, a wide variety of cells (endothelial cells, macrophages, hypertrophic cells, keratinocytes) also respond to this chemokine. The primary function of IL-8 is to induce chemotaxis of its target cells, such as neutrophil cells. In neutrophils, a series of cellular physiological responses required for migration and its major functional phagocytosis are also triggered, such as increased intracellular Ca2+, extracellular secretion (such as histamine release), and respiratory bursts. IL-8 can be secreted by any cell having a TLR associated with an innate immune response. The primary function of IL-8 is to recruit neutrophils to phagocytose the antigen that causes the antigenic pattern TLR. By delivering IL-8 target cells to the endothelium and other target tissues, in addition to its role in body defense, Q IL-8 is involved in the expansion and progression of many of the pathological effects of TNF-α. The movement and regression of white blood cells is regulated not only by chemotactic hormones and their receptors, but also by several adhesion molecules. Among these adhesion molecules, the selectin CD62L is considered to be a therapeutic target for preventing leukocytes from moving to the lymph nodes, and thus is considered as an evaluation parameter for the in vitro effect of arranging non-steroidal anti-inflammatory drugs (NSAIDs). Many immunosuppressive agents and anti-TNF-α molecules affect normal immune -8- 201010988 \ · defense mechanisms because they enhance cytotoxicity against immune cells or inhibit cell line expansion before successful effector immune response Proliferation mechanism. Given the importance of TNF-α secreted by extracellular space (whether soluble in the plasma membrane insertion or secretion), many studies have been designed to block extracellular TNF-α and TNF-receptors I and/or A therapeutic agent for the interaction of TNF-receptor II. The most suitable method is to use a soluble decoy φ TNF-receptor to capture TNF-α and prevent the interaction of the proinflammatory ligand with the cellular receptor due to the long time required for dissociation. The second strategy is to produce humanized anti-human TNF-α antibodies in a conventional manner or to produce bispecific single-chain molecules that are also labeled with other molecules associated with a given disease (eg, in rheumatoid arthritis). Anti-VEGF/anti-TNF-a). Although the above-mentioned molecules are TNF-α antagonists, they limit their mechanism of action to block extracellular secretion of TNF-α. A detailed but incomplete standard that can drive the production and secretion of TNF-α may be: (a) a molecule that drives transcription of TNF-a; (b) drives TNF-a-RNA transport from the nucleus to the cytoplasm and RNA. a molecule that cleaves; (c) a molecule that directs translation of TNF-α; (d) a molecule that regulates TNF-cz-mRNA stability; (e) a cell membrane that directs high-bass cells to the surface of the pre-TNF-α anchoring site Molecules; (f) molecules associated with secretion of TNF-α secreting molecules such as TNF-α converting enzyme (TACE) and (g) molecules that regulate the internalization of the surface form of pre-TNF-α and its signals. All of these are related to the cell labeling of TNF-α production and secretion. Although there are different ways to block the treatment of TNF-o: 201010988, if it can be found that selective blockade is not just TNF-α production, there are other key pro-inflammatory interleukins (such as IFN-r). The new drugs produced will be highly valued. SUMMARY OF THE INVENTION The authors of the present invention have surprisingly discovered that the compounds of formula (I) exhibit some highly interesting immunomodulatory effects which may be useful in controlling the pathological mechanisms of acute and chronic inflammatory diseases and therefore have potential clinical utility. Specifically, the compound used in the present invention can inhibit the production of TNF-α from peripheral blood mononuclear cells (PBMC) of patients with chronic inflammatory diseases such as rheumatoid arthritis, and also inhibits the passage of these cells. IFN-Τ secretion after T cell stimulation. Furthermore, the compounds of formula (I) have been able to modulate the bioreactive properties of secreted chemokines (i.e., IL-8) and regulatory interleukins (i.e., IL-10) and are regulated in different ways in health and disease states. The overall immunomodulatory effects of the compounds of formula (I) do not cause any toxic effects on monocytes from peripheral blood. In addition, activation and proliferative responses following stimulation of cell division are not regulated or even increased by these compounds. In this small molecule, it also has the inhibitory effect on several proinflammatory interleukins such as TNF-α and IFN-r and regulates IL-8 chemokines and/or IL-1 0 regulatory interleukins (all The potential of systemic and organ-specific autoimmune disorders, transplantation, acute and chronic inflammatory diseases, certain metabolic and degenerative diseases, and the pathophysiology of atherosclerosis, Compounds can be classified in the novel immunomodulatory 201010988 class of agents and are exemplified by a cascade of proinflammatory/regulatory interleukins and chemokines prior to various possible reprogramming levels of clinical and therapeutic importance. It is apparent that the immune cell line is sessile and enters the organ to patrol body tissue that is infiltrated in an inflammatory condition in which the immune cells accumulate in lesions distributed according to disease activity, target organ, and degree of damage. Since the compounds used in the present invention are expected to modulate several morphological forms of the pathophysiological process, development assays can be utilized to characterize the number of receptors, binding efficiency, receptor occupancy, and drug probe concentration. Since the white blood cells have a regression characteristic, detecting the therapeutic target can also obtain information about the location of the target cell and the lesion site. Thus, the compounds of formula (I) also exhibit a use as a biomarker for development in drug development, clinical trials and individualized medicine, which provides information not only on pharmacokinetics, distribution and administration, but also in preclinical and Important data on individualized response patterns in clinical trials. The latter may Φ result in a surrogate biomarker defining a validated, reliable, individualized clinical endpoint for administration to a patient in need of the prognostic and individualized assessment of an effective amount of a compound of formula (I) or a pharmaceutical composition thereof , to define the precise bio-developing technology known to those skilled in the art. According to a first aspect, the invention relates to a compound of formula (I): -11 - 201010988

(D 或其醫藥上可接受之鹽、前藥及/或溶劑合物於製備 用於治療急性或慢性發炎疾病之藥物上之用途,其中:Use of (D or a pharmaceutically acceptable salt, prodrug and/or solvate thereof for the manufacture of a medicament for the treatment of an acute or chronic inflammatory disease, wherein:

Ri係選自氫、經取代或未經取代之CrCe烷基、經取 © 代或未經取代之環烷基、經取代或未經取代之芳基或經取 代或未經取代之雜環基; 、 r2係選自經取代或未經取代之C!-c6烷基、經取代或 未經取代之環烷基或N(R’R”),其中R’和R”各別爲氫或 經取代或未經取代之烷基;且 R3係未經取代之Ci-6烷基。在特定實施態樣中,該 用於治療急性或慢性發炎疾病之藥物的作用係藉由抑制至 少一種選自 TNF-α或IFN-r之前發炎細胞介素之產生, ® 或藉由免疫調節IL-8趨化激素及/或IL-10調節性細胞介 素。 在本發明之另一特定實施態樣中,達成該抑制前發炎 細胞介素TNF- α之產生係至少部份於TNF- a mRNA之表 現量上。 在本發明之特定態樣中,該急性或慢性發炎疾病係選 自急性和慢性血清陽性或血清陰性寡關節炎和多發性關節 炎、脊椎關節病、腎小球性腎炎、膠原病、腎小管間質性 -12- 201010988 腎炎、代謝性徵候群、動脈粥樣硬化、骨關節炎、氣喘、 慢性阻塞性肺病、間質性肺病、多發性硬化症、脫髓鞘病 、腦膜炎、腦炎、腦膜腦炎、發炎性神經根病變和週邊神 經病變、發炎性腸病、硬化、肝炎、心衰竭、缺血性疾病 、腎衰竭、發炎性膀胱炎、良性前列腺增生、前列腺炎、 心肌炎、心包膜炎、葡萄膜炎、異位性皮膚炎、濕疹、奪 麻疹、牛皮癣、玫瑰痤瘡、過敏性鼻炎、敗血症、敗血性 〇 休克、多重器官衰竭、全身性自體免疫疾病(諸如全身性 紅斑性狼瘡)、血管炎、皮膚肌炎、澱粉樣變性症或類肉 瘤病、器官特異性自體免疫疾病(諸如重症肌無力)、甲狀 腺炎或胰島炎、器官移植、感染和腫瘤引起之發炎、 TNF- α依賴性細胞退化、壞死、細胞凋亡、移植物抗宿 主病、惡病質或自體分泌和旁分泌病理性細胞生長。 這些治療適應症係至少異常之免疫反應、免疫調節異 常、免疫干擾、免疫發病機制、免疫療法、免疫抑制或免 〇 疫調節性生物反應之後果。 在另一態樣中,本發明關於式(Γ )化合物Ri is selected from hydrogen, substituted or unsubstituted CrCe alkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted aryl or substituted or unsubstituted heterocyclic ; r2 is selected from substituted or unsubstituted C!-c6 alkyl, substituted or unsubstituted cycloalkyl or N(R'R"), wherein R' and R" are each hydrogen or A substituted or unsubstituted alkyl group; and R3 is an unsubstituted Ci-6 alkyl group. In a particular embodiment, the medicament for treating an acute or chronic inflammatory disease is effected by inhibiting the production of at least one inflammatory interleukin selected from TNF-[alpha] or IFN-r, or by immunomodulating IL -8 Chemokines and/or IL-10 regulatory interleukins. In another specific embodiment of the invention, the production of the inflammatory interleukin TNF-[alpha] prior to the inhibition is at least partially expressed in terms of the amount of TNF-a mRNA. In a particular aspect of the invention, the acute or chronic inflammatory disease is selected from the group consisting of acute and chronic seropositive or seronegative oligoarthritis and polyarthritis, spondyloarthropathy, glomerulonephritis, collagen disease, renal tubules Interstitial -12- 201010988 Nephritis, metabolic syndrome, atherosclerosis, osteoarthritis, asthma, chronic obstructive pulmonary disease, interstitial lung disease, multiple sclerosis, demyelinating disease, meningitis, encephalitis , meningoencephalitis, inflammatory radiculopathy and peripheral neuropathy, inflammatory bowel disease, cirrhosis, hepatitis, heart failure, ischemic disease, renal failure, inflammatory cystitis, benign prostatic hyperplasia, prostatitis, myocarditis, heart Encapsulation, uveitis, atopic dermatitis, eczema, measles, psoriasis, rose acne, allergic rhinitis, sepsis, septic shock, multiple organ failure, systemic autoimmune disease (such as systemic Lupus erythematosus), vasculitis, dermatomyositis, amyloidosis or sarcoma-like disease, organ-specific autoimmune diseases (such as myasthenia gravis), Goiter or insulitis, organ transplantation, infection and tumor-induced inflammation, TNF-α-dependent cell degeneration, necrosis, apoptosis, graft versus host disease, cachexia or autocrine and paracrine pathological cell growth. These therapeutic indications are at least abnormal immune response, immunoregulatory abnormalities, immune interference, immune pathogenesis, immunotherapy, immunosuppression or quarantine-free biological response. In another aspect, the invention relates to a compound of formula (Γ)

或其醫藥上可接受之鹽、前藥及/或溶劑合物, -13- 201010988 其中: I係選自氫、經取代或未經取代之Ci_c6烷基、經取 代或未經取代之環烷基、經取代或未經取代之芳基或經取 代或未經取代之雜環基;且 R3係未經取代之Ci-Q烷基。 在第三態樣中,本發明之目的爲一種醫藥組成物,該 醫藥組成物包含如上述定義之式(I’)化合物或其醫藥上可 接受之鹽、前藥及/或溶劑合物,及至少一種醫藥上可接 受之載劑、佐劑及/或賦形劑。 本發明之另一態樣關於如上述定義之式(Γ)化合物, 其用途係作爲藥物。 最後,本發明之另一態樣關於如上述定義之式(I)化 合物(特別是式(Γ)化合物)於顯影和藥理顯影技術中作爲 顯影生物標記之用途。該些生物標記可被用於找出免疫病 灶、標靶細胞及標的分子。 本發明之詳細說明 在本發明之上下文中,下列用語具有如下詳述之意義 用語「山_6烷基」係指由碳及氫原子所組成之線狀或 分支烴鏈基團’不含不飽和烴,具有一至六個碳原子,且 係以單鍵與其餘分子連接,例如甲基、乙基、n_丙基、 丙基、η-丁基、t-丁基、n-戊基等。Cl-6烷基基團可選擇 性地被一或多個取代基取代,諸如環烷基、芳基、雜環基 • 14- 201010988 \ 、鹵基、羥基、烷氧基、氰基、胺基、硝基或烷硫基。 用語「環烷基」係指飽和或部分飽和之安定3至8元 環狀基團,其僅由碳及氫原子組成,諸如環己基或環戊基 。除非說明書中另外具體說明,用語「環烷基」係意圖包 括可被至少一取代基選擇性地取代之環烷基基團,該取代 基係獨立選自氫、q-6烷基基團、鹵基、羥基、_ N(R3)(R4),其中尺3及R4係獨立選自氫及線狀或分支Cl.6 ❹ 院基基團。 用語「芳基」係指安定之5至8元芳香環狀基團,且 其僅由碳及氫原子組成,諸如苯基或環辛四烯。除非說明 書中另外具體說明,用語「芳基」係意圖包括可被至少一 取代基選擇性地取代之芳基基團,該取代基係獨立選自氫 、(:丨-6烷基基團、鹵基、羥基、-N(R3)(R4),其中R3及 R4係獨立選自氫及線狀或分支烷基基團。 「雜環基」係指安定之3至8元環狀基團,其係由碳 〇 原子及一至五個選自氮、氧及硫之雜原子所組成。就本發 明之目的而言,該雜環可能爲部份或完全飽和或芳香。該 雜環之實例包括但不限於吡咯啶、吡啶、噻吩、呋喃等。 除非說明書中另外具體說明,用語「雜環基」係意圖包括 可被至少一取代基選擇性地取代之雜環基基團,該取代基 係獨立選自氫、Cu烷基基團、鹵基、羥基、_n(r3)(r4) ’其中R3及R4係獨立選自氫及線狀或分支Cl.6烷基基團 〇 用§吾「鹵基」係指漠基、氯基、确基或氣基。 -15- 201010988 用語「急性和慢性血清陽性或血清陰性寡關節炎和多 發性關節炎」係指具有陽性或陰性類風濕性因子之涉及一 或數個動關節之滑膜炎疾病,包括類風濕性關節炎及原發 性及繼發性休格倫氏徵候群。 用語「脊椎關節病」係指涉及骶髂關節及/或脊椎及/ 或週邊關節之免疫病理性HLA-B27相關之發炎疾病,亦 包括葡萄膜炎。 用語「腎小球性腎炎」係指腎小球之發炎病灶。 用語「腎小管間質性腎炎」係指涉及腎小管及腎間質 之發炎疾病。 用語「膠原病」係指具有病理免疫機制之系統性發炎 疾病’包括全身性紅斑性狼瘡(SLE)、皮膚肌炎及硬皮病 〇 用語「發炎性腸病」係指具有病理免疫機制之胃腸道 發炎疾病,不論有或無系統性特徵,包括克隆氏病及潰瘍 性結腸炎。 用語「阻塞性肺病」係指用力吐氣體積(FEV)可逆或 不可逆降低之支氣管疾病,包括氣喘及慢性阻塞性肺病。 用語「間質性肺病」係指涉及肺間質之發炎疾病。 用語「脫髓鞘病」係指具有引發髓鞘溶解之病理免疫 機制之中樞神經系統發炎疾病,包括多發性硬化症及視神 經炎。 用語「腦膜炎、腦炎及腦膜腦炎」係指腦膜及/或中 樞神經系統其他構造之發炎疾病。 -16- 201010988 \ 用語「發炎性神經根病變和週邊神經病變」係指週邊 神經系統之發炎疾病。 用語「發炎性膀胱炎」係指膀胱之發炎疾病。 用語「良性前列腺增生」係指前列腺之非惡性肥大及 /或增生。 用語「異位性皮膚炎、濕疹及蓴麻疹」係指具有不論 有或無關於IgE之免疫病理機制之過敏性皮虜疾病。 φ 用語「牛皮癖」係指與免疫系統病理機制有關之過度 角化及紅斑性皮膚反應》 用語「玫瑰痤瘡」係指皮膚之常見發炎狀況,特徵爲 在臉部中央出現紅斑(潮紅及發紅),亦可橫過臉頰、鼻子 或前額,較少出現在頸部及胸部。 用語「過敏性鼻炎」係指間歇性(亦稱爲季節性)或持 續性(亦稱爲經年性)發炎鼻黏膜疾病,其與免疫過敏病理 機制有關。 # 用語「敗血症、敗血性休克及多重器官衰竭」係指對 微生物劑或其他致病因子之異常免疫反應所媒介之系統性 發炎疾病。 用語「類肉瘤病及澱粉樣變性症」係指器官及/或系 統性涉入且無明確定義原因之自發性免疫疾病,其中可觀 察到異常之免疫反應。 用語「器官特異性自體免疫疾病」係指並無經定義之 致病因子之免疫系統媒介之器官病灶,包括重症肌無力、 甲狀腺炎、腦垂體炎、腎上腺炎及其他。 -17- 201010988 用語「器官移植」係指預防及治療經移植之細胞、組 織及器官之排斥。 用語「感染和腫瘤引起之發炎」係指繼發於微生物劑 或癌症刺激之異常免疫反應。 用語「TNF- α依賴性細胞退化、細胞凋亡或壞死」 係指由TNF-α所引起之組織退化或死亡。 用語「移植物抗宿主病」係指由移植細胞所引起之發 炎免疫反應。 用語「惡病質」係指由發炎或腫瘤性疾病所引起之系 統性厭食或營養不良及體重減輕。 用語「動脈粥樣硬化」係指繼發於粥樣瘤或由發炎細 胞(大部分爲巨噬細胞)及含有脂質之細胞碎片堆積於動脈 壁所引起之任何動脈硬化。 用語「缺血性疾病」係指繼發於組織氧合狀態及/或 血流降低之器官病灶包括心臟及腦血管缺血。 用語「自體分泌和旁分泌病理性細胞生長」係指細胞 利用TNF- a作爲調節細胞活化及增生因子之細胞介素之 惡性或良性疾病。 除非另外說明,本發明所使用之化合物係意圖包括只 有一或多個同位素富集原子存在差異之化合物。舉例來說 ’具有本結構但以氘或氚取代氫,或以13C -或14C -富集碳 或15N-富集氮取代碳之化合物係屬於本發明之範圍內。 用語「其醫藥上可接受之鹽、溶劑合物或前藥」係關 於當被投予至接受者時可(直接或間接)提供化合物諸如此 -18- 201010988 處所描述者之鹽、溶劑合物或前藥。然而,將會發現醫藥 上不可接受之鹽亦屬於本發明之範圍內,因爲它們可被用 於製備醫藥上可接受之鹽。鹽、前藥及衍生物可利用該最 新所知之方法製備。「醫藥上可接受」係較佳地關於當投 予至人或動物時爲生理上可耐受且通常不會造成過敏反應 或類似不利反應諸如胃異常、暈眩及該類似反應之分子實 體及組成物。用語「醫藥上可接受」代表其經過聯邦或州 ❻ 政府之管理機構核准或被包括在美國藥典或其他普遍公認 之用於動物特別是人之藥典。 舉例來說,本發明先前所描述之化合物之醫藥上可接 受之鹽係利用習知化學方法從該含有鹼性或酸性單位之先 前描述之化合物合成。該鹽通常藉由例如使該些化合物之 游離酸性或鹼性形式與化學當量之在水或有機溶劑或二者 之混合物中之適當鹼或酸反應而加以製備。不含水之介質 諸如乙醚、乙酸乙酯、乙醇、異丙醇或乙腈通常係較佳地 〇 。酸添加鹽之實例包括例如礦物酸添加鹽諸如氯化氫、溴 化氫、碘化氫、硫酸鹽、硝酸鹽、磷酸鹽及例如有機酸添 加鹽諸如醋酸鹽、順丁烯二酸鹽、延胡索酸鹽、檸檬酸鹽 、草酸鹽、琥珀酸鹽、酒石酸鹽、蘋果酸鹽、杏仁酸鹽、 甲磺酸鹽及對甲苯磺酸鹽。鹼添加鹽之實例包括例如無機 鹽諸如鈉、鉀、鈣、銨、鎂、鋁、鋰,及例如有機鹼鹽諸 如乙二胺、乙醇胺、N,N -二烯基乙醇胺、還原葡糖胺及鹼 性胺基酸鹽。 用語「前藥」在此處係定義爲一種已經歷化學衍生作 19 · 201010988 用諸如取代或添加額外化學基團以改變(爲了醫藥用途)彼 之一些物理化學特性(諸如溶解性或生物利用率)之化學化 合物,例如衍生自活性化合物之酯或醚在被投予至個體後 產生本身之活性化合物。用於從給定之活性化合物產製前 藥之廣爲週知之方法實例係爲該領域之技藝人士所知,且 可見於例如 Krogsgaard-Larsen et al·,Textbook of Drug Design and Discovery, Taylor & F r an ci s (Apri 1 2002)。根 據本發明,用語「溶劑合物」係用來代表有其他分子(最 可能的是極性溶劑)經由非共價鍵與其結合之任何形式之 本發明之化合物。溶劑合物之實例包括水合物及醇化物例 如甲醇化物。 特別較佳之前藥係當該化合物被投予至病患時可增加 本發明之化合物之生物利用率者(例如使經口投予之化合 物更快地被吸收至血液中)或該些相對於原始化合物可增 加原始化合物分佈至生物隔室(例如腦或淋巴系統)者。 本發明所使用之化合物可呈游離化合物或溶劑合物( 例如水合物)之結晶形式也就是多形體,應了解的是二種 形式皆屬於本發明之範圍內。溶合方法係該領域所普遍知 悉。適當之溶劑合物係醫藥上可接受之溶劑合物。在特定 實施態樣中,該溶劑合物係水合物。 鹽 '溶劑合物及前藥可利用最新已知之方法製備。將 可觀察到醫藥上不可接受之鹽、溶劑合物或前藥亦包括在 本發明之範圍內’因爲它們可被用於製備醫藥上可接受之 鹽、溶劑合物或前藥。 -20- 201010988 式(I)化合物或彼等之鹽或溶劑合物係較佳地呈醫藥 上可接受之形式或實質上純的形式。醫藥上可接受之形式 被了解爲特別是具有排除正常醫藥添加劑諸如稀釋劑及賦 形劑之醫藥上可接受之純度,且不包含任何在正常劑量程 度下被認爲具有毒性之物質。該藥物之純度較佳係高於 5 0 %,更佳爲高於7 0 %,且仍然更佳係高於9 0 %。在較佳 之實施態樣中,其係高於95%之式(I)化合物或其鹽、溶 φ 劑合物或前藥。 以上述式(I)所顯示之本發明之化合物可包括依手性 中心存在而定之鏡像異構物或依多鍵存在而定之異構物( 例如Z、E)。各種異構物、鏡像異構物、非鏡像異構物及 彼等之混合物係屬於本發明之範圍內。 在特定之實施態樣中,爲了供顯影和藥理顯影技術上 之用途,或在藥物發展、臨床試驗及個體化醫藥上作爲生 物標記之替代用途,式(I)化合物可經螢光或發光標籤或 Φ 旗幟標示,該標記利用該領域之技藝人士所知之任何方法 導入。 在本發明之特定實施態樣中,係經取代或未經取 代之c,-c6烷基。較佳之Ri係甲基。 在另一特定之實施態樣中,R2係經取代或未經取代 之烷基、經取代或未經取代之環烷基。較佳的是, R2係經取代或未經取代之環烷基。更佳的是,r2係環戊 基。在另一較佳之實施態樣中’ R2係經取代或未經取代 之CrQ烷基。較佳的是,R2係經雜環基取代之Cl-C6院 -21 - 201010988 基。更佳的是,R2係經環氧乙烷取代之CrQ烷基。 在另一特定之實施態樣中,r3係甲基。 在另一更爲特定之實施態樣中,本發明所使用之式 (I)化合物係選自下列化合物,或其醫藥上可接受之鹽、 前藥及/或溶劑合物。Or a pharmaceutically acceptable salt, prodrug and/or solvate thereof, -13- 201010988 wherein: I is selected from the group consisting of hydrogen, substituted or unsubstituted Ci_c6 alkyl, substituted or unsubstituted naphthenic a substituted or unsubstituted aryl group or a substituted or unsubstituted heterocyclic group; and R3 is an unsubstituted Ci-Q alkyl group. In a third aspect, the object of the present invention is a pharmaceutical composition comprising a compound of the formula (I') as defined above, or a pharmaceutically acceptable salt, prodrug and/or solvate thereof, And at least one pharmaceutically acceptable carrier, adjuvant and/or excipient. Another aspect of the invention pertains to a compound of formula (Γ) as defined above for use as a medicament. Finally, another aspect of the invention pertains to the use of a compound of formula (I) as defined above, particularly a compound of formula (U.S.), as a biomarker for development in development and pharmacological development techniques. These biomarkers can be used to identify immune lesions, target cells, and target molecules. DETAILED DESCRIPTION OF THE INVENTION In the context of the present invention, the following terms have the meanings as detailed below. The term "Mountain-6 alkyl" means a linear or branched hydrocarbon chain group consisting of carbon and hydrogen atoms. a saturated hydrocarbon having from one to six carbon atoms and linked to the remaining molecules by a single bond, such as methyl, ethyl, n-propyl, propyl, η-butyl, t-butyl, n-pentyl, etc. . The Cl-6 alkyl group may be optionally substituted by one or more substituents such as cycloalkyl, aryl, heterocyclyl • 14- 201010988 \ , halo, hydroxy, alkoxy, cyano, amine Base, nitro or alkylthio. The term "cycloalkyl" refers to a saturated or partially saturated diazet 3 to 8 membered cyclic group consisting solely of carbon and hydrogen atoms, such as cyclohexyl or cyclopentyl. Unless the specification specifically dictates otherwise, the term "cycloalkyl" is intended to include a cycloalkyl group which may be optionally substituted with at least one substituent selected independently from hydrogen, q-6 alkyl group, Halo, hydroxy, _N(R3)(R4), wherein ampules 3 and R4 are independently selected from the group consisting of hydrogen and linear or branched Cl.6 院 院. The term "aryl" refers to a 5 to 8 membered aromatic cyclic group of diazepam and is composed only of carbon and hydrogen atoms, such as phenyl or cyclooctatetraene. Unless the specification specifically dictates otherwise, the term "aryl" is intended to include an aryl group which is optionally substituted with at least one substituent selected independently from hydrogen, (: 丨-6 alkyl group, Halo, hydroxy, -N(R3)(R4), wherein R3 and R4 are independently selected from hydrogen and a linear or branched alkyl group. "Heterocyclyl" means a stable 3 to 8 membered cyclic group. It consists of a carbonium atom and one to five heteroatoms selected from the group consisting of nitrogen, oxygen and sulfur. For the purposes of the present invention, the heterocyclic ring may be partially or fully saturated or aromatic. Including, but not limited to, pyrrolidine, pyridine, thiophene, furan, etc. Unless otherwise specifically stated in the specification, the term "heterocyclyl" is intended to include a heterocyclyl group which is optionally substituted with at least one substituent, the substituent Is independently selected from the group consisting of hydrogen, Cu alkyl group, halogen group, hydroxyl group, _n(r3)(r4) ' wherein R3 and R4 are independently selected from hydrogen and linear or branched Cl.6 alkyl group. "Halo" means a desert, a chlorine, a confirmatory or a gas base. -15- 201010988 The term "acute and chronic seropositive or "Keep-negative oligoarthritis and polyarthritis" refers to synovitis diseases involving one or several joints with positive or negative rheumatoid factors, including rheumatoid arthritis and primary and secondary Glenn syndrome. The term "sponge joint disease" refers to an immunopathological HLA-B27-associated inflammatory disease involving the ankle joint and/or the spine and/or surrounding joints, including uveitis. "Acne nephritis" refers to an inflammatory lesion of the glomerulus. The term "tubular interstitial nephritis" refers to an inflammatory disease involving the renal tubules and the renal interstitium. The term "collagen disease" refers to a systemic inflammatory disease with pathological immune mechanisms. 'Including systemic lupus erythematosus (SLE), dermatomyositis, and scleroderma. "Inflammatory bowel disease" refers to gastrointestinal inflammatory disease with pathological immune mechanisms, with or without systemic features, including Crohn's disease. And ulcerative colitis. The term "obstructive pulmonary disease" refers to a bronchial disease that is reversible or irreversibly reduced in forced expiratory volume (FEV), including asthma and chronic obstructive pulmonary disease. "Pulmonary disease" refers to an inflammatory disease involving the interstitial lung. The term "demyelinating disease" refers to a pathogenic immune mechanism that causes myelinolysis to cause central nervous system inflammatory diseases, including multiple sclerosis and optic neuritis. "Inflammation, encephalitis, and meningoencephalitis" refers to inflammatory diseases of the meninges and/or other structures of the central nervous system. -16- 201010988 \ The term "inflammatory radiculopathy and peripheral neuropathy" refers to the inflammatory disease of the peripheral nervous system. The term "inflammatory inflammatory cystitis" refers to an inflammatory disease of the bladder. The term "benign prostatic hyperplasia" means non-malignant hypertrophy and/or hyperplasia of the prostate. The term "atopic dermatitis, eczema and urticaria" means With or without allergic skin disease associated with the immunopathological mechanism of IgE. Φ The term "psoriasis" refers to hyperkeratosis and erythematous skin reactions associated with the pathogenesis of the immune system. The term "rosal acne" refers to the common inflammatory condition of the skin characterized by erythema (redness and redness) in the center of the face. ), can also cross the cheeks, nose or forehead, less in the neck and chest. The term "allergic rhinitis" refers to intermittent (also known as seasonal) or persistent (also known as perennial) inflammatory nasal mucosal disease, which is associated with immunopathic pathogenesis. # The term "sepsis, septic shock and multiple organ failure" refers to a systemic inflammatory disease that is mediated by an abnormal immune response to a microbial agent or other virulence factor. The term "sarcoma-like and amyloidosis" refers to a spontaneous immune disease in which an organ and/or system is involved without a well-defined cause, in which an abnormal immune response is observed. The term "organ-specific autoimmune disease" refers to organ lesions of the immune system that are not defined by the causative agent, including myasthenia gravis, thyroiditis, pituitary gland, adrenalitis, and others. -17- 201010988 The term "organ transplantation" refers to the prevention and treatment of rejection of transplanted cells, tissues and organs. The term "inflammation caused by infection and tumor" refers to an abnormal immune response secondary to microbial agents or cancer stimulation. The term "TNF-α-dependent cell degeneration, apoptosis or necrosis" refers to tissue degradation or death caused by TNF-α. The term "graft versus host disease" refers to an inflammatory immune response caused by a transplanted cell. The term "cachexia" refers to a systemic anorexia or malnutrition and weight loss caused by an inflammatory or neoplastic disease. The term "atherosclerosis" refers to any arteriosclerosis caused by atheroma or by inflammatory cells (mostly macrophages) and accumulation of cell debris containing lipids on the arterial wall. The term "ischemic disease" refers to organ lesions, including cardiac and cerebral ischemia, secondary to tissue oxygenation and/or decreased blood flow. The term "autologous secretion and paracrine pathological cell growth" refers to a cell using TNF-a as a malignant or benign disease that regulates the cellular activation and proliferation of interleukins. Unless otherwise stated, the compounds used in the present invention are intended to include compounds having only one or more isotopically enriched atoms. For example, a compound having the present structure but substituting hydrogen with hydrazine or hydrazine, or substituting carbon with 13C- or 14C-enriched carbon or 15N-enriched nitrogen is within the scope of the present invention. The phrase "a pharmaceutically acceptable salt, solvate or prodrug thereof" relates to a salt or solvate which, when administered to a recipient, provides (directly or indirectly) a compound such as those described in this -18-201010988. Or a prodrug. However, it will be found that pharmaceutically unacceptable salts are also within the scope of the invention as they can be used in the preparation of pharmaceutically acceptable salts. Salts, prodrugs and derivatives can be prepared by the method known from the latest. "Pharmaceutically acceptable" is preferably a molecular entity which is physiologically tolerable when administered to a human or animal and which generally does not cause an allergic reaction or a similar untoward reaction such as gastric abnormalities, dizziness and the like. Composition. The term "pharmaceutically acceptable" means approved by the regulatory agency of the federal or state government or included in the United States Pharmacopoeia or other generally recognized pharmacopoeia for animals, especially humans. For example, a pharmaceutically acceptable salt of a compound previously described herein is synthesized from the previously described compound containing a basic or acidic unit using conventional chemical methods. The salt is usually prepared by, for example, reacting the free acidic or basic form of the compounds with a suitable base or acid in water or an organic solvent or a mixture of the two. Non-aqueous media such as diethyl ether, ethyl acetate, ethanol, isopropanol or acetonitrile are generally preferred. Examples of the acid addition salt include, for example, mineral acid addition salts such as hydrogen chloride, hydrogen bromide, hydrogen iodide, sulfates, nitrates, phosphates, and, for example, organic acid addition salts such as acetate, maleate, fumarate, Citrate, oxalate, succinate, tartrate, malate, mandelate, methanesulfonate and p-toluenesulfonate. Examples of the base-added salt include, for example, inorganic salts such as sodium, potassium, calcium, ammonium, magnesium, aluminum, lithium, and, for example, organic base salts such as ethylenediamine, ethanolamine, N,N-dienylethanolamine, reduced glucosamine, and Basic amino acid salt. The term "prodrug" is defined herein as a chemical derivative that has undergone chemical derivation 19 · 201010988 to alter (for medical use) some of its physicochemical properties (such as solubility or bioavailability) by, for example, substituting or adding additional chemical groups. The chemical compound, for example an ester or ether derived from the active compound, produces the active compound of its own after it is administered to the individual. Examples of well-known methods for producing prodrugs from a given active compound are known to those skilled in the art and can be found, for example, in Krogsgaard-Larsen et al., Textbook of Drug Design and Discovery, Taylor & F r an ci s (Apri 1 2002). According to the invention, the term "solvate" is used to denote a compound of the invention in any form in which other molecules, most likely polar solvents, are bonded thereto via non-covalent bonds. Examples of the solvate include hydrates and alcoholates such as methanolate. It is particularly preferred that the prior drug system increases the bioavailability of the compound of the invention when administered to a patient (eg, allows the orally administered compound to be absorbed into the blood more quickly) or The original compound can increase the distribution of the original compound to the biological compartment (eg, the brain or lymphatic system). The compound used in the present invention may be in the form of a crystalline form of a free compound or a solvate (e.g., a hydrate), i.e., a polymorph, and it is to be understood that both forms are within the scope of the present invention. The method of solubilization is generally known in the art. Suitable solvates are pharmaceutically acceptable solvates. In a particular embodiment, the solvate is a hydrate. Salt 'solvates and prodrugs can be prepared by the latest known methods. It will be observed that pharmaceutically unacceptable salts, solvates or prodrugs are also included within the scope of the invention' because they can be used in the preparation of pharmaceutically acceptable salts, solvates or prodrugs. -20- 201010988 The compounds of formula (I) or their salts or solvates are preferably in pharmaceutically acceptable form or in substantially pure form. The pharmaceutically acceptable form is understood to be, in particular, a pharmaceutically acceptable purity excluding normal pharmaceutical additives such as diluents and excipients, and does not contain any substance which is considered to be toxic under normal dosage levels. The purity of the drug is preferably more than 50%, more preferably more than 70%, and still more preferably more than 90%. In a preferred embodiment, it is greater than 95% of the compound of formula (I) or a salt thereof, a lysate or a prodrug. The compound of the present invention shown by the above formula (I) may include an image isomer or an isomer (e.g., Z, E) depending on the presence of a chiral center. Various isomers, mirror image isomers, non-image isomers, and mixtures thereof are within the scope of the invention. In a particular embodiment, the compound of formula (I) may be fluorescent or luminescent for use in development and pharmacological development techniques, or as an alternative to biomarkers in drug development, clinical trials, and individualized medicine. Or Φ banner designation, which is imported using any method known to those skilled in the art. In a particular embodiment of the invention, the substituted, unsubstituted c,-c6 alkyl group. Preferably, Ri is a methyl group. In another specific embodiment, R2 is a substituted or unsubstituted alkyl, substituted or unsubstituted cycloalkyl. Preferably, R2 is a substituted or unsubstituted cycloalkyl group. More preferably, the r2 is a cyclopentyl group. In another preferred embodiment, 'R2 is a substituted or unsubstituted CrQ alkyl group. Preferably, R2 is substituted by a heterocyclic group of Cl-C6 -21 - 201010988. More preferably, R2 is a CrQ alkyl group substituted with ethylene oxide. In another specific embodiment, r3 is methyl. In another more specific embodiment, the compound of formula (I) used in the present invention is selected from the following compounds, or a pharmaceutically acceptable salt, prodrug and/or solvate thereof.

