TW201249841A - Novel salt forms of 3-cyclopropylcarbonyl-3,6-diazabicyclo[3.1.1]heptane - Google Patents

Novel salt forms of 3-cyclopropylcarbonyl-3,6-diazabicyclo[3.1.1]heptane Download PDF

Info

Publication number
TW201249841A
TW201249841A TW101108527A TW101108527A TW201249841A TW 201249841 A TW201249841 A TW 201249841A TW 101108527 A TW101108527 A TW 101108527A TW 101108527 A TW101108527 A TW 101108527A TW 201249841 A TW201249841 A TW 201249841A
Authority
TW
Taiwan
Prior art keywords
compound
acid
salt
disease
heptane
Prior art date
Application number
TW101108527A
Other languages
Chinese (zh)
Inventor
Jon-Paul Strachan
Timothy J Cuthbertson
David Dale Wirth
Gary Maurice Dull
Sharon Rae Letchworth
Kristen G Jordan
David Kombo
Original Assignee
Targacept Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Targacept Inc filed Critical Targacept Inc
Publication of TW201249841A publication Critical patent/TW201249841A/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/08Bridged systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • A61K31/195Carboxylic acids, e.g. valproic acid having an amino group
    • A61K31/197Carboxylic acids, e.g. valproic acid having an amino group the amino and the carboxyl groups being attached to the same acyclic carbon chain, e.g. gamma-aminobutyric acid [GABA], beta-alanine, epsilon-aminocaproic acid or pantothenic acid
    • A61K31/198Alpha-amino acids, e.g. alanine or edetic acid [EDTA]
    • 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
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/4995Pyrazines or piperazines forming part of bridged ring systems
    • 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
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • A61P25/16Anti-Parkinson drugs

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Medicinal Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Engineering & Computer Science (AREA)
  • Biomedical Technology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Neurosurgery (AREA)
  • Neurology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Psychology (AREA)
  • Epidemiology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)

Abstract

The present invention relates to 3-cyclopropylcarbonyl-3, 6-diazabicyclo[3.1.1]heptane, its salt forms, and novel polymorphic forms of these salts.

Description

201249841 六、發明說明: 【發明所屬之技術領域】 本發明係關於3-環丙基羰基-3,6-二氮雜雙環[3.丨^庚 烷、其鹽形式、及該等鹽之新穎多晶型。本發明亦包括該 等鹽形式之醫藥組合物以及用於治療多種病況及病症之方 法。 【先前技術】 化合物3-環丙基幾基-3,6-二氣雜雙環[3.1.1]庚烧(化合物 A)係神經元菸鹼性受體(NNR)激動劑,其對α4β2*(含α4β2)及 α6β2*(含α6β2) NNR具有選擇性。在左旋多巴(lev〇d〇pa)_ 誘導之運動障礙(LID)之臨床前齧齒類動物模型中,化合 物A證實尤其在預防異常不自主運動(AIm)之完全發作亦 及減弱現有AIM中具有功效。化合物A減輕非人類靈長類 (短尾猴)中之LID且並不會妨礙一般活動或左旋多巴 (L-dopa)對運動缺陷之效應》化合物a呈現抵抗大鼠多巴 胺(dopamine)神經元之原代培養物中的MPP+毒性之神經保 護效應。 3-環丙基羰基-3,6-二氮雜雙環[3.1.1]庚烧具有以下結構 式: γ 諸如3-環丙基羰基-3,6-二氮雜雙環[3.1.1]庚烷等藥物候 選物之商業研發涉及許多步驟,包括適於大規模製程之成 162775.doc 201249841 本有效的合成方法之研發。商業研發亦涉及關於藥物物質 之鹽形式的研究,該藥物物質呈現適宜純度、化學穩定 陘、醫藥性質及有利於方便操作及處理之特性。此外,含 有藥物物質之組合物應具有足夠存架壽命。亦即,其應在 儲存一段相當長的時間時物理化學特性(例如但不限於, 化學組成、水含量'密度、吸水性、及溶解性)不會呈現 顯著變化。另外,在投與患者時藥物之可再現且恆定之血 漿濃度曲線亦係重要因素。 對於經口調配物而言,固體鹽形式由於以優先方式呈現 該等性質之趨勢而通常較佳;且在諸如3_環丙基羰基_3,6_ 一氮雜雙環[3 · Μ ]庚烷等鹼性藥物之情形下,酸加成鹽經 常係較佳鹽形式。然而,鹽形式賦予該等性質之能力變化 顯著’且不可精確地預測該等性質。舉例而言,一些鹽在 環境溫度下係固體,而其他鹽在環境溫度下係液體、黏性 油或膠。此外,一些鹽形式在極端條件下對熱及光穩定且 其他鹽形式在遠更溫和條件下容易分解。因此,用於醫藥 組合物中之鹼性藥物之適宜酸加成鹽形式的研發係高度不 可預測之過程。 化合物Α之一種合成揭示於pCT申請案第PCT/US2〇1〇/ 058836 (WO 201 1/071758)號中,該案件以引用方式併入。 然而,期望3-環丙基羰基-3,6-二氮雜雙環[3.^^庚烷之合 成可放大至大規模生產。此外,需要展示改良性質(包括 純度、穩定性、溶解性、及生物利用度)之鹽形式。該等 新賴鹽形式之優先特性包括彼等可增加活性成份之製造及 162775.doc 201249841 其調配成商業產品之簡易性或效率者。最後,需要增加活 性成份之製造及其調配成商業上可行之產品之簡易性或效 率的該等鹽之穩定多晶型。 【發明内容】 本發明之一個態樣係3-環丙基羰基-3,6-二氮雜雙環 [3.1.1] 庚烷之酸加成鹽。在某些實施例中,酸係選自氫氣 酸、對甲苯績酸、L-天冬胺酸、馬來酸、l-麵胺酸、1_經 基-2-萘曱酸(即,羥萘甲酸鹽)、富馬酸、半乳糖二酸、馬 尿酸、L-扁桃酸、琥珀酸、己二酸或(+)_樟腦酸。在較佳 實施例中’酸加成鹽係對曱苯磺酸鹽、馬來酸鹽、半乳糖 二酸鹽、苯甲酸鹽、馬尿酸鹽、羥萘甲酸鹽或(+)_樟腦酸 鹽。在又一些更佳實施例中,鹽係半乳糖二酸鹽、苯曱酸 鹽、馬尿酸鹽或羥萘甲酸鹽。 本發明之另一態樣包括3-環丙基羰基_3,6-二氮雜雙環 [3.1.1] 庚烧半-半乳糖二酸鹽(11611^£&1&(^&^16)單水合物》 本發明之另一態樣包括3 -環丙基幾基_3,6 -二氮雜雙環 [3.1.1] 庚烷之酸加成鹽’其中該鹽係結晶。 本發明之一個態樣係用於治療或預防一或多種疾病或病 症之方法、用途、化合物、或用於製備用於治療或預防一 或多種疾病或病症之藥劑的用途。在一個實施例中,疾病或 病症係帕金森症(Parkinsonism)、帕金森氏病(Parkins〇n,s201249841 VI. INSTRUCTIONS OF THE INVENTION: TECHNICAL FIELD OF THE INVENTION The present invention relates to 3-cyclopropylcarbonyl-3,6-diazabicyclo[3.丨^heptane, its salt form, and novelties of such salts Polymorphic. The invention also includes pharmaceutical compositions in such salt forms and methods for treating a variety of conditions and disorders. [Prior Art] Compound 3-cyclopropyl-based-3,6-dioxabicyclo[3.1.1]heptane (Compound A) is a neuronal nicotinic receptor (NNR) agonist for α4β2* (containing α4β2) and α6β2* (containing α6β2) NNR is selective. In a preclinical rodent model of levodopa-induced dyskinesia (LID), Compound A has been shown to be particularly effective in preventing the complete onset of abnormal involuntary movement (AIm) and attenuating existing AIMs. Has efficacy. Compound A attenuates LID in non-human primates (cynomolgus) and does not interfere with general activity or the effect of L-dopa on motor deficits. Compound a exhibits resistance to rat dopamine neurons. The neuroprotective effect of MPP+ toxicity in primary cultures. 3-Cyclopropylcarbonyl-3,6-diazabicyclo[3.1.1]heptane has the following structural formula: γ such as 3-cyclopropylcarbonyl-3,6-diazabicyclo[3.1.1]g The commercial development of drug candidates such as alkanes involves a number of steps, including the development of a versatile synthetic method suitable for large-scale processes 162775.doc 201249841. Commercial research and development also involves research on the salt form of a drug substance that exhibits appropriate purity, chemical stability, medical properties, and properties that facilitate handling and handling. In addition, compositions containing drug substances should have sufficient shelf life. That is, it should not exhibit significant changes in physicochemical properties (such as, but not limited to, chemical composition, water content 'density, water absorption, and solubility) for a relatively long period of time. In addition, a reproducible and constant plasma concentration profile of the drug when administered to a patient is also an important factor. For oral formulations, the solid salt form is generally preferred due to the tendency to exhibit such properties in a preferential manner; and in, for example, 3_cyclopropylcarbonyl-3,6-azabicyclo[3·Μ]heptane In the case of an alkaline drug, the acid addition salt is often in a preferred salt form. However, the ability of the salt form to impart such properties varies significantly and does not accurately predict such properties. For example, some salts are solid at ambient temperatures, while others are liquids, viscous oils or gums at ambient temperatures. In addition, some salt forms are stable to heat and light under extreme conditions and other salt forms are readily decomposed under much milder conditions. Therefore, the development of suitable acid addition salt forms for basic drugs in pharmaceutical compositions is a highly unpredictable process. A synthesis of the compound oxime is disclosed in the pCT application No. PCT/US2 〇 1〇 / 058836 (WO 201 1/071758), the disclosure of which is incorporated herein by reference. However, it is expected that the synthesis of 3-cyclopropylcarbonyl-3,6-diazabicyclo[3.^^heptane can be scaled up to mass production. In addition, salt forms that exhibit improved properties including purity, stability, solubility, and bioavailability are required. The preferred characteristics of these new forms of salt include their ability to increase the manufacture of active ingredients and the ease or efficiency with which they are formulated into commercial products. Finally, there is a need to increase the manufacture of active ingredients and their stable polymorphic forms of such salts formulated to the ease or efficiency of commercially viable products. SUMMARY OF THE INVENTION One aspect of the present invention is an acid addition salt of 3-cyclopropylcarbonyl-3,6-diazabicyclo[3.1.1]heptane. In certain embodiments, the acid is selected from the group consisting of hydrogen acid, p-toluic acid, L-aspartic acid, maleic acid, l- faceted acid, 1-cyano-2-naphthoic acid (ie, hydroxy Naphthoate), fumaric acid, galactosidic acid, hippuric acid, L-mandelic acid, succinic acid, adipic acid or (+)-camphoric acid. In a preferred embodiment, the 'acid addition salt is a p-toluenesulfonate, a maleate, a galactate, a benzoate, a horse urate, a hydroxynaphthoate or a (+) 樟 camphor Acid salt. In still other preferred embodiments, the salt is a galactose salt, a benzoate, a hippurate or a hydroxynaphthoate. Another aspect of the invention includes 3-cyclopropylcarbonyl-3-3,6-diazabicyclo[3.1.1] heptanosemi-galactosidate (11611^£&1&(^&^ 16) Monohydrates Another aspect of the present invention includes 3-acid cyclopropyl- 3,6-diazabicyclo[3.1.1] heptane acid addition salt, wherein the salt is crystallized. One aspect of the invention is a method, use, compound, or use for the manufacture of a medicament for treating or preventing one or more diseases or conditions, for use in treating or preventing one or more diseases or conditions. In one embodiment, The disease or condition is Parkinsonism, Parkinson's disease (Parkins〇n, s

Disease)、異常不自主運動、運動障礙及左旋多巴誘導之 運動障礙中之一或多者。在一個實施例中,有需要之患者 正經歷包括左旋多巴之投藥療程之現有療法。在又一實施 162775.doc 201249841 例中’投與化合物A具有協同作用,使得患者能夠減少劑 量,或改變時間進程,或以其他方式修飾左旋多巴療法之 現有投藥療程。因此,本發明之一個態樣包括治療左旋多 巴誘導之運動障礙而不抑制左旋多巴之抗帕金森氏病活 性。另一態樣包括阻滯帕金森氏病進展以便提供較低劑量 之左旋多巴或較遲的投藥時間進程,由此延遲左旋多巴誘 導之運動障礙的發作。化合物A減弱異常不自主運動及左 旋多巴誘導之運動障礙。該等構成本發明之另一態樣。本 發明之又一態樣包括減輕左旋多巴誘導之運動障礙而不妨 礙左旋多巴對運動缺陷之效應。化合物A具有神經保護作 用。 本發明之一個態樣包括用於製造化合物A之新穎合成方 法。另外,本發明包括一或多種用於該合成方法中之新穎 中間體》. 本文將闡述本發明之其他態樣及實施例。本發明之範嘴 包括態樣、實施例及優選物之組合。 【實施方式】 定義 以下定義意欲明確(但不限於)所定義術語。若本文所用 之特定術語未明確加以定義,則該術語不應理解為不確 疋。相反’術s吾在其接受意義内使用。 根據所用命名約定,3_環丙基羰基_3,6二氮雜雙環 [3.1.1]庚烷亦可稱作3,6_二氮雜雙環[311]庚烷_3_基(環 基)甲酮,或可能稱作又一些化學名稱。命名約定之選= 162775.doc 201249841 應不影響本發明之範疇。如本文所述,化合物之結構係:Disease, one or more of abnormal involuntary movements, dyskinesias, and levodopa-induced dyskinesias. In one embodiment, a patient in need is undergoing an existing therapy including a levodopa administration regimen. In yet another embodiment 162775.doc 201249841, the administration of Compound A has a synergistic effect such that the patient is able to reduce the dosage, or change the time course, or otherwise modify the existing administration regimen of levodopa therapy. Thus, one aspect of the invention includes the treatment of levodopa-induced dyskinesia without inhibiting the anti-Parkinsonian activity of levodopa. Another aspect includes retarding the progression of Parkinson's disease to provide a lower dose of levodopa or a later dosing time course, thereby delaying the onset of levodopa-induced dyskinesia. Compound A attenuates abnormal involuntary movements and levodopa-induced dyskinesia. These constitute another aspect of the invention. Yet another aspect of the invention includes alleviating levodopa-induced dyskinesia without hindering the effect of levodopa on motor deficits. Compound A has a neuroprotective effect. One aspect of the invention includes a novel synthetic method for the manufacture of Compound A. Additionally, the invention includes one or more novel intermediates for use in the method of synthesis. Other aspects and embodiments of the invention are set forth herein. The mouthpiece of the present invention includes combinations of aspects, examples, and preferences. [Embodiment] Definitions The following definitions are intended to be clear (but not limited to) defined terms. If a specific term used herein is not explicitly defined, the term should not be construed as inaccurate. Instead, the surgery is used within its accepted meaning. Depending on the naming convention used, 3_cyclopropylcarbonyl-3,6-diazabicyclo[3.1.1]heptane may also be referred to as 3,6-diazabicyclo[311]heptane-3-yl (cyclic group). Ketone, or may be referred to as some chemical name. The choice of naming convention = 162775.doc 201249841 should not affect the scope of the invention. As described herein, the structure of the compound is:

,且為便於參照,在本文中亦可稱作化合物Α。 本文所用片語「本發明化合物」係指3-環丙基羰基-3,6-二氣雜雙環[3.1.1]庚烷或其酸加成鹽。酸係選自氫氯酸、 對甲笨磺酸、L-天冬胺酸、馬來酸、L_麩胺酸、丨_羥基_2_ 萘甲酸(即,經萘曱酸鹽)、富馬酸、半乳糖二酸、馬尿 酸、L-扁桃酸、琥珀酸、己二酸或(+)_樟腦酸。在較佳實 施例中’酸加成鹽係對曱苯磺酸鹽、馬來酸鹽、半乳糖二 酸鹽、苯曱酸鹽、馬尿酸鹽、羥萘曱酸鹽或(+)_樟腦酸 鹽。在又一些更佳實施例中,鹽係半乳糖二酸鹽、苯甲酸 鹽、馬尿酸鹽或羥萘甲酸鹽。 片語「本發明化合物」包括水合或溶劑化鹽形式。 此外,根據上下文,將易於明瞭,本文所用術語「化合And for ease of reference, it may also be referred to herein as a compound hydrazine. The phrase "compound of the present invention" as used herein means 3-cyclopropylcarbonyl-3,6-dioxabicyclo[3.1.1]heptane or an acid addition salt thereof. The acid is selected from the group consisting of hydrochloric acid, p-dosyl sulfonic acid, L-aspartic acid, maleic acid, L-glutamic acid, 丨_hydroxy_2_naphthoic acid (ie, naphthoic acidate), Fuma Acid, galactosedioic acid, hippuric acid, L-mandelic acid, succinic acid, adipic acid or (+)-camphoric acid. In a preferred embodiment, the 'acid addition salt is a p-toluenesulfonate, a maleate, a galactate, a benzoate, a hippurate, a hydroxynaphthoate or a (+) 樟 camphor Acid salt. In still other preferred embodiments, the salt is a galactose salt, a benzoate salt, a hippurate salt or a hydroxynaphthoate salt. The phrase "compound of the invention" includes hydrated or solvated salt forms. In addition, depending on the context, it will be easy to understand, and the term "combination" is used herein.