◎ -22- 201010988◎ -22- 201010988

00 (9) a. ^Xk: H-Ij 。八。⑽ 5卜 a\v 1 7—ch2-cn Ά、 α0 △㈣ Cl N 八(15) α0 ^>-〇 。八。⑽ O.〇 C,\ ό (17) α0 cy^o (18) 本發明所使用之式(I)化合物可利用可得到之合成方 法獲得。舉例來說,它們可利用下列圖示所摘要之方法製 備。 -23- 20101098800 (9) a. ^Xk: H-Ij. Eight. (10) 5 Bu a\v 1 7—ch2-cn Ά, α0 △ (4) Cl N 八(15) α0 ^>-〇 . Eight. (10) O. 〇 C, \ ό (17) α0 cy^o (18) The compound of the formula (I) used in the present invention can be obtained by a synthetic method which can be obtained. For example, they can be prepared by the methods outlined in the following figures. -23- 201010988

此方法首先包含化合物4-R3-苯硫基乙酸之酯化反應 以產生甲基酯衍生物。該反應可利用甲基化劑(諸如Mel) 在鹽(諸如NaHC03)存在時進行,或利用MeOH作爲甲基 化劑在酸性介質存在時進行。 在下一步驟中,該甲基酯衍生物(選擇性地不經任何 純化)利用氧化劑(例如發氧方(oxone))在該硫原子處氧化 ,因此從該硫醚基團得到該楓化合物。 接下來以諸如NBS之劑進行鹵化作用,導致在該酯 基團之α -位置形成鹵化物。以不同之硫醯胺在加熱下進 行此化合物之環化加成作用以產生高純度之Ri-經取代之 5-(4-R3-磺醯基-苯基)-4-羥基噻唑。 用於此環化加成作用之硫醯胺係得自對應如·/. MM. Chem(34) 2158-2165, 1991 及 J. Org. Chem.(65),13, 3P 73, 中所述之勞森試劑之醯胺。該硫醯胺包括其中 R!係選自如上述定義之氫、經取代或未經取代之C^-Ce烷 基、經取代或未經取代之環烷基、經取代或未經取代之芳 基或經取代或未經取代之雜環基之化合物。 經由羥基導入R2基團可利用技藝人士所知之任何方 -24- 201010988 法進行以產生本發明所使用之式(I)化合物。在特定實施 態樣中,該羥基基團係與式R2-Hal之鹵化物(較佳爲R2-Br)反應,以提供式(I)化合物。 在所提到之反應中使用之所有反應物可自商業購得。 在另一態樣中,本發明提供一種式(Γ)化合物This method first comprises an esterification reaction of the compound 4-R3-phenylthioacetic acid to give a methyl ester derivative. The reaction can be carried out using a methylating agent such as Mel in the presence of a salt such as NaHC03 or using MeOH as a methylating agent in the presence of an acidic medium. In the next step, the methyl ester derivative (optionally without any purification) is oxidized at the sulfur atom using an oxidizing agent (e.g., oxone), thereby obtaining the maple compound from the thioether group. Subsequent halogenation with a reagent such as NBS results in the formation of a halide at the alpha position of the ester group. The cyclization addition of this compound is carried out under heating with different thioguanamine to produce high-purity Ri-substituted 5-(4-R3-sulfonyl-phenyl)-4-hydroxythiazole. The thioguanamine used in this cyclization addition is obtained from the corresponding ones as described in /. MM. Chem (34) 2158-2165, 1991 and J. Org. Chem. (65), 13, 3P 73, The lanthanide of the Lawson reagent. The thioguanamine includes those wherein R! is selected from the group consisting of hydrogen, substituted or unsubstituted C^-Ce alkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted aryl, as defined above Or a compound of a substituted or unsubstituted heterocyclic group. Introduction of the R2 group via a hydroxyl group can be carried out by any of the methods known to those skilled in the art from -24 to 201010988 to produce a compound of formula (I) for use in the present invention. In a particular embodiment, the hydroxyl group is reacted with a halide of the formula R2-Hal, preferably R2-Br, to provide a compound of formula (I). All of the reactants used in the reactions mentioned are commercially available. In another aspect, the invention provides a compound of formula (Γ)

cAo (Γ) 或其醫藥上可接受之鹽、前藥及/或溶劑合物, 其中:cAo (Γ) or a pharmaceutically acceptable salt, prodrug and/or solvate thereof, wherein:

Ri係選自氫、經取代或未經取代之Ci-Ce烷基、經取 代或未經取代之環烷基、經取代或未經取代之芳基或經取 代或未經取代之雜環基;且Ri is selected from hydrogen, substituted or unsubstituted Ci-Ce alkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted aryl or substituted or unsubstituted heterocyclic And

Rs係未經取代之(^-Ce烷基基團。 在特定之實施態樣中,1係經取代或未經取代之Ci-C6焼基。較佳之1^係甲基。 在另一特定之實施態樣中,R3係甲基。 在另一特定之實施態樣中,本發明之式(Γ)化合物係 選自下列’或其醫藥上可接受之鹽、前藥及/或溶劑合物 -25- 201010988Rs is an unsubstituted (^-Ce alkyl group. In a specific embodiment, 1 is a substituted or unsubstituted Ci-C6 fluorenyl group. Preferably, 1 is a methyl group. In another embodiment, R3 is methyl. In another specific embodiment, the compound of the formula (Γ) of the present invention is selected from the following 'or a pharmaceutically acceptable salt, prodrug and/or solvate thereof物-25- 201010988

本發明另提供用於投予至病患之醫藥組成物’該醫藥 組成物包含該新穎之本發明之式(Γ)化合物,或其醫藥上 可接受之鹽、溶劑合物或前藥,及至少一種醫藥上可接受 之載劑、佐劑及/或賦形劑。 在特定之實施態樣中,該式(I)化合物、其醫藥上可 -26- 201010988 接受之鹽、前藥及/或溶劑合物將以治療上之有效量與一 或多種醫藥上可接受之載劑、佐劑及/或賦形劑一起被調 製成適當之醫藥組成物,以用於投予以預防及/或治療急 性或慢性發炎疾病。 用語「載劑、佐劑及/或賦形劑」係關於與活性成分 一起投予之分子實體或物質。該醫藥載劑、佐劑或賦形劑 可爲無菌液體,諸如水及油(包括該些來自石油或動物、 Q 植物或合成來源者,諸如花生油、大豆油、礦物油、芝麻 油及該類似物)、賦形劑、崩解劑、濕潤劑或稀釋劑。適 當之醫藥載劑係描述於 E.W. Martin之“Remington’s Pharmaceutical Sciences” 〇 該醫藥組成物可藉由任何適當之投予方法投予,例如 經口、非經腸(例如皮下、腹腔內、靜脈內、肌肉內等)、 經直腸投予等,通常係經口投予因爲該被治療之疾病具慢 性特徵。 Φ 在特定實施態樣中,該醫藥組成物可呈經口投予之醫 藥形式,不論是固體或液體形式。經口投予醫藥形式之說 明性實例包括錠劑、膠囊、顆粒、溶液、懸浮液等,且可 包含習知之賦形劑諸如結合劑、稀釋劑、崩解劑、潤滑劑 、濕潤劑等,且可以習知方法製備。該醫藥組成物亦可呈 例如無菌凍乾產品、懸浮液或溶液之適當劑型以適於彼等 之非經腸投予;在此例中,該醫藥組成物將包括適當之賦 形劑,諸如緩衝劑、界面活性劑等。在任何例中,該賦形 劑將根據該選擇之投予醫藥形式加以選擇。有關投予藥物 -27- 201010988 之不同醫藥形式及彼等之製劑之回顧文獻可見於“ Tratado de Farmacia Galinica'' by C. Fauli i Trillo, 1 0th Edition, 1993, Luzan 5, S. A. de Ediciones。 就其在治療上之應用,式(I)化合物將較佳地呈醫藥 上可接受或實質上純的形式,也就是式(I)化合物具有排 除醫藥上可接受之賦形劑之醫藥上可接受之純度,且不包 含在正常劑量程度下被認爲具有毒性之物質。式(I)化合 物之純度較佳係高於5 0%,更佳爲高於70%,更佳係高於 90%。在較佳之實施態樣中,純度高於95%。 欲被投予之式(I)化合物之治療上之有效量一般來說 將視所欲治療之個體、該個體罹患之疾病之嚴重性、所選 擇之投予方法等其他因素而定。因此,本發明所提到之劑 量必須僅被視爲該領域之技藝人士之參考原則,技藝人士 必須根據前述變異參數調整劑量。然而,式(I)化合物可 一天投予一次或多次,例如一天1、2、3或4次,典型之 每曰總量從每天0.1至1000毫克/公斤體重,較佳爲每天 10毫克/公斤體重。 式(I)化合物、其醫藥上可接受之鹽、前藥及/或溶劑 合物以及包含彼等之醫藥組成物可與其它用於治療急性和 慢性發炎疾病之額外藥物一起使用。該額外藥物可形成該 相同醫藥組成物之部分,或者以分開之組成物之形式提供 以與該包含式(I)化合物或其醫藥上可接受之鹽、前藥或 溶劑合物之醫藥組成物同時或非同時投予。 在本發明之範圍內’用語「急性和慢性發炎疾病」係 -28- 201010988 關於起因於在異常共刺激係病理機制之狀況下活化及病理 涉及發炎/免疫細胞及發炎性細胞介素級聯之任何疾病、 異常或狀況。主要牽涉之細胞係發炎/免疫細胞諸如單核 細胞、巨噬細胞、APC、Τ細胞、B細胞、自然殺手(NK) 細胞、漿細胞、顆粒細胞及肥大細胞,或與欲治療之疾病 有關之上述細胞亞群之組合。 用語「細胞介素」係指會影響其他細胞功能之分泌蛋 〇 白質,特別是關於調控免疫系統之細胞之間或與發炎反應 有關之細胞之間之交互反應。該等細胞介素係TNF-α (腫 瘤壞死因子- a)、IFN-r(干擾素- r)、IL-8(介白素-8)趨 化激素及調節性細胞介素IL-10(介白素-10)。IL-10之產 製係由高量TNF-α引起,其藉由阻斷前發炎細胞介素之 轉錄作用促使形成對TNF- α產製之負向回饋。 在本發明之特定態樣中,該急性或慢性發炎疾病係選 自急性和慢性血清陽性或血清陰性寡關節炎和多發性關節 ® 炎、脊椎關節病、腎小球性腎炎、膠原病、腎小管間質性 腎炎、代謝性徵候群、動脈粥樣硬化、骨關節炎、氣喘、 慢性阻塞性肺病、間質性肺病、多發性硬化症、脫髓鞘病 、腦膜炎、腦炎、腦膜腦炎、發炎性神經根病變和週邊神 經病變、發炎性腸病、硬化、肝炎、心衰竭、缺血性疾病 、腎衰竭、發炎性膀胱炎、良性前列腺增生、前列腺炎、 心肌炎、心包膜炎、葡萄膜炎、異位性皮膚炎、濕疹 '蓴 麻疹、牛皮癬、玫瑰痤瘡、過敏性鼻炎、敗血症、敗血性 休克、多重器官衰竭、全身性自體免疫疾病(諸如全身性 -29- 201010988 紅斑性狼瘡)、血管炎、皮膚肌炎、澱粉樣變性症或類肉 瘤病、器官特異性自體免疫疾病(諸如重症肌無力)、甲狀 腺炎或胰島炎、器官移植、感染和腫瘤引起之發炎、 TNF_ α依賴性細胞退化、壞死、細胞凋亡、移植物抗宿 主病、惡病質或自體分泌和旁分泌病理性細胞生長。 在較佳之實施態樣中,該急性或慢性發炎疾病係血清 陽性或血清陰性慢性多發性關節炎,更佳的是類風濕性關 節炎。 本發明之另一態樣係一種用於治療急性或慢性發炎疾 病之方法,該方法包含投予治療上有效量之如上定義之式 (I)化合物或其醫藥組成物至需要該治療病患。 在本說明書之上下文中用語「治療」代表投予具有本 發明之化學式之化合物以預防、減輕或消除疾病或一或多 種與該疾病有關之徵狀。「治療」亦包括預防、減輕或消 除該疾病之生理後果。 在本發明之上下文中用語「減輕」被認爲是代表該接 受治療病患之狀況之任何改進,包括主觀性地(病患之感 覺或給人之感覺)及客觀性地(測量參數)。 本發明之另一態樣係指如上述定義之式(I)化合物於 顯影和藥理顯影技術中作爲顯影生物標記之用途;特別是 用以找出免疫病灶、標靶細胞及標的分子。在特定之實施 態樣中,本發明係指如上述定義之式(Γ)化合物於顯影和 藥理顯影技術中作爲顯影生物標記之用途;較佳的是用以 找出免疫病灶、標靶細胞及標的分子。 -30- 201010988 藥理顯影技術將快速成長之適當臨床前(分子、細胞 、器官及全動物追蹤及槪念機制驗證、療效檢驗等)及臨 床(人醫學)活體內顯影技術組合所得到生物標記之範圍擴 展至該些此處描述且作爲醫藥使用之化合物所產生之有用 訊息資料。 本發明利用實施例另外解說如下。此解釋不得被視爲 對本發明之範圍之限制,因爲其係於申請專利範圍中定義 ❹ 【實施方式】 實施例 合成 實施例1.合成(4-甲基苯硫基)-乙酸甲酯 COOH NaHCO; .XT' 將1.82克(10毫莫耳)之4-甲基苯硫基乙酸溶解於30 毫升之DMF中,接著加入1.34克(16毫莫耳)之NaHC03 :該攪拌混合物係經攪拌約15分鐘。接下來’添加1.9 毫升之ICH3同時在室溫中持續攪拌24小時。待此時間結 束後,將該混合物倒在水/冰上。由於沒有沉源發生’添 加醚以萃取。該有機相經水清洗及無水硫酸鈉乾燥後’該 有機相在旋轉蒸發器中濃縮以得到I.92克(9·7毫莫耳’ 201010988 97%產率)之無色油’經光譜資料證實具有該期待之結構。 此產物將不經純化地用於下一個合成方法之步驟,因爲其 在TLC(洗出液二氯甲烷/甲醇9/1)上得到單一斑點。 RMN ^(CDCh) : 7.2(s,4H) 3.7(s,3H) 3.5(s,2H) 2.4(s,3H CH3S) RMN 13C(CDC13) : 1 71.5 136.9 1 30.4 1 29.4 126.4 51.6 40.2 15.5The present invention further provides a pharmaceutical composition for administration to a patient, the pharmaceutical composition comprising the novel compound of the formula (Γ) of the present invention, or a pharmaceutically acceptable salt, solvate or prodrug thereof, and At least one pharmaceutically acceptable carrier, adjuvant and/or excipient. In a particular embodiment, the compound of formula (I), a salt, prodrug and/or solvate thereof, which is pharmaceutically acceptable from -26 to 201010988, will be in a therapeutically effective amount with one or more pharmaceutically acceptable The carrier, adjuvant and/or excipient are formulated together into a suitable pharmaceutical composition for administration in the prevention and/or treatment of an acute or chronic inflammatory condition. The phrase "carrier, adjuvant and/or excipient" is a molecular entity or substance that is administered with the active ingredient. The pharmaceutical carrier, adjuvant or excipient can be a sterile liquid such as water and oil (including those from petroleum or animal, Q plants or synthetic sources such as peanut oil, soybean oil, mineral oil, sesame oil and the like) ), excipients, disintegrants, wetting agents or diluents. Suitable pharmaceutical carriers are described in "Remington's Pharmaceutical Sciences" by EW Martin. The pharmaceutical composition can be administered by any suitable method of administration, for example, orally or parenterally (e.g., subcutaneously, intraperitoneally, intravenously, Intramuscular, etc., rectal administration, etc., usually by oral administration because of the chronic characteristics of the disease being treated. Φ In certain embodiments, the pharmaceutical composition can be in the form of a medical form for oral administration, whether in solid or liquid form. Illustrative examples of oral administration of pharmaceutical forms include tablets, capsules, granules, solutions, suspensions and the like, and may contain conventional excipients such as binding agents, diluents, disintegrating agents, lubricants, wetting agents, and the like, And can be prepared by a conventional method. The pharmaceutical composition may also be in a suitable dosage form, such as a sterile lyophilized product, suspension or solution, suitable for parenteral administration; in this case, the pharmaceutical composition will include suitable excipients, such as Buffer, surfactant, etc. In any case, the excipient will be selected according to the selected form of administration. A review of the different forms of medicines and their preparations for the administration of the drug -27- 201010988 can be found in "Trtado de Farmacia Galinica'' by C. Fauli i Trillo, 1 0th Edition, 1993, Luzan 5, SA de Ediciones. For therapeutic use, the compound of formula (I) will preferably be in a pharmaceutically acceptable or substantially pure form, i.e., the compound of formula (I) has pharmaceutically acceptable excluding pharmaceutically acceptable excipients. Purity, and does not contain substances considered to be toxic at normal dosage levels. The purity of the compound of formula (I) is preferably above 50%, more preferably above 70%, more preferably above 90%. In a preferred embodiment, the purity is greater than 95%. The therapeutically effective amount of the compound of formula (I) to be administered will generally depend on the individual being treated, the severity of the disease the individual is suffering from. Other factors depending on the method of administration chosen, etc. Therefore, the dosages referred to in the present invention must be considered only as a reference principle for those skilled in the art, and the skilled person must adjust the dosage according to the aforementioned variation parameters. I) compounds can One or more times a day, for example 1, 2, 3 or 4 times a day, typically from 0.1 to 1000 mg/kg body weight per day, preferably 10 mg/kg body weight per day. Compounds of formula (I) The pharmaceutically acceptable salts, prodrugs and/or solvates thereof and pharmaceutical compositions comprising the same can be used in combination with other additional drugs for the treatment of acute and chronic inflammatory diseases. The additional drugs can form the same medicine. A portion of the composition, or in the form of a separate composition, is administered simultaneously or non-simultaneously with the pharmaceutical composition comprising the compound of formula (I) or a pharmaceutically acceptable salt, prodrug or solvate thereof. In the context of the present invention, the term "acute and chronic inflammatory diseases" -28-201010988 relates to any activation and pathology resulting from the pathogenesis of abnormal co-stimulatory lines involving any combination of inflammatory/immune cells and inflammatory interleukin cascades Disease, abnormality or condition. The main cell line involved are inflammatory/immune cells such as monocytes, macrophages, APCs, sputum cells, B cells, natural killer (NK) cells, plasma cells, granulosa cells, and mast cells, or are associated with the disease to be treated. A combination of the above subpopulations of cells. The term "interleukin" refers to the secretion of egg white that affects the function of other cells, particularly the interaction between cells that regulate the immune system or between cells involved in the inflammatory response. These interleukins are TNF-α (tumor necrosis factor-a), IFN-r (interferon-r), IL-8 (interleukin-8) chemokine and regulatory interleukin IL-10 ( Interleukin-10). The production of IL-10 is caused by high levels of TNF-[alpha], which promotes the formation of negative feedback to TNF-[alpha] production by blocking the transcription of proinflammatory interleukins. In a particular aspect of the invention, the acute or chronic inflammatory disease is selected from the group consisting of acute and chronic seropositive or seronegative oligoarthritis and multiple joint inflammation, spondyloarthropathy, glomerulonephritis, collagen disease, kidney Tubulointerstitial nephritis, metabolic syndrome, atherosclerosis, osteoarthritis, asthma, chronic obstructive pulmonary disease, interstitial lung disease, multiple sclerosis, demyelinating disease, meningitis, encephalitis, meningeal brain Inflammation, inflammatory radiculopathy and peripheral neuropathy, inflammatory bowel disease, cirrhosis, hepatitis, heart failure, ischemic disease, renal failure, inflammatory cystitis, benign prostatic hyperplasia, prostatitis, myocarditis, pericarditis , uveitis, atopic dermatitis, eczema 'urticaria, psoriasis, rose acne, allergic rhinitis, sepsis, septic shock, multiple organ failure, systemic autoimmune diseases (such as systemic -29- 201010988 Lupus erythematosus), vasculitis, dermatomyositis, amyloidosis or sarcoma-like disease, organ-specific autoimmune diseases (such as myasthenia gravis), Prostatitis or insulitis, inflammation caused by the organ transplantation, infections and tumors, TNF_ α-dependent cellular degeneration, necrosis, apoptosis, graft versus host disease, cachexia or autologous autocrine and paracrine pathological cell growth. In a preferred embodiment, the acute or chronic inflammatory disease is seropositive or seronegative chronic polyarthritis, more preferably rheumatoid arthritis. Another aspect of the invention is a method for treating an acute or chronic inflammatory disease, comprising administering a therapeutically effective amount of a compound of formula (I), or a pharmaceutical composition thereof, as defined above, to a patient in need thereof. The term "treating" in the context of the present specification means administering a compound having the formula of the present invention to prevent, alleviate or eliminate the disease or one or more symptoms associated with the disease. "Treatment" also includes the prevention, alleviation or elimination of the physiological consequences of the disease. The term "alleviation" in the context of the present invention is considered to mean any improvement in the condition of the patient being treated, including subjectively (feeling or giving a feeling to the patient) and objectively (measuring parameters). Another aspect of the invention refers to the use of a compound of formula (I) as defined above as a developing biomarker in development and pharmacological development techniques; in particular, to identify immune foci, target cells and target molecules. In a particular embodiment, the invention refers to the use of a compound of formula (Γ) as defined above as a developing biomarker in development and pharmacological development techniques; preferably for identifying immune foci, target cells, and The target molecule. -30- 201010988 Pharmacological development technology will rapidly grow the appropriate preclinical (molecular, cell, organ and whole animal tracking and commemorative mechanism verification, efficacy test, etc.) and clinical (human medicine) in vivo development technology combination of biomarkers The scope extends to useful information generated by the compounds described herein and used as pharmaceuticals. The invention is further illustrated below using the embodiments. This explanation should not be construed as limiting the scope of the invention as it is defined in the scope of the claims. [Examples] Examples Synthesis Example 1. Synthesis of (4-methylphenylthio)-methyl acetate COOH NaHCO ;XT' 1.82 g (10 mmol) of 4-methylphenylthioacetic acid was dissolved in 30 ml of DMF, followed by 1.34 g (16 mmol) of NaHC03: the stirred mixture was stirred. 15 minutes. Next, 1.9 ml of ICH3 was added while stirring was continued for 24 hours at room temperature. After this time has elapsed, the mixture is poured onto water/ice. Since no sinking source occurred, 'addition of ether to extract. After the organic phase was washed with water and dried over anhydrous sodium sulfate, the organic phase was concentrated in a rotary evaporator to give I.92 g (9·7 mmoles of '201010988 97% yield) of colorless oil'. It has the structure of expectation. This product was used in the next step of the synthesis without purification because it gave a single spot on TLC (eluent dichloromethane/methanol 9/1). RMN ^(CDCh) : 7.2(s,4H) 3.7(s,3H) 3.5(s,2H) 2.4(s,3H CH3S) RMN 13C(CDC13) : 1 71.5 136.9 1 30.4 1 29.4 126.4 51.6 40.2 15.5

實施例2.合成(4-甲基磺醯基-苯基)-乙酸甲醒Example 2. Synthesis of (4-methylsulfonyl-phenyl)-acetic acid

將30_7克(50毫莫耳)之發氧方(〇x〇ne)溶液(於80 毫升之水中)逐滴添加至含有3.4克(17.3毫莫耳)實施例1 所得到之化合物之於100毫升甲醇之溶液中,保持該反應 在水/冰浴中進行。待添加完成後,持續攪拌5小時,允 許溫度上升至室溫。接著,部分溶劑在減壓下濃縮,該沉 澱固體以水重複過濾清洗。待乾燥後,此固體重3克。過 濾出來之水以二氯甲烷萃取,該有機相經水清洗並在無水 硫酸鈉上乾燥,在旋轉蒸發器中濃縮以得到一多於預期產 出4克(17毫莫耳,100%產率)之產物。此固體於57°C熔 化分解,其純度經HPLC測定爲99%。光譜資料證實該期 待之結構: RMN ^(CDCls) : 7.9(d,2H) 7.5(d,2H) 3.7(s,s 3 + 2H) -32- 201010988 3 .1 (s,3H) RMN 13C(CDC13) : 1 70.80 1 40.1 3 1 39 1 30.3 3 1 27.63 52.3 1 44.49 40.82 實施例3.合成溴基_(4·甲基磺醯基-苯基)-乙酸甲酯30_7 g (50 mmol) of an oxygenated solution (in 80 ml of water) was added dropwise to a compound containing 3.4 g (17.3 mmol) of the compound obtained in Example 1. The solution was maintained in a water/ice bath in milliliters of methanol. After the addition was completed, stirring was continued for 5 hours to allow the temperature to rise to room temperature. Next, a part of the solvent was concentrated under reduced pressure, and the precipitated solid was washed repeatedly with water. After drying, the solid weighed 3 grams. The filtered water was extracted with methylene chloride. The organic phase was washed with water and dried over anhydrous sodium sulfate and concentrated on a rotary evaporator to give a yield of more than 4 g (17 m. ) the product. This solid was melt-decomposed at 57 ° C and its purity was determined by HPLC to be 99%. Spectroscopic data confirmed the expected structure: RMN ^(CDCls) : 7.9(d,2H) 7.5(d,2H) 3.7(s,s 3 + 2H) -32- 201010988 3 .1 (s,3H) RMN 13C( CDC13) : 1 70.80 1 40.1 3 1 39 1 30.3 3 1 27.63 52.3 1 44.49 40.82 Example 3. Synthesis of bromo-(4.methylsulfonyl-phenyl)-methyl acetate

BrBr

❹ 〇 將20克(87.62毫莫耳)之該酯溶解於300毫升之四氯 化碳中.;按比例添加19克(1〇5毫莫耳)之溴琥珀醯亞 胺、2克(12.18毫莫耳)之偶氮雙異丁腈及0.1毫升之溴。 該反應混合物係於80°C加熱3小時,接著冷卻、過濾並 以二氯甲烷清洗。該濾液經水然後經濃鹽水清洗。令該溶 液經無水硫酸鈉.乾燥,在真空中濃縮以產生28克之油, 其利用快速層析法以庚烷/乙酸乙酯(1/1)作爲洗提液進行 春 純化。濃縮該更純之餾份產出19.63克(72.94 %產率)之溴 代衍生化合物。❹ 20 Dissolve 20 g (87.62 mmol) of the ester in 300 ml of carbon tetrachloride. Add 19 g (1 〇 5 mmol) of bromide amber imine, 2 g (12.18) Monomolar azobisisobutyronitrile and 0.1 ml of bromine. The reaction mixture was heated at 80 ° C for 3 hours, then cooled, filtered and washed with dichloromethane. The filtrate was washed with water and then with brine. The solution was dried over anhydrous sodium sulfate and concentrated in vacuo to give <RTI ID=0.0>>> Concentration of the purer fraction yielded 19.63 g (72.94% yield) of the brominated derivative compound.

熔點:8 1.8 - 8 3 . 3 °C NMR ^(CDCh) : 7.9(d,2H) 7.75(d,2H) 5.4(s,lH) 3.8(s,3H) 3.1(s,3H) NMR 13C(CDC13) : 168.0(C) 141.6(C) 141.1(C) 129.8(CH) 127.8(CH) 53.7(CH) 44.7(CH3)及 44.2(CH3) ppm -33- 201010988 實施例4.合成2-甲基-5-(4-甲基磺醯基-苯基)_4_經基 噻唑Melting point: 8 1.8 - 8 3 . 3 °C NMR ^(CDCh) : 7.9 (d, 2H) 7.75 (d, 2H) 5.4 (s, lH) 3.8 (s, 3H) 3.1 (s, 3H) NMR 13C ( CDC13): 168.0(C) 141.6(C) 141.1(C) 129.8(CH) 127.8(CH) 53.7(CH) 44.7(CH3) and 44.2(CH3) ppm -33- 201010988 Example 4. Synthesis of 2-methyl -5-(4-methylsulfonyl-phenyl)_4_yl-thiazole

將實施例3所獲得之16.7克(54.3毫莫耳)之該醋溶 解於4 00毫升之甲苯中,接著添加19毫升之耻啶及4.〇8 克(5 4.34毫莫耳)之硫代醯胺。該反應混合物於8(rc (水浴 溫度)加熱同時攪拌2小時。接著,令該混合物靜置冷卻 ,將該沉澱固體過濾並以水(2x50毫升)然後以醚(2x30毫 升)清洗。令該產物在真空中乾燥以產生7克(2 6毫莫耳, 47.86%產率)之膏狀固體,熔點2 1 6-226°C。 光譜資料證實該期待產物之結構。 RMN ^(d^MSO) <5 : 1 1 · 8 (s,1 Η) 7.8 (m,4H) 3 · 1 (s,3 Η) 2.6(s,3H) ppm RMN 13C(d6DMSO): 162.9(C) 159.4(C) 146.2(C) 143.0(C) 128_0(CH) 126.1 (CH) 104.4(C) 44_2(CH3) 1 9.9(CH3) ppm 實施例5.合成4-環戊氧基-5-(4_甲基磺酸基-苯基)_2 甲基-噻唑16.7 g (54.3 mmol) of the vinegar obtained in Example 3 was dissolved in 400 ml of toluene, followed by the addition of 19 ml of susceptine and 4. 〇 8 g (5 4.34 mmol) of thio Guanamine. The reaction mixture was stirred at 8 (rc (water bath temperature) while stirring for 2 hours. Then, the mixture was allowed to stand for cooling, and the precipitated solid was filtered and washed with water (2×50 ml) and then ether (2×30 ml). Drying in vacuo to give 7 g (2 6 mmol, 47.86% yield) of a cream solid, m.p. 2 166-226 ° C. Spectroscopic data confirm the structure of the desired product. RMN ^(d^MSO) <5 : 1 1 · 8 (s, 1 Η) 7.8 (m, 4H) 3 · 1 (s, 3 Η) 2.6 (s, 3H) ppm RMN 13C (d6 DMSO): 162.9 (C) 159.4 (C) 146.2(C) 143.0(C) 128_0(CH) 126.1 (CH) 104.4(C) 44_2(CH3) 1 9.9(CH3) ppm Example 5. Synthesis of 4-cyclopentyloxy-5-(4-methylsulfonate) Acid-phenyl)_2 methyl-thiazole

201010988 將實施例4所獲得之7.〇克(26毫莫耳)之羥基噻唑與 8.8克(63.67毫莫耳)之碳酸鉀溶解於2〇0毫升之dmF中 。逐滴添加14毫升(130毫莫耳)之溴環戊烷。該反應混合 物係於80°C加熱3小時’接著冷卻,將該混合物倒在冰/ 水上’並以乙酸乙酯萃取。該有機相經水清洗(3 χ1〇〇毫 升)’以無水硫酸鈉乾燥,並經過庚烷/醚(1/1)結晶濃縮以 產生12克之粗物質。利用快速層析法以庚烷/乙酸乙酯 (2/1)作爲洗提液純化該產物,以得到6.7克(19.85毫莫耳 ,76.39%產率)之化合物12。201010988 The 7. gram (26 millimolar) hydroxythiazole obtained in Example 4 and 8.8 grams (63.67 millimoles) of potassium carbonate were dissolved in 2 mM of dmF. 14 ml (130 mmol) of bromocyclopentane was added dropwise. The reaction mixture was heated at 80 ° C for 3 hours' followed by cooling, the mixture was poured onto ice / water and extracted with ethyl acetate. The organic phase was washed with water (3 χ 1 mL) and dried over anhydrous sodium sulfate and concentrated by heptane/ether (1/1) to yield 12 g of crude material. The product was purified by flash chromatography using heptane / ethyl acetate (2/1) as eluent to afford 6.7 g (19.85 m.

熔點:124.6-125.2 °C NMR 1H(CDC13) : 7.9(dd,4H) 5.4(m,lH) 3.0(s,3H) 2.6(s,3H) 1 . 7 - 2.0 (m, 8 H) ppm NMR 13C(CDC13) : 162.6(C) 159.5(C) 138.3(C) 137.1(C) 128.1(CH) 126.8(CH) 109.1(C) 83.7(CH) 45.0(CH3) 33.5(CH2) 24.1(CH2) 20.4(CH3) ppm 生物測定 材料及方法 1.個體 該試驗係以靜脈穿刺採集健康志願者與類風濕性關節 炎病患之肝素化週邊血液進行。 健康志願者係醫院業務內之例行控制下之捐血者。 被納入此試驗之類風濕性關節炎病患符合美國風濕病 學會之診斷標準,在納入訪視時呈現臨床活性疾病,且經 -35- 201010988 20毫克/週口服甲胺喋呤治療至少6個月。疾病活性之程 度定義如下:a) DAS2823.2及/或b) 6個或更多之腫脹 關節及6個或更多之疼痛關節。排除標準如下考量:a)在 納入訪視時患有主動傳染性疾病;b)在納入前患有腫瘤疾 病;c)患有其他在納入訪視前至少一年尙未完全緩解之系 統性或器官特異性自體免疫疾病;<0患有嚴重之腎、心或 肝狀況,該狀況與該些由主要疾病之類風濕性關節炎所引 起之變化程度無關;e)因爲與主要疾病無關之狀況導致整 體狀態嚴重惡化;f)在納入訪視前的一年內已接受皮質類 固醇、免疫抑制劑、細胞靜止劑或任何其他對免疫系統具 有已知活性之藥物之治療,但上述劑量之甲胺喋呤除外; g)納入訪視時懷孕或在產後期。 該試驗係經西班牙馬德里阿爾卡拉大學調查委員會核 准。 2.材料 -MultiGuard Barrier吸管尖(美國猶他州鹽湖城索羅 森(Sorenson)公司) -MicroAmp® PCR 0.2毫升反應試管(美國加州福斯特 市應用生物系統(Applied Biosystems)公司目錄編號 N8010540) -Fast Optical 96孔條碼反應板(應用生物系統公司) -丟棄式細胞刮勺(美國麻州劍橋市科斯達(Costar)公 司 3010) 201010988 -組織培養板,6孔平底低蒸發蓋(美國紐澤西州富 蘭克林湖市貝克頓迪更生實驗室用品公司(Becton Dickinson Labware) 353046 Falcon) -組織培養板,96孔平底低蒸發蓋(美國紐澤西州 074 1 7- 1 886富蘭克林湖市貝克頓迪更生實驗室用品公司 (Becton Dickinson Labware) 3 53 073 Falcon) -非滅菌板,96孔V型板(西班牙馬德里市索里亞格 ❹ 雷納公司(Soria Greiner)格雷納) -丟棄式巴斯德吸管(德國商標) -體積可調式微量移液器20、200、1000及5000微 升(法國吉爾森公司(Gilson)) -艾本德(Eppendorf)多道移液器4780(德國漢堡) -艾本德無菌吸管尖(德國漢堡) -Virgin丙烯無菌吸管尖(西班牙馬德里達斯實驗室 (Daslab)) ❹ -無菌塑膠試管5及10毫升(西班牙馬德里達斯實驗 室) -無菌離心管15及50毫升(美國富蘭克林湖BD Falcon) -細胞計數聚苯乙烯試管5毫升(美國富蘭克林湖Bd Falcon) -肝素採血管10毫升(比利時泰爾茂歐洲公司 (Terumo Europe) Venojet) •血獎分離管1〇毫升(英國普利矛斯BDVacutainer) -37- 201010988 -Pipetboy plus移液管吸注器(德國福羅實驗室(Flow Lab.)) -Millex-GS 22微米無菌過濾器(法國莫爾賽姆密里博 (Millipore)公司) -透明蓋玻片(德國赫希曼(Hirschman)公司) -Neubauer計算盤(德國薩里吉亞(Saaringia)公司) -Herasafe HS12垂直層流排氣櫃(德國賀利氏 (Heraeus)公司) ▲ -Heracell 150二氧化碳細胞培養箱(德國賀利氏公司 ) -貝克曼(Beckman)冷凍離心機 -Multifuge 3SR離心機(德國賀利氏公司) --7(TC冷凍櫃(西班牙特拉薩市塞拉塔(S electa)公司) -奧林巴斯(Olympus) CHS-2顯微鏡(日本東京奧林巴 斯公司) -流式細胞儀FACScalibur(美國山景城貝克頓迪更生 q 公司),分析軟體 BD Cell quest Pro v. 5.5.1 版 -BD FACS陣列生物分析儀 - 酵素免疫分析讀取儀(福羅實驗室 Titertek Multiscan Plus) -貝克曼放射活性貝他計數器 -AutoMACS分選儀(德國貝爾吉施-格拉德巴赫美天 旎生物技術公司(Myltenyi Biotec)) -Nanodrop分光光度計(美國麻州沃爾瑟姆賽默飛世 -38- 201010988 爾科技公司(Thermo Scientific)) -GeneAmp 9700 PCR系統(應用生物系統公司) -7900 HT快速即時Q-PCR系統(應用生物系統公司) 3.用於分離及識別細胞亞群之試劑 -簡單氯化物鹽水(SSF) Apiroserum(西班牙馬德里艾 比斯(Ibys)公司) 〇 - Lymphoprep淋巴細胞分離液(挪威奧斯陸奈格特 (Nyegaard)公司 Ficoll-Hypaque 梯度溶液) _台盼藍(德國布克斯福祿克(Flucke)公司) -PBS FACS FLOW溶液(貝克頓迪更生公司) -HEPES緩衝液(西格瑪(Sigma)公司和百瑞克 (Panreac)公司之試劑) -甲基-3H胸腺核苷(3[H]-T),特定活性60居禮/毫莫 耳(西班牙馬德里ITISA美國放射化學公司) ^ - CliniMACS CD 14試劑(美天旎生物技術公司) -用於免疫螢光及流式細胞試驗之單株抗體:表1 表1·用於該試驗中之單株抗體 CD* Ac Mo 細胞亞群 亞型 螢光色素 62L SK11 白血球L-選擇素 IgG2a 藻紅素(PE) 3 SK7 T淋巴細胞TCR/CD3抗原受器 IgGl FITC 14 M5E2 LBP-LPS之單核細胞TLR4共受器 IgG2a 藻紅素(PE) *於「第七屆人類白血球分化抗原硏討會」中定義 -39- 201010988 4.用於細胞培養中之試劑 -由RPMI-1640(比利時韋爾維耶坎布雷克斯生物科 技(Cambrex BioSciences)公司 B4800)構成之添加 1% L -麩 醯胺酸200毫莫耳和25毫莫耳Hepes(美國加州爾灣福羅 實驗室)之完全培養基 -胎牛血清(FCS)(美國紐約州格蘭德島吉勃可 (Gibco)公司) -培養基:使用添加10% FCS之完全培養基’加熱 至57 °C 45分鐘以去除所有血清中之補體 -植物血球凝集素M(PHA)(西班牙馬德里西格瑪公司 產品批號115K4132編號L-8902) -LPS(美國加州 92121聖地牙哥市英維偉基因 (InvivoGene)公司大腸桿菌0111:B4株) -抗生素混合物。胺苄青黴素鈉10毫克/毫升(西班 牙托利多比徹姆A(Beecham A.)公司商品名Britapen)、硫 酸建它黴素1.6毫克/毫升(西班牙馬德里伊斯蒂夫SA博 士(Dr. Esteve SA)塔馬諦(Tamadit)實驗室)及兩性黴素B 0.5微克/毫升(西班牙巴塞隆納埃斯普盧格斯(Esplugues) 施貴寶(Squibb)公司商品名Fungizona)之混合物被加入至 培養基中。 -抗CD3單株抗體(美國紐澤西州力登奧多醫藥公司 (Orthopharmaceutical Corporation)商品名 Orthoclone OKT3) -抗 CD28單株抗體(西班牙馬德里梅納里尼 201010988 (Menarini)公司商品名 Cion 15E8) 5.用於mRNA分離、逆轉錄及即時定量PCR(Q-PCR) 之試劑 -Illustra RNAspin mRNA純化套組(英國白金漢郡GE 健康照護公司目錄編號25-0 500-72) -高產量cDNA逆轉錄套組(應用生物系統目錄編號 ❹ 4368814) -TaqMan®基因表現通用混合液(應用生物系統目錄編 號 4369016) -用於人 TNF-α ID 編號 HS001 74128_ml [FAM]之 TaqMan®基因表現分析(應用生物系統目錄編號4331182) -人ACTB(冷-肌動蛋白)內源對照[VIC](應用生物系 統目錄編號43263 1 5E) 〇 6.取得生物樣本 a) 靜脈血液:單核細胞係自肘前靜脈穿刺採集之靜脈 血液獲得。採集50毫升之血液並儲存於肝素鋰試管 (Venojet),接著以食鹽水1/1(體積/體積)稀釋,所有皆在 無菌條件下進行。 b) 收集人細胞亞型。爲了分離PBMC,其餘之血液成 分係以Ficoll上之密度梯度分離。此方法係基於血液細胞 之密度上的差異。在含有經稀釋及肝素化之血液的50毫 升離心管中,小心地在其上加入 15毫升之 Ficoll- -41 - 201010988Melting point: 124.6-125.2 °C NMR 1H (CDC13): 7.9 (dd, 4H) 5.4 (m, lH) 3.0 (s, 3H) 2.6 (s, 3H) 1 . 7 - 2.0 (m, 8 H) ppm NMR 13C(CDC13) : 162.6(C) 159.5(C) 138.3(C) 137.1(C) 128.1(CH) 126.8(CH) 109.1(C) 83.7(CH) 45.0(CH3) 33.5(CH2) 24.1(CH2) 20.4 (CH3) ppm Bioassay Materials and Methods 1. Individuals The test was performed by venipuncture to collect heparinized peripheral blood from healthy volunteers and patients with rheumatoid arthritis. Healthy volunteers are blood donors under routine control within the hospital business. Patients with rheumatoid arthritis included in this trial meet the diagnostic criteria of the American College of Rheumatology and present clinically active disease at the time of enrollment, and at least 6 patients treated with oral methotrexate at -35-201010988 20 mg/week month. The extent of disease activity is defined as follows: a) DAS2823.2 and/or b) 6 or more swollen joints and 6 or more painful joints. Exclusion criteria were as follows: a) active infectious disease at the time of inclusion; b) tumor disease prior to enrollment; c) other systemic or non-complete remission at least one year prior to inclusion in the visit Organ-specific autoimmune disease; <0 suffering from severe kidney, heart or liver conditions, regardless of the degree of change caused by rheumatoid arthritis such as major diseases; e) because it is not associated with major diseases The condition causes a serious deterioration of the overall condition; f) has received treatment for corticosteroids, immunosuppressive agents, cytostatic agents or any other drug with known activity in the immune system within one year prior to the inclusion of the visit, but the above doses Except for methotrexate; g) pregnancy at the time of the visit or in the postpartum period. The trial was approved by the Commission of Inquiry of the University of Alcala, Madrid, Spain. 2. Material - MultiGuard Barrier Pipette Tip (Sorenson, Salt Lake City, Utah, USA) - MicroAmp® PCR 0.2 ml reaction tube (Applied Biosystems, Inc., Catalog No. N8010540, USA) - Fast Optical 96-well Barcode Reaction Plate (Applied Biosystems) - Discarded Cell Scrape (Costar, Inc., 3010, USA) 201010988 - Tissue Culture Plate, 6-well Flat-bottomed Low Evaporation Cap (New Jersey, USA) Becton Dickinson Labware 353046 Falcon) - Tissue culture plate, 96-well flat bottom low evaporation cap (Berton Dickens, Franklin Lake, New Jersey, USA 074 1 7- 1 886) Becton Dickinson Labware 3 53 073 Falcon) - Non-sterilized plate, 96-well V-plate (Soria Greiner Grena, Madrid, Spain) - Discarded Pasteur pipette (German trademark) - Volume adjustable micropipettes 20, 200, 1000 and 5000 microliters (Gilson, France) - Eppendor f) Multichannel pipette 4780 (Hamburg, Germany) - Ai Bend sterile straw tip (Hamburg, Germany) - Virgin propylene sterile pipette tip (Daslab, Madrid, Spain) ❹ - Aseptic plastic test tubes 5 and 10 ml ( Madrid Das Labs, Spain - Aseptic centrifuge tubes 15 and 50 ml (Franklin Lake BD Falcon, USA) - Cell count polystyrene tube 5 ml (B Frank Falcon, USA) - Heparin blood collection tube 10 ml (Belgium, Belgium) Company (Terumo Europe) Venojet) • Blood Prize Separation Tube 1 〇 ml (British Plymouth BDVacutainer) -37- 201010988 -Pipetboy plus Pipette Aspirator (Flow Lab.) -Millex -GS 22 micron sterile filter (Millipore, France) - Transparent coverslip (Hirschman, Germany) - Neubauer calculation disk (Saaringia, Germany) -Herasafe HS12 vertical laminar flow venting cabinet (Heraeus, Germany) ▲ -Heracell 150 carbon dioxide cell incubator (Herly, Germany) -Beckman refrigerated centrifuge - Multifuge 3 SR Centrifuge (Herly, Germany) - 7 (TC Freezer (S electa), Trassa, Spain - Olympus CHS-2 microscope (Olympus, Tokyo, Japan) Company) - Flow Cytometry FACScalibur (Bakerton Dickens, Inc., Mountain View, USA), Analysis Software BD Cell quest Pro v. Version 5.5.1 - BD FACS Array Bioanalyzer - Enzyme Immunoassay Reader (Fro Laboratory Titertek Multiscan Plus) - Beckman radioactive beta counter - AutoMACS sorter (Myltenyi Biotec, Bergisch Gladbach, Germany) - Nanodrop spectrophotometer (Wars, MA, USA) Thermo Scientific -38- 201010988 Thermo Scientific) -GeneAmp 9700 PCR System (Applied Biosystems) -7900 HT Fast Instant Q-PCR System (Applied Biosystems) 3. For separation and identification Reagents for Cell Subsets - Simple Chloride Brine (SSF) Apiroserum (Ibys, Madrid, USA) 〇- Lymphoprep Lymphocyte Separation Solution (Nyegaard, Norway) coll-Hypaque Gradient Solution) _Trypan Blue (Flucke, Germany) - PBS FACS FLOW Solution (Beckton Dickens) - HEPES Buffer (Sigma and Panreac) Company's reagent) -Methyl-3H thymidine (3[H]-T), specific activity 60 rit / millimole (ITISA American Radiochemical Company, Madrid, Spain) ^ - CliniMACS CD 14 Reagent (Mitsubishi Bio Technology company) - Monoclonal antibodies for immunofluorescence and flow cytometry: Table 1 Table 1 · Individual antibodies for this test CD* Ac Mo Cell subgroup subtypes Fluorescent pigment 62L SK11 White blood cells L- Selectin IgG2a phycoerythrin (PE) 3 SK7 T lymphocyte TCR/CD3 antigen receptor IgGl FITC 14 M5E2 LBP-LPS monocyte TLR4 co-receptor IgG2a phycoerythrin (PE) * in "The 7th human leukocyte Definition of Differentiation Antigens Conference-39-201010988 4. Reagents for cell culture - 1% added by RPMI-1640 (Cambrex BioSciences B4800, Belgium) L-glutamic acid 200 mM and 25 mM Hepes (US plus Complete medium of Irvine Froe Laboratories - Fetal Bovine Serum (FCS) (Gibco, Grand Island, NY) - Medium: heated to 57 °C using complete medium supplemented with 10% FCS 45 minutes to remove all of the serum complement-phytohemagglutinin M (PHA) (Spain Madrid Sigma product lot number 115K4132 number L-8902) -LPS (California 92921 San Diego, United Kingdom Invigo Gene (InvivoGene) large intestine Bacillus 0111: strain B4) - antibiotic mixture. Ampicillin sodium 10 mg/ml (Beitham A., trade name Britapen, Spain), Jiantianmycin sulfate 1.6 mg/ml (Dr. Esteve SA, Madrid, Spain) A mixture of Tamadit Laboratories and amphotericin B 0.5 μg/ml (Esplugues, Squibb, Fungizona, Spain) was added to the medium. - Anti-CD3 monoclonal antibody (Orthopharmaceutical Corporation, Orthoclone OKT3, USA) - Anti-CD28 monoclonal antibody (Minini, Spain 201010988 (Menarini), trade name Cion 15E8) 5. Reagents for mRNA isolation, reverse transcription, and real-time quantitative PCR (Q-PCR) - Illustra RNAspin mRNA Purification Kit (GE Health Care, UK, Buckinghamshire, Cat. No. 25-0 500-72) - High Yield cDNA Inverse Transcription kit (Applied Biosystems Catalog number ❹ 4368814) -TaqMan® Gene Expression Universal Mixture (Applied Biosystems Cat. No. 4369016) - TaqMan® Gene Performance Analysis for Human TNF-α ID No. HS001 74128_ml [FAM] Biosystems catalog number 4331182) - Human ACTB (cold-actin) endogenous control [VIC] (Applied Biosystems Catalog No. 43263 1 5E) 〇 6. Obtained biological samples a) Venous blood: Monocyte lineage from the elbow The venous blood collected by venipuncture is obtained. 50 ml of blood was collected and stored in a lithium heparin tube (Venojet), followed by dilution with saline 1/1 (vol/vol), all under sterile conditions. b) Collect human cell subtypes. To isolate the PBMC, the remaining blood components were separated by a density gradient on Ficoll. This method is based on differences in the density of blood cells. In a 50 ml centrifuge tube containing diluted and heparinized blood, carefully add 15 ml of Ficoll- -41 - 201010988

Hipaque(密度1.077克/毫升)。在離心(400xg) 45分鐘後 ,得到由二個介面分開之三層(紅血球、Ficoll及血漿): PBMC係包含在介於稀釋血漿及Ficoll之間的介面層。其 可利用巴斯德吸管吸出收集。以此方式收集到之細胞被重 懸於SSF並離心(300xg離心10分鐘);丟棄上清液(清洗 步驟),該細胞團塊被重懸於含有10% PBS之RPMI 164〇 培養基。 c)純化PBMC單核細胞:新鮮循環之單核細胞係以帶 有CD14特異性抗體之順磁微球利用自動磁性細胞分選儀 自PBMC分離。CD14係LPS受器複合物之次單位,該 LPS受器複合物位於習知循環人單核細胞之表面上,其表 現被發展性地控制限制於單核細胞而非其他PBMC亞群, 允許提供快速且有效之單一步驟陽性純化人單核細胞,利 用商業購得之AutoMACS分選儀(德國貝爾吉施-格拉德巴 赫美天旎生物技術公司D-5 1429)將它們從其餘之PBMC 亞型中分選出來。該程序係遵照製造商之詳細說明進行。 簡言之,將1〇8 PBMC重懸於5毫升無菌塑膠試管中之完 全培養基,於 4°C與帶有 CD14抗體之磁性微球 (CliniMACS CD14試劑,美天旎生物技術公司)培養20分 鐘後,加至Aut〇MACSTM細胞分選儀(美天旎生物技術公 司)並使用雙陽性選擇分選程式。單核細胞會先被留置於 AutoMACS的二個管柱中,其他細胞群被洗脫並與單核細 胞分離。單核細胞接著在釋放施用於管柱之順磁場後被洗 脫流出,並以5毫升無菌塑膠管收集。 -42- 201010988 7.計數及存活性試驗 所有細胞懸浮液之細胞濃度及存活性係利用0.1 %稀 釋之台盼藍及Neubauer計算盤顯微鏡計算決定。活細胞 之百分比係根據染料排出之能力而定。只有當細胞存活率 高於95%才繼續該試驗。 φ 8.血清細胞介素之ELISA定量 爲了定量細胞介素之血清濃度,將採集之血液儲存於 含有抗凝劑及不含抗凝劑之試管中(分別利用肘前靜脈穿 刺及動脈導管採血)。令血液在實驗室室溫中凝集,接著 離心以分離血清(60Oxg離心20分鐘)。收集上清液,過濾 ,等量分裝並儲存於-7 0 °C。利用表2描述之商業套組進 行不同細胞介素之濃度測定。 表2 .此試驗所使用之f g業E LI S A套組 細胞介素 定量上清液(皮克/毫升) PM(千道爾頓) 公司 TNF-a 3.83皮克/毫升 17 班德(Bender)Hipaque (density 1.077 g/ml). After 45 minutes of centrifugation (400 x g), three layers (red blood cells, Ficoll and plasma) separated by two interfaces were obtained: The PBMC system contained an interface layer between the diluted plasma and Ficoll. It can be collected by suction using a Pasteur pipette. The cells collected in this manner were resuspended in SSF and centrifuged (300 x g for 10 minutes); the supernatant was discarded (washing step), and the cell pellet was resuspended in RPMI 164 培养基 medium containing 10% PBS. c) Purification of PBMC monocytes: Freshly circulating mononuclear cell lines were isolated from PBMC using paramagnetic microspheres with CD14-specific antibodies using an automated magnetic cell sorter. The subunit of the CD14-based LPS receptor complex, which is located on the surface of conventional circulating human monocytes, whose performance is developmentally restricted to monocytes rather than other PBMC subpopulations, allowing for A fast and efficient single-step positive purification of human monocytes using a commercially available AutoMACS sorter (D-5 1429, Bergisch Gladbach Biotech, Germany) to remove them from the remaining PBMC subtypes Sorted out. This procedure is carried out in accordance with the manufacturer's detailed instructions. Briefly, 1〇8 PBMC was resuspended in complete medium in 5 ml sterile plastic tubes and incubated with magnetic microspheres with CD14 antibody (CliniMACS CD14 reagent, Meitian Biotech Co., Ltd.) for 20 minutes at 4 °C. After that, it was added to the Aut〇MACSTM Cell Sorter (Mitsubishi Biotech) and used a double positive selection sorting program. Monocytes are first placed in the two columns of AutoMACS, and other cell populations are eluted and separated from mononuclear cells. The monocytes were then eluted and released after release of the paramagnetic field applied to the column and collected in 5 ml sterile plastic tubes. -42- 201010988 7. Counting and viability tests The cell concentration and viability of all cell suspensions were determined using a 0.1% diluted trypan blue and Neubauer disk microscopy. The percentage of viable cells is based on the ability of the dye to be discharged. The trial was continued only when the cell viability was above 95%. φ 8. Serum Interferon ELISA Quantification In order to quantify the serum concentration of interleukin, the collected blood was stored in a test tube containing anticoagulant and no anticoagulant (using anterior venous puncture and arterial catheter to collect blood, respectively) . The blood was allowed to aggregate in the laboratory at room temperature, followed by centrifugation to separate the serum (60 o xg for 20 minutes). The supernatant was collected, filtered, and aliquoted and stored at -7 0 °C. The concentration of different interleukins was determined using the commercial kits described in Table 2. Table 2. F g industry E LI S A kit for this test. Interleukin quantitative supernatant (pick/ml) PM (kiloton) Company TNF-a 3.83 pg/ml 17 Bender

使該上清液解凍回溫至室溫,其與被固定在培養板底 部及經不同時間培養之個別抗細胞介素抗體反應。接著, 進行清洗’加入該相關公司所建議之物質以容許比色反應 發生,其與存在上清液中之細胞介素之量成比例。該結果 係由酵素免疫分析讀取儀 Multiscan plus(福羅實驗室 Titertek Multiscan Plus)測定。這些結果係與標準曲線比 -43- 201010988 較,該標準曲線得自公司所提供之已知濃度之各細胞介素 0 標準曲線之分析及後續之內插法係由軟體Delta Soft II 4.1版(生物金屬公司(Biometallics,Inc·))在蘋果麥金塔 電腦上進行。細胞介素濃度被轉換成莫耳數以計算各例之 分子比例及可溶性分子之數量。 9.免疫表現型測定 使用FITC或藻紅素標示(PE)之二種顏色之單株抗體 之直接免疫螢光技術。FITC係可被488奈米波長之雷射 光激發而發射525奈米波長之螢光物質,其可被商業流式 細胞儀偵測並輕易地與雷射光及PE發射波長區別。PE係 可被488奈米波長之雷射光激發及發射波長爲570奈米之 螢光物質,其可被流式細胞儀偵測並輕易地區分。利用表 1所述之抗體進行直接標示: PBMC標示:該方法利用96孔V型板進行標示。在 各孔中加入5x1 04細胞,之後以400xg離心5分鐘。經過 清洗後,細胞被重懸,並加入5微升之對應單株抗體。細 胞經再次重懸,令該孔板在4°C暗處培養20分鐘。接著 加入150微升之PBS,並清洗二次。最後,細胞於150微 升PBS中重懸並收集在細胞儀之試管中,加入PBS使終 體積成爲0.3毫升。 在進行計算時,使用配備氬雷射(調至488奈米波長) 之FACSCalibur流式細胞儀以誘發FITC及藻紅素螢光。 • 44- 201010988 該儀器除了螢光通道之外,亦提供有關大小(FSC)及細胞 複雜度(SSC)之資訊。 不經標示及其他以和該試驗所使用之單株抗體(18〇1_ FITC、PE、TC,IgG2a FITC、PE、TC)相等同型之單株抗 體不相關標示之試驗細胞被用來作爲陰性對照組。 使用 Flow Jo軟體 V8.4(美國奧勒岡州艾許蘭市 97520樹星公司(Tree Star,Inc.))進行流式細胞儀資料之 分析,以評估經AutoMACS分離之週邊血液單核細胞之純 度。 10. 細胞培養及功能試驗 一般培養條件:所有細胞培養均在垂直層流排氣櫃中 以無菌條件操作,使用無菌丟棄式材料或以環氧乙烷或滅 菌裝置滅菌。培養物被保存於5%二氧化碳及95%相對濕 度之3 7 °C培養箱中。 11. 3[H]-胸腺核苷增生試驗 在受到分裂原刺激時,淋巴細胞經歷胚細胞樣轉變及 細胞分化過程。用於定量細胞增生之方法係3[H]-胸腺核 苷倂入DNA重新合成之測定,在其結束及收集之前偵測 從細胞培養乾燥萃取物(該氚化基被加入之物)發射之貝他 放射線。在硏究增生之不同實驗中,經純化之細胞製劑在 含有不同濃度之各種分裂原的情形下,以5x 1 04細胞/孔 (2 00微升終體積)之濃度培養於96微量孔之平底板中4天 -45- 201010988 並重複三次。 對特定剌激之反應取決於密度及所硏究之細胞種類, 以及培養時間及該致分裂劑之濃度。 在細胞培養完成前的20至24個小時,在各孔中加入 1微居禮之1[H]-胸腺核苷;使用特定培養收集器經玻璃 過濾器抽取以收集培養物。 DNA合成係以每分鐘計數(cpm)表示。各試驗重複進 行三次,三次重複之平均値之變異性高於10%之該些資料 被拒絕,因爲可能表示技術誤差或培養物污染。培養係以 每孔固定量之細胞以及固定體積2 00微升進行。所有分裂 原及細胞介素首先以劑量反應曲線及時間反應進行測試。 1 表3及4顯示此實驗發展所使用之各試劑之目錄編號 及批號。樣本之取得係由BD FACS陣列生物分析儀(美國 加州聖荷西市貝克頓迪更生公司FACSArray)進行,結果 係於PC個人電腦以軟體FCAP Array(貝克頓迪更生公司) 完成分析。 簡言之,該實驗流程包括以清洗緩衝液預先清洗培養 板。在傾倒後,捕捉微球之混合物經振盪重懸。在該適當 201010988 之稀釋液中加入標準物及樣本。經永磁攪拌培養1小時後 ,加入PE偵測試劑並再次於室溫中培養2小時。經過清 洗後,在流式細胞儀進行取樣。 表3 .Kit CB A人可溶性蛋白質Multiplex Flex Set系統 試劑 目錄編號 批號 萬用緩衝液套組500測定 58265 95098 測定稀釋液 51-90003992 86192 捕捉微球稀釋液 51-90003804 90262 偵測試劑稀釋液 51-90003993 95811 清洗緩衝液 51-90003798 85964 儀器設定微球A1 51-90003855 87987 儀器設定微球A9 51-90003858 95933 儀器設定微球Π 51-90003851 89231 儀器設定微球F9 51-90003854 94229 PE儀器設定微球F1 51-9005038 94498 ΓΕ陽性對照偵測劑 51-9005065 92154 CBA Flex Set 套組 敏感性 IL-8(A9) 558277 85109 1.2皮克/毫升 IL-10(B7) 558274 97196 0.13皮克庵升 TNF-a(D9) 558273 85106 0.7皮克/¾升 IFN- r (E7) 558269 81562 1.8皮克/¾升 表4.Kit CBA Flex Set之設計中所包括之試劑批號 試劑 標準品 捕捉微球 螢光試劑(ΡΕ) IL-8(A9) 84854 87670 77948 IL-10(B7) 70333 96280 95934 TNF-a(D9) 65908 87667 77294 IFN- γ (E7) 80390 87664 77136The supernatant was thawed back to room temperature and reacted with individual anti-interleukin antibodies that were fixed at the bottom of the plate and cultured at different times. Next, the cleaning is carried out to the substances recommended by the relevant company to allow the colorimetric reaction to occur, which is proportional to the amount of interleukin present in the supernatant. The results were determined by the enzyme immunoassay reader Multiscan plus (Fuertek Multiscan Plus). These results are compared with the standard curve ratio -43-201010988, which is obtained from the analysis of the known concentration of the interleukin 0 standard curve provided by the company and the subsequent interpolation method by the software Delta Soft II version 4.1 ( Biometallics (Inc.) is carried out on an Apple Macintosh computer. The interleukin concentration was converted to a molar number to calculate the molecular ratio and the number of soluble molecules in each case. 9. Immunophenotypic assay Direct immunofluorescence technique using monoclonal antibodies of two colors of FITC or phycoerythrin (PE). The FITC system is excited by 488 nm wavelength laser light to emit 525 nm wavelength phosphors, which can be detected by commercial flow cytometry and easily distinguished from laser light and PE emission wavelengths. The PE system is excited by a 488 nm laser light and emits a phosphor of 570 nm, which can be detected by a flow cytometer and easily distinguished. Direct labeling using the antibodies described in Table 1: PBMC labeling: This method was labeled using a 96-well V-plate. 5x1 04 cells were added to each well, followed by centrifugation at 400 xg for 5 minutes. After washing, the cells were resuspended and 5 μl of the corresponding monoclonal antibody was added. The cells were resuspended again and the plates were incubated for 20 minutes in the dark at 4 °C. Then 150 μl of PBS was added and washed twice. Finally, the cells were resuspended in 150 μl of PBS and collected in a tube of a cytometer, and PBS was added to make a final volume of 0.3 ml. For calculations, a FACSCalibur flow cytometer equipped with an argon laser (adjusted to a wavelength of 488 nm) was used to induce FITC and phycoerythrin fluorescence. • 44- 201010988 This instrument provides information on size (FSC) and cell complexity (SSC) in addition to the fluorescent channel. Test cells unlabeled and other unlabeled monoclonal antibodies of the same type as the monoclonal antibodies (18〇1_ FITC, PE, TC, IgG2a FITC, PE, TC) used in the test were used as negative controls. group. Analysis of flow cytometry data using Flow Jo software V8.4 (Tree Star, Inc., Ashland, OH) to evaluate peripheral blood mononuclear cells isolated by AutoMACS purity. 10. Cell culture and functional assays General culture conditions: All cell cultures are run under sterile conditions in a vertical laminar exhaust cabinet, using sterile disposable materials or sterilized with ethylene oxide or a sterilization device. The culture was stored in a 3 7 ° C incubator with 5% carbon dioxide and 95% relative humidity. 11. 3[H]-thymidine proliferation assay When stimulated by mitogens, lymphocytes undergo blast-like transformation and cell differentiation. The method for quantifying cell proliferation is a measurement of 3[H]-thymidine nucleoside cleavage into DNA resynthesis, and detection of the dried extract from the cell culture (the sulfhydryl group is added) is detected before the end and collection thereof. Beta radiation. In the different experiments in which the proliferation was carried out, the purified cell preparation was cultured in a 96-well flat bottom plate at a concentration of 5×10 4 cells/well (200 μl final volume) in the presence of various concentrations of various mitogens. 4 days - 45 - 201010988 and repeated three times. The response to a particular stimulus depends on the density and the type of cell being studied, as well as the culture time and concentration of the mitogen. One [H]-thymidine was added to each well 20 to 24 hours before the completion of the cell culture; the culture was collected by a glass filter using a specific culture collector. DNA synthesis is expressed in counts per minute (cpm). Each test was repeated three times, and the data with an average 値 variability of more than 10% of the three replicates was rejected because it may indicate technical error or culture contamination. The culture was performed with a fixed amount of cells per well and a fixed volume of 200 microliters. All mitogens and interleukins were first tested in a dose response curve and time response. 1 Tables 3 and 4 show the catalog number and lot number of each reagent used in the development of this experiment. Samples were obtained from a BD FACS Array Bioanalyzer (FACSArray, Bakerton Dickens, San Jose, Calif.) and the results were analyzed on a PC PC with a software FCAP Array (Becktondi Rehabilitation). Briefly, the experimental procedure involves pre-cleaning the plate with a wash buffer. After pouring, the mixture of captured microspheres was resuspended by shaking. Standards and samples are added to the appropriate 201010988 dilution. After incubation for 1 hour with permanent magnet stirring, PE detection reagent was added and cultured again at room temperature for 2 hours. After washing, sampling was performed on a flow cytometer. Table 3. Kit CB A human soluble protein Multiplex Flex Set system reagent catalog number batch number universal buffer kit 500 determination 58265 95098 assay diluent 51-90003992 86192 capture microsphere dilution 51-90003804 90262 detection reagent dilution 51- 90003993 95811 Washing buffer 51-90003798 85964 Instrument setting microsphere A1 51-90003855 87987 Instrument setting microsphere A9 51-90003858 95933 Instrument setting microsphere Π 51-90003851 89231 Instrument setting microsphere F9 51-90003854 94229 PE instrument setting microsphere F1 51-9005038 94498 ΓΕ positive control detection agent 51-9005065 92154 CBA Flex Set kit sensitivity IL-8 (A9) 558277 85109 1.2 picogram / ml IL-10 (B7) 558274 97196 0.13 picogram soar TNF- a(D9) 558273 85106 0.7Pick/3⁄4L IFN-r (E7) 558269 81562 1.8Pick/3⁄4L Table 4. Kits included in the design of Kit CBA Flex Set Reagents Standards Reagents Capture Microsphere Fluorescent Reagents (ΡΕ) IL-8(A9) 84854 87670 77948 IL-10(B7) 70333 96280 95934 TNF-a(D9) 65908 87667 77294 IFN- γ (E7) 80390 87664 77136