形劑或鹽形式。Form or salt form.

現一定程度之穩定 162775.doc 201249841 性以便使得其適於製造及商業化目的。 本文所用術語「有效量」、「治療量」或「有效劑量」係 指活性成份足以引發期望藥理或治療效應、由此可有效預 防或治療病症的量。病症之預防可表現為延遲或預防病症 之進展、以及延遲或預防與病症相關之症狀的發作。病症 之治療可表現為減少或消除症狀、抑制或逆轉病症之進 展、以及使患者幸福感之任一其他因素。 有效劑量可端視諸如以下等因素而定:患者之病況、病 症之症狀的嚴重程度、及投與醫藥組合物之方式。通常, 欲以有效劑量投與之化合物需要以小於5 mg/kg患者體重 之量投與》該等化合物經常可以小於約i mg/kg患者體重 至小於約100 Pg/kg患者體重、且偶爾介於約10 pg/kg至小 於約1〇〇 Kg/kg患者體重之間之量投與。上述有效劑量通常 代表以單一劑量形式、或以經24 h時段投與之一或多個劑 量形式投與之量。對於人類患者而言,化合物之有效劑量 可旎需要以至少約1 mg/24 hr/患者、但不超過約1000 mg/24 hr/患者、且經常不超過約5〇〇 μ hr/患者之量投 與化。物。潛在劑量可在500 Hg至2 mg範圍内,以游離鹼 等效物形式。 本文所用片言吾「實質上結晶」包括大於2〇%、較佳大於 30/。且更佳大於40% (例如,大於5〇%、6〇%、7〇0/〇、 8〇%或9〇%中之任-者)之結晶。 如本文所定義術語「穩定性」包括化學穩定性及固態穩 Ϊ _甘_. rl·* L1 -» 五「 ^ δ〇 化學穩定性」包括以分離形式、或以與 162775.doc 201249841 ㈣物开之載劑、稀釋劑、賦形劑或佐劑混合提供之 類以口服劑型,例如錠劑、膠囊或諸如此 )在私準儲存條件下儲存本發明鹽且 :不:顯的潛能,且片語「固態穩定性」包括= 與醫樂上可接受之載劑、稀釋劑、賦形劑或佐劑 混。提供之固體調配物形式(例如以口服劑型,例如錠 劑、膠囊或諸如此類)在標準儲存條件下儲存本發明鹽且 固體轉變(例如結晶、重結晶、固態相轉變、水合、脫 水、溶劑化或去溶劑化)程度不明顯的潛能。 「標準儲存條件」之實例包括以下t之-或多者:溫度 介於-80C與50。(:之間,較佳介於〇t:與赋之間且更佳環 i見皿度,例如15C至30°C ;壓力介於o.i巴與2巴之間,較 佳大氣壓;相對濕度介於5%與95%之間,較佳1〇%至 60〇/〇 ;且於460 1UX或更小之uv/可見光中暴露延長時段, 例如大於或等於6個月。在該等條件下,可發現本發明之 鹽(若適當)的5%以下、更佳2%以下且尤其1%以下化學降 解或分解或固態轉變。熟習此項技術者應瞭解,上述溫 度、壓力及相對濕度之上限及下限代表標準儲存條件之極 端’且在標準儲存(例如,50°c之溫度及〇1巴之壓力)期間 將不經歷該等極端之某些組合。 化合物 本發明之一個實施例包括3-環丙基羰基_3,6_二氮雜雙環 [3.1.1]庚烷(式I)或其醫藥上可接受之鹽。 162775.doc 201249841 ΗThere is now a certain degree of stability to make it suitable for manufacturing and commercial purposes. The terms "effective amount", "therapeutic amount" or "effective amount" as used herein mean an amount of the active ingredient which is sufficient to elicit the desired pharmacological or therapeutic effect, thereby effectively preventing or treating the condition. Prevention of a condition can be manifested by delaying or preventing the progression of the condition, as well as delaying or preventing the onset of symptoms associated with the condition. Treatment of a condition can be manifested as any reduction or elimination of symptoms, inhibition or reversal of the progression of the condition, and any other factor that contributes to the patient's well-being. The effective dose may depend on factors such as the condition of the patient, the severity of the symptoms of the condition, and the manner in which the pharmaceutical composition is administered. In general, the compound to be administered in an effective dose needs to be administered in an amount of less than 5 mg/kg of the patient's body weight. These compounds can often be less than about i mg/kg of patient body weight to less than about 100 Pg/kg of patient body weight, and occasionally Administration is carried out in an amount between about 10 pg/kg to less than about 1 Kg/kg of patient body weight. The above effective doses generally represent amounts administered in a single dose or in one or more doses administered over a 24 hour period. For human patients, an effective dose of the compound may be required to be at least about 1 mg/24 hr/patient, but no more than about 1000 mg/24 hr/patient, and often no more than about 5 〇〇μ hr/patient. Investment. Things. Potential doses can range from 500 Hg to 2 mg in the form of the free base equivalent. As used herein, the phrase "substantially crystallized" includes greater than 2%, preferably greater than 30%. More preferably, it is more than 40% (for example, greater than 5%, 6%, 7%, 8%, or 9%). The term "stability" as defined herein includes chemical stability and solid state stability. _ Gan_. rl·* L1 -» Five "^ δ 〇 chemical stability" is included in isolated form, or in combination with 162775.doc 201249841 (four) Mixing the carrier, diluent, excipient or adjuvant to provide an oral dosage form, such as a tablet, capsule or the like, for storing the salt of the invention under private storage conditions and without: significant potential, and The phrase "solid state stability" includes = mixed with medically acceptable carriers, diluents, excipients or adjuvants. Providing a solid formulation form (for example, in an oral dosage form such as a troche, capsule or the like) for storage of a salt of the invention under standard storage conditions and solids transformation (eg, crystallization, recrystallization, solid phase transformation, hydration, dehydration, solvation or Desolvation) The potential for an insignificant degree. Examples of "standard storage conditions" include - or more of the following: temperatures between -80C and 50. (between, preferably between 〇t: and Fu and better ring i, such as 15C to 30 ° C; pressure between oi and 2 bar, preferably atmospheric pressure; relative humidity between Between 5% and 95%, preferably 1% to 60 〇/〇; and an extended period of exposure in uv 1 / uv or less visible light, for example greater than or equal to 6 months. Under these conditions, It is found that the salt of the present invention (if appropriate) is less than 5%, more preferably less than 2% and especially less than 1% chemically degraded or decomposed or solid state converted. Those skilled in the art should be aware of the above upper limits of temperature, pressure and relative humidity and The lower limit represents the extreme of standard storage conditions and will not undergo some combination of such extremes during standard storage (eg, a temperature of 50 ° C and a pressure of 1 bar). Compounds One embodiment of the invention includes a 3-ring Propylcarbonyl-3,6-diazabicyclo[3.1.1]heptane (Formula I) or a pharmaceutically acceptable salt thereof. 162775.doc 201249841 Η

式ι 化合物A 一個實施例包括3-環丙基羰基-3,6-二氮雜雙環[3丄^庚 院或其醫藥上可接受之鹽的用途,其用於製造藥劑。 本發明之一個實施例包括用於治療或預防多種病症及功 能障礙之方法,其包含向需要該治療之哺乳動物投與治療 有效量之3-環丙基羰基_3,6_二氮雜雙環[3丄丨]庚烷或其醫 藥上可接受之鹽。 更具體而言’該病症或功能障礙可選自由Cns病症(包 括AIMS及LID)、或本文進一步詳細闡述之其他病症組成 之群。本發明之另一實施例包括用作診斷試劑且用於如本 文所述受體結合研究中之化合物。 本發明之一個實施例包括包含治療有效量之3_環丙基幾 基-3,6·二氮雜雙環[3· 1.1]庚烷或其醫藥上可接受之鹽及一 或多種醫藥上可接受之載劑的醫藥組合物。本發明之另一 實施例包括本發明醫藥組合物之用途,其用於製造用於治 療中樞神經系統病症及功能障礙之藥劑。參照實例中之任 一者,本發明之另一實施例包括3-環丙基羰基_3,6_二氮雜 雙環[3.1.1]庚烷或其醫藥上可接受之鹽。本發明之另—實 施例包括3-環丙基羰基-3,6-二氮雜雙環[3.1.1]庚烷或其醫 藥上可接受之鹽’其用作活性治療物質。本發明之另—實 162775.doc •10· 201249841 鉍例包括3-環丙基羰基_3,6_二氮雜雙環[3丨u庚烷或其醫 藥上可接觉之鹽,其用於調節有需要之個體之NNR。本發 月之另貫施例包括3-環丙基羰基_3,6_二氮雜雙環[3.1.1] 庚烧或其醫藥上可接受之鹽,其用於治療或預防由一或多 種NNR ’丨導之病況或病症。本發明之另一實施例包括3 _環 丙基羰基_3,6·二氮雜雙環[3.1.1]庚烷或其醫藥上可接受之 鹽的用途,其帛於製造用於調節有需要之個體之一或多種 _R的藥劑。本發明之另-實施例包括3-環丙基羰基_3,6_ 二氣雜雙環[3.1.1]庚烷或其醫藥上可接受之鹽的用途,其 用於製k用於治療或預防由_或多種顺尺介導之病況或病 症的藥劑本發明之另一實施例包括經由投與3 ·環丙基叛 基-3,6-二氮雜雙環[3.M]庚烷或其醫藥上可接受之鹽調節 有需要之個體之一或多的方法。 除非另有說明,否則本文所描述之結構亦意欲包括僅在 或多個同位素畐集原子存在方面不同之化合物。舉例而 :’具4有本發明結構、只是由氘或氣替代氫原子、或由 13C或14C替代碳原子、或由15N替代氮原子、或用17〇或18〇 替代氧化子之化合物在本發明範嗨内。該等同位素標記之 化合物可用作研究或診斷工具。 如本文所述,本發明包括具體代表性化合物,本文特別 對其進行鑑別。本發明化合物可藉由各種方法(包括眾所 周知之標準合纟方法)製得。了文闡$闡釋性—般合成方 法且隨後在工作實例中製備本發明之具體化合物。 在下述所有實例中,若需要,根據合成化學之一般原則 162775.doc •11· 201249841 針對敏感性或反應性基團採用保護基團。根據有機合成之 才示準方法操縱保護基團(T. W. Green及P. G. M. Wuts, ecting Groups in Organic Synthesis > ^ 3 M ' JohnFormula ι Compound A One embodiment includes the use of 3-cyclopropylcarbonyl-3,6-diazabicyclo[3丄^^, or a pharmaceutically acceptable salt thereof, for the manufacture of a medicament. One embodiment of the invention includes a method for treating or preventing a plurality of conditions and dysfunction comprising administering to a mammal in need of such treatment a therapeutically effective amount of 3-cyclopropylcarbonyl-3,6-diazabicyclo [3丄丨]heptane or a pharmaceutically acceptable salt thereof. More specifically, the condition or dysfunction may be selected from a group of Cns disorders (including AIMS and LID), or other disorders as further detailed herein. Another embodiment of the invention encompasses use as a diagnostic reagent and for a compound in a receptor binding assay as described herein. An embodiment of the invention comprises a therapeutically effective amount of 3-cyclopropyl-based-3,6-diazabicyclo[3·1.1]heptane or a pharmaceutically acceptable salt thereof and one or more pharmaceutically acceptable salts A pharmaceutical composition that accepts a carrier. Another embodiment of the invention encompasses the use of a pharmaceutical composition of the invention for the manufacture of a medicament for the treatment of disorders and dysfunctions of the central nervous system. According to any one of the examples, another embodiment of the present invention includes 3-cyclopropylcarbonyl-3,6-diazabicyclo[3.1.1]heptane or a pharmaceutically acceptable salt thereof. Another embodiment of the present invention includes 3-cyclopropylcarbonyl-3,6-diazabicyclo[3.1.1]heptane or a pharmaceutically acceptable salt thereof, which is used as an active therapeutic substance. Another example of the present invention is 162775.doc •10·201249841 Examples include 3-cyclopropylcarbonyl-3,6-diazabicyclo[3丨uheptane or a pharmaceutically acceptable salt thereof, which is used for Adjust the NNR of the individual in need. Further examples of this month include 3-cyclopropylcarbonyl-3-3,6-diazabicyclo[3.1.1]heptane or a pharmaceutically acceptable salt thereof for use in the treatment or prevention by one or more NNR's condition or condition. Another embodiment of the invention includes the use of 3- _cyclopropylcarbonyl-3,6-diazabicyclo[3.1.1]heptane or a pharmaceutically acceptable salt thereof, which is suitable for use in conditioning for conditioning One or more of the individual's agents. Further embodiments of the invention include the use of 3-cyclopropylcarbonyl-3,6-dioxabicyclo[3.1.1]heptane or a pharmaceutically acceptable salt thereof for the treatment or prevention of Agents of a condition or disorder mediated by _ or multiple cissicles. Another embodiment of the invention comprises administering 3·cyclopropylheptyl-3,6-diazabicyclo[3.M]heptane or A pharmaceutically acceptable salt modulates one or more of the individuals in need. Unless otherwise indicated, structures depicted herein are also intended to include compounds that differ only in the presence of one or more isotope collection atoms. For example: 'A compound having the structure of the present invention, replacing hydrogen atoms by helium or gas, or replacing a carbon atom by 13C or 14C, or replacing a nitrogen atom with 15N, or replacing an oxidant with 17〇 or 18〇 Within the scope of the invention. Such isotopically labeled compounds can be used as research or diagnostic tools. As described herein, the invention includes specific representative compounds, which are specifically identified herein. The compounds of the present invention can be prepared by a variety of methods, including standard well-known methods. The specific synthetic compounds of the present invention are prepared in the working examples by way of illustration. In all of the following examples, if necessary, according to the general principles of synthetic chemistry 162775.doc •11·201249841 Protective groups are employed for sensitive or reactive groups. The protecting group is manipulated according to the method of organic synthesis (T. W. Green and P. G. M. Wuts, ecting Groups in Organic Synthesis > ^ 3 M ' John

Wiley & Sons,New York (1999))。在化合物合成之方便階 段使用對彼等熟習此項技術者顯而易見之方法去除該等基 園。方法以及反應條件及其執行次序之選擇應與本發明化 合物之製備一致。 本發明亦提供可用作中間體之化合物之合成方法。 一般合成方法 可合成化合物A之方式可有所變化。在一種方法中,化 合物A可經由以下製備:6位氮原子經保護(防止與醯化劑 反應)之3,6-二氮雜雙環[3.1.1]庚烷與適宜環丙基羰基衍生 物(例如環丙基甲醯氣)偶合,之後去除保護基團(通常利用 酸)。環丙基甲醯氯可藉由尤其用亞硫醯氣或草醯氣等試 劑處理環丙基曱酸來製備。 適宜保護之3,6-二氮雜雙環[3.1.1]庚烷、6·(第三丁氧基 幾基)-3,6-一氮雜雙環[3.1.1]庚烧的合成揭示於頒予pjnna 專人之WO 2005/108402中。(關於該合成教示以引用方式 併入)。彼等熟習有機合成技術者應瞭解,其他適宜保護 之3,6-二氮雜雙環[3.1.1]庚烧亦可用於製備化合物a (參 見’例如 ’ T· W. Greene 及 P.G. M. Wuts,Protective Groups in Organic Synthesis,第 3版,John Wiley & Sons, NewWiley & Sons, New York (1999)). These libraries are removed at a convenient stage of compound synthesis using methods apparent to those skilled in the art. The method and the choice of reaction conditions and their order of execution should be consistent with the preparation of the compounds of the invention. The present invention also provides a method of synthesizing a compound useful as an intermediate. General Synthetic Methods The manner in which Compound A can be synthesized can vary. In one method, Compound A can be prepared by protecting 3,6-diazabicyclo[3.1.1]heptane with a suitable cyclopropylcarbonyl derivative protected at the 6-position nitrogen atom (preventing reaction with a oximation agent). Coupling (e.g., cyclopropylmethylhydrazine), followed by removal of the protecting group (usually with an acid). Cyclopropylguanidinium chloride can be prepared by treating cyclopropyl decanoic acid, in particular, with a reagent such as sulfoxide or oxalate. Synthesis of a suitably protected 3,6-diazabicyclo[3.1.1]heptane, 6·(t-butoxymethyl)-3,6-azabicyclo[3.1.1]heptane is disclosed in Presented to WO 2005/108402 by pjnna. (About this synthetic teaching is incorporated by reference). Those skilled in the art of organic synthesis should be aware that other suitable 3,6-diazabicyclo[3.1.1] heptane can also be used to prepare compound a (see 'T' W. Greene and PGM Wuts, Protective Groups in Organic Synthesis, 3rd edition, John Wiley & Sons, New