13.純化 mRNA -47- 20101098813. Purification of mRNA -47- 201010988

mRNA分離係利用Illustrat RNAspin純化套組根據製 造商之詳細說明(目錄編號24-0500-72)進行。簡言之’藉 由離心收集5x 1 06細胞,添加緩衝液RA 1及劇烈振盪以溶 解細胞。在該溶解物中加入一份體積之70%乙醇,經劇烈 混合,裝塡至RNA結合管柱上。管柱以8000xg離心30 秒,以允許mRN A附著至該管柱基質。藉由添加膜脫鹽緩 衝液(MDB)及1 1 000 G離心1分鐘以移除管柱中之鹽。污 染DNA之移除係藉由在基質上添加95微升之DNA酶反 應混合物並於室溫中培養1 5分鐘。管柱基質經緩衝液 RA2清洗一次及緩衝液RA3清洗二次。最後,該純化之 RN A被洗脫於100微升之H20中,以Nano drop分光光度 計使用Nanodrop 1.2版本軟體定量後,立即儲存於-80°CThe mRNA isolation was performed using the Illustrat RNAspin purification kit according to the manufacturer's detailed instructions (catalog number 24-50-72). Briefly, 5x106 cells were collected by centrifugation, buffered with RA1 and vigorously shaken to dissolve the cells. A volume of 70% ethanol was added to the lysate, and the mixture was vigorously mixed and loaded onto an RNA-binding column. The column was centrifuged at 8000 xg for 30 seconds to allow mRN A to adhere to the column matrix. The salt in the column was removed by adding membrane desalting buffer (MDB) and centrifugation at 1 000 G for 1 minute. The removal of contaminating DNA was carried out by adding 95 μl of the DNase reaction mixture to the substrate and incubating for 15 minutes at room temperature. The column matrix was washed once with buffer RA2 and twice with buffer RA3. Finally, the purified RN A was eluted in 100 μl of H20 and quantified using a Nano drop spectrophotometer using Nanodrop 1.2 software and immediately stored at -80 °C.

14.自mRNA逆轉錄成cDNA mRNA逆轉錄成cDNA係利用高產量cDNA逆轉錄套 組(應用生物系統目錄編號43 688 1 4)根據製造商之詳細說 明進行。簡言之,在0.2毫升之PCR試管中混合0.5微克 之全mRNA與2微升之10倍RT緩衝液、0.8微升之25 倍dNTP混合物(100毫莫耳)、2微升之10倍RT隨機引 子、1微升之MultiScribe逆轉錄酶、4.2微升之不含核酸 酶之水,終體積成爲20微升。逆轉錄反應係於GenAmp 97〇0 Thermal Cycler(應用生物系統公司)中進行,使用下 列程式:251 10分鐘之步驟,37°C 120分鐘之步驟及最後 -48- 201010988 8 5 t: 5秒之步驟。 15.即時Q-PCR分析 人TNF-α mRNA表現專用之定量PCR係利用TNF TaqMan®基因表現分析(應用生物系統 ID 編號 118 00 1 74 1 28_1111)及丁&91^11©基因表現通用混合液(應用生 物系統目錄編號4369016)根據製造商之說明進行。人冷-φ 肌動蛋白內源對照(目錄編號4326315E)被用於相對定量 TNF- α基因轉錄活化時之TNF- α倍數調控。簡言之,反 應係於Fast Optical 96孔條碼反應板(應用生物系統目錄 編號43469 06)中進行,終體積爲20微升/孔。每孔中包括 之反應混合物爲5微升之cDNA、10微升之2倍Taqman® 基因表現通用混合液、2微升之20倍TNF- α (及/或石-肌 動蛋白)之TaqMan®基因表現分析與3微升之不含核酸酶 之水。該光學板係於7900 HT快速即時Q-PCR系統(應用 ❷ 生物系統目錄編號435 1 4〇5)中進行反應,使用下列熱循 環條件:第1步:保持50°C之溫度2分鐘;第2步:保 持95°C之溫度10分鐘;35個循環:溫度95°C 1〇秒鐘, 溫度60°C 1分鐘。所有樣本均作爲技術對照重複測定以避 免操作誤差。擴大反應之相對定量係利用包括於Q-PCR 儀器(應用生物系統)中之RQ Manager 1.2軟體分析。 統計分析 實驗測試之結果係以平均値及平均値之標準與估計誤 -49- 201010988 差(根據分佈種類)表示。 就統計分析而言,首先利用薛卜若-維克(shaPiro_ Whilk)之常態分佈檢定分析分佈之特性。曼-惠特尼 (Mann-Whitney)之非參數U檢驗被用來建立比較。在所有 例子中,低於5%顯著程度爲有意義的(Ρ <〇·〇5 )。 統計分析係利用軟體SPSS 1 1 ·0(伊利諾州60606芝加 哥南瓦克大道233號11樓SPSS公司總部)進行。 化合物4、5、6、9、10、12、15、16及17之免疫調 節效應試驗結果 對健康志願者及類風濕性關節炎病患之週邊血液單核 細胞(PBMC)產製TNF-α、IFN- r及其他細胞介素之免疫 調節效應。 實施例6.化合物5及12對TNF-α產製之影響 首先硏究化合物5及12在LPS存在或不存在之情況 下對於健康志願者之PBMC產製TNF- α之影響。來自健 康志願者之PBMC(5xl04細胞/孔,200微升)係於96孔平 底低蒸發蓋組織培養板(美國紐澤西州074 1 7- 1 886富蘭克 林湖市貝克頓迪更生實驗室用品公司353072 Falcon)平行 重覆二次培養,使用添加胎牛血清(原美國,吉勃可編號 261 40-079)及濃度1CT6莫耳之單獨溶劑或添加10_6莫耳、 1〇_7莫耳或1(Γ8莫耳之所示化合物之完全培養基(含麩醯 胺酸之RPΜΙ-1640,比利時Β4800韋爾維耶坎布雷克斯生 -50- 201010988 物科技公司編號BE-12-70F),在LPS(10微克/毫升;美國 加州92121聖地牙哥市英維偉基因大腸桿菌LPS 01ιι:Β4 株)存在及不存在之情況下培養24小時。培養上清液被冷 凍於·20°(:,以物種專一性ELISA定量TNF-α之濃度(用 於定量偵測腫瘤壞死因子-α之酵素連結免疫吸附試驗: A TNF- a ELISA BM S223/4TEUCE,力口州 94010 柏林格 姆班德醫學系統公司)。 表5.化合物5存在時健康志願者之PBMC產製TNF- α之情況。結果以該分泌細胞介 素之中位數±標準誤差表示。統計顯著結果係以粗體表示,它們相對應之ρ値包括在 _ 括弧中(各實驗條件相較於載劑之資料)。 健康志願者PBMC之TNF- α產製(皮克/¾升) n—D 綱 10·6莫耳 ΙΟ·7莫耳 ΙΟ"8 期 培養基 141.4±277.2 847.15±206.7 (0.893) 263.21140.72 (0.225) 736.2±265 (0.893) LPS (10毫克/¾升) 500.3±226.9 7421228.88 (0.893) 256.91146.49 (0.043) 679.11279.1 (0.500) 如表5所示’當化合物5以10·7莫耳之濃度存在於 培養物中時,顯著降低受到LPS剌激之健康志願者PBMC 之 TNF- α 產製(ρ<0.05)。 圖2顯示化合物12以10·6莫耳或1〇_7莫耳之濃度存 在於培養物中時,以統計顯著之方式降低受到LPS剌激 之PBMC培養物之上清液中經定量之TNF-α濃度(ρ<〇.〇5) 。另外,1〇_6莫耳之化合物12亦降低PBMC自發性之 TNF- α產製。 -51 - 201010988 表6.化合物12存在時健康志願者之PBMC產製TNF-α:之情況。結果以該分泌細胞 介素之中位數±標準誤差表示。統計顯著結果係以粗體表示,它們相對應之P値包括 在括弧中(各實驗條炉 湘較於載劑之雜)。 n=10 健康志願者PBMC之TNF- α產製 〔皮克/毫升) 載劑 ΚΓ6莫耳 1〇-7莫耳 培養基 1118.00±800.00 551.60±324.17 (0.009) 1121.00±100.48 (0.114) LPS(10微克/毫升) 3084.631450.00 1064.91+52.82 (0.005) 1938.00+487.74 (0.028) 化合物12之免疫調節效應被進一步以PBMC之細胞 @ 介素產製情況加以特徵化,該單核細胞受到LPS刺激或 以單株抗體抗CD3與抗CD2 8之組合活化淋巴細胞。此試 驗係於來自健康志願者及類風濕性關節炎病患之PBMC培 養物中,以滴定劑量之化合物12存在或不存在的情況下 進行。 來自13名健康志願者之PBMC(5xl04細胞/孔)係於添 加最高溶劑濃度(1(Τ6莫耳)、或10·6莫耳及1(Γ7莫耳之化 合物12之200微升之完全培養基中,在LPS(10微克/毫 © 升)存在或不存在的情況下培養(重複二次)24小時。培養 上清液係冷凍於-20 °C,且利用同時測定該指定細胞介素 濃度之特定試劑(CBA Flex Multiplex Set,BDTM流式細胞 小球微陣列術,CBA : IL-8、IL-10、TNF-α 及 IFN- γ, 加州9 2 1 2 1聖地牙哥貝克頓迪更生生物科技醫藥公司)以 B D F A C S陣列生物分析儀(加州9 2 1 2 1聖地牙哥B D生物 科技公司目錄編號340128)進行定量。 以濃度莫耳及10·7莫耳存在於培養基中之化合 -52- 201010988 物12以統計顯著方式(p<〇.〇5)減少受到LPS刺激之健康 志願者PBMC之TNF- α產製(圖3)。相反的,不論是經單 株抗體抗CD3及抗CD28刺激或PBMC自發性(無外源性 刺激存在)之TNF- α分泌均未受到化合物1 2之調節。 表7.化合物12存在時健康志願者產製TNF- (ϊ之情況。結果以該分泌細胞介素之平 均値±標準誤差表示。統計顯著結果係以粗體表示,它們相對應之卩値包括在括弧中( _各實驗條件相較於載劑之資料)。_ η=13 健康志願者PBMC之TNF- α產製(皮克/¾升) 載劑 HT6莫耳 1〇·7莫耳 1〇·8莫耳 培養基 163.84130.00 148.31150.00 (0.422) 197.81±40.00 (0.249) 168.49±16.00 (0.65) LPS (10微克/¾升) 923.93+160.00 602.12±100.00 (0.002) 746.841125.00 (0.046) 738.81±100.00 (0.055) 抗CD3 (12.5奈克庵升)+ 抗 CD28(l/3xl05) 435.81±60.00 334.21163.00 (0.116) 404.11土97.00 (0.753) 355.62±65.00 (0.422) 在另一實驗中,來自7名類風濕性關節炎病患之 Φ PBMC(每孔5χ104細胞)係於添加最高溶劑濃度(1〇·6莫耳) 或10_6莫耳、1(Γ7莫耳及1(Γ8莫耳之化合物12之200微 升之完全培養基中,在LPS(10微克/毫升)或抗CD3(12.5 奈克/毫升)(美國紐澤西州力登奧多醫藥公司 Orthoclone OKT3) +抗CD28(l/3xl05)(西班牙馬德里梅納里尼公司 Cion 15E8)存在或不存在的情況下培養(重複二次)24小時 。培養上清液係冷凍於-20 °C,且利用專門測定該細胞介 素濃度之CBA Flex Set(貝克頓迪更生)以BD FACS陣列 生物分析儀根據製造商說明加以定量TNF- α之濃度。 -53- 201010988 在經LPS刺激之類風濕性關節炎病患之PBMC中, TNF-α之產製受到濃度1(Γ6莫耳之化合物12之顯著抑制 (ρ<0.05)(圖4)。培養基中所含有之化合物12並不抑制類 風濕性關節炎病患之PBMC之自發性TNF- α產製,亦不 抑制經單株抗體抗CD3及抗CD28刺激後之TNF- α產製 在括弧中(各實驗條件相較於載劑之資料)。 n=7 類風濕性關 節炎病患PBMC之TNF- α產製(皮克/毫升) 載劑 10-6莫耳 1〇—7莫耳 10-8莫耳 培養基 100.64140.00 177.32±95.00 (0.735) 191.33180.00 (0.735) 131.95±85.00 (0.735) LPS (10微克庵升) 700.39±180.00 383.75±75.00 (0.05) 626.571150.00 (0.237) 683.401120.00 (0.499) 抗CD3 (12_5奈克/¾升)+ 抗 CD28(l/3xl05) 648.72±90.00 532.07±80.00 (0.31) 568.47±97.00 (0.612) 695.81179.00 (0.398) 表8.化合物12存在時類風濕性關節炎病患產製TNF- α之情況。結果以該分泌細胞 介素之平均値±標準誤差表示。統計顯著結果係以粗體表示,它們相對應之Ρ値包括 ©14. Reverse transcription of mRNA into cDNA mRNA is reverse transcribed into a cDNA line using a high yield cDNA reverse transcription kit (Applied Biosystems Catalog No. 43 688 1 4) according to the manufacturer's detailed instructions. Briefly, mix 0.5 μg of total mRNA with 2 μl of 10x RT buffer, 0.8 μl of 25 fold dNTP mix (100 millimolar), 2 microliters of 10 times RT in a 0.2 ml PCR tube. A random primer, 1 μl of MultiScribe reverse transcriptase, 4.2 μl of nuclease-free water, and a final volume of 20 μl. The reverse transcription reaction was performed in a GenAmp 97〇0 Thermal Cycler using the following program: 251 10 minute step, 37 ° C 120 minute step and finally -48- 201010988 8 5 t: 5 seconds step. 15. Real-time Q-PCR analysis of human TNF-α mRNA expression-specific quantitative PCR system using TNF TaqMan® gene expression analysis (Applied Biosystem ID No. 118 00 1 74 1 28_1111) and Ding & 91^11© gene expression universal mixture The liquid (Applied Biosystems Catalog No. 4369016) was carried out according to the manufacturer's instructions. The human cold-φ actin endogenous control (catalog number 4326536E) was used to quantify the TNF-[alpha] fold regulation of transcriptional activation of the TNF-[alpha] gene. Briefly, the reaction was performed on a Fast Optical 96-well barcode reaction plate (Applied Biosystems Catalog No. 43469 06) with a final volume of 20 μl/well. The reaction mixture included in each well is 5 μl of cDNA, 10 μl of 2-fold Taqman® Gene Universal Mix, 2 μl of 20-fold TNF-α (and/or stone-actin) TaqMan® Gene performance analysis with 3 μl of nuclease-free water. The optical plate was reacted in a 7900 HT Rapid Instant Q-PCR System (Applied Biosystems Catalog No. 435 1 4〇5) using the following thermal cycling conditions: Step 1: Maintain a temperature of 50 °C for 2 minutes; 2 steps: maintain a temperature of 95 ° C for 10 minutes; 35 cycles: temperature 95 ° C 1 〇 seconds, temperature 60 ° C 1 minute. All samples were repeated as technical controls to avoid operational errors. The relative quantification of the expanded response was performed using RQ Manager 1.2 software analysis included in the Q-PCR instrument (Applied Biosystems). Statistical Analysis The results of the experimental tests are expressed as the difference between the average 値 and the mean 与 and the estimated error -49- 201010988 (according to the type of distribution). For statistical analysis, the characteristics of the distribution are first analyzed using the normal distribution of shaPiro_ Whilk. Mann-Whitney's nonparametric U test was used to establish comparisons. In all cases, a significance of less than 5% is significant (Ρ <〇·〇5). Statistical analysis was performed using software SPSS 1 1 ·0 (SPSS Corporate Headquarters, 11th Floor, 233, Southwark Avenue, Chicago, Illinois 60606). Results of immunomodulatory effects of compounds 4, 5, 6, 9, 10, 12, 15, 16 and 17 Production of TNF-α in peripheral blood mononuclear cells (PBMC) in healthy volunteers and patients with rheumatoid arthritis , immunomodulatory effects of IFN-r and other interleukins. Example 6. Effect of Compounds 5 and 12 on TNF-α Production First, the effects of Compounds 5 and 12 on the production of TNF-α by PBMCs in healthy volunteers in the presence or absence of LPS were investigated. PBMCs from healthy volunteers (5xl04 cells/well, 200 μl) were plated in 96-well flat-bottomed low-evaporation cap tissue culture plates (Berton Dickens Laboratory Supplies, Franklin Lake, New Jersey, USA 074 1 7- 1 886) 353072 Falcon) Parallel repeated secondary culture, using added fetal bovine serum (formerly US, Ji Boco No. 261 40-079) and a single solvent of 1 CT6 molar or adding 10_6 mol, 1〇_7 mol or 1 (Complete medium for the compound shown in Γ8 Moer (RPΜΙ-1640 containing glutamic acid, Belgian Β4800 Vervier Can Blakesson-50-201010988 Material Technology Company No. BE-12-70F), at LPS (10 μg/ml; cultured in E. coli LPS 01ιι: Β4 strain of Invitrogen, 92021, San Diego, CA, USA), cultured for 24 hours in the presence and absence of culture. The culture supernatant was frozen at ·20° (:, Species-specific ELISA for quantification of TNF-α concentration (enzyme-linked immunosorbent assay for quantitative detection of tumor necrosis factor-α: A TNF- a ELISA BM S223/4TEUCE, Likouzhou 94010 Berlin Gemband Medical Systems Table 5. Health in the presence of Compound 5 The results of PBMC production of TNF-α. The results are expressed as the median ± standard error of the secreted interleukin. Statistically significant results are shown in bold, and their corresponding ρ 値 is included in _ brackets (each experiment Conditions compared to the carrier data. TNF-α production of healthy volunteers PBMC (Pick/3⁄4 liters) n-D 纲10·6 莫ΙΟ·7莫耳ΙΟ"8-stage medium 141.4±277.2 847.15 ±206.7 (0.893) 263.21140.72 (0.225) 736.2±265 (0.893) LPS (10 mg/3⁄4 liter) 500.3±226.9 7421228.88 (0.893) 256.91146.49 (0.043) 679.11279.1 (0.500) as shown in Table 5 When Compound 5 was present in culture at a concentration of 10.7 moles, TNF-α production (p < 0.05) of PBMCs stimulated by LPS was significantly reduced. Figure 2 shows Compound 12 at 10.6. The concentration of TNF-α in the supernatant of PBMC cultures stimulated by LPS is reduced in a statistically significant manner when molar or 1 〇 7 molar concentrations are present in the culture (ρ<〇.〇) 5) In addition, 1〇_6 mol of compound 12 also reduces the spontaneous production of TNF-α in PBMC. -51 - 201010988 Table 6. Compounds In the presence of substance 12, PBMC of healthy volunteers produced TNF-α:. Results are expressed as the median ± standard error of the secreted interleukin. Statistically significant results are shown in bold, and their corresponding P値 is included in parentheses (the experimental strips are more complex than the carrier). n=10 TNF-α production of healthy volunteers PBMC [pick/ml) Carrier ΚΓ6 mol 1〇-7 molar medium 1118.00±800.00 551.60±324.17 (0.009) 1121.00±100.48 (0.114) LPS (10 μg /ml) 3084.631450.00 1064.91+52.82 (0.005) 1938.00+487.74 (0.028) The immunomodulatory effects of Compound 12 are further characterized by the cellular production of PBMC, which is stimulated by LPS or The strain antibody anti-CD3 binds to anti-CD2 8 to activate lymphocytes. This test was performed in PBMC cultures from healthy volunteers and patients with rheumatoid arthritis in the presence or absence of a titration dose of Compound 12. PBMC (5xl04 cells/well) from 13 healthy volunteers were added to the highest solvent concentration (1 (Τ6 mol), or 10.6 mol and 1 (Γ7 mol of compound 12 of 200 μl of complete medium) In the presence or absence of LPS (10 μg/ml liter), culture (repeated twice) for 24 hours. The culture supernatant was frozen at -20 °C, and the specified interleukin concentration was simultaneously measured. Specific reagents (CBA Flex Multiplex Set, BDTM Flow Cellular Microspheres, CBA: IL-8, IL-10, TNF-α, and IFN-γ, California 9 2 1 2 1 San Diego Beckton Dickens Biotech Pharmaceuticals Inc. was quantified using a BDFACS Array Bioanalyzer (California 9 2 1 2 San Diego BD Biotech Company Catalog No. 340128). The concentration of Mox and 10.7 Mo in the medium was combined -52 - 201010988 Event 12 reduced the TNF-α production of PBMC in healthy volunteers stimulated by LPS in a statistically significant manner (p<〇.〇5). Conversely, whether it was monoclonal antibody anti-CD3 and anti-CD28 Stimulation or TNF-α secretion of PBMC spontaneously (without exogenous stimuli) Subject to modulation by Compound 12. Table 7. Production of TNF- in healthy volunteers in the presence of Compound 12. (Results are expressed as mean 値± standard error of the secreted interleukin. Statistically significant results are in bold, Their corresponding 卩値 are included in parentheses ( _ each experimental condition compared to the carrier data). _ η = 13 TNF-α production of healthy volunteer PBMC (Pick / 3⁄4 liter) Carrier HT6 Mo 1〇·7 Mo Er 1〇·8 Mourn Medium 163.84130.00 148.31150.00 (0.422) 197.81±40.00 (0.249) 168.49±16.00 (0.65) LPS (10 μg/3⁄4 liter) 923.93+160.00 602.12±100.00 (0.002 746.841125.00 (0.046) 738.81±100.00 (0.055) Anti-CD3 (12.5 ng liters) + anti-CD28 (l/3xl05) 435.81±60.00 334.21163.00 (0.116) 404.11 soil 97.00 (0.753) 355.62±65.00 (0.422 In another experiment, Φ PBMC (5χ104 cells per well) from 7 patients with rheumatoid arthritis were added at the highest solvent concentration (1〇·6 mol) or 10_6 mol, 1 (Γ7 mol) And 1 (Γ8 mol of compound 12 in 200 μl of complete medium, in LPS (10 μg/ml) or anti-CD3 (12.5 Ng/升) (Orthoclone OKT3, Riden Otto Pharmaceuticals, New Jersey, USA) + Anti-CD28 (l/3xl05) (Cion 15E8, Menarini, Spain) culture in the presence or absence of (repeated twice) 24 hour. The culture supernatant was frozen at -20 °C, and the concentration of TNF-α was quantified by a BD FACS array bioanalyzer according to the manufacturer's instructions using a CBA Flex Set specifically measuring the concentration of the interleukin. -53- 201010988 In PBMC of patients with rheumatoid arthritis stimulated by LPS, the production of TNF-α was significantly inhibited by concentration 1 (Γ6 mol of compound 12 (ρ < 0.05) (Fig. 4). Compound 12 contained in the compound does not inhibit the spontaneous TNF-α production of PBMC in rheumatoid arthritis patients, nor does it inhibit the production of TNF-α after stimulation with monoclonal antibodies against CD3 and anti-CD28 in brackets. (The experimental conditions are compared with the carrier data.) n=7 TNF-α production of PBMC in patients with rheumatoid arthritis (pick/ml) Carrier 10-6 mol 1〇-7 mol 10 -8 molar medium 100.64140.00 177.32±95.00 (0.735) 191.33180.00 (0.735) 131.95±85.00 (0.735) LPS (10 micrograms soar) 700.39±180.00 383.75±75.00 (0.05) 626.571150.00 (0.237) 683.401120. 00 (0.499) Anti-CD3 (12_5 Nike/3⁄4 L) + Anti-CD28 (l/3xl05) 648.72±90.00 532.07±80.00 (0.31) 568.47±97.00 (0.612) 695.81179.00 (0.398) Table 8. In the presence of compound 12 TNF-α was produced in patients with rheumatoid arthritis. The results were expressed as the mean 値± standard error of the secreted interleukin. Statistically significant results were obtained. Bold, they correspond to the Ρ Zhi include ©