York (1999))。 製備化合物A之另一方式係使適宜6-保護之·3,6·二氮雜 162775.doc 201249841 雙環[3.1.1]庚烷與環丙基甲酸偶合,之後去除保護基團。 此方法通常需要使用適宜活化劑,例如N,N’-二環己基碳 化二亞胺(DCC)、六氟磷酸(苯并三唑-1-基氧基)叁(二曱基 胺基)鎸(BOP)、六氟磷酸(苯并三唑-1-基氧基)三吼咯啶基 鱗(PyBOP)、六氟磷酸0(苯并三唑-1-基)-Ν,Ν,Ν',Ν1-雙(四 亞曱基)脲鏽鹽(HBPyU)、六氟磷酸0-(苯并三唑-1-基)-队队:^’,;^-四甲基脲鏽鹽(1^丁1;)、四氟硼酸0-(苯并三唑-1-基)-Ν,Ν,Ν·,Ν’-四甲基脲鏽鹽(TBTU)或(1-乙基-3-(3-二甲 基胺基丙基)碳化二亞胺)(EDCI)與1-羥基苯并三唑 (HOBt) 〇其他活化劑已為彼等熟習此項技術者所熟知,例 如,參見Kiso 及 Yajima,Peptides,第 39-91 頁,Academic Press, San Diego, CA (1995) 0 反應圓1:化合物A之合成 cioCs^/N^coa 1York (1999)). Another way of preparing Compound A is by coupling a suitable 6-protected 3,6-diazepine 162775.doc 201249841 bicyclo[3.1.1]heptane with cyclopropylcarboxylic acid, followed by removal of the protecting group. This method usually requires the use of a suitable activator such as N,N'-dicyclohexylcarbodiimide (DCC), hexafluorophosphate (benzotriazol-1-yloxy)indole (didecylamino) hydrazine. (BOP), hexafluorophosphate (benzotriazol-1-yloxy)triazolidine-based scale (PyBOP), hexafluorophosphate 0 (benzotriazol-1-yl)-oxime, oxime, Ν' , Ν1-bis(tetradecyl)urea rust salt (HBPyU), hexafluorophosphate 0-(benzotriazol-1-yl)-team: ^',;^-tetramethylurea rust salt (1 ^丁1;), tetrafluoroboric acid 0-(benzotriazol-1-yl)-indole, hydrazine, hydrazine, Ν'-tetramethylurea rust salt (TBTU) or (1-ethyl-3- (3-Dimethylaminopropyl)carbodiimide) (EDCI) and 1-hydroxybenzotriazole (HOBt) Other activators are well known to those skilled in the art, for example, see Kiso And Yajima, Peptides, pp. 39-91, Academic Press, San Diego, CA (1995) 0 Reaction circle 1: Synthesis of compound A cioCs^/N^coa 1

BraBra

BnOH 醚 Na 日 H4 thf/ch3ohBnOH ether Na day H4 thf/ch3oh

Bn〇2〇Bn〇2〇

BnMH2, dmfBnMH2, dmf

Bn—N- i-C〇2Bn ^C〇2Bn 4Bn-N- i-C〇2Bn ^C〇2Bn 4

Msa TEA, DCM 10% Ρύ/C, HC02NH4 C2H5OH yV Br Bf CO^Bn 3Msa TEA, DCM 10% Ρύ/C, HC02NH4 C2H5OH yV Br Bf CO^Bn 3

化合物ACompound A

NaH, NMP Bn,NaH, NMP Bn,

Bn=苄基;Ms=甲烷磺醯基;DMF=二曱基甲醯胺;THF=四 氫呋喃;TEA =三乙胺;DCM =二氣甲烷;NMP=N-曱基-2-吡 洛咬酮。 或者,且有利地,化合物A可使用類似於反應圖1中所示 反應順序製得。此方法涉及環丙基曱醯胺之陰離子與雙- 162775.doc -13- 201249841 親電子劑(例如中間體6)反應之閉環過程,從而與前述合成 (例如’ WO 2011/071758)相比減少合成的總體長度。合成 化合物A之此方法的具體實例於實例部分令給出且概述於 反應圖1中。然而,用於完成此方法之轉變之試劑可有所 變化。舉例而言,酯化反應可使用多種醇,從而提供多種 二酯產物(實例為二酯3)。特定二酯之物理性質可提供純化 或操作方面之優點。同樣,多種胺及溶劑可用於4員(氮雜 環丁烷)環之環閉合,特定胺/溶劑混合物提供反應性或產 物純度(包括立體化學純度)方面之優點。可藉由存於多種 溶劑中之多種試劑(例如,硼氫化物及鋁氫化物試劑)完成 一烧基氮雜環丁基-2,4-二甲酸酯(例如,中間體4)至相應 二醇(例如二醇5)之還原。重要的是,選擇保存氮雜環丁烷 環周圍之順式相對立體化學的還原試劑。可藉由彼等熟習 此項技術者已知之多種試劑完成二醇至適宜雙_親電子劑 之轉化。將醇轉化為相應齒化物(例如,氣化物、溴化 物、碘化物)之反應、以及彼等將醇轉化為磺酸酯、磷酸 酯及諸如此類者均在化學文獻中眾所周知。亦可利用多種 試劑(包括多種用於產生陰離子之驗及多種溶劑)完成6員 (六氫。比咬)環與環丙基甲醯胺之閉合。最後,根據保護基 圈之性質’可W多種條件完減雜環丁基氮之去保護。 彼等熟習有機合成技術者應瞭解,有多種產生一或多個 原子經放射性同位素標記且適於各種用途之化合物續方 式。舉例而言,使用上文所列料化劑巾之_者使標 記之環丙基甲酸與6-(第:r 丁氯甚铛萁、 V乐一丁乳暴羰基)-3,6•二氮雜雙環 162775.doc 14 201249841 [3.1.1] 庚烷偶合將產生3_uc·標記之_環丙基羰基第三 丁氧基羰基二氮雜雙環[3.1.1]庚烷。如上文所述,隨 後去除第—丁氧基魏基保護基團將產生適用於正電子發射 斷層攝影術中之化合物。同樣,如上文所述3H_4Mc_標記 之環丙基甲酸與6_(第三丁氧基羰基)_3,6_二氮雜雙環 [3.1.1] 庚烷偶合、之後去除保護基團將產生同位素修飾之 化合物A,其適用於受體結合及機制研究或適用作替代治 療化合物。 如上文所提及,對於經口調配物而言,固體鹽形式由於 以優先方式呈現該等性質之趨勢而通常較佳;且在諸如3_ 環丙基羰基-3,6-二氮雜雙環[^^庚烷等鹼性藥物之情形 下’酸加成鹽經常係較佳鹽形式。 不同鹽形式賦予該等性質之能力變化顯著’且不可精確 地預測該等性質。舉例而言’ 一些鹽在環境溫度下係固 體,而其他鹽在環境溫度下係液體、黏性油或膠。此外, -些鹽形式在極端條件下對熱及光穩定且其他鹽形式在遠 f溫和條件下容易分解H用於醫藥組合物中之驗性 樂物之適宜酸加a盛形式的研發係高度不可預測之過程。 需要呈現改良性質(包括純度、穩定性、溶解性、及生物 利用度)之鹽形式。該等新穎鹽形式之優先特性包括彼等 可增加活性成份之製造及其調配成商業產品之簡易性或效 率者。最後,需要可增加活性成份之製造及其調配成商業 產品之簡易性或效率的該等鹽之穩定多晶型。 σ由’’·、I itb項技術者使用χ射線粉末繞射(XRPD)測定結 162775.doc •15· 201249841 晶程度(%)。亦可使用其他技術,例如固態NMR、FT-IR、 拉曼光譜、示差掃描熱量測定法(DSC)及微量熱法。對於 本發明化合物而言’已發現可產生呈實質上結晶形式之 P场〇 該等結晶鹽中之若干證實足以確立其在醫藥製劑生產中 之前景的穩定性。可以多種方式證實該穩定性。可藉由動 態蒸氣吸附(DVS)分析獲得並釋放大氣水分之傾向。可藉 由將固體鹽於40°C/75。/。RH下儲存高達8天,且隨後藉由重 量、顯微鏡下之外觀及XRPD各自重新檢驗來研究對高溫 及濕度之穩定性。 多晶型 本發明化合物可以一種以上形式結晶,即稱作多態性之 特徵,且該等多晶形式(「多晶型」)屬於本發明之範疇。 多態性通常可作為溫度、壓力或二者變化之響應發生。多 態性亦可因結晶過程之變化而產生。多晶型之不同之處可 在於業内已知之各種物理特性(例如’ XRPD圖案(繞射 圖)、於不同溶劑中之溶解度及熔點)。 本發明包括3·環丙基羰基_3,6•二氮雜雙環[3丄丨]庚烷之 鹽形式之不同多晶型(包括鹽之水合物及溶劑合物)。該等 多晶型由其X射線粉末繞射(XRPD)圖案(繞射圖)表徵。 如上所述,3_環丙基羰基·3,6·二氮雜雙環[3.1.1]庚烷之 鹽形式可以溶劑化(例如水合)以及非溶劑化形式存在。本 發明涵蓋所有該等形式。 本發明亦包括同位素標記之化合物,其中-或多個原子 I62775.doc 201249841 經原子量或質量數不同於自然界中通常所發現之原子量或 質量數的原子替代。可納入本發明化合物中之同位素之實 例包括包括氫、碳、氮及氧之同位素,例如2H ' 4、 im丨%及”〇。該等同位素標記化合物可用作 研究或診斷工具。 醫藥组合物 儘管可以體活性化學物質形式投與本發明化合物,但較 佳以醫藥組合物或調配物形式投與該化合物。因此,本發 明之-個態樣包括包含本發明化合物及—或多種醫藥上可 接受之載劑、稀釋劑或賦形劑的醫藥組合物。本發明之另 -態樣提供製備醫藥組合物之方法,纟包含混合本發明化 口物與-或多種醫藥上可接受之載劑、稀釋劑或賦形劑。 投與本發明化合物之方式可有所變化。較佳經口投與本 發明化合物。用於經口投與之較佳醫藥組合物包括錠劑、 谬囊、膜衣錠'«、溶液及料液。本發明之醫藥組合 物可以經改良釋放劑型(例如延時釋放㈣及膠㈣配物) 提供。 該等醫藥組合物亦可經由、、:^鉍„ .. ▲田庄射,即靜脈内、肌内、皮 下、腹膜腔内、動脈内、勒内及腦室内投與。靜脈内投與 係較佳主射方法。適用於注射之載劑已為彼等熟習此項技 術者所熟知且包括5%右旋糖、、交、放 , 也 疋糖/合液、鹽水及磷酸鹽緩衝鹽 水。 亦可使用其他方式投與調配物 腸投與之調配物(例如栓劑)已為彼等 ’例如直腸投與。用於直 熟習此項技術者所熟 162775.doc 201249841 知m合物亦可藉由吸人’例如以氣溶膠形式;局 °p例如以洗液形4 ;經皮,例如使用經皮貼劑(例如, 藉由使用可自N〇Vartis and仙公司靖得之技術);藉由粉 末注射;或藉由經頰、舌下或鼻内吸收投與。 可以單位劑型或多個或亞單位劑量調配醫藥組合物。 本文所述醫藥組合物之投與可間歇、或以逐漸、連續、 怪定或受控速率投與。可將醫藥組合物投與溫血動物,例 如哺乳動物,例如小鼠、大鼠、猶、兔、狗、豬、牛或狼 子;但有利地投與人類。另外,每天投與醫藥組合物之白 天時間及次數可有所變化。 本發明化合物可用於治療多種病症及病況,且因此可與 多種適用於治療或預防彼等病纟或病況之其他治療劑組合 使用。因此,本發明之一個實施例包括組合投與本發明化 合物與其他治療化合物。舉例而言,本發明化合物可與以 下物質組合使用:其他NNR配體(例如伐尼克蘭 (varenicline))、抗氧化劑(例如自由基清除劑)、抗細菌劑 (例如青黴素(penicillin)抗生素)、抗病毒劑(例如核苷類似 物,如齊多夫定(zidovudine)及阿昔洛維(acyclovir))、抗 凝血劑(例如殺鼠靈(warfarin))、抗炎劑(例如NSAID)、解 熱藥、鎮痛藥、麻醉藥(例如用於外科手術)、乙醯膽鹼酯 酶抑制劑(例如杜尼匹次(d〇nepezii)及加蘭他敏 (galantamine))、抗精神病藥(例如氟哌啶醇(hal〇perid〇1)、 氯氮平(clozapine) '奥蘭紫平(〇ianzapjne)、及喧硫平 (quetiapine))、免疫抑制劑(例如環胞素(cyci〇sp〇rine)及胺 162775.doc •18· 201249841 曱蝶呤(methotrexate))、神經保護劑(例如A2a抑制劑及咖 啡因(caffeine))、血腦屏障渗透增強劑、類固醇(例如類固 醇激素)、腎上腺皮質類固醇(例如地塞米松 (dexamethasone)、潑尼松(predisone)及氫化可的松 (hydrocortisone))、維生素、礦物質、營養製品、興奮劑 (例如米帕明(imipramine)、敗西;丁(fluoxetine)、帕羅西 ί丁 (paroxetine)、依他普命(escitalopram)、舍曲林(sertraline)、 文拉法辛(venlafaxine)、及杜洛西;丁(duloxetine))、安眠藥 (例如阿普唾侖(alprazolam)及丁螺環_ (buspirone))、抗痙 攣劑(例如苯妥英(phenytoin)及加巴喷丁(gabapentin))、血 管擴張劑(例如0底。坐嗪(prazosin)及昔多芬(sildenafil))、情 緒穩定劑(例如丙戊酸鹽(valproate)及阿立'»底°坐 (aripiprazole))、抗癌藥物(例如抗增殖劑)、抗高血壓藥劑 (例如阿替洛爾(atenolol)、可樂定(clonidine)、氨氣地平 (amlopidine)、異搏定(verapamil)、及奥美沙坦(olmesartan))、 輕满藥、大便軟化劑、利尿劑(例如吱塞米(furosemide))、 解痙攣藥(例如雙環維林(dicyclomine))、抗運動障礙劑及 抗潰瘍藥劑(例如伊索派°坐(esomeprazole)) ^本發明化合物 之一種較佳用途係治療及預防帕金森氏病、AIM及LID, 且因此,本發明化合物可與用於治療帕金森氏病、AIM及 LID之醫藥劑組合使用。該等藥劑包括NNR激動劑(α4β2、 α7等)、多巴胺前體(例如左旋多巴-卡比多巴(carbidopa)、 左旋多巴-苄絲肼(benserazide)、及複方左旋多巴(duodopa))、 多巴胺激動劑(例如溴隱亭(bromocriptine)、卡麥角林 162775.doc 19 201249841 (cabergoline)、利舒腺(lisuride)、硫丙麥角林(perg〇iide)、 普拉克索(pramipexole)、羅匹尼羅(popinirole)、他利克索 (talipexole)、 羅替高灯(rotigotine)、及阿撲嗎。非 (apomorphine))、多巴羧化酶抑制劑、MAO-B抑制劑(例如 司來吉蘭(selegiline)、雷沙吉蘭(rasag〇iine)、及沙非醯胺 (safinamide))、COMT抑制劑(例如諾康停(entacap〇ne)及托 卡朋(tolcapone))、抗麵胺酸能藥劑(例如金剛烧胺 (amantadine))、抗膽鹼激導性藥劑(例如苯海索 (trihexyphenidyl)、苯紮托品(benztropine)、及比派立登 (biperiden))、抗癡呆藥劑(例如利斯的明(rivastigmine)、 杜尼匹次(donepezil)、加蘭他敏、及美滿町(memantine))、 抗精神病藥劑(例如唾硫平(qUetiapine)、氯氮平 (clozapine)、及利培酮(resperidone)) '抗癲癇劑(例如唑尼 沙胺(zonisamide))、去甲腎上腺素前體(例如屈昔多巴 (droxidopa))、mGluR4 激動劑、mGluR5 抑制劑、 5HT1A/1B激動劑、5HT2A #抗劑/反向激動劑、類鴻片物 質拮抗劑(μ、δ、κ) '及AMPA受體阻斷劑。醫藥活性劑之 該組合可-起或分開投與,且當分開投與時,彳同時或依 序、或依任何次序進行投藥》應選擇投與化合物或試劑之 量及相對時間,點’以達成所期望治療效應。本發明化合物 與其他治療劑之組合投與可藉由以以下形式同時投與來組 合.⑴整體醫藥組合物’丨包括兩種化合物;或⑺單獨 醫藥組合物’其各自包括化合物中之一者。或者可以依 序方式分開投與該組合,其中首先投與—種治療劑,再投 162775.doc •20· 201249841 與另一第二治療劑。該依序投藥法可在接近時間點或在距 離一段時間後投藥。 治療方法 3-環丙基羰基-3,6-二氮雜雙環[3.1.1]庚烷、其醫藥上可 接受之鹽、或含有其之醫藥組合物可用於預防或治療已提 出或顯示其他類型之菸鹼化合物可用作治療劑的各種病況 或病症,例如CNS病症(包括神經退化症)、發炎、與細菌 及/或病毒感染相關之發炎反應、疼痛、糖尿病、代謝症 候群、自體免疫性病症、皮膚病況、成癮症、肥胖症或本 文進一步詳述之其他病症。此化合物亦可在受體結合研究 (活體外及活體内)中用作診斷劑。該等治療及其他教示闡 述於(例如)本文先前列舉之參考文獻中,包括wiUiams等 人,Drwg iVews 尸eripec· 7(4): 205 (1994),Arneric等人, CiVS Drwg 1(1): 1-26 (1995),Arneric等人,五χρ· /πναί· DrMgj 5(1): 79-100 (1996),Yang 等人,dcia P/zarmiico/· 57«. 30(6): 740-751 (2009),Bencherif等人,丄 Pharmacol. Exp. Ther. 279: 1413 (1996), Lippiello^ A > J. ·Ελτ?· 77zer. 279: 1422 (1996),Damaj等人,《/. P/zizrmaco/. 77ier. 291: 390 (1999) ; Chiari 等人,Bn=benzyl; Ms=methanesulfonyl; DMF=dimercaptocaramine; THF=tetrahydrofuran; TEA=triethylamine; DCM=di-methane; NMP=N-mercapto-2-pyrrolidone . Alternatively, and advantageously, Compound A can be prepared using a reaction sequence similar to that shown in Reaction Scheme 1. This method involves a ring closure process in which the anion of cyclopropylguanamine reacts with a bis-162775.doc-13-201249841 electrophile (e.g., intermediate 6) to reduce the synthesis (e.g., 'WO 2011/071758) The overall length of the synthesis. Specific examples of this method of synthesizing Compound A are given in the Examples section and are summarized in Reaction Scheme 1. However, the reagents used to accomplish the conversion of this method may vary. For example, a plurality of alcohols can be used in the esterification reaction to provide a plurality of diester products (examples are diesters 3). The physical properties of a particular diester can provide advantages in purification or handling. Similarly, a wide variety of amines and solvents can be used for ring closure of a 4-membered (azetidine) ring, and specific amine/solvent mixtures provide advantages in reactivity or product purity, including stereochemical purity. The monoalkyl azepanyl-2,4-dicarboxylate (for example, intermediate 4) can be completed by a plurality of reagents (for example, borohydride and aluminum hydride reagent) present in a plurality of solvents. Reduction of a diol such as diol 5. Importantly, a reducing reagent that preserves the cis relative stereochemistry around the azetidin ring is selected. The conversion of the diol to a suitable bis-electrophile can be accomplished by a variety of reagents known to those skilled in the art. The conversion of alcohols to the corresponding dentates (e.g., vapors, bromides, iodides), and their conversion of alcohols to sulfonates, phosphates, and the like are well known in the chemical literature. The closure of a 6-member (hexahydrogen-to-bite) ring with cyclopropylcarbamamine can also be accomplished using a variety of reagents, including a variety of reagents for the generation of anions and a variety of solvents. Finally, the deprotection of the heterocyclic butyl nitrogen can be reduced under a variety of conditions depending on the nature of the protective ring. Those skilled in the art of organic synthesis should be aware that there are a number of compound continuations that produce one or more atoms that are radiolabeled and suitable for a variety of uses. For example, using the granules listed above, the labeled cyclopropyl formic acid and 6-(the:r-butyl chloride, V-butyl emulsifiable carbonyl)-3,6•2 Azabicyclohexene 162775.doc 14 201249841 [3.1.1] Heptane coupling will give 3_uc·labeled _cyclopropylcarbonylt-butoxycarbonyldiazabicyclo[3.1.1]heptane. Subsequent removal of the first butoxywei group protecting group will result in a compound suitable for use in positron emission tomography, as described above. Similarly, the coupling of 3H_4Mc_labeled cyclopropyl formic acid with 6-(t-butoxycarbonyl)-3,6-diazabicyclo[3.1.1]heptane as described above, followed by removal of the protecting group will result in isotopic modification Compound A, which is suitable for receptor binding and mechanistic studies or as an alternative therapeutic compound. As mentioned above, for oral formulations, the solid salt form is generally preferred due to the tendency to exhibit such properties in a preferential manner; and in, for example, 3_cyclopropylcarbonyl-3,6-diazabicyclo[ In the case of an alkaline drug such as heptane, the acid addition salt is often in a preferred salt form. The ability of different salt forms to impart such properties varies significantly' and these properties are not accurately predictable. For example, some salts are solid at ambient temperatures, while others are liquid, viscous or glue at ambient temperatures. In addition, some salt forms are stable to heat and light under extreme conditions and other salt forms are easily decomposed under far f mild conditions. H is used in pharmaceutical compositions for the determination of suitable acid additions in the form of a suitable acid. Unpredictable process. Salt forms that exhibit improved properties including purity, stability, solubility, and bioavailability are required. The preferred characteristics of these novel salt forms include those which increase the manufacture of the active ingredient and the ease or efficiency of its formulation into a commercial product. Finally, there is a need for stable polymorphs of such salts which increase the manufacture of the active ingredients and their ease of formulation or efficiency into commercial products. σ is determined by the technique of '’·, I itb using X-ray powder diffraction (XRPD). 162775.doc •15· 201249841 Crystal degree (%). Other techniques such as solid state NMR, FT-IR, Raman spectroscopy, differential scanning calorimetry (DSC), and microcalorimetry can also be used. For the compounds of the present invention, it has been found that P field in the form of a substantially crystalline form can be produced. Several of these crystalline salts are confirmed to be sufficient to establish their stability in the production of pharmaceutical preparations. This stability can be confirmed in a variety of ways. The tendency to obtain and release atmospheric moisture can be analyzed by dynamic vapor adsorption (DVS). The solid salt can be used at 40 ° C / 75. /. The storage was carried out under RH for up to 8 days, and then the stability against high temperature and humidity was studied by weight, microscopic appearance and XRPD respective retest. Polymorphs The compounds of the invention may be crystallized in more than one form, i.e., characterized by polymorphisms, and such polymorphic forms ("polymorphs") are within the scope of the invention. Polymorphism can usually occur as a response to temperature, pressure, or both. Polymorphism can also result from changes in the crystallization process. Polymorphs can differ in a variety of physical properties known in the art (e.g., 'XRPD patterns (diffraction patterns), solubility in different solvents, and melting point). The present invention includes different polymorphic forms (including salt hydrates and solvates) of the salt form of 3·cyclopropylcarbonyl-3-3,6-diazabicyclo[3丄丨]heptane. These polymorphs are characterized by their X-ray powder diffraction (XRPD) pattern (diffraction pattern). As noted above, the salt form of 3-cyclopropylcarbonyl-3,6-diazabicyclo[3.1.1]heptane can be present in solvated (e.g., hydrated) as well as unsolvated forms. The present invention covers all such forms. The invention also includes isotopically labeled compounds wherein - or more atoms I62775.doc 201249841 are substituted by atoms having an atomic mass or mass number different from the atomic mass or mass number normally found in nature. Examples of isotopes which may be incorporated into the compounds of the invention include isotopes including hydrogen, carbon, nitrogen and oxygen, such as 2H '4, im丨% and "〇. These isotopically labeled compounds are useful as research or diagnostic tools. While the compounds of the invention may be administered in the form of a physically active chemical, it is preferred to administer the compound in the form of a pharmaceutical composition or formulation. Thus, aspects of the invention include the inclusion of a compound of the invention and/or a plurality of pharmaceutically acceptable A pharmaceutical composition for receiving a carrier, diluent or excipient. Another aspect of the invention provides a method of preparing a pharmaceutical composition comprising mixing a mouthfeel of the invention with - or a plurality of pharmaceutically acceptable carriers , diluent or excipient. The manner of administration of the compound of the present invention may vary. Preferably, the compound of the present invention is orally administered. Preferred pharmaceutical compositions for oral administration include lozenges, sacs, and films. Ingredients ', solutions and stock solutions. The pharmaceutical compositions of the present invention may be provided by modified release dosage forms such as extended release (tetra) and gum (tetra) formulations. : Bi ^ ".. ▲ Grange shot, i.e. intravenous, intramuscular, subcutaneous, intraperitoneal, intraarterial, intraventricular, and Rene administered. Intravenous administration is a preferred method of primary injection. Carriers suitable for injectable use are well known to those skilled in the art and include 5% dextrose, bismuth, excipient, sucrose/liquid, saline and phosphate buffered saline. Formulations may also be administered in other ways. Intestine-administered formulations (e.g., suppositories) have been administered to them, e.g., rectal. For those skilled in the art, 162,775.doc 201249841 can also be obtained by inhaling 'for example, in the form of an aerosol; for example, in the form of a lotion; transdermal, for example, using a transdermal patch. (for example, by using techniques available from N〇Vartis and Xian); by powder injection; or by buccal, sublingual or intranasal absorption. Pharmaceutical compositions can be formulated in unit dosage form or in multiple or subunit dosages. Administration of the pharmaceutical compositions described herein can be administered intermittently, or at a gradual, continuous, ambiguous or controlled rate. The pharmaceutical composition can be administered to a warm-blooded animal, such as a mammal, such as a mouse, rat, juvenile, rabbit, dog, pig, cow or wolf; but is advantageously administered to a human. In addition, the daily time and number of times the pharmaceutical composition is administered per day may vary. The compounds of the invention are useful in the treatment of a variety of conditions and conditions, and thus can be used in combination with a variety of other therapeutic agents suitable for the treatment or prevention of such conditions or conditions. Accordingly, one embodiment of the invention encompasses the combined administration of a compound of the invention with other therapeutic compounds. For example, the compounds of the invention can be used in combination with other NNR ligands (eg, varenicline), antioxidants (eg, free radical scavengers), antibacterial agents (eg, penicillin antibiotics), Antiviral agents (such as nucleoside analogs such as zidovudine and acyclovir), anticoagulants (such as warfarin), anti-inflammatory agents (such as NSAIDs), Antipyretics, analgesics, anesthetics (for example for surgery), acetylcholinesterase inhibitors (such as d〇nepezii and galantamine), antipsychotics (eg Haloperidol (hal〇perid〇1), clozapine 'Olan Ziping (〇ianzapjne), and quetiapine), immunosuppressants (eg, cyclocytosine (cyci〇sp〇) Rine) and amine 162775.doc •18· 201249841 methotrexate, neuroprotective agents (eg A2a inhibitors and caffeine), blood-brain barrier penetration enhancers, steroids (eg steroids), adrenal glands Corticosteroids (eg, cloches) Dexamethasone, predisone and hydrocortisone, vitamins, minerals, nutraceuticals, stimulants (eg imipramine, sulphonic; fluoxetine, paro) Paroxetine, escitalopram, sertraline, venlafaxine, and duloxetine, sleeping pills (eg alprazolam) ) and snail ring _ (buspirone), anti-caries agents (such as phenytoin and gabapentin), vasodilators (such as 0 bottom, prazosin and sildenafil), mood stabilizers (eg valproate and aripiprazole), anticancer drugs (eg antiproliferative agents), antihypertensive agents (eg atenolol, clonidine) ), amlopidine, verapamil, and olmesartan, light fullness, stool softener, diuretic (such as furosemide), antispasmodic (for example) Dicyclomine) Anti-dystonia and anti-ulcer agents (e.g., esomeprazole). A preferred use of the compounds of the invention is in the treatment and prevention of Parkinson's disease, AIM and LID, and thus, the compounds of the invention may be used It is used in combination with medical agents for treating Parkinson's disease, AIM and LID. Such agents include NNR agonists (α4β2, α7, etc.), dopamine precursors (eg, levodopa-carbidopa, levodopa-benserazide, and compound levodopa (duodopa) ), dopamine agonists (eg bromocriptine, cabergoline 162775.doc 19 201249841 (cabergoline), lisuride, perg〇iide, pramipexole ), ropinirole (popinirole), taliexole, rotigotine, and apomorphine, apomorphine, dopa carboxylase inhibitor, MAO-B inhibitor ( For example, selegiline, rasag〇iine, and safinamide, COMT inhibitors (such as entacap〇ne and tolcapone) , anti-facial acid agents (such as amantadine), anticholinergic agents (such as trihexyphenidyl, benztropine, and biperiden) Anti-dementia agents (such as rivastigmine, doneipzi (donepezi) l), galantamine, and memantine), antipsychotic agents (eg, qUetiapine, clozapine, and resperidone) anti-epileptic agents (eg, azoles) Zonisamide, norepinephrine precursor (eg droxidopa), mGluR4 agonist, mGluR5 inhibitor, 5HT1A/1B agonist, 5HT2A #antibody/inverse agonist, class Tablet substance antagonist (μ, δ, κ) ' and AMPA receptor blockers. The combination of pharmaceutically active agents can be administered separately or separately, and when administered separately, sputum is administered simultaneously or sequentially, or in any order. The amount and relative time of administration of the compound or agent should be selected, Achieve the desired therapeutic effect. The combination of a compound of the present invention and other therapeutic agents can be combined by simultaneous administration in the following forms: (1) the whole pharmaceutical composition '丨 includes two compounds; or (7) a single pharmaceutical composition 'each of which includes one of the compounds . Alternatively, the combination can be administered separately in a sequential manner, wherein a therapeutic agent is administered first, followed by another 162775.doc •20·201249841 with another second therapeutic agent. The sequential administration can be administered at a time point close to or after a period of time. Therapeutic Method 3-Cyclopropylcarbonyl-3,6-diazabicyclo[3.1.1]heptane, a pharmaceutically acceptable salt thereof, or a pharmaceutical composition containing the same can be used for prevention or treatment, has been proposed or displayed other Types of nicotine compounds useful as therapeutic agents for various conditions or conditions, such as CNS disorders (including neurodegenerative diseases), inflammation, inflammatory responses associated with bacterial and/or viral infections, pain, diabetes, metabolic syndrome, autoimmunity Sexual condition, skin condition, addiction, obesity or other conditions as further detailed herein. This compound can also be used as a diagnostic agent in receptor binding studies (in vitro and in vivo). Such treatments and other teachings are set forth, for example, in the references previously cited herein, including wiUiams et al, Drwg iVews corpus eripec 7(4): 205 (1994), Arneric et al, CiVS Drwg 1(1): 1-26 (1995), Arneric et al., χρ· /πναί· DrMgj 5(1): 79-100 (1996), Yang et al., dcia P/zarmiico/· 57«. 30(6): 740- 751 (2009), Bencherif et al., 丄 Pharmacol. Exp. Ther. 279: 1413 (1996), Lippiello^ A > J. · Ελτ?· 77zer. 279: 1422 (1996), Damac et al., /. P/zizrmaco/. 77ier. 291: 390 (1999) ; Chiari et al.