此外,接著探討化合物12對於來自9名健康志願者 之PBMC所純化分離出之單核細胞在LPS存在或不存在 的情況下產製 TNF- α之影響。來自各健康志願者之 PBMC係經分離,利用自動磁活化細胞分選術(MACS)純化 他們的單核細胞。將1〇8 PBMC重懸於5毫升無菌塑膠試 管中之完全培養基,於4°C與帶有CD 14抗體之磁性微球 (CliniMACS CD14試劑,德國貝爾吉施-格拉德巴赫美天 旎生物技術公司 D-5 1 429)) —起培養,加至 Aut〇MACSTW -54- 201010988 細胞分選儀(美天旎生物技術公司)以純化他們的CD 1 4 + PBMC亞群(習知之單核細胞)。該經分離之單核細胞在經 FITC標示之CD3及經PE標示之CD14特異性抗體染色後 於5毫升之細胞計數聚苯乙烯試管中利用直接免疫螢光及 多色流式細胞儀來測定它們的純度。定量該指定之T細胞 (CD3)及單核細胞(CD14)表面受器之表面分佈係於 FACScalibur分析儀(貝克頓迪更生)中進行。結果之分析 © 係利用Flow Jo流式細胞儀軟體(美國奧勒岡州樹星公司) 進行。該經分離之PBMC CD3—CD14 +單核細胞之純度係例 行性地> 9 8 %之活細胞,單核細胞根據它們特別的正向散 射光(FSC)及側向散射光(SCC)特性被圈選,以允許定義明 顯之單核細胞群集和其餘之淋巴細胞(圖5)。單核細胞 (1·25χ105細胞/孔,5毫升)係培養於6孔平底低蒸發蓋之 組織培養板(353046 Falcon,貝克頓迪更生),使用添加胎 牛血清及濃度1(Γ6莫耳之單獨溶劑或添加1〇_6莫耳之化 〇 合物12之完全培養基,並在10克/毫升之LPS存在或不 存在之情況下培養經過動力學實驗所示之時間期。不含細 胞之上清液及細胞被同時儲存於如圖6及7所示之動力學 實驗中,以利用細胞中之mRNA之量及上清液中之蛋白質 之量來測定TNF- α基因產物之表現。如上所述,上清液 被冷凍儲存於-20°C,該分泌之TNF- α蛋白質之濃度係利 用BD TNF-α之CBA試劑(貝克頓迪更生生物科學醫藥公 司)以BD FACS陣列生物分析儀(貝克頓迪更生)根據製造 商說明而加以測量。該黏附之單核細胞係利用丟棄式細胞 -55- 201010988 刮勺(美國麻州021 39劍橋市科斯達公司3010)從培養板上 收集,並立即進行細胞mRNA分離、mRNA定量及逆轉錄 反應與定量即時聚合酶連鎖反應(Q-PCR)試驗,以於7900 HT快速即時Q-PCR系統(美國加州福斯特市應用生物系 統)中測定TNF-α基因之mRNA表現量。 圖5顯示高度富集之單核細胞(98.3 0±0.17%)被用於 此包含9名健康志願者之試驗中,以分析此PBMC亞群中 TNF- α之mRNA表現及蛋白質分泌之動力學。如預期地 ,圖6顯示TNF-α之mRNA表現在LPS刺激之試管內人 單核細胞中被強烈誘導(4 8.35+17.46倍,ρ<0· 00 1),在基 準期之活體外狀況中(〇小時)實質上不表現,非常早期地 活化基因表現之動力學特性在2.5小時達到高峰,而短暫 轉錄產生快速下降因此TNF- α之mRNA在24小時下降至 接近基準期之水準(圖6方塊)。在缺乏LPS之刺激時, TNF- a之基因表現不被活化,且TNF - α特定mRNA在此 試管內系統維持其基準期之活體外水準(圖6菱形)。値得 注意的是,化合物12顯著降低該經LPS刺激之TNF-a mRNA高峰誘導,其中純化之單核細胞係來自健康志願者 ,並於7900 HT快速即時Q-PCR系統中以人TNF-cr專用 之TaqMan®基因表現分析測量(33.72±15.88%,ρ<〇·〇1)(圖 6三角形)。另外,TNF-α基因表現mRNA之量在LPS刺 激該純化之單核細胞的5及8小時後亦顯著降低(分別爲 27.14±9·07%, p<0.01 ;及 36·45±11.58%, ρ<〇.〇1)。我們 得到的結論是,化合物12在來自健康志願者之PBMC純 201010988 化單核細胞中顯著降低該LPS刺激之TNF- ct mRNA誘導 ,且在所有該試驗之期間仍發生誘導之變化而不影響 TNF- α基因產物表現之向上及向下調節之高峰動力學。 和未經LPS刺激之人單核細胞中非常低之TNF-α基因表 現與mRNA誘導之快速動力學所觀察到一致的是,TNF-α蛋白質之分泌啓動快速,但伴隨著從mRNA轉譯成蛋白 質、其處理、分泌及釋放至培養基之完整生物過程所需之 @ 延遲時間,使得預期之高峰產量在LPS刺激該缺乏化合 物12之對照培養基中之P BMC純化單核細胞之8小時後 出現(圖7方形)。在只含有溶劑載劑但不含LPS及化合物 12之單核細胞培養不誘發TNF- α之分泌(圖7菱形)。値 得注意的是,化合物12顯著減少該經LPS刺激之TNF- α 分泌蛋白質之高峰生產(8小時),其中純化之單核細胞係 來自健康志願者,並於FACS陣列生物生物分析儀中以人 TNF-α專用之 CBA試劑套組測量(31.52±14.38%, ^ ρ<〇·〇1)(圖7三角形)。如同上面顯示在TNF-amRNA誘 導之動力學所發生的情況,TNF-a分泌蛋白質之量從0 小時刺激該培養之後的產製動力學監測之時間點均較低。 事實上,統計分析顯示,化合物1 2在2.5小時、5小時及 24小時亦顯著抑制由LP S刺激之來自健康志願者之純化 PBMC單核細胞之 TNF- α分泌蛋白質(分別爲 45.56 土 16.25% , ρ<0.01 ; 41.33 ± 13.37〇/〇 , ρ<0.01 ;及 51.17± 15.62%)。由於該伴隨之資料顯示,化合物12顯著地抑制 來自9名健康捐贈者之PBMC純化單核細胞中TNF- α基 -57- 201010988 因產物表現之mRNA及蛋白質之量,我們接下來要硏究的 是,由化合物12所促使之TNF- a mRNA誘導之抑制量與 TNF-α分泌蛋白質之減少量之間是否具有統計顯著相關 性,以它們個別之表現高峰進行回歸分析測定。値得注意 的是,我們發現化合物12對於該純化單核細胞之TNF-cr 基因產物表現之mRNA與分泌蛋白質之間的免疫調節效應 具有顯著相關性(r2 = 0.7952)。綜合上述結果,這顯示化合 物12抑制LPS刺激之純化單核細胞之TNF- a mRNA誘導 量是一個可用來說明化合物12對TNF- α分泌抑制之免疫 調節效果之實質部份之機制。 實施例 7.化合物 4、5、6、9、10、12、15、16 及 17 對IFN- r產製之影響 使來自健康志願者之PBMC(每孔5X104細胞)於僅添 加最高溶劑濃度(1〇_δ莫耳),或添加10·6莫耳、10_7莫耳 或1(Γ8莫耳之測試化合物之200微升完全培養基中,在 LPS(10微克/毫升)或抗CD3(12.5奈克/毫升)+抗CD28(l/3 xlO5)存在及不存在之情況下培養(重覆二次)24小時。培 養上清液被冷凍於-20°C,利用專門用於測定IFN- r濃度 之CBA Flex Set(貝克頓迪更生)根據製造商之說明以Bd FACS陣列生物分析儀定量rFN_ 7之濃度。 201010988 表9.化合物16存在時健康志願者之PBMC產製IFN-之情況。結果以該分泌細胞 介素之中位數±標準誤差表示。統計顯著結果係以粗體表示,它們相對應之p値包括 在括弧中(各實驗條件相較於載劑之資料)〇 n=5 健康志願者PBMC之IFN- «τ產製(皮克/毫升) 載劑 ίο·6莫耳 1〇-7莫耳 1〇·8莫耳 培養基 94.5124.5 45.46±6.89 (0.043) 50.215.2 (0.043) 44.3±3.56 (0.043) LPS (10微克/«升) 115+25.1 68.38±24.83 (0.138) 62.84±4 (0.043) 69.77110.34 (0.043) 抗CD3 (12.5奈克/¾升)+ 抗 CD28(l/3xl05) 256611819.3 1075±1239.04 (0.043) 169111921.1 (0.138) 1097±1665.03 (0.080) 濃度1〇_7莫耳或1(Γ8莫耳之化合物16顯著地抑制經 LPS刺激之健康志願者PBMC之IFN- r產製(ρ<0·05)。另 外,1(Τ6莫耳之濃度抑制經單株抗體抗CD3及抗CD2 8所 誘發之IFN- 7產製(ρ<0.05)。10_6莫耳、10·7莫耳及10·8 莫耳之化合物16顯著抑制健康志願者P BMC之IFN- r自 發性分泌(Ρ<〇·〇5)。 ❹ 表10.化合物4存在時健康志願者之PBMC產製IFN- r之情況。結果以該分泌細胞 介素之中位數±標準誤差表示。統計顯著結果係以粗體表示,它們相對應之P値包括 _在括弧中(各實驗條件相較於載劑之資料)。__ n=5 健康志原 頁者PBMC之IFN- r產製(皮克/¾升) 載劑 ΗΓ6莫耳 1〇-7莫耳 10·8莫耳 培養基 94.5±24.5 56.38±9.98 (0.080) 74.51±10.75 (0.225) 57.54±2.59 (0.080) LPS (10微克/毫升) 115±25.1 67.82+4.62 (0.043) 45.2411.29 (0.043) 46.25+3.45 (0.043) 抗CD3 (12.5奈克/毫升)+ 抗 CD28(l/3xl05) 2566±1819.3 1084土 672.2 (0.043) 2622土 1283.57 (0.686) 3797土 775.88 (0.500) -59- 201010988 濃度1(Γό莫耳、1(Γ7莫耳及1(Γ8莫耳之化合物4顯 著地抑制經LPS刺激之健康志願者PBMC之IFN- r產製 (p<0.05)。另外,濃度1CT6莫耳之化合物4亦顯著抑制經 單株抗體抗CD3及抗CD28所誘發之IFN- r產製(Ρ<〇·〇5) 。化合物4並不顯著調節健康志願者PBMC之IFN-^之 自發性分泌。 表11.化合物5存在時健康志願者之PBMC產製IFN- r之情況。結果以該分泌細胞 @ η=5 健康志願者PBMC之IFN- r產製(皮克/毫升) 載劑 nr6莫耳 HT7莫耳 10_8莫耳 培養基 94.5±24.5 34.8±2.9 (0.043) 30.21±5.64 (0.043) 49.7±60 (0.500) LPS (10微克/毫升) 115±25.1 32.56±2.42 (0.043) 35.33±12.15 (0.043) 81.4137.9 (0.500) 抗CD3 (12.5奈克/¾升)+ 抗 CD28(l/3xl05) 2566±1819_3 193411002.79 (0.043) 13201803.97 (0.080) 152811271.3 (0.500) 介素之中位數土標準誤差表示。統計顯著結果係以粗體表示,它們相對應之P値包括 在括弧中(各實驗條件相較於載劑之資料)。_ _ 濃度1(Γ6莫耳或1CT7莫耳之化合物5顯著地抑制經 LPS刺激之健康志願者PBMC之IFN-r產製(ρ<0.05)。另 外,1(Γ6莫耳濃度之化合物5亦顯著抑制經單株抗體抗 CD3及抗CD28所誘發之IFN-r產製(ρ<0.05)。濃度10_6 莫耳及1(Γ7莫耳之化合物16亦抑制健康志願者PBMC之 IFN- r自發性分泌(Ρ<〇·〇5)。 -60- 201010988 表12.化合物6存在時健康志願者之PBMC產製IFN-r之情況。結果以該分泌細胞 介素之中位數±標準誤差表示。統計顯著結果係以粗體表示,它們相對應之p値包括 在括弧中(各實驗條件相較於載劑之資料)。 n=5 健康志願者PBMC之IFN- r產製(皮克/¾升) 載劑 HT6莫耳 1〇-7莫耳 ι〇-8莫耳 培養基 94.5±24.5 70.9816.37 (0.080) 55.7±18.4 (0.043) 77.44±15.5 (0.043) LPS (10微克/«升) 115±25.1 80.6 土 6.89 (0.043) 104.2120.1 (0.080) 82.8±16.2 (0.080) 抗CD3 (12.5奈克/¾升)+ 抗 CD28(l/3xl05) 256611819.3 850±2396.16 (0.500) 147112031.5 (0.893) 328212584.7 (0.225) 濃度1 (Γ6莫耳之化合物6顯著地抑制經LPS刺激之 健康志願者PBMC之IFN-r產製(p<0.05)。其於1〇·7莫耳 及1(Γ8莫耳顯著降低健康志願者PBMC之IFN· 7自發性 分泌(Ρ<〇·〇5)。 表13.化合物9存在時健康志願者之PBMC產製IFN- r之情況。結果以該分泌細胞 介素之中位數±標準誤差表示。統計顯著結果係以粗體表示,它們相對應之p値包括 在括弧中(各實驗條件相較於載劑之資料)。 n=5 健康志願者PBMC之IFN- r產製(皮克/¾升) 載劑 ίο·6莫耳 1〇·7莫耳 10-8莫耳 培養基 94.5±24.5 75.80112.86 (0.080) 66.20±9.80 (0.043) 45.50±9.20 (0.043) LPS (10微克/毫升) 115125.1 62.60±24.88 (0.138) 46.80115.10 (0.080) 61.00±12.10 (0.080) 抗CD3 (12.5奈克/¾升)+ 抗 CD28(l/3xl05) 2566± 1819.3 701.00土 1877.04 (0.138) 1176.00土 1954.90 (0.500) 2764.00土 1281.70 (0.686) 濃度1(Γ7莫耳及1(Γ8莫耳之化合物9顯著地抑制來 -61 - 201010988 自5名健康志願者之PBMC之IFN- r自發性分泌(p<0.05) 表14.化合物10存在時健康志願者之PBMC產製IFN- «τ之情況。結果以該分泌細胞 介素之中位數土標準誤差表示。統計顯著結果係以粗體表示,它們相對應之p値包括 在括弧中(各實驗條件相較於載劑之資料)。 n=5 健康志願者PBMC之IFN- γ產製(皮克/¾升) 載劑 10_6莫耳 1〇-7莫耳 1〇-8莫耳 培養基 94.5±24.5 84.15129.43 (0.225) 113.32+21.96 (0.500) 126.10133.60 (0.686) LPS (10微克/毫升) 115125.1 66.20115.91 (0.080) 124.88120.47 (0.080) 114.60±27.40 (0.043) 抗CD3 (12.5奈克/¾升)+ 抗 CD28(l/3xl05) 256611819.3 1345±1379.13 (0.043) 1841±2135.94 (0.893) 214911224.50 (0.225) 濃度1 〇_8莫耳之化合物1 〇顯著地抑制經LPS刺激之 健康志願者PBMC之IFN-r產製(P<〇.〇5)。另外,在濃度 1(Γ6莫耳時,其亦顯著抑制經單株抗體抗CD3及抗CD28 所刺激之PBMC IFN-r產製(Ρ <〇.〇5)。化合物10並不調 節健康志願者PBMC之IFN- r之自發性分泌。 -62- 201010988 表15.化合物17存在時健康志願者之PBMC產製IFN- 7之情況。結果以該分泌細胞 介素之中位數±標準誤差表示。統計顯著結果係以粗體表示,它們相對應之p値包括 在括弧中(各實驗條件相較於載劑之資料)。 η=5 健康志願者PBMC之IFN- r產製(皮克/毫升) 載劑 ΗΓ6莫耳 ΙΟ·7莫耳 ΗΓ8莫耳 培養基 94.5±24.5 20.60122.80 (0.080) 75.15115.44 (0.043) 67.40113.80 (0.138) LPS (10微克/毫升) 115±25.1 33.10±22.20 (0.080) 84.60±15.29 (0.043) 90.00±43.90 (0.500) 抗CD3 (12.5奈克庵升)+ 抗 CD28(l/3xl05) 2566±1819.3 2389±2480.70 (0.893) 272111241.75 (0.500) 123811237.60 (0.345) 濃度1〇_7莫耳之化合物17顯著地抑制來自5名健康 志願者之經LPS刺激之PBMC之IFN- r產製(ρ<0·05)。其 於10_7莫耳顯著抑制志願者PBMC之IFN- r自發性分泌 表16.化合物15存在時健康志願者之PBMC產製IFN- γ之情況。結果以該分泌細胞 介素之中位數增準誤差表示。統計顯著結果係以粗體表示,它們相對應之Ρ値包括 ® _在括弧中(各實驗條件相較於載劑之資料)。_ n=5 健康志願者PBMC之IFN- γ產製(皮克/¾升) 載劑 10·6莫耳 ΚΓ7莫耳 10-8莫耳 培養基 94.5124.5 78.60±1.60 (0.225) 119.52±5.88 (0.893) 89.40±18.80 (0.893) LPS (10微克/¾升) 115125.1 73.30±7.80 (0.043) 90.49124.35 (0.225) 41.70114.00 (0.043) 抗CD3 (12.5奈克/¾升)+ 抗 CD28(l/3xl05) 2566±1819.3 249011654.30 (0.345) 3831±1547.58 (0.686) 316511514.10 (0.686) 濃度1(Γ8莫耳之化合物15顯著地抑制來自5名健康 -63- 201010988 志願者之經LPS刺激之PBMC之IFN- r產製(P<〇.〇5)。其 不顯著調節健康志願者 PBMC之IFN- r之自發性或 CD3 + CD28刺激分泌。 表Π.化合物12存在時健康志願者之PBMC產製IFN- γ之情況。結果以該分泌細胞 介素之平均値土標準誤差表示》統計顯著結果係以粗體表示,它們相對應之P値包括 __在括弧中(各實驗條件相較於載劑之資料)。_ n=13 健康志願者PBMC之IFN- 7產製(皮克/¾升) 載劑 ΙΟ"6莫耳 1〇-7莫耳 10_8莫耳 培養基 283.78±90.00 189.72140.00 (0.279) 296.28190.00 (0.807) 279.16±60.00 (0.807) LPS (10微魏升) 860.42±105.00 649.61±120.00 (0.011) 919.53土130.00 (0.552) 1049.85i230.00 (0.382) 抗CD3 (12.5奈克/¾升 )+抗 CD28(l/3 xlO5) 11802.22土 1300.00 7927.17± 1003.00 (0.019) 11552.08士 1600.00 (0.917) 10254.66土 1500.00 (0.196) 濃度1 〇_6莫耳之化合物1 2顯著地抑制經LP S刺激或 經單株抗體抗CD3及抗CD28刺激之健康志願者PBMC之 _ 〇 IFN- τ 產製(p<〇.〇5)(圖 8)。 化合物12並不調節健康志願者PBMC之IFN- r之自 發性分泌。 使來自8名類風濕性關節炎病患之PBMC(每孔5x1 04 細胞)於添加最高溶劑濃度(10_6莫耳),及1〇·6莫耳、1〇·7 莫耳及1〇_8莫耳之化合物12之200微升完全培養基中, 在LPS(10微克/毫升)或抗CD3(12.5奈克/毫升)+抗 CD28(l/3xl05)存在及不存在之情況下培養(重覆二次)24 小時。培養上清液被冷凍於-2 0 °C,利用專門用於測定 -64- 201010988 IFN- 7濃度之CBA Flex Set根據製造商之說明以BD FACS陣列生物分析儀測定IFN- r之濃度。 以1(Γ6莫耳、1(Γ7莫耳及1〇·8莫耳之濃度存在的化 合物12顯著地降低來自類風濕性關節炎病患之PBMC在 經單株抗體抗CD3及抗CD28刺激時所誘發之1?]^-7產 製(ρ<0.05)(圖9)。化合物12不調節來自類風濕性關節炎 病患之PBMC之自發性IFN- r分泌或LPS誘發之分泌。 表18.化合物12存在時類風濕性關節炎病患之PBMC產製IFN- 7"之情況。結果以該 分泌細胞介素之平均値±標準誤差表示。統計顯著結果係以粗體表示,它們相對應之 _P値包括在括弧中(各實驗條件相較於載劑之資料)。 n=8 類風濕性1 哥節炎病患PBMC之IFN- τ產製(皮克/毫升) 載劑 HT6莫耳 HT7莫耳 ΗΓ8莫耳 培養基 145.49i28.00 193.68145.00 (0.779) 203.72150.00 (0.208) 235.10160.00 (0.674) LPS (10微克/¾升) 461.14185.00 205.26±46.00 (0.484) 146.50133.00 (0.208) 248.73±35.00 (0.889) 抗CD3(12.5奈克/ 毫升)+抗D28 (1/3x105) 8406.1½ 1000.00 5115.27土 600.00 (0.05) 6411.7士 700.00 (0.05) 7122.07土 950 (0.017) 實施例8。化合物5、6、12及16對介白素8(1 L-8)產 製之影響 進一步硏究化合物5、6、12及16對於來自健康志願 者之PBMC產製IL-8之影響,同時在LPS刺激及單株抗 體抗CD3及抗CD28存在及不存在時評估。使來自健康志 願者之PBMC(每孔5xl04細胞)於僅添加最高溶劑濃度(1〇_ 6莫耳),或添加1〇_6莫耳、1〇_7莫耳或1〇_8莫耳之測試 -65- 201010988 化合物之200微升完全培養基中,在LPS(10微克/毫升) 或抗CD3(12.5奈克/毫升)+抗CD28(l/3xl05)存在及不存 在之情況下培養(重覆二次)24小時。培養上清液被冷凍 於-20°C,利用專門用於測定IL-8濃度之CBA Flex Set根 據製造商之說明以BD FACS陣列生物分析儀定量IL-8之 濃度。 表19.化合物16存在時健康志願者之PBMC產製IL-8之情況。結果以該分泌細胞介 素之中位數±標準誤差表示。統計顯著結果係以粗體表示,它們相對應之P値包括在 括弧中(各實驗條件相較於載劑之資料)。 η=5 健康志願者PBMC之IL-8產製(皮克/毫升) 載劑 HT6莫耳 10.7莫耳 10-8莫耳 培養基 80961±2255 65232129577 (0.138) 53645130467 (0.043) 55043129843 (0.080) LPS(10微克/¾升) 119800土 23353 66988±29703 (0.138) 65232±32229 (0.138) 60278±33874 (0.225) 抗CD3(12.5奈克/毫 升)+抗CD28 (1/3χ105) 90653±16263 59496126929 (0.345) 56850128160 (0.686) 59496126425 (0.345) 在所有剌激條件及化合物之測試濃度下,化合物1 6 明顯降低來自5名捐贈者PBMC之IL-8產製。另外,在 1(Γ7莫耳之濃度下,其顯著抑制健康志願者PBMC之IL-8 自發性分泌(Ρ<〇.〇5)。 201010988 表20.化合物5存在時健康志願者之PBMC產製IL-8之情況。結果以該分泌細胞介 素之中位數±標準誤差表示。統計顯著結果係以粗體表示,它們相對應之p値包括在 _括弧中(各實驗條件相較於載劑之資料)。 n=5 健康志願者PBMC之IL-8產製(皮克/1 !升) 載劑 10·6莫耳 1〇·7莫耳 10·8莫耳 培養基 8096112255 48453±25626 (0.043) 62698±28354 (0.138) 58725128788 (0.138) LPS(10微克/毫升) 119800±23353 51626133558.86 (0.043) 59886+33157 (0.138) 58725132519 (0.138) 抗CD3(12.5奈克/ 毫升)+抗CD28 (1/3χ105) 90653+16263 70202±27558 (0.500) 62698127532 (0.686) 61072127502 (0.686) 在所有刺激條件及化合物之測試濃度下,化合物1 6 明顯降低來自5名捐贈者PBMC之IL-8產製。另外,在 1(Γ7莫耳之濃度下,其顯著抑制健康志願者PBMC之IL-8 自發性分泌(Ρ < 〇 . 〇 5 )。 表21.化合物6存在時健康志願者之PBMC產製IL-8之情況。結果以該分泌細胞介 素之中位數±標準誤差表示。統計顯著結果係以粗體表示,它們相對應之p値包括在 Φ _括弧中(各實驗條件相較於載劑之資料)。 n=5 健康志願者PBMC之IL-8產製(皮克/¾升) 載劑 10-6莫耳 ΙΟ·7莫耳 1〇·8莫耳 培養基 80961±2255 56528土 20562.02 (0.043) 680623121088 (0.225) 115156±17433 (0.500) LPS (10微克/毫升) 119800123353 120889±18073 (0.345) 120889119096 (0.345) 120889±19725 (0.225) 抗 CD3(12.5 奈 克/«升)+抗 CD28(l/3xl〇5) 90653116263 67547116971 (0.893) 73902±16943 (0.500) 65317+19671 (0.500) -67- 201010988 在所有化合物之測試濃度下,化合物6明顯降低5名 捐贈者之PBMC在CD3 + CD28抗體刺激及自發性釋放條件 下之IL-8產製。另外,在後者條件下,其以1(Γό莫耳之 濃度顯著抑制健康志願者PBMC之IL-8自發性分泌 (ρ<0·05) ° 表22.化合物12存在時健康志願者之PBMC產製IL-8之情況。結果以該分泌細胞介 素之平均値±標準誤差表示。統計顯著結果係以粗體表示,它們相對應之P値包括在 括弧中(各實驗條件相較於載劑之資料)。 η=13 健康志願者PBMC之IL-8產製(皮克/¾升) 載劑 ίο·6莫耳 ΗΓ7莫耳 1〇-8莫耳 培養基 72814.17土 10647.16 45634.31土 6311.13 (0.016) 53732.66±7 952.75 (0.006) 57773.60土 6667.57 (0.087) LPS(10微克/¾升) 173877.36土 8474.49 168467.15土 9656.96 (0.807) 162075.59土 10258.41 (0.249) 169609.11土 9060.90 (0.861) 抗CD3(12.5奈克/ 毫升)+抗CD28 (1/3x105) 107008.56土 8351.21 99779.72土 20000 (0.311) 97968.61士 10220.21 (0.055) 92989.29土 8463.47 (0.028) 在1(Γ6莫耳、1(Γ7莫耳及1(Γ8莫耳之濃度下,化合 物12顯著抑制IL-8之自發性產製(ρ<0.05)(圖10)。1(Γ7 莫耳及1(Γ8莫耳濃度之化合物12顯著抑制由單株抗體抗 CD3及抗CD28所誘發之IL-8產製。然而,沒有觀察到 對LPS刺激PBMC之IL-8產製之影響。 亦硏究對於類風濕性關節炎病患之PBMC產製IL-8 之影響。使來自8名類風濕性關節炎病患之PBMC(每孔5 X104細胞)於添加最高溶劑濃度(1(Γ6莫耳),及ΐ(Γ6莫耳 -68- 201010988 、10_7莫耳及1(Γ8莫耳之化合物12之200微升完全培養 基中,在LPS(10微克/毫升)或抗CD3(12_5奈克/毫升)+抗 CD28(1/3 xlO5)存在及不存在之情況下培養(重覆二次)24 小時。培養上清液被冷凍於-20°C,利用專門用於測定IL-8濃度之CBA Flex Set根據製造商之說明以BD FACS陣 列生物分析儀測定IL-8之濃度。 以1(Γ6莫耳及1〇_8莫耳之濃度存在於培養中之化合 物12顯著地調節來自類風濕性關節炎病患之PBMC中由 LPS所誘發之IL-8產製,但是無法調節自發性及單株抗 體抗CD3及CD28之組合所誘發之IL-8產製(圖11)。所 有結果顯示化合物12對IL-8之調節可能在明顯之健康及 定義疾病條件下顯示明確之調節模式。 表23.類風濕性關節炎病患之IL-8產製。結果以該分泌細胞介素之平均値±標準誤差 表示。統計顯著結果係以粗體表示,它們相對應之ρ値包括在括弧中(各實驗條件相 _碰載劑之資料)。 n=8 類風濕性關節炎病患PBMC之IL-8產製(皮克/¾升) 載劑 KT6莫耳 1〇-7莫耳 1〇-8莫耳 培養基 57877.64土 11143.52 98012.08土 8395.97 (0.208) 96498.50土 10784.62 (0.161) 93977.67土 9247.38 (0.123) LPS(10微克/¾升) 150759.65土 22966.06 185536.78土 22000.00 (0.012) 174156.23土 13385.66 (0.208) 184S41J7土 21500.00 (0.012) 抗CD3(12.5奈克/ 毫升)+抗CD28 (1/3χ105) 118017.97土 21116.00 124576.09土 14605.02 (0.263) 109607.00土 18310.15 (0.889) 121587.88土 16117.25 (0.575) 實施例9。化合物5、12及16對介白素10(IL-10)產 -69- 201010988 製之影響 硏究化合物5、12及16對於來自健康志願者及類風 濕性關節炎病患(化合物12)之PBMC,在LPS刺激及單株 抗體抗CD3及抗CD28之組合存在及不存在時對IL-10產 製之影響。使來自健康志願者之PBMC(每孔5xl04細胞) 於僅添加最高溶劑濃度(10_6莫耳),及添加1 莫耳、1〇-7莫耳或1(Γ8莫耳之測試化合物之200微升完全培養基中 ’在LPS(10微克/毫升)或抗CD3(12.5奈克/毫升)+抗 CD28(l/3xl〇5)存在及不存在之情況下培養(重覆二次)24 小時。培養上清液被冷凍,並利用CBA Flex Set(貝克頓 迪更生)根據製造商之說明測定人IL-10之濃度。 表24.化合物16存在時健康志願者之PBMC產製IL-10之情況。結果以該分泌細胞 介素之中位數±標準誤差表示。統計顯著結果係以粗體表示,它們相對應之p値包括 _在括弧中(各實驗條件相較於載劑之資料)。 _ n=5 健康志原 畐者PBMC之IL-10產製(皮克/«升) 載劑 I。·6莫耳 1〇-7莫耳 ΗΓ8莫耳 培養基 367±188 328±266 (0.225) 4181238 (0.138) 307±271 (0.686) LPS(10微克/毫升) 425±314 494±337 (0.686) 4861351 (0.225) 4411376 (0.138) 抗CD3(12.5奈克/毫升)+ 抗 CD28(l/3xl05) 366±217 3171297 (0.138) 432±255 (0.043) 379±237 (0.043) 在培養中添加化合物16無法調節來自健康捐贈者 PBMC之自發性及LPS誘發之IL-10產製。依此方法,在 1(Γ7莫耳及1(Γ8莫耳之濃度下,其顯著地向上調節或向下 調節健康志願者PBMC在經CD3 + CD28刺激後之IL-10分 201010988 泌(p<0.05),這與在相同條件下之Multiplex分析中所觀 察到之ΠΓ6莫耳顯著抑制IFN- r產製形成對照(p<0.05)。 表25.化合物5存在時健康志願者之PBMC產製IL-10之情況。結果以該分泌細胞介 素之中位數±標準誤差表示。統計顯著結果係以粗體表示,它們相對應之p値包括在 _I舌弧中(各實驗條件相較於載劑之資料)。 n=5 健康志願者PBMC之IL-10產製(皮克/¾升) 載劑 ΗΓ6莫耳 KT7莫耳 1〇-8莫耳 培養基 367.4±187.5 299.60土 240.8 (0.893) 333.40土 211.71 (0.500) 339.10土 420.30 (0.893) LPS(10微克/毫升) 424.9±313.9 427.20土 310.49 (0.500) 402.35土 370.67 (0.686) 441.30士 505.40 (0.686) 抗CD3(12.5奈克/毫 升)+抗 CD28(l/3xl05) 365.50±217 418.00土 196.46 (0.893) 391.45土 241.22 (0.043) 394.30土 390.40 (0.345)Further, the effect of Compound 12 on the production of TNF-α in the presence or absence of LPS in the presence of mononuclear cells purified from PBMC from 9 healthy volunteers was examined. PBMCs from various healthy volunteers were isolated and their mononuclear cells were purified by automated magnetic activated cell sorting (MACS). Resuspend 1〇8 PBMC in complete medium in 5 ml sterile plastic test tubes and magnetic microspheres with CD 14 antibody at 4 °C (CliniMACS CD14 reagent, Bios technology of Bergisch-Gradbach, Germany) Company D-5 1 429)) - Cultured, added to Aut〇MACSTW -54- 201010988 Cell Sorter (Mitsubishi Biotech) to purify their CD 1 4 + PBMC subpopulations (known monocytes) ). The isolated monocytes were stained with FITC-labeled CD3 and PE-labeled CD14-specific antibodies and assayed in 5 ml cell count polystyrene tubes using direct immunofluorescence and multicolor flow cytometry. Purity. Quantification of the surface distribution of the designated T cells (CD3) and monocyte (CD14) surface receptors was performed in a FACScalibur analyzer (Beckondi Rex). Analysis of the results © using the Flow Jo flow cytometer software (Treestar Inc., Oregon, USA). The purity of the isolated PBMC CD3-CD14+ monocytes is routinely > 98% of viable cells, monocytes according to their particular forward scattered light (FSC) and side scatter light (SCC) The characteristics were circled to allow for the definition of distinct monocyte clusters and the remaining lymphocytes (Figure 5). Mononuclear cells (1·25χ105 cells/well, 5 ml) were cultured in a 6-well flat-bottomed low-evaporation cover tissue culture plate (353046 Falcon, Becktondi Rex), supplemented with fetal bovine serum and concentration 1 (Γ6 mol The solvent alone or the complete medium of 1 〇 6 mol of the chelate 12 was added and cultured in the presence or absence of 10 g/ml of LPS for the period of time indicated by the kinetic experiments. The supernatant and cells were simultaneously stored in the kinetic experiments as shown in Figures 6 and 7 to determine the expression of the TNF-α gene product using the amount of mRNA in the cells and the amount of protein in the supernatant. The supernatant is stored frozen at -20 ° C, and the concentration of the secreted TNF-α protein is BD FACS array bioanalyzer using BD TNF-α CBA reagent (Becktondi Rehabilitation Bioscience Pharmaceutical Co., Ltd.) (Beckton Dickens) was measured according to the manufacturer's instructions. The adherent mononuclear cell line was collected from the culture plates using a disposable cell-55-201010988 spatula (Costa Corporation, Massachusetts, 021 39, USA). Cellular mRNA immediately Isolation, mRNA quantification and reverse transcription reactions and quantitative instant polymerase chain reaction (Q-PCR) assays for the determination of TNF-α genes in the 7900 HT Rapid Instant Q-PCR System (Applied Biosystems, Foster City, CA) mRNA expression. Figure 5 shows that highly enriched monocytes (98.3 0 ± 0.17%) were used in this 9 healthy volunteers to analyze mRNA and protein expression of TNF-α in this PBMC subpopulation. Kinetics of secretion. As expected, Figure 6 shows that mRNA expression of TNF-α is strongly induced in human mononuclear cells stimulated by LPS (4 8.35 + 17.46 times, ρ < 0 00 1), in the baseline period In the in vitro condition (〇 hours), the kinetics of the very early activation gene showed a peak at 2.5 hours, while the transient transcription produced a rapid decrease, so the mRNA of TNF-α decreased to near the baseline period at 24 hours. Level (Figure 6 square). In the absence of LPS stimulation, TNF-a gene expression was not activated, and TNF-α specific mRNA maintained its in vitro level in this in vitro system (Figure 6 diamond). I have to pay attention to Compound 12 significantly reduced the peak induction of LPS-stimulated TNF-a mRNA from healthy volunteers and the TaqMan® gene specific for human TNF-cr in the 7900 HT rapid real-time Q-PCR system. Performance analysis measurements (33.72 ± 15.88%, ρ < 〇 · 〇 1) (Figure 6 triangle). In addition, the amount of mRNA expressed by the TNF-α gene was also significantly reduced after 5 and 8 hours of LPS stimulation of the purified monocytes. (27.14±9·07%, p<0.01; and 36.45±11.58%, respectively, ρ<〇.〇1). We conclude that Compound 12 significantly reduced LPS-stimulated TNF- ct mRNA induction in PBMC pure 201010988 monocytes from healthy volunteers, and induced changes during all of the trials without affecting TNF - The peak kinetics of up- and down-regulation of alpha gene products. The very low TNF-α gene expression in human monocytes that have not been stimulated with LPS is consistent with the rapid kinetics of mRNA induction. The secretion of TNF-α protein is initiated rapidly, but is accompanied by translation from mRNA to protein. The @delay time required for the complete biological process of handling, secretion and release into the medium, such that the expected peak yield occurs 8 hours after PBMC purified monocytes in the control medium lacking Compound 12 by LPS (Figure 7 square). Mononuclear cell culture containing only a solvent carrier but no LPS and Compound 12 did not induce secretion of TNF-[alpha] (Fig. 7 diamond). It is noted that Compound 12 significantly reduced peak production of the LPS-stimulated TNF-α secreted protein (8 hours), with purified monocyte lines from healthy volunteers and in a FACS array bio-biological analyzer. The CBA reagent kit for human TNF-α was measured (31.52±14.38%, ^ρ<〇·〇1) (Fig. 7 triangle). As shown above in the kinetics of TNF-a mRNA-induced kinetics, the amount of TNF-a secreted protein was lower from the time point of 0 hour stimulation of the production kinetics monitoring after the culture. In fact, statistical analysis showed that Compound 1 2 also significantly inhibited TNF-α secreted protein from purified PBMC monocytes from healthy volunteers stimulated by LP S at 2.5 hours, 5 hours, and 24 hours (45.56 soil 16.25%, respectively) , ρ <0.01; 41.33 ± 13.37 〇 / 〇, ρ <0.01; and 51.17 ± 15.62%). As the accompanying data showed that Compound 12 significantly inhibited the amount of mRNA and protein expressed by TNF-α-based-57-201010988 in PBMC-purified monocytes from nine healthy donors, we will continue to study Yes, whether there is a statistically significant correlation between the amount of inhibition of TNF-a mRNA induction by Compound 12 and the amount of TNF-α secreted protein, and regression analysis was performed at their individual peaks. It is noted that Compound 12 was found to have a significant correlation with the immunomodulatory effects between the mRNA expressed by the TNF-cr gene product of the purified monocytes and the secreted protein (r2 = 0.7952). Taken together, the above results show that the inhibition of TNF-a mRNA induction by LPS-stimulated purified monocytes by Compound 12 is a mechanism which can be used to explain the substantial part of the immunomodulatory effect of Compound 12 on the inhibition of TNF-α secretion. Example 7. Effect of Compounds 4, 5, 6, 9, 10, 12, 15, 16 and 17 on IFN-R Production PBMCs from healthy volunteers (5X104 cells per well) were spiked with only the highest solvent concentration ( 1〇_δ莫耳), or add 10·6 moles, 10_7 moles or 1 (Γ8 moles of test compound in 200 μl of complete medium, in LPS (10 μg/ml) or anti-CD3 (12.5 Nai)克/ml) + anti-CD28 (l/3 xlO5) in the presence and absence of culture (repeated twice) for 24 hours. The culture supernatant was frozen at -20 ° C, specifically for the determination of IFN-r Concentration of CBA Flex Set quantifies the concentration of rFN_7 using a Bd FACS array bioanalyzer according to the manufacturer's instructions. 201010988 Table 9. PBMC production of IFN- in healthy volunteers in the presence of compound 16. Results The median ± standard error of the secreted interleukin is expressed. The statistically significant results are shown in bold, and their corresponding p値 is included in parentheses (each experimental condition is compared with the carrier) 〇n=5 Healthy volunteer PBMC IFN- «Tau production system (Pick / ml) Carrier ίο·6 Mo Er 1〇-7 Mo Er 1〇·8 Mo Medium 94.5124.5 45.46±6.89 (0.043) 50.215.2 (0.043) 44.3±3.56 (0.043) LPS (10 μg/«L) 115+25.1 68.38±24.83 (0.138) 62.84±4 (0.043) 69.77110.34 (0.043 Anti-CD3 (12.5 Ng/3⁄4 liter) + Anti-CD28 (l/3xl05) 256611819.3 1075±1239.04 (0.043) 169111921.1 (0.138) 1097±1665.03 (0.080) Concentration 1〇_7mol or 1(Γ8莫耳Compound 16 significantly inhibited the IFN-R production of PBMC stimulated by LPS (ρ < 0.05). In addition, 1 (Τ6 molar concentration inhibition induced by monoclonal antibody anti-CD3 and anti-CD2 8 IFN- 7 production (ρ < 0.05). 10_6 Mo, 10.7 Mo and 10.8 More Compound 16 significantly inhibited the spontaneous secretion of IFN-r from healthy B BMC in healthy volunteers (Ρ<〇·〇5 10 Table 10. Production of IFN-r by PBMCs from healthy volunteers in the presence of Compound 4. Results are expressed as the median ± standard error of the secreted interleukin. Statistically significant results are shown in bold, they are Corresponding P値 includes _ in parentheses (data for each experimental condition compared to the carrier). __ n=5 Health Physician PBMC IFN- r production (Pick / 3⁄4 liter) Carrier ΗΓ 6 Mo 1 -7 Mo 10 8 8 Mo medium 94.5 ± 24.5 56.38 ± 9.98 (0.080) 74.51 ±10.75 (0.225) 57.54±2.59 (0.080) LPS (10 μg/ml) 115±25.1 67.82+4.62 (0.043) 45.2411.29 (0.043) 46.25+3.45 (0.043) Anti-CD3 (12.5 Ng/ml) + resistant CD28(l/3xl05) 2566±1819.3 1084 soil 672.2 (0.043) 2622 soil 1283.57 (0.686) 3797 soil 775.88 (0.500) -59- 201010988 Concentration 1 (Γό莫耳, 1 (Γ7莫耳 and 1 (Γ8莫耳之Compound 4 significantly inhibited the IFN- r production of PBMC stimulated by LPS (p<0.05). In addition, Compound 1 at a concentration of 1CT6 Moline also significantly inhibited IFN induced by monoclonal antibody against CD3 and anti-CD28. - r production (Ρ <〇·〇5). Compound 4 did not significantly regulate the spontaneous secretion of IFN-^ from healthy volunteers PBMC. Table 11. PBMC of healthy volunteers in the presence of Compound 5 produced IFN-r The results were obtained by the secretory cell @η=5 healthy volunteer PBMC IFN-r (pick/ml) carrier nr6 mol HT7 mol 10_8 molar medium 94.5±24.5 34.8±2.9 (0.043) 30.21±5.64 (0.043) 49.7±60 (0.500) LPS (10 μg/ml) 115±25.1 32.56±2.42 (0.043) 35.33±12.15 (0.043) 81.4137.9 (0.500) Anti-CD3 (12.5 NEK/ 3⁄4 liters) + anti-CD28 (l/3xl05) 2566±1819_3 193411002.79 (0.043) 13201803.97 (0.080) 152811271.3 (0.500) median soil standard error expression. Statistically significant results are shown in bold, they correspond to P値 is included in parentheses (each experimental condition is compared to the carrier). _ _ concentration 1 (Γ6 mol or 1CT7 mol compound 5 significantly inhibits IFN-r production by PBMC stimulated by LPS in healthy volunteers (ρ < 0.05) In addition, 1 (Γ6 molar concentration of Compound 5 also significantly inhibited the production of IFN-r induced by monoclonal antibodies against CD3 and anti-CD28 (ρ < 0.05). The concentration of 10_6 Mo and 1 (Γ7 molar compound 16 also inhibited the spontaneous secretion of IFN-r from healthy volunteers PBMC (Ρ<〇·〇5). -60- 201010988 Table 12. Healthy volunteers in the presence of Compound 6 PBMC produced IFN-r. The results were expressed as the median ± standard error of the secreted interleukin. Statistically significant results were shown in bold, and their corresponding p値 was included in parentheses (compared to each experimental condition) Information on the carrier) n=5 IFN- r production of PBMC in healthy volunteers (Pick / 3⁄4 liter) Carrier HT6 Mo 1 -7 Mole ι 〇 8 molar medium 94.5 ± 24.5 70.9816. 37 (0.080) 55.7±18.4 (0.043) 77.44±15.5 (0.043) LPS (10 μg/« liter) 115±25.1 80.6 Soil 6.89 (0.043) 104.2120.1 (0.080) 82.8±16.2 (0.080) Anti-CD3 (12.5克/3⁄4 liters) + anti-CD28 (l/3xl05) 256611819.3 850±2396.16 (0.500) 147112031.5 (0.893) 328212584.7 (0.225) Concentration 1 (Γ6 mol of compound 6 significantly inhibited IFN of PBMC in healthy volunteers stimulated by LPS -r production system (p < 0.05). It significantly reduced the spontaneous secretion of IFN·7 in healthy volunteers PBMC at 1〇7 mol and 1 (Γ8 mol) (Ρ&lt 〇·〇5). Table 13. IFN-r production by PBMCs from healthy volunteers in the presence of Compound 9. Results are expressed as the median ± standard error of the secreted interleukin. Statistically significant results are in bold It is indicated that their corresponding p値 is included in parentheses (data of each experimental condition compared to the carrier). n=5 IFN- r production of PBMC in healthy volunteers (Pick/3⁄4 liter) Carrier ίο·6 Moer 1〇·7 Molar 10-8 More Medium 94.5±24.5 75.80112.86 (0.080) 66.20±9.80 (0.043) 45.50±9.20 (0.043) LPS (10 μg/ml) 115125.1 62.60±24.88 (0.138) 46.80115 .10 (0.080) 61.00±12.10 (0.080) Anti-CD3 (12.5 NEK/3⁄4 liter) + Anti-CD28 (l/3xl05) 2566± 1819.3 701.00 Soil 1875.04 (0.138) 1176.00 Soil 1954.90 (0.500) 2764.00 Soil 1281.70 (0.686) Concentration 1 (Γ7 mol and 1 (Γ8 mol of compound 9 significantly inhibited -61 - 201010988 spontaneous secretion of IFN-r from PBMC of 5 healthy volunteers (p<0.05) Table 14. In the presence of compound 10 PBMC from healthy volunteers produced IFN-[τ. The results are expressed as the median soil standard error of the secretory interleukin. Statistically significant results are shown in bold and their corresponding p値 is included in parentheses (data for each experimental condition compared to vehicle). n=5 IFN-γ production of PBMC in healthy volunteers (Pick/3⁄4 liter) Carrier 10_6 Mo 1〇-7 Mo 1〇-8 Moule medium 94.5±24.5 84.15129.43 (0.225) 113.32+21.96 (0.500) 126.10133.60 (0.686) LPS (10 μg/ml) 115125.1 66.20115.91 (0.080) 124.88120.47 (0.080) 114.60±27.40 (0.043) Anti-CD3 (12.5 Ng/3⁄4 liter) + Anti-CD28 (l /3xl05) 256611819.3 1345±1379.13 (0.043) 1841±2135.94 (0.893) 214911224.50 (0.225) Concentration 1 〇 _8 mol of compound 1 〇 significantly inhibited IFN-r production of PBMC stimulated by LPS (P<;〇.〇5). In addition, at a concentration of 1 (Γ6 mol, it also significantly inhibited the production of PBMC IFN-r stimulated by monoclonal antibodies against CD3 and anti-CD28 (Ρ <〇.〇5). Compound 10 does not regulate health volunteers. Spontaneous secretion of IFN-r from PBMC. -62- 201010988 Table 15. Production of IFN-7 by PBMCs from healthy volunteers in the presence of compound 17. Results are expressed as the median ± standard error of the secreted interleukin Statistically significant results are shown in bold, and their corresponding p値 is included in parentheses (data for each experimental condition compared to vehicle). η=5 IFN- r production of healthy volunteer PBMC (Pick/毫升) Carrier ΗΓ6 Mo ΙΟ·7 Mo ΗΓ 8 Moule Medium 94.5±24.5 20.60122.80 (0.080) 75.15115.44 (0.043) 67.40113.80 (0.138) LPS (10 μg/ml) 115±25.1 33.10±22.20 (0.080) 84.60±15.29 (0.043) 90.00±43.90 (0.500) Anti-CD3 (12.5 Nike )) + Anti-CD28 (l/3xl05) 2566±1819.3 2389±2480.70 (0.893) 272111241.75 (0.500) 123811237.60 (0.345) Concentration 1〇_7 mol of compound 17 significantly inhibited IFN-r of LPS-stimulated PBMC from 5 healthy volunteers ρ(ρ<0·05). It significantly inhibited the spontaneous secretion of IFN-r from volunteer PBMC at 10-7 mol. Table 16. The production of IFN-γ by PBMC in healthy volunteers in the presence of compound 15. The result was secretion. Interleukin quantification error is indicated. Statistically significant results are shown in bold, and their corresponding Ρ値 includes ® _ in brackets (each experimental condition is compared to the carrier). _ n=5 IFN-γ production of healthy volunteer PBMC (Pick/3⁄4 liter) Carrier 10·6 Molar 7 Moer 10-8 Mo culture medium 94.5124.5 78.60±1.60 (0.225) 119.52±5.88 (0.893) 89.40± 18.80 (0.893) LPS (10 μg / 3⁄4 liter) 115125.1 73.30 ± 7.80 (0.043) 90.49124.35 (0.225) 41.70114.00 (0.043) Anti-CD3 (12.5 NEK / 3⁄4 liter) + Anti-CD28 (l/3xl05) 2566 ±1819.3 249011654.30 (0.345) 3831±1547.58 (0.686) 316511514.10 (0.686) Concentration 1 (Γ8 mol of compound 15 significantly inhibited IFN- r production from LPS-stimulated PBMC from 5 healthy-63-201010988 volunteers (P<〇.〇5). It did not significantly regulate the spontaneous or IFN-r stimulation of PBMC in healthy volunteers. Table Π. In the presence of Compound 12, PBMC of healthy volunteers produced IFN-γ. The results are expressed as the mean standard error of the secretory interleukin. The statistically significant results are shown in bold, and their corresponding P値 includes __ in parentheses (data for each experimental condition compared to the carrier). _ n=13 IFN- 7 production of healthy volunteer PBMC (Pick / 3⁄4 liter) Carrier ΙΟ "6 Mo Er 1〇-7 Molar 10_8 Mo culture medium 283.78±90.00 189.72140.00 (0.279) 296.28190.00 (0.807) 279.16±60.00 (0.807) LPS (10 micro Wei liters) 860.42±105.00 649.61±120.00 (0.011) 919.53 soil 130.00 (0.552) 1049.85i230.00 (0.382) Anti-CD3 (12.5 NEK/3⁄4 liter) + anti-CD CD28(l/3 xlO5) 11802.22 soil 1300.00 7927.17± 1003.00 (0.019) 11552.08士1600.00 (0.917) 10254.66 soil 1500.00 (0.196) Concentration 1 〇_6 mol of compound 1 2 significantly inhibited by LP S stimulation or by single plant Anti-CD3 and anti-CD28-stimulated healthy volunteers PBMC _ 〇 IFN- τ production system (p < 〇. 〇 5) (Figure 8). Compound 12 did not modulate the spontaneous secretion of IFN-r from healthy volunteer PBMC. PBMC from 8 rheumatoid arthritis patients (5x1 04 cells per well) were added at the highest solvent concentration (10_6 mol), and 1 〇·6 mol, 1 〇·7 mol and 1〇_8 Moultaneous compound 12 in 200 μl of complete medium, cultured in the presence and absence of LPS (10 μg/ml) or anti-CD3 (12.5 Ng/ml) + anti-CD28 (l/3xl05) (repeated Second) 24 hours. The culture supernatant was frozen at -200 ° C, and the concentration of IFN-r was measured by a BD FACS array bioanalyzer using a CBA Flex Set specifically for measuring the concentration of -64-201010988 IFN-7 according to the manufacturer's instructions. Compound 12, which is present at a concentration of 1 (Γ6 mol, 1 (Γ7 mol and 1 〇·8 mol), significantly reduced PBMC from rheumatoid arthritis patients when stimulated with monoclonal antibodies against CD3 and anti-CD28 The induced 1?]^-7 production (ρ < 0.05) (Fig. 9). Compound 12 did not regulate spontaneous IFN-r secretion or LPS-induced secretion of PBMC from rheumatoid arthritis patients. In the presence of Compound 12, PBMC of rheumatoid arthritis patients produced IFN-7" The results are expressed as the mean 値± standard error of the secreted interleukin. Statistically significant results are shown in bold, and they correspond. _P値 is included in parentheses (data for each experimental condition compared to vehicle). n=8 rheumatoid 1 IFN- τ production of PBMC in patients with gangfire (Pick/ml) Carrier HT6 Mo Ear HT7 Moule 8 More Medium 145.49i28.00 193.68145.00 (0.779) 203.72150.00 (0.208) 235.10160.00 (0.674) LPS (10 μg / 3⁄4 liter) 461.14185.00 205.26 ± 46.00 (0.484) 146.50133.00 (0.208) 248.73±35.00 (0.889) Anti-CD3 (12.5 Ng/ml) + Anti-D28 (1/3x105) 8406.11⁄2 1000.00 5115.27 Soil 600.00 (0.05 6411.7士700.00 (0.05) 7122.07土950 (0.017) Example 8. Effect of compounds 5, 6, 12 and 16 on the production of interleukin 8 (1 L-8) Further study of compounds 5, 6, 12 and 16 The effect of IL-8 production by PBMC from healthy volunteers was evaluated at the same time as LPS stimulation and the presence and absence of monoclonal antibody anti-CD3 and anti-CD28. PBMC from healthy volunteers (5xl04 cells per well) Add only the highest solvent concentration (1〇_ 6m), or add 1〇_6mol, 1〇_7mol or 1〇_8mol test-65- 201010988 Compound in 200 μl of complete medium , cultured (repeated twice) in the presence and absence of LPS (10 μg / ml) or anti-CD3 (12.5 Ng / ml) + anti-CD28 (l / 3xl05) for 24 hours. The culture supernatant was frozen The concentration of IL-8 was quantified by a BD FACS array bioanalyzer at -20 ° C using a CBA Flex Set dedicated to determining IL-8 concentration according to the manufacturer's instructions. Table 19. PBMC of healthy volunteers in the presence of Compound 16 Production of IL-8. The results are expressed as the median ± standard error of the secretory cell. Statistically significant results are shown in bold and their corresponding P値 is included in parentheses (data for each experimental condition compared to vehicle). η=5 IL-8 production of PBMC in healthy volunteers (pick/ml) Carrier HT6 Molex 10.7 Molar 10-8 More medium 80961±2255 65232129577 (0.138) 53645130467 (0.043) 55043129843 (0.080) LPS( 10 μg / 3⁄4 liter) 119800 soil 23353 66988 ± 29703 (0.138) 65232 ± 32229 (0.138) 60278 ± 33874 (0.225) Anti-CD3 (12.5 Ng / ml) + anti-CD28 (1/3 χ 105) 90653 ± 16263 59496126929 (0.345 56850128160 (0.686) 59496126425 (0.345) Compound 16 significantly reduced IL-8 production from 5 donor PBMCs under all stimulation conditions and test concentrations of the compounds. In addition, at 1 (Γ7 molar concentration, it significantly inhibited the spontaneous secretion of IL-8 from healthy volunteers PBMC (Ρ<〇.〇5). 201010988 Table 20. Production of PBMC in healthy volunteers in the presence of Compound 5 The case of IL-8. The results are expressed as the median ± standard error of the secreted interleukin. The statistically significant results are shown in bold, and their corresponding p値 is included in the brackets (the experimental conditions are compared with the Information of the agent) n=5 IL-8 production of PBMC in healthy volunteers (Pick / 1 ! liter) Carrier 10·6 Mo Er 1〇·7 Mo Er 10·8 Mo Er Medium 8096112255 48453±25626 ( 0.043) 62698±28354 (0.138) 58725128788 (0.138) LPS (10 μg/ml) 119800±23353 51626133558.86 (0.043) 59886+33157 (0.138) 58725132519 (0.138) Anti-CD3 (12.5 Ng/ml) + anti-CD28 (1 /3χ105) 90653+16263 70202±27558 (0.500) 62698127532 (0.686) 61072127502 (0.686) Compound 16 significantly reduced IL-8 production from 5 donor PBMCs under all stimulation conditions and test concentrations of the compound. At 1 (Γ7 molar concentration, it significantly inhibited IL-8 spontaneously in healthy volunteer PBMC Secretion (Ρ < 〇. 〇5). Table 21. Production of IL-8 by PBMCs in healthy volunteers in the presence of Compound 6. Results are expressed as the median ± standard error of the secreted interleukin. Statistically significant results They are indicated in bold and their corresponding p値 is included in Φ _ brackets (data for each experimental condition compared to vehicle). n=5 IL-8 production of healthy volunteer PBMC (Pick/3⁄4 liter) Carrier 10-6 Moer·7 Mo Er 1〇·8 Moule Medium 80961±2255 56528 Soil 20562.02 (0.043) 680623121088 (0.225) 115156±17433 (0.500) LPS (10 μg/ml) 119800123353 120889±18073 (0.345) 120889119096 (0.345) 120889±19725 (0.225) Anti-CD3 (12.5 NEK / «L) + anti-CD28 (l/3xl〇5) 90653116263 67547116971 (0.893) 73902±16943 (0.500) 65317+19671 (0.500) -67- 201010988 Compound 6 significantly reduced IL-8 production by 5 donors of PBMC under conditions of CD3 + CD28 antibody stimulation and spontaneous release at the test concentrations of all compounds. In addition, under the latter conditions, it significantly inhibited the spontaneous secretion of IL-8 in healthy volunteers by a concentration of 1 (Γό莫耳(ρ<0·05) ° Table 22. Production of PBMC in healthy volunteers in the presence of Compound 12 The results of IL-8 were obtained. The results were expressed as the mean 値± standard error of the secreted interleukins. Statistically significant results are shown in bold, and their corresponding P値 is included in parentheses (each experimental condition is compared to the carrier) Information)) η=13 IL-8 production of PBMC in healthy volunteers (Pick/3⁄4 liter) Carrier ίο·6 Mo ΗΓ7 Mo Er 1〇-8 Mo Er Medium 72814.17 Soil 10647.16 45634.31 土6311.13 (0.016) 53732.66±7 952.75 (0.006) 57773.60 soil 6667.57 (0.087) LPS (10 μg / 3⁄4 liter) 173877.36 soil 8744.49 168467.15 soil 9596.96 (0.807) 162075.59 soil 10258.41 (0.249) 169609.11 soil 9060.90 (0.861) anti-CD3 (12.5 Ng / ml ) + anti-CD28 (1/3x105) 107008.56 soil 8351.21 99779.72 soil 20000 (0.311) 97968.61 士10220.21 (0.055) 92989.29 土8463.47 (0.028) at 1 (Γ6莫耳, 1 (Γ7莫耳和1(Γ8莫耳的浓度 concentration) Compound 12 significantly inhibits the spontaneous production of IL-8 (ρ≪ 0.05) (Fig. 10). 1 (Γ7 Mo and 1 (Γ8 molar concentration of compound 12 significantly inhibited IL-8 production induced by monoclonal antibodies against CD3 and anti-CD28. However, no observation was observed. LPS stimulates the effects of IL-8 production of PBMC. Also investigates the effects of PBMC on the production of IL-8 in patients with rheumatoid arthritis. PBMC from 8 patients with rheumatoid arthritis (5 per well) X104 cells) were added at the highest solvent concentration (1 (Γ6 mol), and ΐ(Γ6莫尔-68-201010988, 10_7 mol and 1 (Γ8 mol of compound 12 in 200 μl of complete medium in LPS ( 10 μg/ml) or anti-CD3 (12_5 Ng/ml) + anti-CD28 (1/3 xlO5) in the presence and absence of culture (repeated twice) for 24 hours. The culture supernatant was frozen at -20 °C, using a CBA Flex Set specifically for the determination of IL-8 concentration, the concentration of IL-8 was determined by a BD FACS array bioanalyzer according to the manufacturer's instructions. A concentration of 1 (Γ6 mol and 1〇_8 mol) Compound 12 present in culture significantly regulates LPS-induced IL-8 production in PBMC from rheumatoid arthritis patients, but does not regulate spontaneous and monogenic resistance The combination of anti-CD3 induced CD28 and IL-8 production of manufactured (FIG. 11). All results show that modulation of IL-8 by Compound 12 may show a clear regulatory pattern under significant health and defined disease conditions. Table 23. IL-8 production in patients with rheumatoid arthritis. The results are expressed as the mean 値± standard error of the secreted interleukin. Statistically significant results are shown in bold, and their corresponding ρ値 is included in parentheses (data for each experimental condition _ collision carrier). n=8 IL-8 production of PBMC in patients with rheumatoid arthritis (pick / 3⁄4 liter) Carrier KT6 Mo 1 -7 Mo 1 -8 Mole medium 57877.64 soil 11143.52 98012.08 soil 8395.97 (0.208 96498.50土10784.62 (0.161) 93977.67 soil 9243.38 (0.123) LPS (10 μg / 3⁄4 liter) 150759.65 soil 22966.06 185536.78 soil 22000.00 (0.012) 174156.23 soil 13385.66 (0.208) 184S41J7 soil 21500.00 (0.012) anti-CD3 (12.5 Ng / ml ) + anti-CD28 (1/3χ105) 118017.97 soil 21116.00 124576.09 soil 14605.02 (0.263) 109607.00 soil 18310.15 (0.889) 121587.88 soil 16117.25 (0.575) Example 9. Effects of Compounds 5, 12 and 16 on the Production of Interleukin 10 (IL-10) -69-201010988 Studies Compounds 5, 12 and 16 for healthy volunteers and patients with rheumatoid arthritis (Compound 12) PBMC, the effect on IL-10 production in the presence and absence of LPS stimulation and the combination of monoclonal antibody anti-CD3 and anti-CD28. PBMCs from healthy volunteers (5xl04 cells per well) were spiked with only the highest solvent concentration (10_6 mol), and 1 mol, 1 〇-7 mol or 1 (Γ8 mol of test compound 200 μl) Culture (repeated twice) in the presence or absence of LPS (10 μg/ml) or anti-CD3 (12.5 Ng/ml) + anti-CD28 (l/3xl〇5) in complete medium for 24 hours. The supernatant was frozen and the concentration of human IL-10 was determined using a CBA Flex Set according to the manufacturer's instructions. Table 24. The production of IL-10 by PBMCs of healthy volunteers in the presence of Compound 16. The results are expressed as the median ± standard error of the secreted interleukin. Statistically significant results are shown in bold, and their corresponding p値 includes _ in parentheses (data for each experimental condition compared to the carrier). n=5 IL-10 production of PBMC of Health Zhiyuan (Pick/«L) Carrier I.·6 Mo Er 1〇-7 MoΗΓ8 Mo Er Medium 367±188 328±266 (0.225) 4181238 (0.138) 307±271 (0.686) LPS (10 μg/ml) 425±314 494±337 (0.686) 4861351 (0.225) 4411376 (0.138) Anti-CD3 (12.5 g/ml) + anti-CD28 (l/3xl05) 366±217 3171297 (0.138) 432±255 (0.043) 379±237 (0.043) Addition of compound 16 to culture does not regulate spontaneous and LPS induction from healthy donor PBMC According to this method, at 1 (Γ7 mol and 1 (Γ8 mol concentration, it significantly up-regulates or down-regulates the IL-in the healthy volunteer PBMC after stimulation with CD3 + CD28) 10 points 201010988 secretion (p < 0.05), which contrasted with the observation that ΠΓ 6 mole significantly inhibited IFN- r production in the multiplex analysis under the same conditions (p < 0.05). Table 25. Health in the presence of compound 5 The PBMC of volunteers produced IL-10. The results were expressed as the median ± standard error of the secreted interleukin. The statistically significant results were shown in bold, and their corresponding p値 was included in the _I tongue arc. (The experimental conditions are compared with the carrier data.) n=5 IL-10 production of PBMC in healthy volunteers (Pick/3⁄4 liter) Carrier ΗΓ6 Mo Er KT7 Mo Er 1〇-8 Mo Er Medium 367.4± 187.5 299.60 soil 240.8 (0.893) 333.40 soil 211.71 (0.500) 339.10 soil 420.30 (0.893) LPS (10 μg / ml) 424.9 ± 31 3.9 427.20 soil 310.49 (0.500) 402.35 soil 370.67 (0.686) 441.30 505.40 (0.686) anti-CD3 (12.5 ng / ml) + anti-CD28 (l / 3xl05) 365.50 ± 217 418.00 soil 196.46 (0.893) 391.45 soil 241.22 (0.043 ) 394.30 soil 390.40 (0.345)

在培養中添加化合物5對於來自5名捐贈者PBMC之 IL- 1 0產製造成極小變化,在該化合物之所有測試濃度中 ,不論LPS刺激或CD3 + CD28抗體誘發產製之細胞介素 分泌量非常輕微的增加偶而降低。依此方法,在1 (Γ7莫 耳之濃度下,經CD3+CD28刺激之健康志願者PBMC所分 泌之IL-10被增加(p<0.05)。 -71 - 201010988 表26.化合物12存在時健康志願者之PBMC產製IL-10之情況。結果以該分泌細胞 介素之平均値±標準誤差表示。統計顯著結果係以粗體表示,它們相對應之p値包括 _在括弧中(各實驗條件相較於載劑之資料)〇 n=13 健康志願者PBMC之IL-10產製(皮克/毫升) 載劑 1〇_6莫耳 10-7莫耳 10_8莫耳 培養基 490.46土 87.16 559.06土 112.20 (0.753) 609.10± 108.74 (0.701) 578.66土 106.20 (0.861) LPS(10微克/¾升) 2342.20土 384.02 2420.68土 455.50 (0.507) 2392.99土 427.86 (0.507) 23 89.63土 366.01 (0.701) 抗 CD3(12.5 奈克/¾ 升)+抗 CD28(l/3xl05) 1042.64土 162.36 1239.47土 251.24 (0.507) 1132.69± 213.32 (0.382) 1122.51土 211.03 (0.507) 另一方面’使來自8名類風濕性關節炎病患之 PBMC(每孔5xl04細胞)於添加最高溶劑濃度(10·6莫耳), 及1(Γ6莫耳、10_7莫耳及1〇·8莫耳之化合物12之200微 升完全培養基中,在LPS(1 0微克/毫升)或抗CD3(12.5奈 克/毫升)+抗CD2 8(1 /3 X1 05)存在及不存在之情況下培養( 重覆二次)24小時。培養上清液被冷凍於_20乞,利用專 門用於測定IL-10濃度之CB A Flex Set根據製造商之說明 以B D F AC S陣列生物分析儀測定il- 1 0之濃度。 -72- 201010988 表27.化合物12存在時類風濕性關節炎病患之PBMC產製IL-10之情況。結果以該 分泌細胞介素之平均値增準誤差表示。統計顯著結果係观體表示,它們相對應之 P値包括在括弧中(各實驗條件相較於載劑之資料)。_ 類風濕性圈 藝節炎病患PBMC之IL-10產製(皮克/¾升) n=8 載劑 10·6莫耳 10·7莫耳 HT8莫耳 培養基 452.69± 107.47 682.50土 113.65 (0.674) 798.92土 144.46 (0.401) 468.14± 65.80 (0.263) LPS(10微克/¾升) 2388.64土 300.00 2787.12土 416.91 (0.069) 2597.85土 513.19 (0.327) 3000.00土 400.00 (0.025) 抗CD3(12.5奈克/毫 升)+抗 CD28(l/3xl05) 836.39土 189.09 976.96土 187.99 (0.484) 805.41土 141.23 (1.000) 872.06土 150.87 (0.674) 化合物12對於來自健康志願者之p BMC在二種測試 刺激存在及不存在之情形下之IL-10產製並無影響(圖12) 。然而’化合物12以1〇·8莫耳之濃度存在七培養中,顯 著向上調節類風濕性關節炎病患之PBMC在受到LPS刺 0 激後之IL·10產製(p<0.05)。化合物12對於類風濕性關節 炎病患PBMC之自發性或單株抗體抗CD3及抗CD28刺激 後之IL-10產製並無顯著效果(圖13)。 對免疫系統細胞之其他效果 實施例10.化合物4、5、6、9、12及16對PBMC增 生反應之影響 硏究化合物4、5、6、9、12及16在植物血球凝集素 (PHA) 1〇微克/毫升;PHA 2微克/毫升加外源性IL-2添 加劑(25國際單位/毫升),及單株抗體抗CD-3(12.5奈克/ -73- 201010988 毫升)與抗CD-28(1:320.00 0)之組合之刺激存在或不存在 下對來自健康志願者之PBMC增生反應之影響。 PBMC(每孔50000細胞)係於完全培養基中以37t及 5%二氧化碳大氣(細胞培養箱HERA Cell 1 50,德國朗根 塞爾博德63 5 05賽默飛世爾科技)培養4天。培養刺激條 件爲含有或不含植物血球凝集素(PHA,西班牙馬德里西 格瑪編號L-8902) 10微克/毫升;PHA 2微克/毫升加外源 性IL-2添加劑(25國際單位/毫升,人重組IL-2 Macrolin ,西班牙馬德里凱龍伊比利亞(Chiron Iberica)公司批號 SA753228/4),及單株抗體抗CD-3(12.5奈克/毫升)與抗 CD-28(1 :320.000)。在培養的最後 8小時加入甲基-3H胸 腺核苷(西班牙馬德里ITISA美國放射化學公司,甲基·3Η 胸腺核苷60居禮/毫莫耳)。細胞以玻璃纖維濾膜收集並 埋入閃爍培養基中(1 450-441 Meltilex A,de Wallac Oy, Ref#1450-441)。DNA倂入放射活性係利用貝他粒子計數 器(華雷克奧伊(Wallac Oy)公司)定量。作爲培養細胞存活 性之對照組,這些在顯微鏡(德國哥廷根卡爾蔡司顯微圖 像有限公司Zeiss Axiovert 40CFL)下根據含有培養基但無 其他信號之細胞培養的密度及型態學觀察,以偵測及定量 該硏究分子對PBMC之可能的致分裂直接作用,並評估培 養後之細胞存活性。 -74- 201010988 表28.來自6名健康志願者之PBMC在滴定量之化合物16(此處被稱爲藥物)存在或不 存在時對所示刺激之增生反應。結果係以甲基-3H胸腺核苷吸收(cpm :每分鐘計數) 之中位數±標準誤差表示。統計顯著結果係以粗體表示(此表中無),它們相對應之P 値包括在括弧中(各實驗條件相較於載劑之資料)。_ F-30125-RR 10_6莫耳 1(Τ7莫耳 1〇-8莫耳 10_9莫耳 1(Τ1()莫耳 培養基 載 劑 4173±750 31341731 10751559 786±781 42111395 藥 物 2355+668 (0.249) 2101±1006 (0.917) 1932±421 (0.463) 20401287 (0.345) 2456±212 (0.345) PHA-P (10微克/¾升) 載 劑 132842土 12078 133400土 16114 149886土 21502 151690土 17207 125060土 15270 藥 物 101935土 16298 (0.753) 150870土 19554 (0.463) 148142土 19742 (0.917) 142231土 12403 (0.600) 151052土 16849 (0.075) PHA-P(2ug/ml) +IL2 (25單位/¾升) 載 劑 75594土 7429 92185± 11446 85481土 9600 87486土 9689 72575土 7773 藥 物 71695± 8080 (0.463) 95472士 8034 (0.463) 85565土 9363 (0.753) 92553土 8892 (0.753) 96128土 9283 (0.075) TCE(微取細胞 比 2:1) 載 劑 107000土 13172 177461土 25149 174961± 24024 178210土 23219 165498土 25428 藥 物 100537土 17868 (0.917) 1815001 28947 (0.116) 189561土 27195 (0.463) 170159土 19945 (0.917) 183824士 23708 (0.173)Addition of Compound 5 to the culture produced minimal changes in IL-10 production from 5 donor PBMCs, regardless of the amount of interleukin secretion induced by LPS stimulation or CD3 + CD28 antibody in all tested concentrations of the compound. Very slight increases are occasionally reduced. According to this method, IL-10 secreted by PBMC stimulated by CD3+CD28 was increased (p<0.05) at a concentration of 1 (Γ7 mol). -71 - 201010988 Table 26. Health in the presence of Compound 12 The PBMC of volunteers produced IL-10. The results were expressed as the mean 値± standard error of the secreted interleukins. The statistically significant results were shown in bold, and their corresponding p値 included _ in brackets (each experiment) Conditions compared to the carrier data) 〇n=13 IL-10 production of PBMC in healthy volunteers (pick/ml) Carrier 1〇_6 Molar 10-7 Molar 10_8 Mo culture medium 490.46 Soil 87.16 559.06 Soil 112.20 (0.753) 609.10± 108.74 (0.701) 578.66 soil 106.20 (0.861) LPS (10 μg/3⁄4 liter) 2342.20 soil 382.02 2420.68 soil 455.50 (0.507) 2392.99 soil 427.86 (0.507) 23 89.63 soil 366.01 (0.701) anti-CD3 ( 12.5 Nike / 3⁄4 liter) + anti-CD28 (l/3xl05) 1042.64 soil 162.36 1239.47 soil 251.24 (0.507) 1132.69 ± 213.32 (0.382) 1122.51 soil 211.03 (0.507) On the other hand 'make 8 rheumatoid arthritis diseases PBMC (5xl04 cells per well) with the highest solvent concentration (10·6 mol) And 1 (Γ6 mol, 10_7 mol and 1 〇·8 mol of compound 12 in 200 μl of complete medium, in LPS (10 μg/ml) or anti-CD3 (12.5 Ng/ml) + anti-CD2 8 (1 / 3 X1 05) cultured (repeated twice) in the presence and absence of 24 hours. The culture supernatant was frozen at -20 Torr, using a CB A Flex Set dedicated to the determination of IL-10 concentration. The concentration of il-10 was measured by a BDF AC S array bioanalyzer according to the manufacturer's instructions. -72- 201010988 Table 27. Production of IL-10 by PBMC in rheumatoid arthritis patients in the presence of Compound 12. Results The average 値 値 误差 误差 。 。 。 。 。 。 。 。 。 。 。 。 。 。 。 。 。 。 。 。 。 。 。 。 。 。 。 。 。 。 。 。 。 。 。 。 。 。 。 。 。 。 。 。 。 。 。 。 Arthritis patients with PBMC IL-10 production (pick / 3⁄4 liters) n = 8 carriers 10 · 6 moles 10 · 7 moles HT8 molar medium 452.69 ± 107.47 682.50 soil 113.65 (0.674) 798.92 soil 144.46 (0.401) 468.14± 65.80 (0.263) LPS (10 μg/3⁄4 liter) 2388.64 soil 300.00 2787.12 soil 416.91 (0.069) 2597.85 soil 513.19 (0.327 3000.00 soil 400.00 (0.025) anti-CD3 (12.5 Ng/ml) + anti-CD28 (l/3xl05) 836.39 soil 189.09 976.96 soil 187.99 (0.484) 805.41 soil 141.23 (1.000) 872.06 soil 150.87 (0.674) Compound 12 for health Volunteer p BMC had no effect on IL-10 production in the presence and absence of both test stimuli (Figure 12). However, Compound 12 was present in seven cultures at a concentration of 1 〇·8 mol, and significantly increased the IL-10 production of PBMC in patients with rheumatoid arthritis after LPS puncturing (p<0.05). Compound 12 had no significant effect on the spontaneous or monoclonal antibody anti-CD3 and anti-CD28 stimulation of IL-10 production in patients with rheumatoid arthritis (Fig. 13). Other Effects on Cells of the Immune System Example 10. Effect of Compounds 4, 5, 6, 9, 12, and 16 on PBMC Proliferative Response Studies Compounds 4, 5, 6, 9, 12, and 16 in Plant Hemagglutinin (PHA) 1 μg/ml; PHA 2 μg/ml plus exogenous IL-2 additive (25 IU/ml), and monoclonal antibody anti-CD-3 (12.5 Ng/-73-201010988 ml) and anti-CD The effect of a combination of -28 (1:320.00 0) on the proliferative response of PBMC from healthy volunteers in the presence or absence of stimulation. PBMC (50,000 cells per well) were cultured in complete medium for 4 days in a 37t and 5% carbon dioxide atmosphere (cell incubator HERA Cell 150, Deutsche Sölberg 63 5 05 Thermo Fisher Scientific). The culture conditions were stimulated with or without phytohemagglutinin (PHA, Madrid Sigma number L-8902) 10 μg/ml; PHA 2 μg/ml plus exogenous IL-2 additive (25 IU/mL, human recombination) IL-2 Macrolin, Chiron Iberica, Madrid, Spain, batch number SA753228/4), and monoclonal antibodies against CD-3 (12.5 ng/ml) and anti-CD-28 (1:320.000). Methyl-3H thymidine was added during the last 8 hours of culture (ITISA American Radiochemical Company, Madrid, Spain), methyl 3 thymidine nucleoside 60 mus / millimolar. The cells were collected on a glass fiber filter and embedded in scintillation medium (1 450-441 Meltilex A, de Wallac Oy, Ref #1450-441). DNA intrusion radioactivity was quantified using a beta particle counter (Wallac Oy). As a control group for cell viability, these were observed under microscope (Zeiss Axiovert 40CFL, Göttingen, Germany) based on the density and morphology of cell cultures containing medium but no other signals. The effect of the study molecule on the possible mitogenic division of PBMC was measured and quantified, and the cell viability after culture was evaluated. -74- 201010988 Table 28. Proliferative response of the PBMCs from 6 healthy volunteers to the indicated stimuli in the presence or absence of titrated compound 16 (referred to herein as a drug). Results are expressed as median ± standard error of methyl-3H thymidine uptake (cpm: counts per minute). Statistically significant results are shown in bold (none in this table) and their corresponding P 値 are included in parentheses (data for each experimental condition compared to vehicle). _ F-30125-RR 10_6 Mo 1 (Τ 7 Mo 1 〇 8 Mo 10_9 Mo 1 (Τ 1 () Mo medium carrier 4173 ± 750 31341731 10751559 786 ± 781 42111395 Drug 2355 + 668 (0.249) 2101 ±1006 (0.917) 1932±421 (0.463) 20401287 (0.345) 2456±212 (0.345) PHA-P (10 μg/3⁄4 liter) Carrier 132842 Soil 12078 133400 Soil 16114 149886 Soil 21502 151690 Soil 17207 125060 Soil 15270 Drug 101935 Soil 16298 (0.753) 150870 soil 19554 (0.463) 148142 soil 19742 (0.917) 142231 soil 12403 (0.600) 151052 soil 16849 (0.075) PHA-P (2ug / ml) + IL2 (25 units / 3⁄4 liters) carrier 75594 soil 7429 92185± 11446 85481 soil 9600 87486 soil 9689 72575 soil 7773 drug 71695± 8080 (0.463) 95472士8034 (0.463) 85565 soil 9936 (0.753) 92553 soil 8892 (0.753) 96128 soil 9283 (0.075) TCE (micro-cell ratio 2:1) Carrier 107000 soil 13172 177461 soil 25149 174961 ± 24024 178210 soil 23219 165498 soil 25428 drug 100537 soil 17868 (0.917) 1815001 28947 (0.116) 189561 soil 27195 (0.463) 170159 soil 19945 (0.917) 183824 ± 23708 (0.173 )