Anesthesiology 91: 1447 (1999), Lavand’homme andAnesthesiology 91: 1447 (1999), Lavand’homme and

Eisenbach,91: 1455 (1999),Holladay 等 人,·/. Mei Chw. 40(28): 4169-94 (1997),Bannon等人, Science 279: 77 (1998),PCT WO 94/08992、PCT WO 96/31475、PCT WO 96/40682、及頒予 Bencherif 等人之美 162775.doc -21 - 201249841 國專利第5,583,140號、頒予Dull等人之第5,597,919號、頒 予Smith等人之第5,604,231號、及頒予Cosford等人之第 5’852,041 號。 CNS病症 該等化合物及其醫藥組合物可用於治療或預防多種Cns 病症’包括神經退化症、神經精神病症、神經性病症及成 癮症。該等化合物及其醫藥組合物可用於治療或預防年齡 相關性及其他認知缺陷及功能障礙、注意力障礙及癡呆症 (包括彼等由於感染物或代謝紊亂造成者);提供神經保 護,治療驚厥及多發性大腦梗塞;治療心境障礙、強迫行 為及成癮行為;提供痛覺缺失;控制發炎,例如藉由細胞 因子及核因子κ B介導之發炎;治療發炎性病症;提供疼 痛減輕;及作為用於治療細菌、真菌及病毒感染之抗感染 劑治療感染。可使用本發明化合物及醫藥組合物治療或預 防之病症、疾病及病況係:年齡相關性記憶缺損 (AAMI)、輕度認知缺損(MCI)、年齡相關性認知衰退 (ARCD)、早老性癡呆、早期發作之阿茲海默氏病 (Alzheimer's disease)、老年性癡呆、阿兹海默氏型癡呆 (dementia 〇f the Alzheimer’s type)、阿茲海默氏病、無癡 呆型認知缺損(CIND)、路易體癡呆(Lewy b〇dy dementia)、 HIV癡呆、AiDS癡呆複合症、血管性癡呆、唐氏症候群 (Down syndrome)、頭部創傷、創傷性腦損傷(tbi)、拳擊 員癡呆 '克雅二氏病(Creutzfeld_Jac〇b Disease)及朊病毒 疾病、中風、中心缺血(central ischemia)、外周缺血注 162775.doc •22· 201249841 意力缺陷障礙、注意力缺陷運動障礙、誦讀障礙、精神分 裂症、精神分裂症樣精神障礙、分裂情感障礙、精神分裂 症中之或知功能障礙、精神分裂症中之認知缺陷、帕金森 症(包括帕金森氏病、腦炎後帕金森症、關島型帕金森症_ 癡呆(parkinsonism-dementia of Gaum) '帕金森氏型額顯骨 癡呆(FTDP))、皮克氏病(Pick's disease)、尼曼皮克氏病 (Niemann-Pick's disease)、亨庭頓氏病(Huntington's Disease)、 予庭頓氏舞蹈症(Huntington's chorea)、異常不自主運動、 運動障礙、左旋多巴誘導之運動障礙、遲發性運動障礙、 痙攣性肌張力異常、運動過度、進行性核上性麻痒、進行 性核上性麻痒性癡呆、下肢不寧症候群、克雅二氏病 (Creutzfeld-Jakob disease)、多發性硬化、肌萎縮性脊髓側 索硬化症(ALS)、運動神經元疾病(Mnd)、多系統萎縮症 (MSA)、皮質基底退化症、格·巴二氏症候群(GuiUain· BaW Syndr〇me)(GBS)、及慢性發炎性脫髓勒多神經病 (CIDP)、癲癇、常染色體顯性遺傳夜發性額葉癲癇、狂 躁、焦慮症、抑鬱症、經前焦慮、恐慌症、貪食症、食欲 減退、嗜眠症、白天睡眠過多、雙相性情感障礙、廣泛性 焦慮障礙、強迫症、暴怒症(rage 〇utbursts)、行為失調 症、對立性違抗障礙、妥瑞氏症候群、自閉症、藥物及酒 精成癮症、菸草成瘾症、強迫性暴食及性功能障礙。 認知缺損或功能障礙可與以下相關:精神病症或病況, 例如精神分裂症及其他精神障礙(包括但不限於精神障 礙、精神分裂症樣精神障礙、分裂情感障礙、妄想症、短 162775.doc •23· 201249841 時精神障礙'共有型精神障礙、及由於一般醫學病況之精 神障礙)、癡呆症及其他認知障礙(包括但不限於輕度認知 缺損、早老性癡呆、阿茲海默氏病、老年性癡呆、阿茲海 默氏型癡呆、年齡相關性記憶缺損、路易體癡呆、血管性 癡呆、AIDS癡呆複合症、誦讀障礙、帕金森症(包括帕金 森氏病)、運動障礙、左旋多巴誘導之運動障礙、異常不 自主運動(AIM)、認知缺損及帕金森氏病型癡呆、多發性 硬化之認知缺損、由創傷性腦損傷造成之認知缺損、由於 其他一般醫學病況之癡呆症)、焦慮症(包括但不限於非陌 生環境恐懼之恐慌症、陌生環境恐懼之恐慌症、無恐慌症 病史之陌生環境恐懼、特定恐懼症、社交恐懼症、強迫 症、創傷後應激障礙、急性應激障礙、廣泛性焦慮障礙及 由於一般醫學病況之廣泛性焦慮障礙)、心境障礙(包括但 不限於重度抑鬱症、心境惡劣、雙相性抑鬱症、雙相性狂 钿、雙相性I型情感障礙、與躁狂、抑鬱或混合發作相關 之抑鬱症、雙相性II型情感障礙、循環性情感障礙、及由 於一般醫學病況之心境障礙)、睡眠障礙(sleep dis〇rder)(包括但限於睡眠障礙(dyss〇mnia dis〇rder)、原發 性失眠症、原發性嗜睡症、嗜眠症 '睡眠過度症、惡夢 症、夜驚症及夢遊症)、智力發育遲緩、學習障礙、運動 技能障礙、交流障礙、全身性發育遲緩、注意力缺陷及破 壞性行為障礙、注意力缺陷障礙、注意力缺陷運動障礙、 嬰兒、兒童或成人之餵食及進食障礙、抽動障礙、排泄障 礙、物質相關性病症(包括但不限於物質依賴、物質濫用、 162775.doc • 24· 201249841 物質中毒、物質戒斷、酒精相關性病症、安非他命 (amphetamine)或安非他命樣相關性病症、咖啡因相關性病 症、大麻相關性病症、可卡因(cocaine)相關性病症、致幻 劑相關性病症、吸入劑相關性病症、菸鹼相關性病症、類 鴉片物質相關性病症、苯環利定(phencyclidine)或苯環利 定樣相關性病症)、及鎮靜-、催眠-或抗焦慮相關性病症、 人格障礙(包括但不限於強迫性人格障礙及衝動控制障礙)。 可利用確認認知量表(例如,Alzheimer's Disease Assessment Scale之認知子量表(ADAS_c〇g))分析認知性 能。本發明化合物改良§忍知之效率的一種量測可包括根據 該量表量測患者之變化程度。 關於強迫行為及成瘾行為,本發明化合物可作為治療劑 用於菸鹼成癮症及其他腦愉悅病症,例如物質濫用(包括 酒精成癮症、非法及處方藥物成瘾症)、進食障礙(包括肥 胖症)、及行為成瘾症(例如賭博)或成癮症之其他類似行為 表現。 以上病況及病症進一步詳細論述於(例如)Amedcan Psychiatric Association: Diagnostic and Statistical Manual of Mental Disorders,第 4版,文本修訂,Washington, DC, American Psychiatric Association, 2000中。此手冊亦可更 詳細提及與物質使用、濫用及依賴相關之症狀及診斷特 徵。不時更新並修訂此手冊且意欲使本文中之疾病及病況 的說明與該修訂本^一致。 較佳地’可治療或預防疾病、病症及病況而無明顯不利 162775.doc -25- 201249841 副作用,其包括(例如)血壓及心率顯著增大胃腸道上之 顯著副作用、及骨骼肌上之顯著作用。 據信本發明化合物在以有效量使用時可調節他*及/或 _2*證之活性,而與表徵人類神經節之㈣亞型無明 顯相互作用,如由無引發腎上腺嗜鉻組織或骨路肌中之终 鹼功能的能力證實,進一步由無引發表現肌肉型菸鹼受體 之細胞製劑中的菸鹼功能的能力證實。因此,據信該等化 合物能夠治療或預防疾病、病症及病況,而不會在神經節 及神經肌肉位點處引發顯著副作用相關性活性,因此,據 信化合物之投與可提供治療窗,其中提供某些疾病、病症 及病況之治療,且避免某些副作用。亦即,據信化合物之 有效劑量可足以對疾病、病症或病況提供期望作用,但據 信不足以(即,不會以足夠高程度)提供不期望副作用。 因此,本發明提供本發明化合物或其醫藥上可接受之鹽 的用途’其用於療法(例如上述療法)中。 在再一態樣中,本發明提供本發明化合物或其醫藥上可 接受之鹽的用途’其用於製造用於治療CNS病症(例如上述 病症、疾病或病況)之藥劑。 發炎 已知主要穿過迷走神經之神經系統可藉由抑制巨噬細胞 腫瘤壞死因子(TNF)之釋放來調控先天性免疫應答之強 度。此生理機制稱作「膽鹼激導性抗炎途徑」(參見,例 如,「The Inflammatory Reflex」,420: 853-9 (2002))。發炎及腫瘤壞死因子之過度合成會在各種疾病中 162775.doc •26· 201249841 造成發病及甚至死亡。該等疾病包括(但不限於)内毒素血 症、類風濕性關節炎、骨關節炎、牛皮癬、哮喘、動脈粥 樣硬化、特發性肺纖維化及發炎性腸病。 可藉由投與本文所述化合物治療或預防之發炎病況包括 (但不限於)慢性及急性發炎、牛皮癬、内毒素血症、痛 風、急性假痛風、急性痛風性關節炎、關節炎、類風濕性 關節炎、骨關節炎、同種異體移植排斥、慢性移植物排 斥 '哮喘、動脈粥樣硬化、單核_吞噬細胞依賴性肺損 傷、特發性肺纖維化、特應性皮炎、慢性阻塞性肺病、成 人型呼吸窘迫症候群、鐮狀細胞疾病中之急性胸腔症候 群、發炎性腸病、刺激性腸症候群、克羅恩氏病(Crohn,s disease)、潰瘍、潰瘍性結腸炎、急性膽管炎、口瘡性口 炎、惡病質、回腸囊袋炎、腎小球腎炎、狼瘡性腎炎、血 栓症及移植物抗宿主反應。 與細菌及/或病毒感染相關之發炎反應 許多細菌及/或病毒感染與由毒素形成、及身體對細菌 或病毒及/或毒素之自然反應導致的副作用相關。如上文 所論述,身體對感染之反應經常涉及產生大量TNF&amp;/或其 他細胞因子。該等細胞因子之過度表現可產生明顯損傷, 例如敗血性休克(當細菌係敗血症細菌時)、内毒素性休 克、尿膿毒病、病毒性肺炎及中毒性休克症候群。 細胞因子表現係由NNR介導,且可藉由投與該等受體之 激動劑或部分激動劑得以抑制。因此,作為該等受體之激 動劑或部分激動劑之本文所述彼等化合物可用於使與細菌 162775.doc -27- 201249841 感染、及與病毒及真菌感染相關之發炎反應最小化。該等 細菌感染之實例包括炭疽熱、肉毒桿菌病及敗血症。其中 有些該等化合物亦可能具有抗微生物性質。 該等化合物亦可作為辅助療法與現有療法組合使用,以 管控細菌、病毒及真菌感染,例如抗生素、抗病毒劑及抗 真菌劑。抗毒素亦可用於與感染物所產生之毒素結合,且 可使所結合毒素通過身體,而不產生發炎反應。抗毒素劑 之實例揭示於(例如)頒予Bundle等人之美國專利第 6,310,043號中。有效抗細菌及其他毒素之其他藥劑亦有效 且可藉由與本文所述化合物共同投與,達到互補之治療效 應。 疼痛 可投與化合物以治療及/或預防疼痛,其包括急性、神 經性(neurologic)、發炎性、神經性(neur〇pathic)及慢性疼 痛°該等化合物可與麻醉劑結合使用以使麻醉劑成癮症之 可能性最小化(例如’嗎啉保護療法)❶可以持續發炎疼痛 及神經性疼痛模式證實本文所述化合物之鎮痛活性,如美 國公開專利申請案第20010056084 A1號(Allgeier等人)中所 述實施(例如,發炎性疼痛之完全弗氏佐劑(Freund,s adjuvant)大鼠模型中的機械性痛覺過敏及神經性疼痛之小 鼠部分坐骨神經結紮模型中的機械性痛覺過敏)。 鎮痛作用適於治療各種成因或病源學之疼痛、具體而言 治療發炎性疼痛及相關性痛覺過敏、神經性疼痛及相關性 痛覺過敏、慢性疼痛(例如,嚴重慢性疼痛、手術後疼痛 162775.doc -28- 201249841 及與包括癌症之各種病況相關的疼痛、心絞痛、腎或膽道 絞痛、月經痛、偏頭痛及痛風)。發炎性疼痛可具有不同 成因’包括關節炎及類風濕性疾病、腱鞘炎及血管炎。神 經性疼痛包括三叉神經痛或皰疹性神經痛、神經病變(例 如糖尿病性神經病變疼痛)、灼性神經痛、下背疼痛及去 傳入症候群(例如臂叢撕脫傷)。 其他病症 除治療CNS病症、發炎及血管增生及疼痛外,本發明化 合物亦可用於預防或治療NNR起作用之某些其他病況、疾 病及病症。實例包括自身免疫性病症(例如狼瘡)、與細胞 因子釋放相關之病症、感染繼發性惡質病(例如,如 AIDS、AIDS相關性複合症及贅瘤形成中所出現)、肥胖 症、天皰瘡(pemphigus)、尿失禁、膀胱過度活動症、腹 瀉、便秘、視網膜病、感染疾病、肌無力、Eat〇n_Lamben 症候群、高血壓、先兆子癇、骨質疏鬆症、血管收縮、血 管擴張、心律不齊、Ϊ型糖尿病、„型糖尿病、貪食症、食 欲減退及性功能障礙、以及彼等闞述於公開pCT申請案 WO 98/25619中之適應症。亦可投與本發明化合物以治療 驚厥(例如彼等具有癲癇症狀者)及治療諸如梅毒及克雅二 氏病等病況》最後,本發明化合物可用於治療多種皮膚病 症’其包括但不限於牛皮癬、皮炎、痤瘡 '膿包病、白斑 病及諸如此類。 診斷用途 該等化合物可用於診斷組合物(例如探針)中,尤其當其 162775.doc • 29· 201249841 經4飾以包括適當標記時。探針可用於(例如)測定特異性 受體、尤其α4β2*及/或含α6之受體亞型的相對數量及/或功 能。出於此目#’最佳地,用可使用電子發射斷層攝影術 (PET)檢測之放射性同位素部&amp; (例如π〇標記本發明化合 物。期望间特異活性以在非飽和濃度下顯現受體亞型。所 杈與劑量通常低於毒性範圍且提供高對比圖像。期望化合 物能夠以無毒性程度投與。以熟習放射性標記成像技術者 已知之方式實施劑量之測定。參見,例如,頒予L〇nd〇n等 人之美國專利第5,969,144號》 可使用已知技術投與化合物。參見,例如,頒予L〇nd〇n 等人之美國專利第5,969,144號,如上文所述。可以納入其 他成份(例如可用於調配診斷組合物之彼等類型的成份)之 調配組合物形式投與化合物。根據實施本發明使用之化合 物最佳以高純度形式採用。參見頒予Elrnalch等人之美國 專利第5,853,696號。 在將化合物投與個體(例如,人類個體)後,可藉由適當 技術使個體内存在之該化合物成像並對其進行定量以指示 所選NNR亞型之存在、量及功能。除人類外,亦可將化合 物投與動物,例如小鼠、大鼠、狗及猴子。可使用任一適當 技術及設備實施PET成像。參見Villemagne等人,In: Arneric 等人(編輯)Neuronal Nicotinic Receptors: Pharmacology and Therapeutic Opportunities, 235-250 (1998)及頒予 Elmalch 等人之美國專利第5,853,696號,關於代表性成像技術之揭 示内容’其各自以引用方式併入本文中。 162775.doc • 30· 201249841 放射性標記之化合物以高親和力結合選擇性nNR亞型 (例如,α4β2*及/或含α6)且較佳對其他菸鹼膽鹼激導性受 體亞型(例如,彼等與肌肉及神經節相關之受體亞型)呈現 可忽略之非特異性結合。因此,該等化合物可作為藥劑用 於個體體内、尤其腦内之菸鹼膽鹼激導性受體亞型的非侵 入性成像用於與多種CNS疾病及病症相關之診斷。 在一個態樣中,診斷組合物可用於診斷個體(例如人類 患者)之疾病的方法中。該方法包括投與該患者本文所述 以可檢測方式標記之化合物、及檢測該化合物與所選nnr 亞型(例如,α4β2*及/或含α6之受體亞型)之結合。彼等使 用診斷工具(例如ΡΕΤ)之熟習此項技術者可使用本文所述 放射性標記之化合物以診斷多種病況及病症,其包括與中 框及自主神經系統功能障礙相關之病況及病症。該等病症 包括多種CNS疾病及病症,其包括阿茲海默氏病、帕金森 氏病及精神分裂症。可評價之該等及其他代表性疾病及病 症包括彼等闡述於頒予Bencherif等人之美國專利第 5,952,339號中者。 在另一態樣中,診斷組合物可用於監測個體(例如人類 患者)之選擇性菸鹼受體亞型的方法十。該方法包括投與 該患者本文所述以可檢測方式標記之化合物、及檢測該化 合物與所選菸鹼受體亞型(即α4β2*及/或含α6之受體亞型) 之結合。 受體結合 本發明化合物可在結合NNR亞型、尤其及/或α4β2*及/或 含α6之受體亞型之化合物的結合分析中用作參照配體。出 162775.doc •31- 201249841 於此目的,較佳用放射性同位素部分(例如,!4C)標記 本發明化合物。 實例 以下實例提供用以闡釋本發明,且不應理解為對其具有 限制性。在該等實例中,除非另有說明,否則所有份數及 百分數均係以重量計。 實例1 .用於表徵3-環丙基羰基_3,6-二氮雜雙環【311】庚烧 之鹽形式之儀錶及實驗方案 X射線粉末繞射(XRPD) 在使用CuKa輻射(40 kV,40 mA)、自動χγζ座標台、用 於自動取樣定位之雷射視頻顯微鏡及HiStar 2維區域檢測 器之Bruker AXS C2 GADDS繞射儀上收集X射線粉末繞射 圖案。X射線光學器件由與0.3 mm之針孔型準直器偶聯之 單一 G5be丨多層鏡組成。光束發散度(即,試樣上之χ射線 光束之有效尺寸)係約4 mm。採用θ·θ連續掃描模式與2〇 cm之試樣-檢測器距離’其產生3.2。至29.7。之有效2Θ範 圍。試樣通常可於X射線光束中暴露120秒。使用如接收原 樣而未研磨之粉末將環境條件下運行之試樣製備為平板樣 品。將約1 -2 mg試樣輕輕地壓在玻璃載片上以獲得平表 面。將非環境條件下運行之試樣安裝於具有導熱化合物之 矽晶圓上《隨後將試樣以約l〇&lt;t /min加熱至適當溫度且隨 後等溫保持約5分鐘’之後開始數據收集。峰位置報告為 °2Θ ’精確度為±〇1。。 單晶 XRD (SXD) 在配備 Oxford Cryosystems Cryostream 冷卻裝置之 162775.doc 32· 201249841Eisenbach, 91: 1455 (1999), Holladay et al., /. Mei Chw. 40(28): 4169-94 (1997), Bannon et al, Science 279: 77 (1998), PCT WO 94/08992, PCT No. 5,583,140 to Dull et al. 5,604,231 and 5'852,041 to Cosford et al. CNS Disorders The compounds and pharmaceutical compositions thereof are useful in the treatment or prevention of a variety of Cns conditions including neurodegenerative disorders, neuropsychiatric disorders, neurological disorders, and addictions. The compounds and pharmaceutical compositions thereof are useful for treating or preventing age-related and other cognitive deficits and dysfunctions, attention disorders, and dementia (including those caused by infections or metabolic disorders); providing neuroprotection, treating convulsions And multiple cerebral infarction; treatment of mood disorders, obsessive-compulsive behaviors and addictive behaviors; provision of analgesia; control of inflammation, such as inflammation mediated by cytokines and nuclear factor kappa B; treatment of inflammatory conditions; provision of pain relief; An anti-infective agent for the treatment of bacterial, fungal and viral infections to treat infections. Conditions, diseases, and conditions that can be treated or prevented using the compounds of the invention and pharmaceutical compositions: age-related memory impairment (AAMI), mild cognitive impairment (MCI), age-related cognitive decline (ARCD), Alzheimer's disease, Early onset of Alzheimer's disease, Alzheimer's disease, Dementia 〇f the Alzheimer's type, Alzheimer's disease, Dementia-free cognitive impairment (CIND), Lewy b〇dy dementia, HIV dementia, AiDS dementia complex, vascular dementia, Down syndrome, head trauma, traumatic brain injury (tbi), boxer dementia 'Kaya II Creutzfeld_Jac〇b Disease and prion diseases, stroke, central ischemia, peripheral ischemia 162775.doc •22· 201249841 Stress deficit disorder, attention deficit dyskinesia, dyslexia, schizophrenia Symptoms, schizophrenia-like mental disorders, schizoaffective disorders, schizophrenia or cognitive dysfunction, cognitive deficits in schizophrenia, Parkinson's disease (including Parkinson's disease, Parkinson's disease after encephalitis, Guam Parkinson's disease _ dementia (parkinsonism-dementia of Gaum) 'Parkinson's type of osteogenic dementia (FTDP)), Pick's disease, Niemann Niemann-Pick's disease, Huntington's Disease, Huntington's chorea, abnormal involuntary movement, dyskinesia, levodopa-induced dyskinesia, late onset Sexual dyskinesia, spastic dystonia, hyperkinesia, progressive supranuclear palsy, progressive supranuclear palsy, restless leg syndrome, Creutzfeld-Jakob disease, multiple Sclerosing, amyotrophic lateral sclerosis (ALS), motor neuron disease (Mnd), multiple system atrophy (MSA), cortical basal degeneration, and Gua Bain Syndrome (GuiUain· BaW Syndr〇me) GBS), and chronic inflammatory demyelinating neurosis (CIDP), epilepsy, autosomal dominant nocturnal frontal lobe epilepsy, mania, anxiety, depression, premenstrual anxiety, panic disorder, bulimia, loss of appetite , narcolepsy Excessive sleep during the day, bipolar disorder, generalized anxiety disorder, obsessive-compulsive disorder, rage 〇utbursts, behavioral disorders, oppositional defiant disorder, Tourette's syndrome, autism, drug and alcohol addiction , tobacco addiction, obsessive overeating and sexual dysfunction. Cognitive impairment or dysfunction can be related to a psychiatric condition or condition, such as schizophrenia and other mental disorders (including but not limited to mental disorders, schizophrenia-like mental disorders, schizoaffective disorders, delusions, short 162775.doc • 23· 201249841 Mental disorders 'common mental disorders, and mental disorders due to general medical conditions), dementia and other cognitive disorders (including but not limited to mild cognitive impairment, Alzheimer's disease, Alzheimer's disease, old age Dementia, Alzheimer's type dementia, age-related memory impairment, Lewy body dementia, vascular dementia, AIDS dementia complex, dyslexia, Parkinson's disease (including Parkinson's disease), dyskinesia, levodopa Induced dyskinesia, abnormal involuntary movement (AIM), cognitive impairment and Parkinson's disease type dementia, cognitive impairment of multiple sclerosis, cognitive impairment caused by traumatic brain injury, dementia due to other general medical conditions, Anxiety disorders (including but not limited to panic disorder in fear of unfamiliar environment, panic disorder in fear of unfamiliar environment, Unfamiliar environmental fears of panic disorder history, specific phobias, social phobias, obsessive-compulsive disorder, post-traumatic stress disorder, acute stress disorder, generalized anxiety disorder and generalized anxiety disorder due to general medical conditions), mood disorders (including But not limited to major depression, mood disorder, bipolar depression, bipolar mania, bipolar I type affective disorder, depression associated with mania, depression or mixed seizures, bipolar type II affective disorder, cyclical emotions Disorders, and mood disorders due to general medical conditions), sleep dis〇rder (including but limited to sleep disorders (dyss〇mnia dis〇rder), primary insomnia, primary narcolepsy, narcolepsy Hypersomnia, nightmares, night terrors and sleepwalking), mental retardation, learning disabilities, motor skills disorders, communication disorders, systemic developmental delay, attention deficits and destructive behavioral disorders, attention deficit disorder, attention Defective dyskinesia, feeding and eating disorders in infants, children or adults, tic disorder, excretory disorders, substance-related Symptoms (including but not limited to substance dependence, substance abuse, 162775.doc • 24·201249841 substance poisoning, substance withdrawal, alcohol-related disorders, amphetamine or amphetamine-related disorders, caffeine-related disorders, cannabis-related Sexual disorders, cocaine-related disorders, hallucinogen-related disorders, inhalation-related disorders, nicotine-related disorders, opioid-related disorders, phencyclidine or phencyclidine-like Related disorders), and sedative-, hypnotic- or anti-anxiety-related disorders, personality disorders (including but not limited to obsessive-compulsive disorder and impulsive disorder). Cognitive performance can be analyzed using a recognition cognitive scale (e.g., the Alzheimer's Disease Assessment Scale's Cognitive Subscale (ADAS_c〇g)). A measure of the compound of the invention that improves the efficiency of § tolerance can include measuring the degree of change in the patient based on the scale. With regard to compulsive behavior and addictive behavior, the compounds of the invention are useful as therapeutic agents for nicotine addiction and other brain-sustainable conditions, such as substance abuse (including alcohol addiction, illegal and prescription drug addiction), eating disorders ( Other obesity behaviors including obesity, behavioral addiction (eg gambling) or addiction. The above conditions and conditions are discussed in further detail in, for example, the Amedcan Psychiatric Association: Diagnostic and Statistical Manual of Mental Disorders, 4th Edition, Text Revision, Washington, DC, American Psychiatric Association, 2000. This handbook also refers in more detail to the symptoms and diagnostic features associated with substance use, abuse and dependence. This manual is updated and revised from time to time and is intended to be consistent with the revision of this document. Preferably, it can treat or prevent diseases, conditions and conditions without significant disadvantages. 