培養中存在之化合物16並不顯著調節來自6名健康 志願者之PBMC在各種硏究之致分裂刺激存在或不存在下 之增生反應(表 28)。高劑量之IL-10已知可抑制 CD3 + CD28抗體誘發之增生反應,表示調節IL-1含量不 是這裡分析之明確實驗條件所作用之主要機制。其不像許 多一般之免疫抑制劑爲負向影響T細胞增生之免疫抑制化 合物。 -75- 201010988 表29.來自ό名健康志願者之PBMC在滴定量之化合物4(此處被稱爲藥物)存在或不 存在時對所示刺激之增生反應。結果係以甲基-3Η胸腺核苷吸收(cpm :每分鐘計數) 之中位數±標準誤差表示。統計顯著結果係以粗體表示(此表中無),它們相對應之p ί 直包括在括弧中(各實驗條件相_ 嫌載劑之資料)。 F-30217-RS 10·6莫耳 HT7莫耳 1〇-8莫耳 1〇_9莫耳 10_10莫耳 培養基 載 劑 4173±750 3134±731 1075±559 786±781 421±1395 藥 物 3184土 2296 (0.345) 2652士 1544 (0.753) 2577土 1162 (0.116) 2830土 1015 (0.116) 2528±417 (0.345) ΡΗΑ-Ρ (10微克/¾升) 載 劑 132842土 12078 133400土 16114 149886土 21502 151690士 17207 125060土 15270 藥 物 98922土 11888 (0.463) 112615土 13048 (0.173) 112835士 12645 (0.173) 1225041 13667 (0.173) 1189791 9448 (0.917) PHA-P(2ug/ml) +IL2 (25單位/¾升) 載 劑 75594土 7429 92185土 11446 85481土 9600 87486士 9689 72575土 7773 藥 物 70821土 4932 (0.463) 76154土 6307 (0.173) 72169± 6899 (0.345) 86532± 6706 (0.917) 77816土 5686 (0.463) TCE(微麵胞 比 2:1) 載 劑 107000土 13172 177461土 25149 174961土 24024 178210土 23219 1654兕± 25428 藥 物 113360土 18427 (0.753) 183187土 22197 (0.600) 173897土 20637 (0.753) 159689土 22900 (0.463) 182127土 19440 (0.600)Compound 16 present in the culture did not significantly regulate the proliferative response of PBMC from 6 healthy volunteers in the presence or absence of various schizophrenic stimuli (Table 28). High doses of IL-10 are known to inhibit CD3 + CD28 antibody-induced proliferative responses, suggesting that regulation of IL-1 levels is not the primary mechanism by which the experimental conditions of the assay are based. Unlike many general immunosuppressive agents, immunosuppressive compounds that negatively affect T cell proliferation. -75- 201010988 Table 29. Proliferative response of the PBMC from an anonymous health volunteer to the indicated stimulus in the presence or absence of a titrated amount of Compound 4 (herein referred to as a drug). Results are expressed as median ± standard error of methyl-3Η thymidine uptake (cpm: counts per minute). Statistically significant results are shown in bold (none in this table) and their corresponding p ί are included in parentheses (data for each experimental condition _ suspected carrier). F-30217-RS 10·6 Mo Er HT7 Mo Er 1〇-8 Mo Er 1〇_9 Mo Er 10_10 Mo Er Medium Carrier 4173±750 3134±731 1075±559 786±781 421±1395 Drug 3184 Soil 2296 (0.345) 2652士1544 (0.753) 2577土1162 (0.116) 2830 soil 1015 (0.116) 2528±417 (0.345) ΡΗΑ-Ρ (10 μg / 3⁄4 liter) Carrier 132842 soil 12078 133400 soil 16114 149886 soil 21502 151 690 17207 125060 soil 15270 drug 98922 soil 11888 (0.463) 112615 soil 13048 (0.173) 112835 ± 12645 (0.173) 1225041 13667 (0.173) 1189791 9448 (0.917) PHA-P (2ug / ml) + IL2 (25 units / 3⁄4 liters) Carrier 75594 soil 7429 92185 soil 11446 85481 soil 9600 87486 ± 9689 72575 soil 7773 drug 70821 soil 4932 (0.463) 76154 soil 6307 (0.173) 72169 ± 6899 (0.345) 86532 ± 6706 (0.917) 77816 soil 5686 (0.463) TCE ( Micro-cell ratio 2:1) Carrier 107000 soil 13172 177461 soil 25149 174961 soil 24024 178210 soil 23219 1654兕± 25428 Drug 113360 soil 18427 (0.753) 183187 soil 22197 (0.600) 173897 soil 20637 (0.753) 159689 soil 22900 (0.463 ) 182127 soil 19440 (0.600)

培養中存在之化合物4並不顯著調節來自6名健康志 願者之PBMC在各種硏究之致分裂刺激存在或不存在下之 增生反應(表29)。這表示此化合物不像許多一般之免疫抑 制劑爲負向影響T細胞增生之免疫抑制化合物。 -76- 201010988 表30.來自6名健康志願者之PBMC在滴定量之化合物5(此處被稱爲藥物)存在或不 存在時對所示刺激之增生反應。結果係似甲基_3H胸腺核苷吸收(cpm :每分鐘計數) 之中位數±標準誤差表示。統計顯著結果係以粗體表示’它們相對應之P値包括在括 弧中(各實驗條件相較於載劑之資料)°__ F-30218-RS 1〇-6莫耳 1〇·7莫耳 10_8莫耳 10.9莫耳 1(Τ1()莫耳 培養基 載 劑 4173±750 31341731 10751559 7861781 421±1395 藥 物 28281391 (0.116) 2474+581 (0.753) 28961615 (0.075) 23281962 (0.249) 2680±731 (0.345) ΡΗΑ-Ρ (10微克/毫升) 載 劑 132842土 12078 133400土 16114 149886土 21502 151690土 17207 125060土 15270 藥 物 78622土 12398 (0.116) 131549土 11837 (0.753) 151166士 15659 (0.753) 147416士 11912 (0.345) 147352土 15286 (0.075) PHA-P(2ug/ml) +IL2 (25單位/¾升) 載 劑 75594土 7429 92185土 11446 85481土 9600 87486土 9689 72575土 7773 藥 物 45222土 6660 (0.075) 78290土 6748 (0.173) 79679土 5527 (0.345) 83402土 4557 (0.345) 83135士 4607 (0.173) TCE(微糊胞 比 2:1) 載 劑 107000土 13172 177461土 25149 174961士 24024 178210土 23219 165498土 25428 藥 物 57761土 15198 (0.028) 157875± 19319 (0.600) 184083土 21986 (0.753) 183725土 21655 (0.249) 171383土 19208 (0.917) 只有以最高1 〇_6莫耳測試劑量之培養中之化合物5 顯著抑制來自6名健康志願者之PBMC在CD3 + CD28抗體 剌激時之增生反應(Ρ<〇·〇5,表30)。這項特徵區別此化合 物與其它結構相關化合物,它們具有相同之一些結構和功 能特性。 -77- 201010988 表31.來自6名健康志願者之PBMC在滴定量之化合物6(此處被稱爲藥物)存在或不 存在時對所示刺激之增生反應。結果係以甲基-3H胸腺核苷吸收(cpm :每分鐘計數) 之中位數±標準誤差表示。統計顯著結果係以粗體表示(此表中無),它們相對應之p _値包括在括弧中(各實驗條件相較於載劑之資料)。 F-30230-RS HT6莫耳 10·7莫耳 10_8莫耳 ΙΟ-9莫耳 ΙΟ-10莫耳 培養基 載 劑 4173±750 3134+731 1075+559 786±781 421±1395 藥 物 2432±411 (0.173) 2388±388 (0.463) 2252+217 (0.173) 2144±164 (0.345) 24781223 (0.345) PHA-P (10微克/¾升) 載 劑 132842土 12078 133400土 16114 149886土 21502 1516901 17207 125060土 15270 藥 物 111292土 13373 (0.753) 12338½ 10812 (0.753) 123872土 12805 (0.345) 123708土 12430 (0.345) 124177土 13284 (0.753) PHA- P(2ug/ml)+ IL2 (25單位/毫升) 載 劑 75594土 7429 92185士 11446 85481土 9600 87486土 9689 72575土 7773 藥 物 75230土 4053 (0.753) 84630土 7105 (0.345) 83203土 7576 (0.753) 74878土 9183 (0.463) 72810土 10656 (0.249) TCE(微麵胞 比 2:1) 載 劑 107000土 13172 177461± 25149 174961土 24024 1782101 23219 165498土 25428 藥 物 102354土 16275 (0.600) 164461± 17856 (0.917) 177780土 20099 (0.345) 1787(B± 19511 (0.345) 166529± 18415 (0.463)Compound 4 present in the culture did not significantly regulate the proliferative response of PBMC from 6 healthy volunteers in the presence or absence of various schizophrenic stimuli (Table 29). This means that this compound does not resemble many conventional immunosuppressive agents as immunosuppressive compounds that negatively affect T cell proliferation. -76-201010988 Table 30. Proliferative response of the PBMCs from 6 healthy volunteers to the indicated stimuli in the presence or absence of titrated Compound 5 (herein referred to as the drug). The results are expressed as the median ± standard error of methyl _3H thymidine uptake (cpm: counts per minute). Statistically significant results are shown in bold 'their corresponding P値 are included in parentheses (each experimental condition is compared to the carrier).__ F-30218-RS 1〇-6莫耳1〇·7mol 10_8 Mol 10.9 Mo Er 1 (Τ1() More Medium Carrier 4173±750 31341731 10751559 7861781 421±1395 Drug 28281391 (0.116) 2474+581 (0.753) 28961615 (0.075) 23281962 (0.249) 2680±731 (0.345) ΡΗΑ-Ρ (10 μg/ml) Carrier 132842 Soil 12078 133400 Soil 16114 149886 Soil 21502 151690 Soil 17207 125060 Soil 15270 Drug 78622 Soil 12398 (0.116) 131549 Soil 11837 (0.753) 151166士15659 (0.753) 147416士11912 (0.345 ) 147352土15286 (0.075) PHA-P(2ug/ml) +IL2 (25 units/3⁄4 liters) Carrier 75594 soil 7429 92185 soil 11446 85481 soil 9600 87486 soil 9689 72575 soil 7773 drug 45222 soil 6660 (0.075) 78290 soil 6748 (0.173) 79679 soil 5527 (0.345) 83402 soil 4557 (0.345) 83135 ± 4607 (0.173) TCE (micro-powder ratio 2:1) Carrier 107000 soil 13172 177461 soil 25149 174961士24024 178210 soil 23219 165498 soil 25428 drug 57761 soil 15198 (0.028) 157875± 19319 (0.600) 184083 soil 21986 (0.753) 183725 soil 21655 (0.249) 171383 soil 19208 (0.917) Only the compound 5 in culture with a maximum test dose of 1 〇 6 moles significantly inhibited PBMC from 6 healthy volunteers. The proliferative response of CD3 + CD28 antibody when stimulated (Ρ <〇·〇5, Table 30). This feature distinguishes this compound from other structurally related compounds, which have the same structural and functional properties. -77- 201010988 Table 31 PBMC from 6 healthy volunteers responded to the proliferative response of the indicated stimulus in the presence or absence of titrated Compound 6 (herein referred to as the drug). Results are expressed as median ± standard error of methyl-3H thymidine uptake (cpm: counts per minute). Statistically significant results are shown in bold (none in this table) and their corresponding p _ 値 are included in parentheses (data for each experimental condition compared to vehicle). F-30230-RS HT6 Molar 10·7 Molar 10_8 MoΙΟ-9 MoerΙΟ-10 Moter Medium Carrier 4173±750 3134+731 1075+559 786±781 421±1395 Drug 2432±411 (0.173 2388±388 (0.463) 2252+217 (0.173) 2144±164 (0.345) 24781223 (0.345) PHA-P (10 μg/3⁄4 liter) Carrier 132842 Soil 12078 133400 Soil 16114 149886 Soil 21502 1516901 17207 125060 Soil 15270 Drug 111292土13373 (0.753) 123381⁄2 10812 (0.753) 123872 soil 12805 (0.345) 123708 soil 12430 (0.345) 124177 soil 13284 (0.753) PHA- P (2ug/ml) + IL2 (25 units / ml) carrier 75594 soil 7429 92185士11446 85481土9600 87486土9689 72575土7773 drug 75230 earth 4053 (0.753) 84630 soil 7105 (0.345) 83203 soil 7576 (0.753) 74878 soil 9183 (0.463) 72810 soil 10656 (0.249) TCE (micro-face ratio 2 :1) Carrier 107000 soil 13172 177461 ± 25149 174961 soil 24024 1782101 23219 165498 soil 25428 drug 102354 soil 16275 (0.600) 164461 ± 17856 (0.917) 177780 soil 20099 (0.345) 1787 (B ± 19511 (0.345) 166529 ± 18415 ( 0.463)

培養中存在之化合物6並不顯著調節來自6名健康志 願者之PBMC對任何所示刺激之增生反應(p&lt;〇.〇5 ;表31) 。這表示此化合物不像許多一般之免疫抑制劑爲負向影響 T細胞增生之免疫抑制化合物。 -78- 201010988 * 表32.來自健康志願者之PBMC在滴定量之化合物9(此處被稱爲藥物)存在或不存在 時對所示刺激之增生反應。結果係以甲基-3H胸腺核苷吸收(cpm :每分鐘計數)之中 位數土標準誤差表示。統計顯著結果係以粗體表示(此表中無)’它們相對應之P値包 括在括弧中(各實驗條件相較於載劑之資料)。__ F-40221-RS 1〇-6莫耳 1(Τ7莫耳 10_8莫耳 1〇_9莫耳 莫耳 培養基 載 劑 4173±750 31341731 1075±559 786±781 421±1395 藥 物 3902±412 (0.846) 21451287 (0.249) 24321231 (0.249) 27851248 (0.345) 24771229 (0.345) PHA-P (10微克/¾升) 載 劑 132842土 12078 133400± 16114 149886土 21502 1516901 17207 125060± 15270 藥 物 91169± 14336 (0.345) 122456士 24310 (0.173) 143737土 10540 (0.345) 146143土 7862 (0.600) 143807土 10375 (0.345) PHA- P(2ug/ml)+ IL2 (25單位/¾升) 載 劑 75594土 7429 92185土 11446 85481士 9600 87486土 9689 72575土 7773 藥 物 69428士 9105 (0.917) 75861土 16776 (0.173) 83875土 8722 (0.917) 88996土 5048 (0.917) 93063土 7327 (0.116) TCE(微球/細胞 比 2:1) 載 劑 107000土 13172 177461土 25149 174961土 24024 ,178210 ±23219 165498土 25428 藥 物 10095¾ 19317 (0.917) 140678土 29168 (0.345) 162960土 20292 (0.753) 164158土 16818 (0.345) 1665291 21592 (0.600) 培養中存在之化合物9並不顯著抑制健康志願者之 PBMC對任何所示刺激之增生反應(p&lt;0.05 ;表32)。這表 示此化合物不像許多一般之免疫抑制劑爲負向影響T細胞 增生之免疫抑制化合物。 培養中存在之化合物12並不顯著調節健康志願者之 PBMC在各種硏究之致分裂刺激存在或不存在下之增生反 應(圖14a及14b)。 -79- 201010988 實施例1 1.化合物12對PBMC釋放CD62L之影響 硏究化合物12對於來自健康志願者之PBMC之淋巴 細胞及單核細胞釋放CD62L之影響。 當化合物12以1〇·6莫耳之濃度存在時,CD62L (CD62L-藻紅素(PE),cion BD SK11 目錄編號 341012, BD生物科技公司)之釋放顯著增加(p&lt;〇.〇5)。値得注意的 是,在CD62L釋放後所偵測到之CD62L-PE標示之幾何 平均螢光強度(MFI)之降低選擇性地發生在淋巴細胞,但 單核細胞中並未觀察到顯著差異(圖15a及15b)。 表33.淋巴細胞表面CD62L表現之平均螢光強度。p値對應各實驗條件中相較於載劑 之資料。 η=10 淋巴細胞 (平均値+標準誤差)/統計顯著性 載劑 ΗΓ6莫耳 1〇·7莫耳 MFI (P) 112.68113.57 90.01±8.93 (0.47) 116.90+11.02 (0.333) 表34.單核細胞表面CD62L表現之平均螢光強度。p値對應各實驗條件中相較於載劑 之資料。 _ n=10 單核細胞 (平均値+標準誤差)/統計顯著性 載劑 HT6莫耳 1〇·7莫耳 MFI (P) 65.34±8.06 63.90±8.52 (0.721) 72.30110.21 (0.878) 實施例12。鼠內毒血症··化合物12對TNF- α血清 -80- 201010988 濃度之經口活性 使用體重30至35克及飼養無特定病原標準環境下之 CD1公鼠。這些動物禁食(14至16小時;每組12隻;每 籠^ 5 〇 〇平方公分6隻),可任意飮用薦糖(8 %)及 NaCl(0.2%)之營養溶液。經口測試治療係以每公斤10毫 升體積比之懸浮液給予(載劑Tween 80 0·1%_Ν&amp;&lt;:1 〇·9%) 。經過1小時後’藉由腹腔內注射來自大腸桿菌血清型 055:Β5(西格瑪L-2880)之脂多糖(以1毫克/公斤劑量及1〇 毫升/公斤體積比溶解於無菌食鹽水中)以誘發鼠毒血症。 在LPS注射(ip) 90分鐘後,以異氟烷麻醉小鼠,經心臟 穿刺採集0.5至0 · 8毫升之血液;該血液樣本經過離心 (6000 rpm; 10分鐘;4 °C),取二份血清樣本冷凍於-70 °C 直到測量血清TNF- α濃度(EIA :來自 R&amp;D系統之小鼠 TNF/TNFSF1 A Quantikine)。得到之結果摘列於表3 5及圖 16。 ❹ 表35. TNF-α (皮克/¾升) 產品 載劑 洛利普藍 (rolipram) 化雜12 劑Λ途徑) 10毫升/公斤 (經口) 5毫克/公斤 (經口) 100毫克/公斤 (經口) 200毫克/公斤 (經口) η 10 9 8 7 平均値 3211.85 696.83 1386.51 956.42 標準誤差 517.53 65.70 233.06 109.06 抑制(%) --- 78.30 56.83 70.35 Ρ値 (單向 anova) — P&lt;0.001 P&lt;0.01 P&lt;0.01 -81 - 201010988 以100及2 00毫克/公斤/10毫升劑量經口給予之化合 物12在此大腸桿菌LPS(ip)誘發鼠內毒血症之實驗模型中 顯示活性;其相較於載劑組以統計顯著方式降低血清 TNF- α含量。 實施例13.化合物12在小鼠膠原誘發關節炎模型中之 抗關節炎活性 使用飼養於適當環境之8週齡DBA/U公鼠。第〇天 ,動物在尾根部接受100微克之牛膠原二型乳化於完全弗 氏佐劑(1/1)之皮下注射免疫。初次免疫後的16天,以上 述相同程序補強小鼠,但這次以不完全弗氏佐劑乳化膠原 (1/1)。在第19及29天,動物接受40微克之LPS(大腸桿 菌0111 :B4)腹腔注射。藥理治療始於第20天,每天進行 近一個月。關節炎指數每週評估三次,利用雙測量系統: 所有腳掌厚度(數位式游標卡尺)及各肢體之臨床分數(目 視評估〇至4級)。在試驗結束時,取得血清樣本以測定 IL-6濃度(BD生物科技之EIA)。得到之結果摘列於表36 及圖1 7。 -82- 201010988 表36 治療 空白 對照 地塞米松 化合物12 劑量(毫克/公斤) 2(ip) lOO(ip) 300(po) η 4 12 12 12 12 平均厚度(毫米) 2.10 2.59 1.94** 2.30* 2.35* %恢復 132.65 51.02 48.98 臨床分數 0 0.3854 0++ 0.2188+ 0.2404 %恢復 100 43.23 37.62 IL-6(皮克/¾升) 9.37 49.13 1.86** 48.01 26.13* *ρ&lt;0·05 ’ **ρ&lt;〇·〇1與對照組比較之單向ANOVA +p&lt;0.05,++ρ&lt;〇.〇1與對照組比較之克拉斯卡瓦立斯檢定 以100毫克/公斤腹腔注射及300毫克/公斤口服之化 合物12在小鼠膠原誘發之關節炎模型中顯示顯著之抗關 節炎作用。 【圖式簡單說明】 圖1 :經選擇之微球用於流式細胞小球微陣列術 φ (CBA)顯影時之不同螢光特性。 圖2 :化合物12對於受到LPS刺激之健康志願者 (n=10) PBMC產製TNF- α之影響。矩形柱代表各樣本在 不同實驗條件下進行二次培養所得到之中位數及標準誤差 。星號代表各實驗條件之對應資料相較於賦形劑具有統計 顯著差異性(Ρ&lt;〇.〇5)。 圖3 :化合物12對於受到LPS或TCR/CD3 + CD28刺 激之健康志願者(n=13)PBMC產製TNF - α之影響。矩形柱 代表樣本在不同實驗條件下進行二次培養所得到之平均値 -83- 201010988 及標準誤差。星號代表各個不同之實驗條件之對應資料相 較於賦形劑具有統計顯著差異性(P&lt;0. 〇5) ° 圖4 :化合物1 2對於來自類風濕性關節炎病患(n = 7) 之PBMC產製TNF- 〇:之影響。矩形柱代表各樣本在不同 實驗條件下進行二次培養所得到之平均値及標準誤差。星 號代表各個不同之實驗條件之對應資料相較於賦形劑具有 統計顯著差異性(P&lt;〇.〇5)。 圖5:利用FACSCalibur分析儀定量自健康志願者 PBMC分離之AutoMACS分選單核細胞之純度。左圖顯示 FSC(細胞大小)xSSC(顆粒性)分佈之散點圖,其顯示被包 括在圈選出之單核細胞族群閘門內之全活細胞之百分比 (98.5%)。右圖顯示T淋巴細胞與單核細胞標記表現之模 式,其顯示左圖定義之單核細胞閘門內之99.6%之細胞具 有普通人循環單核細胞之CDH + CD3·典型表現型。基準背 景値(象限)係利用同型吻合特異性對照組設定;即使不是 全部之大部分細胞(99.8%)係於左上之「單核細胞象限」 。該顯示之資料係自代表9個獨立進行試驗之志願者之 P BMC純化單核細胞之中位數結果。 圖6:在經LPS刺激之來自健康志願者PBMC純化之 單核細胞中’化合物12對TNF-a mRNA表現動力學之 影響。該曲線表示在單獨與溶劑載劑(♦菱形)或在1〇·6莫 耳之化合物12存在(▲三角形)或不存在(方形)之LPS 刺激培養後’在所示時間點TNF- a mRNA相較於內部 /9-肌動蛋白mRNA家務基因對照之表現量變化之動力學 201010988 ’一種基因產物在各時間點皆於79〇〇ΗΤ快速即時Q-PCR 系統計算。該動力學之時間點(0小時、2 5小時、5小時 、8小時及24小時)係顯示於橫軸。tnF- α /召-肌動蛋白 mRNA表現比之誘導倍數係顯示於縱軸。該顯示之資料係 自代表9個獨立進行試驗之志願者之TNF-amRNA表現 動力學試驗之中位數結果》 圖7:在經LPS刺激之來自健康志願者PbmC純化之 ❹ 單核細胞中’化合物12對TNF-α蛋白質分泌動力學之影 響。該曲線表示在單獨與溶劑賦形劑(♦菱形)或在1〇·6莫 耳之化合物12存在(▲三角形)或不存在(方形)之LPS 刺激培養後,在所示時間點該分泌之T N F - α蛋白質之變 化之動力學。該動力學之時間點(〇小時、2 · 5小時、5小 時、8小時及24小時)係顯示於橫軸。在bD FACSarray 生物分析儀中藉由細胞介素CBA定量所測量之TNF-α蛋 白質之量係根據各時間點計算以顯示於縱軸。該顯示之資 Q 料係自代表9個獨立進行試驗之志願者之TNF-α蛋白質 動力學試驗之中位數結果。 圖8:化合物12對於來自健康志願者(η = ι3)之PBMC 產製IFN-r之影響。矩形柱代表各樣本在不同實驗條件 下進行二次培養所得到之平均値及標準誤差。星號代表該 所示實驗條件之對應資料相較於賦形劑具有統計顯著差異 性(p&lt;0.05)。 圖9:化合物12對於來自類風濕性關節炎病患(n = 8) 之PBMC產製IFN- r之影響。矩形柱代表各樣本在不同 -85- 201010988 實驗條件下進行二次培養所得到之平均値及標準誤差。星 號代表各個不同之實驗條件之對應資料相較於賦形劑具有 統計顯著差異性(p&lt;0.05)。 圖10 :化合物12對於來自健康志願者(n= 13)之 PBMC產製IL-8之影響。矩形柱代表各樣本在不同實驗 條件下進行二次培養所得到之平均値及標準誤差。星號代 表各實驗條件之對應資料相較於賦形劑具有統計顯著差異 性(p&lt;0.05)。 圖11:化合物12對於來自類風濕性關節炎病患(n = 8) 之PBMC產製IL-8之影響。矩形柱代表各樣本在不同實 驗條件下進行二次培養所得到之平均値及標準誤差。星號 代表各實驗條件之對應資料相較於賦形劑具有統計顯著差 異性(p&lt;0.05)。 圖12:化合物12對於來自健康志願者(n= 13)之 PBMC產製IL-10之影響。矩形柱代表各樣本在不同實驗 條件下進行二次培養所得到之平均値及標準誤差。未發現 統計差異性。 圖13 :化合物12對於來自類風濕性關節炎病患(n= 8) 之PBMC產製IL-10之影響。矩形柱代表各樣本在不同實 驗條件下進行二次培養所得到之平均値及標準誤差。星號 代表該對應資料相較於賦形劑具有統計顯著差異性 (P&lt;0.05) ° 圖14 :在PHA、PHA加IL-2或單株抗體抗CD-3(T3) 與抗CD-28之組合之刺激存在或不存在下,化合物12(a) 201010988 與賦形劑(b)對於來自健康志願者(n = 8)之PBMC增生反應 之影響。 圖15:化合物12對於來自健康志願者之PBMC之淋 巴細胞U)及單核細胞(b)釋放CD62L之影響。 圖16:化合物12對於血清TNF-α之量之影響。 圖17:小鼠膠原誘發之關節炎模型中以腳掌平均厚 度(a)及臨床分數(b)評估化合物12之抗關節炎活性。Compound 6 present in the culture did not significantly regulate the proliferative response of PBMC from 6 healthy volunteers to any of the indicated stimuli (p&lt;〇.〇5; Table 31). This means that this compound does not resemble many general immunosuppressive agents as immunosuppressive compounds that negatively affect T cell proliferation. -78-201010988 * Table 32. Proliferative response of PBMCs from healthy volunteers to the indicated stimuli in the presence or absence of titrated Compound 9 (herein referred to as the drug). The results are expressed as the median soil standard error of methyl-3H thymidine uptake (cpm: counts per minute). Statistically significant results are shown in bold (none in this table). Their corresponding P値 is included in parentheses (data for each experimental condition compared to the carrier). __ F-40221-RS 1〇-6莫耳1(Τ7莫耳10_8莫耳1〇_9molecule medium carrier 4173±750 31341731 1075±559 786±781 421±1395 drug 3902±412 (0.846 21451287 (0.249) 24321231 (0.249) 27851248 (0.345) 24771229 (0.345) PHA-P (10 μg / 3⁄4 liter) Carrier 132842 Soil 12078 133400 ± 16114 149886 Soil 21502 1516901 17207 125060 ± 15270 Drug 91169 ± 14336 (0.345) 122456士24310 (0.173) 143737土10540 (0.345) 146143 soil 7862 (0.600) 143807 soil 10375 (0.345) PHA- P (2ug/ml) + IL2 (25 units / 3⁄4 liters) carrier 75594 soil 7429 92185 soil 11446 85481士9600 87486土9689 72575土7773 Drug 69428士9105 (0.917) 75861土16776 (0.173) 83875土8722 (0.917) 88996 soil 5048 (0.917) 93063 soil 7327 (0.116) TCE (microsphere / cell ratio 2:1) Carrier 107000 soil 13172 177461 soil 25149 174961 soil 24024, 178210 ± 23219 165498 soil 25428 drug 100953⁄4 19317 (0.917) 140678 soil 29168 (0.345) 162960 soil 20292 (0.753) 164158 soil 16818 (0.345) 1665291 21592 (0.600) exists in culture Compound 9 does not significantly inhibit health The proliferative response of PBMC to any of the indicated stimuli (p&lt;0.05; Table 32) indicates that this compound is not an immunosuppressive compound that negatively affects T cell proliferation like many common immunosuppressive agents. Compound 12 did not significantly regulate the proliferative response of PBMCs in healthy volunteers in the presence or absence of various schizophrenic stimuli (Figures 14a and 14b). -79- 201010988 Example 1 1. Compound 12 releases CD62L to PBMC The effect of the study compound 12 on the release of CD62L by lymphocytes and monocytes from PBMCs from healthy volunteers was affected. When Compound 12 was present at a concentration of 1 〇·6 mol, the release of CD62L (CD62L-phycoerythrin (PE), cion BD SK11 catalog number 341012, BD Biotech) was significantly increased (p&lt;〇.〇5) . It is noted that the decrease in geometric mean fluorescence intensity (MFI) of the CD62L-PE marker detected after CD62L release occurs selectively in lymphocytes, but no significant difference is observed in monocytes ( Figures 15a and 15b). Table 33. Average fluorescence intensity of CD62L expression on lymphocyte surface. P値 corresponds to the data of each experimental condition compared to the carrier. η=10 lymphocytes (mean 标准+standard error)/statistically significant carrier ΗΓ6 mol 1〇·7 mol MFI (P) 112.68113.57 90.01±8.93 (0.47) 116.90+11.02 (0.333) Table 34. The average fluorescence intensity of CD62L on the surface of nuclear cells. P値 corresponds to the data of each experimental condition compared to the carrier. _ n=10 monocytes (mean 标准 + standard error) / statistically significant carrier HT6 mol 1〇·7 mol MFI (P) 65.34±8.06 63.90±8.52 (0.721) 72.30110.21 (0.878) Example 12. Intravenous toxemia · Compound 12 for oral activity of TNF-α serum -80- 201010988 Concentration CD1 male mice were used with a body weight of 30 to 35 g and a specific pathogen-free environment. These animals were fasted (14 to 16 hours; 12 in each group; 6 in each cage) and 6 nutrient solutions of recommended sugar (8 %) and NaCl (0.2%). The oral test treatment was administered at a volume ratio of 10 ml per kg (carrier Tween 80 0. 1% Ν &amp;&lt;: 1 〇 · 9%). After 1 hour, by intraperitoneal injection of lipopolysaccharide from Escherichia coli serotype 055: Β5 (Sigma L-2880) (dissolved in sterile saline at a dose of 1 mg/kg and 1 〇ml/kg) to induce Rat toxemia. After 90 minutes of LPS injection (ip), the mice were anesthetized with isoflurane and 0.5 to 0.8 ml of blood was collected by cardiac puncture; the blood sample was centrifuged (6000 rpm; 10 minutes; 4 °C), taking two Serum samples were frozen at -70 °C until serum TNF-α concentrations were measured (EIA: mouse TNF/TNFSF1 A Quantikine from the R&amp;D system). The results obtained are summarized in Tables 3 and 16 . ❹ Table 35. TNF-α (Pick/3⁄4 liter) product carrier rolipram (rolipram) 12 doses of sputum) 10 ml / kg (oral) 5 mg / kg (oral) 100 mg / Kg (orally) 200 mg/kg (oral) η 10 9 8 7 Average 値3211.85 696.83 1386.51 956.42 Standard error 517.53 65.70 233.06 109.06 Inhibition (%) --- 78.30 56.83 70.35 Ρ値 (one-way anova) — P&lt; 0.001 P &lt; 0.01 P &lt; 0.01 -81 - 201010988 Compound 12 orally administered at a dose of 100 and 200 mg / kg / 10 ml showed activity in this experimental model of Escherichia coli LPS (ip) induced endotoxemia; It reduced serum TNF-[alpha] levels in a statistically significant manner compared to the vehicle group. Example 13. Anti-arthritic activity of Compound 12 in a mouse collagen-induced arthritis model 8 week old DBA/U male rats were housed in an appropriate environment. On the third day, the animals received a subcutaneous injection of 100 micrograms of bovine collagen type II emulsified in complete Freund's adjuvant (1/1) at the base of the tail. On the 16th day after the initial immunization, the mice were reinforced by the same procedure as above, but this time the collagen (1/1) was emulsified with incomplete Freund's adjuvant. On days 19 and 29, the animals received 40 micrograms of LPS (E. coli 0111: B4) intraperitoneally. Pharmacological treatment began on the 20th day and lasted for nearly a month. The arthritis index was assessed three times a week using a dual measurement system: all foot thickness (digital vernier calipers) and clinical scores for each limb (visual assessment to level 4). At the end of the experiment, serum samples were taken to determine IL-6 concentration (EIA of BD Biotech). The results obtained are summarized in Table 36 and Figure 17. -82- 201010988 Table 36 Treatment blank control dexamethasone compound 12 dose (mg/kg) 2(ip) lOO(ip) 300(po) η 4 12 12 12 12 average thickness (mm) 2.10 2.59 1.94** 2.30* 2.35* % recovery 132.65 51.02 48.98 Clinical score 0 0.3854 0++ 0.2188+ 0.2404 % recovery 100 43.23 37.62 IL-6 (pick / 3⁄4 liters) 9.37 49.13 1.86** 48.01 26.13* *ρ&lt;0·05 ' **ρ&lt ; 〇·〇1 compared with the control group, one-way ANOVA +p&lt;0.05,++ρ&lt;〇.〇1 compared with the control group, the Claska Valis test was administered intraperitoneally at 100 mg/kg and 300 mg/kg. Oral Compound 12 showed a significant anti-arthritic effect in a mouse collagen-induced arthritis model. [Simplified illustration] Figure 1: Selected microspheres for flow cytometry microarray φ (CBA) development of different fluorescence characteristics. Figure 2: Effect of Compound 12 on the production of TNF-α by PBMCs in healthy volunteers (n=10) stimulated by LPS. The rectangular column represents the median and standard error of the secondary culture of each sample under different experimental conditions. The asterisk indicates that the corresponding data for each experimental condition is statistically significantly different from the excipient (Ρ&lt;〇.〇5). Figure 3: Effect of Compound 12 on the production of TNF-α by PBMCs in healthy volunteers (n=13) stimulated by LPS or TCR/CD3 + CD28. The rectangular column represents the average 値 -83- 201010988 and standard error of the sample obtained by secondary culture under different experimental conditions. The asterisk indicates that the corresponding data for each of the different experimental conditions is statistically significantly different from the vehicle (P&lt;0. 〇5) ° Figure 4: Compound 1 2 for patients with rheumatoid arthritis (n = 7) The effect of PBMC on the production of TNF-〇: The rectangular column represents the average enthalpy and standard error obtained by secondary culture of each sample under different experimental conditions. The asterisk indicates that the corresponding data for each of the different experimental conditions is statistically significantly different from the excipient (P&lt;〇.〇5). Figure 5: Quantification of AutoMACS sorting mononuclear cells isolated from healthy volunteer PBMCs using a FACSCalibur analyzer. The left panel shows a scatter plot of the FSC (cell size) xSSC (granularity) distribution showing the percentage of total viable cells (98.5%) included in the circle of selected monocyte populations. The right panel shows the pattern of T lymphocyte and monocyte marker expression, showing that 99.6% of the cells in the monocyte gate defined in the left panel have CDH + CD3 · typical phenotypes of normal human circulating monocytes. The baseline background (quadrant) was set using a homozygous specific control group; even if not all of the cells (99.8%) were in the upper left "monocyte quadrant". The data shown is the median results of purified PBMC purified monocytes from 9 independent volunteers. Figure 6: Effect of Compound 12 on the kinetics of TNF-a mRNA expression in LPS-stimulated monocytes purified from healthy volunteer PBMC. The curve indicates TNF-a mRNA at the indicated time points after incubation with solvent vehicle (♦ rhombohedron) or LPS stimulated culture in the presence of (〇 triangle) or in the absence (square) of 1 〇 6 mol. Kinetics of changes in the amount of expression compared to the internal/9-actin mRNA housekeeping gene control 201010988 'A gene product was calculated at 79 〇〇ΗΤ fast real-time Q-PCR system at each time point. The time points of the kinetics (0 hours, 25 hours, 5 hours, 8 hours, and 24 hours) are shown on the horizontal axis. The tnF-α/call-actin mRNA expression is shown on the vertical axis compared to the induced fold. The data shown are from the median results of the TNF-a mRNA performance kinetics test for 9 independent volunteers. Figure 7: In LPS-stimulated PbmC purified ❹ mononuclear cells from healthy volunteers' Effect of Compound 12 on kinetics of TNF-α protein secretion. The curve indicates that the secretion is expressed at the indicated time points after incubation with solvent excipients (♦ diamonds) or LPS stimulation in the presence of (▲ triangles) or in the absence of (squares) of 1 〇 6 moles of compound 12 Kinetics of changes in TNF-α protein. The time points of the kinetics (〇 hours, 2 · 5 hours, 5 hours, 8 hours, and 24 hours) are shown on the horizontal axis. The amount of TNF-α protein measured by interleukin CBA quantification in the bD FACSarray Bioanalyzer was calculated from each time point to be displayed on the vertical axis. The displayed Q material was based on the median results of the TNF-α protein kinetics test for 9 independent volunteers. Figure 8: Effect of Compound 12 on the production of IFN-r by PBMC from healthy volunteers (η = ι3). Rectangular bars represent the average enthalpy and standard error of each sample subjected to secondary culture under different experimental conditions. The asterisk indicates that the corresponding data for the experimental conditions shown are statistically significantly different (p&lt;0.05) compared to the vehicle. Figure 9: Effect of Compound 12 on IFN-r production by PBMC from rheumatoid arthritis patients (n = 8). The rectangular column represents the average enthalpy and standard error of each sample under different experimental conditions of -85-201010988. The asterisks represent statistically significant differences in the corresponding data for each of the different experimental conditions compared to the vehicle (p&lt;0.05). Figure 10: Effect of Compound 12 on IL-8 production by PBMC from healthy volunteers (n = 13). The rectangular column represents the average enthalpy and standard error obtained by secondary culture of each sample under different experimental conditions. The asterisk represents the statistically significant difference in the corresponding data for each experimental condition compared to the vehicle (p&lt;0.05). Figure 11: Effect of Compound 12 on the production of IL-8 by PBMC from patients with rheumatoid arthritis (n = 8). Rectangular bars represent the average enthalpy and standard error of each sample subjected to secondary culture under different experimental conditions. Asterisks represent statistically significant differences in the corresponding data for each experimental condition compared to vehicle (p&lt;0.05). Figure 12: Effect of Compound 12 on the production of IL-10 by PBMC from healthy volunteers (n=13). The rectangular column represents the average enthalpy and standard error obtained by secondary culture of each sample under different experimental conditions. No statistical differences were found. Figure 13: Effect of Compound 12 on IL-10 production by PBMC from patients with rheumatoid arthritis (n=8). Rectangular bars represent the average enthalpy and standard error of each sample subjected to secondary culture under different experimental conditions. The asterisk indicates that the corresponding data is statistically significantly different from the vehicle (P<0.05). Figure 14: PHA, PHA plus IL-2 or monoclonal antibody anti-CD-3 (T3) and anti-CD-28 The effect of Compound 12(a) 201010988 and excipient (b) on the proliferative response of PBMC from healthy volunteers (n = 8) in the presence or absence of combined stimulation. Figure 15: Effect of Compound 12 on lymphocyte U) and monocyte (b) release of CD62L from PBMC from healthy volunteers. Figure 16: Effect of Compound 12 on the amount of serum TNF-[alpha]. Figure 17: Anti-arthritic activity of Compound 12 was evaluated by mean paw thickness (a) and clinical score (b) in a mouse collagen-induced arthritis model.

-87--87-

Claims (1)

201010988 七、申請專利範圍: 1. 一種式(I)化合物201010988 VII. Patent application scope: 1. A compound of formula (I) 或其醫藥上可接受之鹽、前藥及/或溶劑合物於製備 用於治療急性或慢性發炎疾病之藥物上之用途,其中: @ Ri係選自經取代或未經取代之CrCe烷基、經取代或 未經取代之環烷基、經取代或未經取代之芳基或經取代或 未經取代之雜環基; R2係選自經取代或未經取代之烷基、經取代或 未經取代之環烷基或N(R’R”),其中R’和R”各別爲氫或 經取代或未經取代之C^-Ce烷基;且 R3係未經取代之Ci-6烷基。 2.如申請專利範圍第1項之用途,其中該用於治療急 © 性或慢性發炎疾病之藥物的作用係藉由抑制至少一種選自 TNF- 〇:或IFN- r之前發炎細胞介素之產生,或藉由免疫 調節IL-8趨化激素及/或IL-10調節性細胞介素。 3 ·如申請專利範圍第2項之用途,其中係至少部份於 TNF- a mRNA之表現量上達成該抑制前發炎細胞介素 TNF- α之產生。 4.如申請專利範圍第1至3項中任一項之用途,其中 該急性或慢性發炎疾病係選自急性和慢性血清陽性或血清 -88- 201010988 陰性寡關節炎和多發性關節炎、脊椎關節病、腎小球性腎 炎、膠原病、腎小管間質性腎炎、代謝性徵候群、動脈粥 樣硬化、骨關節炎、氣喘、慢性阻塞性肺病、間質性肺病 、多發性硬化症、脫髓鞘病、腦膜炎、腦炎、腦膜腦炎、 發炎性神經根病變和週邊神經病變、發炎性腸病、硬化、 肝炎、心衰竭、缺血性疾病、腎衰竭、發炎性膀胱炎、良 性前列腺增生、前列腺炎、心肌炎、心包膜炎、葡萄膜炎 @ 、異位性皮虜炎、濕疹、蓴麻疹、牛皮癬、玫瑰痤瘡、過 敏性鼻炎、敗血症、敗血性休克、多重器官衰竭、全身性 自體免疫疾病(諸如全身性紅斑性狼瘡)、血管炎、皮膚肌 炎、澱粉樣變性症或類肉瘤病、器官特異性自體免疫疾病 (諸如重症肌無力)、甲狀腺炎或胰島炎、器官移植、感染 和腫瘤引起之發炎、TNF- α依賴性細胞退化、壞死、細 胞凋亡、移植物抗宿主病、惡病質或自體分泌和旁分泌病 理性細胞生長。 〇 5·如申請專利範圍第4項之用途,其中該慢性發炎疾 病係血清陽性或血清陰性慢性多發性關節炎。 6 ·如申請專利範圍第5項之用途,其中該慢性多發性 關節炎係類風濕性關節炎。 7. 如申請專利範圍第1至3項中任一項之用途,其中 Ri係經取代或未經取代之烷基。 8. 如申請專利範圍第7項之用途,其中1係甲基。 9. 如申請專利範圍第1至3項中任一項之用途,其中 R2係經取代或未經取代之環烷基。 -89- 201010988 10. 如申請專利範圍第9項之用途,其中R2係環戊基 〇 11. 如申請專利範圍第1至3項中任一項之用途,其 中R3係甲基。 12. 如申請專利範圍第1至3項中任一項之用途,其 中該式(I)化合物係選自下列化合物:Or a pharmaceutically acceptable salt, prodrug and/or solvate thereof for use in the manufacture of a medicament for the treatment of an acute or chronic inflammatory disease, wherein: @ Ri is selected from substituted or unsubstituted CrCe alkyl , substituted or unsubstituted cycloalkyl, substituted or unsubstituted aryl or substituted or unsubstituted heterocyclic; R 2 is selected from substituted or unsubstituted alkyl, substituted or Unsubstituted cycloalkyl or N(R'R"), wherein R' and R" are each hydrogen or substituted or unsubstituted C^-Ce alkyl; and R3 is unsubstituted Ci- 6 alkyl. 2. The use of the first aspect of the patent application, wherein the medicament for treating acute or chronic inflammatory diseases is caused by inhibiting at least one inflammatory interleukin selected from the group consisting of TNF-〇: or IFN-r Producing, or by immunomodulating, IL-8 chemokines and/or IL-10 regulatory interleukins. 3. The use of the second aspect of the patent application, wherein the production of the inflammatory interleukin TNF-α is achieved at least in part in the expression amount of TNF-a mRNA. 4. The use of any one of claims 1 to 3, wherein the acute or chronic inflammatory disease is selected from the group consisting of acute and chronic seropositive or serum-88-201010988 negative oligoarthritis and polyarthritis, spine Arthropathy, glomerulonephritis, collagen disease, tubulointerstitial nephritis, metabolic syndrome, atherosclerosis, osteoarthritis, asthma, chronic obstructive pulmonary disease, interstitial lung disease, multiple sclerosis, Demyelinating disease, meningitis, encephalitis, meningoencephalitis, inflammatory radiculopathy and peripheral neuropathy, inflammatory bowel disease, cirrhosis, hepatitis, heart failure, ischemic disease, renal failure, inflammatory cystitis, Benign prostatic hyperplasia, prostatitis, myocarditis, pericarditis, uveitis@, atopic dermatitis, eczema, urticaria, psoriasis, rose acne, allergic rhinitis, sepsis, septic shock, multiple organ failure Systemic autoimmune disease (such as systemic lupus erythematosus), vasculitis, dermatomyositis, amyloidosis or sarcoma-like disease, organ-specific autoimmune disease (such as myasthenia gravis), thyroiditis or insulitis, organ transplantation, infection and tumor-induced inflammation, TNF-α-dependent cell degeneration, necrosis, apoptosis, graft-versus-host disease, cachexia or autocrine and adjacent Secretion of pathological cell growth. 〇 5. The use of the fourth aspect of the patent application, wherein the chronic inflammatory disease is seropositive or seronegative chronic polyarthritis. 6. The use of the fifth aspect of the patent application, wherein the chronic polyarthritis is rheumatoid arthritis. 7. The use of any one of claims 1 to 3 wherein Ri is substituted or unsubstituted alkyl. 8. For the purposes of application No. 7 of the patent scope, 1 is a methyl group. 9. The use of any one of claims 1 to 3 wherein R2 is a substituted or unsubstituted cycloalkyl group. -89- 201010988 10. The use of the ninth aspect of the patent application, wherein R2 is a cyclopentyl hydrazone, wherein the R3 is a methyl group, as claimed in any one of claims 1 to 3. 12. The use of any one of claims 1 to 3, wherein the compound of formula (I) is selected from the group consisting of: -90- 201010988-90- 201010988 201010988201010988 或其醫藥上可接受之鹽、前藥及/或溶劑合物。 13. —種用於治療急性或慢性發炎疾病之醫藥組成物 ,該醫藥組成物包含治療上有效量之如申請專利範圍第1 項所定義之式(I)化合物。 14. 一種式(Γ)化合物Or a pharmaceutically acceptable salt, prodrug and/or solvate thereof. 13. A pharmaceutical composition for the treatment of an acute or chronic inflammatory disease, the pharmaceutical composition comprising a therapeutically effective amount of a compound of formula (I) as defined in claim 1 of the scope of the patent application. 14. A compound of the formula (Γ) 或其醫藥上可接受之鹽、前藥及/或溶劑合物, 其中: R!係選自經取代或未經取代之CKC6烷基、經取代或 未經取代之環烷基、經取代或未經取代之芳基或經取代或 未經取代之雜環基;且 R3係未經取代之Ci-CU烷基。 1 5 ·如申請專利範圍第1 4項之化合物,其中R!係經 -92 - 201010988 取代或未經取代之c , - C 6烷基。 16.如申請專利範圍第15項之化合物,其中Ri係甲 基。 1 7 ·如申請專利範圍第1 4項之化合物,該化合物係選 自下列:Or a pharmaceutically acceptable salt, prodrug and/or solvate thereof, wherein: R! is selected from substituted or unsubstituted CKC6 alkyl, substituted or unsubstituted cycloalkyl, substituted or An unsubstituted aryl group or a substituted or unsubstituted heterocyclic group; and R3 is an unsubstituted Ci-CU alkyl group. 1 5 · A compound of claim 14 wherein R! is a substituted or unsubstituted c, -C 6 alkyl group of -92 - 201010988. 16. A compound according to claim 15 wherein Ri is a methyl group. 1 7 · If the compound of claim 14 is applied, the compound is selected from the following: 或其醫藥上可接受之鹽、前藥及/或溶劑合物。 1 8 · —種醫藥組成物,該醫藥組成物包含如申請專利 範圍第14至17項中任一項所定義之式(I,)化合物或其醫 藥上可接受之鹽、前藥及/或溶劑合物,及至少一種醫藥 -93- 201010988 上可接受之載劑、佐劑及/或賦形劑。 19. 如申請專利範圍第14至17項中任一項之式(1,)化 合物,其用途係作爲藥物。 20. —種如申請專利範圍第1項所定義之式(I)化合物 於顯影和藥理顯影技術中作爲顯影生物標記以找出免疫病 灶、標靶細胞及標的分子上之用途。 21. —種如申請專利範圍第14至17項中任一項之式 (I,)化合物於顯影和藥理顯影技術中作爲顯影生物標記之 用途。Or a pharmaceutically acceptable salt, prodrug and/or solvate thereof. A pharmaceutical composition comprising a compound of the formula (I) as defined in any one of claims 14 to 17 or a pharmaceutically acceptable salt, prodrug thereof and/or A solvate, and at least one carrier, adjuvant, and/or excipient that is acceptable on the pharmaceutical-93-201010988. 19. The compound of the formula (1,) according to any one of claims 14 to 17, which is used as a medicament. 20. Use of a compound of formula (I) as defined in claim 1 of the patent application as a developing biomarker in development and pharmacological development techniques for the identification of immune foci, target cells and target molecules. 21. Use of a compound of formula (I,) according to any one of claims 14 to 17 as a developing biomarker in development and pharmacological development techniques. -94--94-
TW098118927A 2008-06-16 2009-06-06 5-(4-methanesulfonyl-phenyl)-thiazole derivatives for the treatment of acute and chronic inflammatory diseases TWI403506B (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP08380177A EP2135864A1 (en) 2008-06-16 2008-06-16 5-(4-methanesulfonyl-phenyl)-thiazole derivatives for the treatment of acute and chronic inflammatory diseases
US12/139,661 US7781594B2 (en) 2008-06-16 2008-06-16 5-(4-methanesulfonyl-phenyl)-thiazole derivatives for the treatment of acute and chronic inflammatory diseases

Publications (2)

Publication Number Publication Date
TW201010988A true TW201010988A (en) 2010-03-16
TWI403506B TWI403506B (en) 2013-08-01

Family

ID=43414178

Family Applications (1)

Application Number Title Priority Date Filing Date
TW098118927A TWI403506B (en) 2008-06-16 2009-06-06 5-(4-methanesulfonyl-phenyl)-thiazole derivatives for the treatment of acute and chronic inflammatory diseases

Country Status (19)

Country Link
EP (1) EP2313382A1 (en)
JP (1) JP2011524352A (en)
KR (1) KR20110022679A (en)
CN (1) CN102083806A (en)
AR (1) AR072113A1 (en)
AU (1) AU2009259451B2 (en)
BR (1) BRPI0915325A2 (en)
CA (1) CA2728139A1 (en)
CO (1) CO6331429A2 (en)
IL (1) IL209801A0 (en)
MA (1) MA32473B1 (en)
MX (1) MX2010013978A (en)
NZ (1) NZ589894A (en)
RU (1) RU2495031C2 (en)
SA (1) SA109300384B1 (en)
TW (1) TWI403506B (en)
UY (1) UY31896A (en)
WO (1) WO2009153226A1 (en)
ZA (1) ZA201008877B (en)

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FR2961695B1 (en) * 2010-06-29 2012-07-06 Galderma Res & Dev USE OF COMPOUNDS IN THE TREATMENT OR PREVENTION OF SKIN DISORDERS
RU2502513C2 (en) * 2012-01-11 2013-12-27 Государственное бюджетное учреждение здравоохранения Свердловской области "Свердловский областной клинический психоневрологический госпиталь для ветеранов войн" (ГБУЗ СО "СОКП Госпиталь для ветеранов войн") Method of treating patients with multiple organ pathology using ozone therapy
CN111343975A (en) * 2017-11-23 2020-06-26 埃慕尼克股份公司 Dose regimen of vedolol for the prevention or treatment of chronic inflammatory and/or autoimmune diseases
KR102076936B1 (en) * 2019-08-12 2020-02-13 연세대학교 산학협력단 Composition including thiazole or salt thereof as active ingredients for preventing or treating allergic disease or atopic dermatitis

Family Cites Families (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
IE900394L (en) * 1989-02-08 1990-08-08 Abbott Lab Thiazole derivatives
CA2195847A1 (en) * 1994-07-27 1996-02-08 John J. Talley Substituted thiazoles for the treatment of inflammation
WO1996041626A1 (en) * 1995-06-12 1996-12-27 G.D. Searle & Co. Compositions comprising a cyclooxygenase-2 inhibitor and a 5-lipoxygenase inhibitor
AU7559200A (en) * 1999-10-12 2001-04-23 Japan Tobacco Inc. Hypertriglyceridemia remedies and antiobestics
IL160327A0 (en) * 2001-08-13 2004-07-25 Janssen Pharmaceutica Nv 2,4,5-trisubstituted thiazolyl derivatives and their antiinflammatory activity
US20050026902A1 (en) * 2003-01-31 2005-02-03 Timothy Maziasz Methods and compositions for the treatment or prevention of human immunodeficiency virus and related conditions using cyclooxygenase-2 selective inhibitors and antiviral agents

Also Published As

Publication number Publication date
AU2009259451A1 (en) 2009-12-23
WO2009153226A1 (en) 2009-12-23
NZ589894A (en) 2011-08-26
CO6331429A2 (en) 2011-10-20
TWI403506B (en) 2013-08-01
RU2011101439A (en) 2012-07-27
UY31896A (en) 2010-01-29
RU2495031C2 (en) 2013-10-10
JP2011524352A (en) 2011-09-01
CA2728139A1 (en) 2009-12-23
AR072113A1 (en) 2010-08-04
KR20110022679A (en) 2011-03-07
EP2313382A1 (en) 2011-04-27
IL209801A0 (en) 2011-02-28
MX2010013978A (en) 2011-03-29
CN102083806A (en) 2011-06-01
AU2009259451B2 (en) 2013-03-14
ZA201008877B (en) 2012-02-29
BRPI0915325A2 (en) 2015-10-27
SA109300384B1 (en) 2013-04-20
MA32473B1 (en) 2011-07-03

Similar Documents

Publication Publication Date Title
JP6778747B2 (en) Prodrugs of JAK inhibitor compounds for the treatment of gastrointestinal inflammatory bowel disease
ES2586837T3 (en) Method for in vivo expansion of regulatory T lymphocytes
TW201720817A (en) Modulators of chemokine receptors
KR102134171B1 (en) Lanthionine synthetase c-like 2-based therapeutics
US9505798B2 (en) Steroid compounds as RORyt modulators and uses thereof
JP6563947B2 (en) CCR6 compound
JP2022507474A (en) Inhibitors of ARG1 and / or ARG2
JP2021531314A (en) Pyridone A2R antagonist
KR20200128117A (en) Arginase inhibitors
CN103635188A (en) Methods and compositions using PDE4 inhibitors for the treatment and management of autoimmune and inflammatory diseases
WO2009023267A2 (en) Methods for modulating development and expansion of il-17 expressing cells
EP0709097B1 (en) Anti-Fas antibody for rheumatic disease
TW201010988A (en) 5-(4-methanesulfonyl-phenyl)-thiazole derivatives for the treatment of acute and chronic inflammatory diseases
US20210347811A1 (en) Steroid compounds as treg modulators and uses thereof
WO2022111573A1 (en) Pharmaceutical composition for enhancing cell killing, and use thereof
CA3104674A1 (en) Natural killer cells
Tatar et al. Synthesis, characterization and screening of antimicrobial, antituberculosis, antiviral and anticancer activity of novel 1, 3-thiazolidine-4-ones derived from 1-[2-(benzoylamino)-4-(methylthio) butyryl]-4-alkyl/arylalkyl thiosemicarbazides
KR20210040803A (en) Compounds and pharmaceutical compositions for treating autoimmune diseases
KR20220137555A (en) Novel compounds and use thereof for treating psoriasis, asthma, or systemic lupus erythematosus
CA3214244A1 (en) Pyridinyl substituted oxoisoindoline compounds
US8492411B2 (en) 5-(4-methanesulfonyl-phenyl)-thiazole derivatives for the treatment of acute and chronic inflammatory diseases
WO2022226078A1 (en) Therapy for alcohol-related liver disease
US20220323423A1 (en) Oxathiazin_dioxide for treating, preventing, inhibiting or reducing cytokine release
Wang et al. Immunization with autologous T cells enhances in vivo anti-tumor immune responses accompanied by up-regulation of GADD45β
JP3817273B2 (en) Pharmaceutical composition for immunosuppressive treatment, anticancer treatment and antiviral treatment

Legal Events

Date Code Title Description
MM4A Annulment or lapse of patent due to non-payment of fees