162775.doc -25 - 201249841 Side effects including, for example, significant increase in blood pressure and heart rate, significant side effects on the gastrointestinal tract, and significant effects on skeletal muscle . It is believed that the compounds of the invention may modulate the activity of the * and/or _2* when used in an effective amount, but have no significant interaction with the (4) subtype characterizing the human ganglia, such as by the absence of adrenal pheochromic tissue or bone. The ability of the final base function in the road muscle was confirmed to be further confirmed by the ability to induce nicotine function in a cell preparation exhibiting muscle type nicotinic receptor. Accordingly, it is believed that such compounds are capable of treating or preventing diseases, conditions, and conditions without causing significant side-effect-related activity at the ganglion and neuromuscular sites, and therefore, administration of the compound is believed to provide a therapeutic window, wherein Provide treatment for certain diseases, conditions and conditions, and avoid certain side effects. That is, it is believed that an effective dose of the compound may be sufficient to provide the desired effect on the disease, condition or condition, but it is believed that it is insufficient (i.e., not to a sufficiently high degree) to provide undesirable side effects. Accordingly, the invention provides the use of a compound of the invention, or a pharmaceutically acceptable salt thereof, in a therapy (e.g., as described above). In still another aspect, the invention provides the use of a compound of the invention, or a pharmaceutically acceptable salt thereof, for use in the manufacture of a medicament for the treatment of a CNS disorder, such as a condition, disease or condition as described above. Inflammation The nervous system, primarily known to cross the vagus nerve, regulates the strength of the innate immune response by inhibiting the release of macrophage tumor necrosis factor (TNF). This physiological mechanism is called "choline-induced anti-inflammatory pathway" (see, for example, "The Inflammatory Reflex", 420: 853-9 (2002)). Inflammation and over-synthesis of tumor necrosis factor can cause morbidity and even death in various diseases. 162775.doc •26·201249841 Such diseases include, but are not limited to, endotoxemia, rheumatoid arthritis, osteoarthritis, psoriasis, asthma, atherosclerosis, idiopathic pulmonary fibrosis, and inflammatory bowel disease. Inflammatory conditions which may be treated or prevented by administration of a compound described herein include, but are not limited to, chronic and acute inflammation, psoriasis, endotoxemia, gout, acute pseudogout, acute gouty arthritis, arthritis, rheumatoid Arthritis, osteoarthritis, allograft rejection, chronic graft rejection 'asthma, atherosclerosis, mononuclear phagocyte-dependent lung injury, idiopathic pulmonary fibrosis, atopic dermatitis, chronic obstructive Pulmonary disease, adult respiratory distress syndrome, acute thoracic syndrome in sickle cell disease, inflammatory bowel disease, stimulating bowel syndrome, Crohn's disease, ulcer, ulcerative colitis, acute cholangitis , aphthous stomatitis, cachexia, ileal pouchitis, glomerulonephritis, lupus nephritis, thrombosis and graft versus host response. Inflammatory Responses Associated with Bacterial and/or Viral Infections Many bacterial and/or viral infections are associated with side effects caused by the formation of toxins and the natural reaction of the body to bacteria or viruses and/or toxins. As discussed above, the body's response to infection often involves the production of large amounts of TNF&amp;/other cytokines. Excessive performance of these cytokines can cause significant damage, such as septic shock (when bacteria are sepsis bacteria), endotoxin shock, urinary sepsis, viral pneumonia, and toxic shock syndrome. Cytokine expression is mediated by NNR and can be inhibited by administration of agonists or partial agonists of such receptors. Thus, the compounds described herein as agonists or partial agonists of such receptors can be used to minimize inflammatory responses associated with bacterial 162775.doc -27-201249841 infection and viral and fungal infections. Examples of such bacterial infections include anthrax, botulism, and sepsis. Some of these compounds may also have antimicrobial properties. These compounds can also be used as adjuvant therapies in combination with existing therapies to control bacterial, viral and fungal infections such as antibiotics, antivirals and antifungals. The antitoxin can also be used to bind to the toxin produced by the infectious agent and allow the bound toxin to pass through the body without producing an inflammatory response. An example of an anti-toxin agent is disclosed in, for example, U.S. Patent No. 6,310,043 issued to Bundle et al. Other agents effective against bacteria and other toxins are also effective and can be combined to achieve a complementary therapeutic effect by co-administering with the compounds described herein. Pain can be administered to treat and/or prevent pain, including acute, neurological, inflammatory, neuropathic, and chronic pain. These compounds can be combined with anesthetics to add to the anesthetic. Minimization of the likelihood of disease (eg, 'morpholine protection therapy) ❶ can continue to inflammatory pain and neuropathic pain patterns confirm the analgesic activity of the compounds described herein, as in US Published Patent Application No. 20010056084 A1 (Allgeier et al) It is described (for example, mechanical hyperalgesia in a partial sciatic nerve ligation model of a mouse model of mechanical hyperalgesia and neuropathic pain in a complete Freund's septic rat model). Analgesic effects are suitable for the treatment of various causes or pathogenic pains, in particular for the treatment of inflammatory pain and associated hyperalgesia, neuropathic pain and related hyperalgesia, chronic pain (eg, severe chronic pain, post-operative pain 162775.doc) -28- 201249841 and pain, angina pectoris, kidney or biliary colic, menstrual pain, migraine and gout associated with various conditions including cancer. Inflammatory pain can have different causes, including arthritis and rheumatoid diseases, tenosynovitis, and vasculitis. Neuropathic pain includes trigeminal neuralgia or herpetic neuralgia, neuropathy (such as diabetic neuropathic pain), burning neuralgia, lower back pain, and de-inflammation (such as brachial plexus avulsion). Other Conditions In addition to treating CNS disorders, inflammation, and vascular proliferation and pain, the compounds of the invention may also be used to prevent or treat certain other conditions, diseases, and conditions in which NNRs function. Examples include autoimmune disorders (such as lupus), disorders associated with cytokine release, infection with secondary echinococcosis (eg, such as AIDS, AIDS-related complexes, and neoplasia), obesity, days Pemphigus, urinary incontinence, overactive bladder, diarrhea, constipation, retinopathy, infectious disease, muscle weakness, Eat〇n_Lamben syndrome, hypertension, pre-eclampsia, osteoporosis, vasoconstriction, vasodilation, arrhythmia Qi, sputum type diabetes, „type diabetes, bulimia, loss of appetite and sexual dysfunction, and their indications as disclosed in the published pCT application WO 98/25619. Compounds of the invention may also be administered to treat convulsions ( For example, those with epilepsy symptoms and treatment of conditions such as syphilis and Creutzfeldt-Jakob disease. Finally, the compounds of the present invention are useful for treating a variety of skin conditions including, but not limited to, psoriasis, dermatitis, acne pustule disease, leukoplakia and Such as diagnostic uses, such compounds can be used in diagnostic compositions (eg, probes), especially when they are 162775.doc • 29· 201249841 When 4 is included to include the appropriate label, the probe can be used, for example, to determine the relative amount and/or function of a specific receptor, particularly α4β2* and/or a receptor subtype containing α6. Most preferably, the radioisotope moiety &amp; (e.g., π 〇 labeled with the compound of the invention can be labeled with electron emission tomography (PET). The specific activity is expected to visualize the receptor subtype at an unsaturated concentration. The dosage is usually below the toxicity range and provides a high contrast image. The desired compound can be administered in a non-toxic manner. The dosage is determined in a manner known to those skilled in the art of radiolabel imaging. See, for example, L〇nd〇n et al. U.S. Patent No. 5,969,144, the disclosure of which is incorporated herein by reference to U.S. Pat. Compounds of the type of ingredients of the diagnostic composition are formulated to be administered as a compound. The compounds used in accordance with the practice of the invention are preferably employed in high purity form. U.S. Patent No. 5,853,696 to Elrnalch et al.. After administration of a compound to an individual (e.g., a human subject), the compound present in the individual can be imaged and quantified by appropriate techniques to indicate the selected NNR subtype. The presence, amount and function. In addition to humans, compounds can also be administered to animals such as mice, rats, dogs and monkeys. PET imaging can be performed using any suitable technique and equipment. See Villemagne et al., In: Arneric Neuronal Nicotinic Receptors: Pharmacology and Therapeutic Opportunities, 235-250 (1998) and U.S. Patent No. 5,853,696 to Elmalch et al., the disclosure of which is incorporated herein by reference. in. 162775.doc • 30· 201249841 Radiolabeled compounds bind with high affinity to selective nNR subtypes (eg, α4β2* and/or α6) and preferably to other nicotinic choline-exciting receptor subtypes (eg, These receptor subtypes associated with muscle and ganglia exhibit negligible non-specific binding. Thus, such compounds can be used as medicaments for non-invasive imaging of nicotinic choline stimulating receptor subtypes in individuals, particularly in the brain, for use in the diagnosis of a variety of CNS diseases and conditions. In one aspect, the diagnostic composition can be used in a method of diagnosing a disease in an individual, such as a human patient. The method comprises administering to the patient a compound that is detectably labeled as described herein, and detecting binding of the compound to a selected nnr subtype (e.g., α4β2* and/or a receptor subtype containing α6). Those skilled in the art using diagnostic tools (e.g., sputum) can use the radiolabeled compounds described herein to diagnose a variety of conditions and conditions, including those associated with mesenteric and autonomic nervous system dysfunction. Such conditions include a variety of CNS diseases and conditions, including Alzheimer's disease, Parkinson's disease, and schizophrenia. </ RTI> <RTIgt; </ RTI> <RTIgt; </ RTI> <RTIgt; </ RTI> <RTIgt; In another aspect, the diagnostic composition can be used to monitor a selective nicotinic receptor subtype of an individual (e.g., a human patient). The method comprises administering to the patient a compound that is detectably labeled as described herein, and detecting binding of the compound to a selected nicotinic receptor subtype (i.e., a4[beta]2* and/or a [alpha]6 containing receptor subtype). Receptor Binding The compounds of the invention can be used as reference ligands in binding assays for compounds that bind to NNR isoforms, particularly and/or α4β2* and/or receptor subtypes containing α6. 162775.doc • 31- 201249841 For this purpose, the compounds of the invention are preferably labeled with a radioisotope moiety (e.g., !4C). EXAMPLES The following examples are provided to illustrate the invention and are not to be construed as limiting thereof. In the examples, all parts and percentages are by weight unless otherwise indicated. Example 1. Instrument and experimental scheme for characterizing the salt form of 3-cyclopropylcarbonyl-3,6-diazabicyclo[311] heptane. X-ray powder diffraction (XRPD) using CuKa radiation (40 kV, The X-ray powder diffraction pattern is collected on a Bruker AXS C2 GADDS diffractometer with a 40 mA), automatic χγ 标 table, a laser video microscope for automatic sampling and positioning, and a HiStar 2D area detector. The X-ray optics consist of a single G5be丨 multilayer mirror coupled to a 0.3 mm pinhole collimator. The beam divergence (i.e., the effective size of the x-ray beam on the sample) is about 4 mm. A θ·θ continuous scan mode was used with a sample-detector distance of 2 〇 cm, which produced 3.2. To 29.7. Effective 2Θ range. The sample is typically exposed to an X-ray beam for 120 seconds. A sample run under ambient conditions was prepared as a flat sample using a powder that was not ground as received. Approximately 1-2 mg of the sample was gently pressed onto a glass slide to obtain a flat surface. A sample run under non-environmental conditions is mounted on a crucible wafer with a thermally conductive compound. "Data is then collected after the sample is heated to a suitable temperature at about 1 Torr &lt; t /min and then isothermally maintained for about 5 minutes." . The peak position is reported as °2Θ ’ accuracy of ±〇1. . Single crystal XRD (SXD) is equipped with Oxford Cryosystems Cryostream cooling unit 162775.doc 32· 201249841

Bruker AXS IK SMART CCD繞射儀上收集數據。使用 SHELXS或SHELXD程式解析結構且利用作為Bruker AXS SHELXTL套件之部分的SHELXL程式重新精修。除非另有 說明,否則附接至碳之氫原子係按幾何方式安置且利用騎 式各向同性位移參數(riding isotropic displacement parameter)進行精修。附接至雜原子之氫原子位於不同傅裏 葉(Fourier)合成中且使其以各向同性位移參數自由精修。 核磁共振(NMR)譜 在配備有自動取樣器且由DRX400控制器台控制之Varian Unity 300 MHz儀器或 Bruker 400 MHz儀器上收集NMR 譜。使用利用Topspin v 1.3 (修補程式等級8)運行之 ICONNMR V4.0.4 (組建1)使用標準Bruker加載實驗獲得自 動化實驗。對於非常規譜而言,僅經由使用Topspin獲得 數據。 熔點 以對應於約5°C /min之加熱速率之設置使用Fisher-Johns 熱台、溶點設備。 示差掃描熱量測定法(DSC) 在配備有50位自動取樣器之ΤΑ Instruments Q1000或 Mettler DSC 823e上收集DSC數據。使用合格銦針對能量 及溫度校準對儀器進行校準。通常將銷孔式鋁盤中之 0.5-1.5 mg每一試樣以10°C/min自25°C加熱至175°C至 200°C。在試樣上以30 mL/min維持氮吹掃。 熱重分析法(TGA) 在配備有16位自動取樣器之TA Instruments Q500 TGA或 配備有34位自動取樣器之Mettler TGA/SDTA 851e上收集 162775.doc -33- 201249841 TGA數據。ΤΑ Instruments Q5〇〇 :使用合格鋁鎳合金 (Alumel)校準儀器之溫度。通常將5_1〇 mg每一試樣加載於 預稱量之姑掛堝及鋁DSC盤上,並以i〇°c /min自環境溫度 加熱至350 C。在試樣上以60 mL/min維持氣吹掃。Mettler TGA/SDTA 851e :使用合格銦校準儀器之溫度。通常將5_ 10 mg每一試樣加載於預稱量之鋁坩堝上並以1〇&lt;t/min自 環境溫度加熱至350°C。在試樣上以50 mL/min維持氮吹 掃。 偏光顯微鏡法(PLM) 利用具有用於捕獲影像之數位視訊相機的Leica LM/DM 偏光顯微鏡研究試樣。將少量每一試樣放置於玻璃載片 上’安裝於浸沒油中並用玻璃片覆蓋,個別粒子盡可能地 分開。以適當放大觀察試樣且使光部分偏振,偶合至入假 滤色器。 熱台顯微銳法(HSM) 使用與Mettler-Toledo MTFP82HT熱台及用於捕獲影像 之數位視訊相機組合的Leica LM/DM偏光顯微鏡實施熱台 顯微鏡法。將少量每一試樣放置於玻璃載片上,個別粒子 盡可能地分開。以適當放大觀察試樣且使光部分偏振,偶 合至λ假濾色器,同時通常以10〇c /min自環境溫度加熱。 動態蒸氣吸附(DVS) 使用由SMS分析套件軟體控制之SMS DVS Intrinsic水份 吸收分析儀測定吸附等溫線。藉由儀器控制將試樣溫度維 持於25°C下。藉由混合乾與濕氮流控制濕度,總流速為 200 mL/min。藉由位於試樣附近之經校準R〇tr〇nic探針量 162775.doc •34- 201249841 測相對濕度(動態範圍為1.0-100% RH)。藉由微量天平(精 確度±0.005 mg)持續監測隨RH%變化之試樣之重量變化(質 量鬆弛)。 通常在環境條件下將5-20 mg試樣放置於稱量之絲網不 銹鋼籃上。於40% RH及25°C (典型環境條件)下加載試樣及 不加載。如下文概述實施水份吸附等溫線(2次掃描產生1 個完整循環)。於25°C下以10% RH間隔在0-90% RH範圍内 實施標準等溫線。 DVS Generic方法參數 參數 值 吸附-掃描1 40-90 解吸附/吸附-掃描2 90 -乾,乾-40 間隔(RH%) 10 掃描次數 2 流速(mL/min) 200 溫度(°C) 25 穩定性(°C/min) 0.2 吸附時間(小時) 6小時暫停 在完成等溫線後回收試樣並藉由XRPD重新分析。 藉由卡爾·費希爾法(KF)之水測定 利用 Mettler Toledo DL39 Coulometer 使用 Hydranal Coulomat AG試劑及氬吹掃量測每一試樣之水含量。將稱 重固體試樣引入鉑TGA盤上之容器中,該盤與subaseal連 接以避免水進入。每次滴定使用約1 〇 mg試樣且一式兩份 地進行測定。 162775.doc -35- 201249841 藉由HPLC之熱力學水溶解性 藉由將足夠化合物懸浮於水中以產生210 mg/mL母體游 離形式之化合物的最大最終濃度來測定水溶解性。將懸浮 液於25°C下平衡24 h,且隨後量測pH。隨後經由玻璃纖維 C過濾器將懸浮液過濾至96孔板中。隨後將濾液稀釋101 倍。參照存於DMSO中之約0.1 mg/mL標準溶液藉由HPLC 進行定量。注射不同體積之標準、經稀釋及未經稀釋溶 液。使用藉由對與標準注射中之主要峰相同之保留時間下 發現之峰進行積分測定的峰面積計算溶解性。若濾板中存 在足夠固體,則採集XRPD。 熱力學水溶解性方法之HPLC方法參數 方法類型: 利用梯度洗脫之反相 管柱: Phenomenex Luna, C18 (2) 5 μηι, 5〇x4.6 mm 管柱溫度(°c): 25 標準注射(μ!〇 : 1、2、3、5、7、10 測試注射(jiL) · 卜 2、3、10、20、50 檢測: 波長、帶寬(nm): 260、80 流速(mL/min): 2 相A : 存於水中之0.1%TFA 相B : 存於乙腈中之0.085% TFA 時間表: 時間(min) 相A% 相B% 0.0 95 5 1.0 80 20 2.3 5 95 3.3 5 95 3.5 95 5 4.4 . 95 5 162775.doc -36· 201249841 在配備有二極體陣列檢測器且使用ChemStation軟體 vB.02.01-SR1之Agilent HP 1100系列系統上實施分析。 藉由HPLC之化學純度 在配備有二極體陣列檢測器且使用ChemStation軟體 vB.02.01-SR1之Agilent HP 11 〇〇系列系統上實施純度分 析。 化學純度測定之HPLC方法參數 試樣製備 0.5 mg/mL ’ 存於乙腈:水i:i (v/v)中, 管枉: Phenomenex Luna C18 (2), 150x4.6 mm, 5 μπι 管柱溫度(°c): 25 注射(pL): 5 檢測: 波長、帶寬:(nm): 255、90 流速(mL/min): 1 相A : 存於水中之0.1%TFA 相B : 存於乙腈中之0.085% TFA 時間表: 時間(min) 相A% 相B% 0 95 5 25 5 95 25.2 95 5 30 95 5 離子層析 在使用 IC Net軟體 v2.3之 Metrohm 761 Advanced Compact 1C (用於陽離子)及 Metrohm 861 Advanced Compact 1C (用 於陰離子)上收集數據。將試樣製備為存於DMSO中之1000 ppm儲液。在測試之前將試樣用DMSO稀釋至100 ppm。藉 162775.doc •37· 201249841 由與已知濃度之正分析離子的標準溶液比較達成定量。 陰離子之離子層析方法 方法類型 陰離子變化 管柱: Metrosep A Supp 5 - 250 (4.0x250 mm) 管柱溫度(°c): 環境溫度 注射(0): 20 檢測: 傳導性檢測器 流速(mL/min): 0.7 洗脫劑: 3.2 mM碳酸鈉、 1.0 mM碳水氫納,存於水中 陽離子之離子層析方法 方法類型 陽離子變化 管柱: Metrosep C 2 - 250 (4.0x250 mm) 管柱溫度(°C): 環境溫度 注射(HL): 20 檢測: 傳導性檢測器 流速(mL/min): 1.0 洗脫劑: 4.0 mM酒石酸、 0.75 mM二0比咬缓酸,存於水中 pKa測定及預測 利用附接有D-PAS之Sirius GlpKa儀器收集數據。在25°C 下於水溶液中藉由UV且於甲醇水混合物中藉由電位測定 法進行量測》利用0.15 M KC1 (水溶液)對滴定介質進行離 子強度調節(ISA)。經由Yasuda-Shedlovsky外推將甲醇水 混合物中發現之值校正至〇%共溶劑。使用Refinement Pro 162775.doc -38- 201249841 軟體vl.O精修數據。使用ACD pKa預測軟體v9預測pKa 值。Data was collected on a Bruker AXS IK SMART CCD diffractometer. Resolve the structure using SHELXS or SHEXLD programs and refine it with the SHELXL program that is part of the Bruker AXS SHELXTL suite. Unless otherwise stated, the hydrogen atom attached to carbon is geometrically placed and refined using a riding isotropic displacement parameter. The hydrogen atoms attached to the heteroatoms are located in different Fourier synthesis and are freely refined with isotropic displacement parameters. Nuclear Magnetic Resonance (NMR) Spectroscopy NMR spectra were collected on a Varian Unity 300 MHz instrument or a Bruker 400 MHz instrument equipped with an autosampler and controlled by a DRX400 controller station. Automation experiments were performed using a standard Bruker loading experiment using ICONNMR V4.0.4 (form 1) running with Topspin v 1.3 (patch level 8). For unconventional spectra, data is obtained only by using Topspin. Melting Point A Fisher-Johns hot stage, melting point apparatus was used at a setting corresponding to a heating rate of about 5 ° C / min. Differential Scanning Calorimetry (DSC) DSC data was collected on a Q Instruments Q1000 or Mettler DSC 823e equipped with a 50-bit autosampler. Calibrate the instrument for energy and temperature calibration using qualified indium. 0.5-1.5 mg of each sample in a pin-hole aluminum pan is usually heated from 25 ° C to 175 ° C to 200 ° C at 10 ° C/min. A nitrogen purge was maintained at 30 mL/min on the sample. Thermogravimetric Analysis (TGA) 162775.doc -33 - 201249841 TGA data was collected on a TA Instruments Q500 TGA equipped with a 16-bit autosampler or a Mettler TGA/SDTA 851e equipped with a 34-position autosampler. ΤΑ Instruments Q5〇〇 : Calibrate the temperature of the instrument with a qualified aluminum-alloy (Alumel). Each sample of 5_1 〇 mg is typically loaded onto a pre-weighed gutta-percha and aluminum DSC pan and heated to 350 C from ambient temperature at i〇°c / min. A gas purge was maintained at 60 mL/min on the sample. Mettler TGA/SDTA 851e: Calibrate the instrument with a qualified indium. Typically 5-10 mg of each sample was loaded onto a pre-weighed aluminum crucible and heated from ambient temperature to 350 °C at 1 Torr &lt; t/min. A nitrogen purge was maintained at 50 mL/min on the sample. Polarized Microscopy (PLM) Samples were studied using a Leica LM/DM polarizing microscope with a digital video camera for capturing images. A small amount of each sample was placed on a glass slide&apos; mounted in immersion oil and covered with a glass slide, with individual particles being separated as much as possible. The sample was observed with appropriate magnification and the light was partially polarized and coupled to the dummy filter. Hot Stage Microsharp (HSM) Hot stage microscopy was performed using a Leica LM/DM polarizing microscope in combination with a Mettler-Toledo MTFP82HT hot stage and a digital video camera for capturing images. A small amount of each sample was placed on a glass slide and individual particles were separated as much as possible. The sample is observed with appropriate magnification and the light is partially polarized, coupled to a lambda false color filter, while typically heating at ambient temperature from 10 〇c / min. Dynamic Vapor Sorption (DVS) Adsorption isotherms were determined using an SMS DVS Intrinsic Moisture Absorption Analyzer controlled by the SMS Analysis Suite software. The sample temperature was maintained at 25 ° C by instrument control. The humidity was controlled by mixing dry and wet nitrogen streams at a total flow rate of 200 mL/min. Relative humidity (dynamic range 1.0-100% RH) was measured by the amount of calibrated R〇tr〇nic probe located near the sample 162775.doc •34- 201249841. The weight change (mass relaxation) of the sample as a function of RH% was continuously monitored by a microbalance (accuracy ± 0.005 mg). A 5-20 mg sample is typically placed on a weighed wire mesh stainless steel basket under ambient conditions. The sample was loaded at 40% RH and 25 ° C (typical environmental conditions) and was not loaded. The water adsorption isotherm is implemented as outlined below (2 scans yield 1 full cycle). Standard isotherms were performed at 25 °C at 10% RH intervals in the range of 0-90% RH. DVS Generic Method Parameter Parameter Value Adsorption - Scan 1 40-90 Desorption / Adsorption - Scan 2 90 - Dry, Dry - 40 Interval (RH%) 10 Number of Scans 2 Flow Rate (mL/min) 200 Temperature (°C) 25 Stable Sex (°C/min) 0.2 Adsorption time (hours) 6 hours pause The sample was recovered after completion of the isotherm and reanalyzed by XRPD. Water Determination by Karl Fischer Method (KF) The water content of each sample was measured using a Mettler Toledo DL39 Coulometer using Hydranal Coulomat AG reagent and argon purge. The weighed solid sample was introduced into a vessel on a platinum TGA pan which was connected to the subaseal to avoid water ingress. Approximately 1 〇 mg sample was used for each titration and assayed in duplicate. 162775.doc -35- 201249841 Thermodynamic Water Solubility by HPLC Water solubility was determined by suspending sufficient compound in water to produce a maximum final concentration of 210 mg/mL parent free form compound. The suspension was equilibrated at 25 ° C for 24 h and then the pH was measured. The suspension was then filtered through a glass fiber C filter into a 96 well plate. The filtrate was then diluted 101 times. Quantification was performed by HPLC with reference to a standard solution of about 0.1 mg/mL in DMSO. Standard, diluted and undiluted solutions of different volumes are injected. Solubility was calculated using the peak area determined by integrating the peaks found at the same retention time as the main peak in the standard injection. If sufficient solids are present in the filter plate, XRPD is collected. HPLC method for thermodynamic water solubility method Parameter method Type: Reversed-phase column with gradient elution: Phenomenex Luna, C18 (2) 5 μηι, 5〇x4.6 mm Column temperature (°c): 25 Standard injection ( μ!〇: 1, 2, 3, 5, 7, 10 Test injection (jiL) · Bu 2, 3, 10, 20, 50 Detection: Wavelength, bandwidth (nm): 260, 80 Flow rate (mL/min): 2 Phase A: 0.1% TFA in water Phase B: 0.085% TFA in acetonitrile Schedule: Time (min) Phase A% Phase B% 0.0 95 5 1.0 80 20 2.3 5 95 3.3 5 95 3.5 95 5 4.4 . 95 5 162775.doc -36· 201249841 The analysis was carried out on an Agilent HP 1100 series system equipped with a diode array detector and using ChemStation software vB.02.01-SR1. The chemical purity by HPLC is equipped with a diode Purity analysis was performed on a body array detector using an Agilent HP 11® series system using ChemStation software vB.02.01-SR1. HPLC method for chemical purity determination Specimen sample preparation 0.5 mg/mL ' stored in acetonitrile: water i:i ( In v/v), tube: Phenomenex Luna C18 (2), 150x4.6 mm, 5 μπι Column temperature (°c): 25 Injection (pL): 5 Detection: Wavelength, Bandwidth: (nm): 255, 90 Flow Rate (mL/min): 1 Phase A: 0.1% TFA in Water Phase B: 0.085% TFA in Acetonitrile Schedule: Time (min) Phase A% phase B% 0 95 5 25 5 95 25.2 95 5 30 95 5 Ion chromatography on Metrohm 761 Advanced Compact 1C (for cations) and Metrohm 861 Advanced Compact 1C (for anions) using IC Net software v2.3 The data was collected. The sample was prepared as a 1000 ppm stock solution in DMSO. The sample was diluted to 100 ppm in DMSO prior to testing. By 162775.doc •37·201249841 Positive ions were analyzed with known concentrations Standard solution comparison is achieved. Anion ion chromatography method method type anion change column: Metrosep A Supp 5 - 250 (4.0x250 mm) Column temperature (°c): Ambient temperature injection (0): 20 Detection: Conductivity Detector flow rate (mL/min): 0.7 Eluent: 3.2 mM sodium carbonate, 1.0 mM sodium bicarbonate, ion chromatography in cations in water Method Type Cation change column: Metrosep C 2 - 250 (4.0x250 mm Column temperature (°C): ambient temperature Shot (HL): 20 Detection: Conductivity detector flow rate (mL/min): 1.0 Eluent: 4.0 mM tartaric acid, 0.75 mM dioxin-like bite acid, stored in water pKa determination and prediction using attached D- PAS's Sirius GlpKa instrument collects data. The titration medium was subjected to ion intensity adjustment (ISA) using a 0.15 M KC1 (aqueous solution) in an aqueous solution by UV and in a methanol water mixture at 25 °C. The values found in the methanol water mixture were corrected to the 〇% cosolvent via Yasuda-Shedlovsky extrapolation. Use the Refinement Pro 162775.doc -38- 201249841 software vl.O refinement data. The pKa value was predicted using the ACD pKa prediction software v9.

Log P測定 藉由在Sirius GlpKa儀器上電位滴定使用辛醇:離子強度 調節(ISA)水之三種比率以產生L〇g p、Log P離子及Log D值 來收集數據。使用ReHnement Pro軟體vl.O精修數據。使用 ACD 9 版及 Syracuse KOWWIN 1.67版軟體預測 Log P值。 實例2. 3·環丙基羰基-3,6-二氮雜雙環[3.1.1]庚烷(化合物A) 之合成 向配備有機械攪拌器、兩個回流冷凝器、冷凝管、溫度 計、氮入口及排放出口(引導至水性亞硫酸氫鈉/氫氧化鈉 分水器)之三頸燒瓶中裝填戊二醯氣(35〇 g,2.07 m〇1),之 後裝填溴(160 mL,3.12 mol)。將所得混合物於氮下攪拌並 於緩慢回流下加熱,此時經7 h時段初始溫度自58〇c逐漸 增加至91°C。在加熱時段期間且在觀察到溴消耗/損失 時,添加兩次額外溴(第一次9〇 mL且稍後12〇 mL卜隨後 使反應混合物逐漸冷卻至環境溫度,同時於氮.氣氛下攪拌 過夜。於甲醇中中止反應之反應混合物之等份試樣的分析 指不完全轉化為2,4-二溴戊二酸二甲基酯(基於(5(:]^8及 LCMS分析得&gt;98 %)。此2,4-二溴戊二醯氯未經進一步純化 即用於下一步驟。 向配備有機械攪拌器、氮入口、添加漏斗及溫度計之三 頸燒瓶中裝填無水醚(7 L),之後裝填苄醇(493 g,4 56 m〇1)。將溶液於氮下攪拌並在冰水浴中冷卻直至達到8t 162775.doc •39· 201249841 之内部溫度為止。隨後經〗11至1.5 h經由添加漏斗將2,心二 溴戊二醯氣添加至攪拌溶液中(借助於無水醚沖洗),在此 時間期間出現12°c之最大觀察放熱曲線。在添加後,將反 應混合物在冷浴液中攪拌丨h (觀察到内部溫度為12它)。 隨後使反應混合物在氮氣氛下攪拌,同時逐漸升溫至環境 溫度過夜。稀釋於甲醇中之反應混合物之等份試樣的基於 LCMS之分析指示完全消耗2,4_二溴戊二醯氣(即,未觀察 到2,4-二溴戊二酸二甲基酯)。將反應混合物轉移至分液漏 斗(借助於謎沖洗)且將有機溶液用水洗滌兩次,用1 硫酸氫鈉水溶液洗滌兩次,用飽和碳酸氫鈉水溶液洗滌兩 次(第二次洗滌用pH試紙測試為鹼性)且最後用飽和氣化鈉 水溶液洗務一次。將所收集有機相經無水硫酸鈉乾燥,過 濾並在減壓下濃縮至乾燥’從而得到淺黃色油狀中間體 2,4-一 &gt;臭戊一酸二节基醋(1.034 kg,定量粗產率),其未經 進一步純化即用於下一步驟。NMR (CDC13,300 MHz): δ 7.38 (s, 10Η), 5.23 (s, 4H), 4.59-4.41 (m, 2H), 2.97-2.64 (m,2H)。 向配備有機械攪拌器、氮入口、溫度計、添加漏斗及回 流冷凝器之三頸燒瓶中裝填2,4-二溴戊二酸二苄基酯(中間 體3)(1.034 kg,2.199 mol)存於二甲基甲醯胺(2.93 L)中之 溶液。於氮及環境溫度(内部溫度為18。〇下攪拌混合物, 其後藉由添加漏斗一次性添加苄胺(707 g ’ 720 mL,6.60 mol)(添加時間&lt; 5 min),在此時間期間觀察到55〇c至⑼它 之最大溫度之放熱曲線。在完成添加後即刻將反應混合物 162775.doc -40· 201249841 於93°C至95°C下加熱4.5 h稀釋於曱醇中之反應混合物之 等份試樣的LCMS分析指示完全消耗中間體3且形成產物 (以及其反式對等部分)。停止加熱且在氮下將反應混合物 逐漸冷卻至環境溫度過夜。隨後將反應溶液倒入1:1乙酸 乙酯/己烷(8 L)與水(6 L)之攪拌二相混合物中。攪動後, 分離有機相與水相。用水、飽和碳酸氫鈉水溶液(水用pH 試紙測試為鹼性)及飽和氣化鈉水溶液連續洗滌所收集有 機相。將有機層在無水硫酸鈉上乾燥,過濾並在減壓下濃 縮至乾燥,從而提供790 g深色油β將粗物質加載於矽膠 塞上(借助於最少量之二氣甲烷洗滌來加載),其後用15〇/〇 乙酸乙酯/己烷洗脫《在減壓下濃縮所選部分,從而產生 543 g N-节基氮雜環丁烷_2,4-二甲酸二节基醋,其具有 82:17之順式(中間體4):反式立體化學之比及92%之總體 化學純度。自二乙醚結晶並風乾,從而提供31〇 g (自戊二 醯氣之總體產率為3 6%)白色固體狀順式苄基氮雜環丁 烷-2,4-二甲酸二苄基酯(中間體4)(基於LCMS純度&gt;96%)。 H NMR (CDC13, 300 MHz): δ 7.30 (m, 10Η), 5.07 (dd, 4H) 3.89 (s, 2H), 3.67 (dd, 2H), 2.55 (dd, 1H), 2.36 (dd, 1H); LCMS (m/z): 416 (M+l)。 向配備有機械攪拌器、溫度計及氮入口之三頸燒瓶中裝 填順式-N-苄基氮雜環丁烷-2,4-二甲酸二苄基g旨(173 g, 416 mmol),之後裝填甲醇(1.14 L)及四氫呋喃(56〇 mL)。 於環境溫度及氮下攪拌所得混合物直至獲得均句為止。@ 後在乾冰/丙酮浴液中冷卻溶液直至達到-4°C之内部溫度為 162775.doc • 41 · 201249841 止。經1 h至1·5 h—次性添加硼氫化鈉(79 〇呂,2⑽ 叫,在此時間期間觀察到最大溫度之輕微放熱曲 線。在添加後’在氮下授拌所得混合物,同時逐漸升溫至 環境溫度過夜[注意··在升溫期間,觀察到高達27t之最 大溫度之第二放熱曲線且係藉由使用冰浴加以控制]。自 於甲醇中稀釋之反應混合物中取等份試樣進行的lcms分 析指示起始物質已消耗並形成產物(及苄醇)(&gt; 隨後在冰水 浴中冷卻反應混合物(内部溫度為8。〇並經由滴加漏斗逐滴 添加水(100 mL)中止反應。在減壓下濃縮所得混合物,以 去除大部分揮發物。將剩餘部分分配在丨0〇/〇曱醇/二氣甲烷 (500 mL)與水(200 mL)之間。相分離後,收集有機相(上 層)及水相。用10%甲醇/二氣甲烷(2x5〇〇 mL;現在有機相 成為下層)再次萃取水相。此階段時之水相之等份試樣的 分析顯示不存在產物。用水及飽和氣化納水溶液連續洗滌 合併有機萃取物。此水相(水加上飽和氣化鈉水溶液)之分 析顯示有產物(加苄醇)存在,因此用10〇/〇甲醇/二氯甲烧 (3 X)反萃取水相。合併所有曱醇/二氣曱烷萃取物並經無水 硫酸鈉乾燥。過濾及減壓濃縮後,獲得177 g黃色油狀 物。在75°C水浴及高度真空下進一步濃縮此殘餘物,從而 去除大部分苄醇。在觀察到重量恆定後,剩餘100 g油狀 物。將此物質溶解於二氣甲烷中並在減壓下濃縮,從而產 生半固體。將半固體懸浮於己烷中兩次且隨後在減壓下濃 縮。將剩餘固體懸浮於己烷(260 mL)中且將所得懸浮液攪 162775.doc • 42· 201249841 拌過夜。隨後過濾懸浮液並用己烷(2χ)洗滌所收集固體β 溶解於甲醇中之固體試樣之LCMS指示產物純度為約 90°/〇(含苄醇雜質)。將固體再次懸浮於己烷(2〇〇 mL)中並 將懸浮液於30°C下攪拌(在旋轉蒸發器上)1小時。過濾懸浮 液,並再用己烷(lx)洗滌所收集固體。溶解於曱醇中之固 體試樣之LCMS分析指示產物純度&gt;96%。將固體進一步風 乾至恆定重量,從而得到81.9 g (95%產率)白色固體狀順 式-N-节基-2,4-雙(經基曱基)氮雜環丁烷(中間體5)。iH NMR (CDC13, 300 MHz): δ 7.28 (m, 5H), 4.24 (dd, 2H), 3.63 (s, 2H), 3.12 (dd, 4H), 3.05 (m, 2H), 2.00 (dd, 1H), 1.57 (dd, 1H); LCMS (m/z): 208 (M+l) » 向配備有機械攪拌器、溫度計、氮入口及添加漏斗之三 頸燒瓶中裝填順式-N-苄基-2,4-雙(羥基曱基)氮雜環丁烷 (81·9 g ’ 395 mmol),之後裝填無水二氯甲烷(820 mL)。在 氮氣下攪拌所得溶液,其後一次添加全量三乙胺(160 g, 220 mL,1.58 mol)。將所得溶液在乾冰/丙酮浴液中冷卻直 至達到-6 C之内部溫度為止。隨後經4 5 m i η藉由添加漏斗 逐滴添加曱炫•績醯氣(109 g,73.4 mL,948 mmol),在此時 間期間出現高達4°C之最大觀察溫度的放熱曲線。在添加 後,使反應混合物逐漸升溫至環境溫度,同時在氮下擾拌 過夜。稀釋於乙腈中之反應混合物之等份試樣的LCMS分 析指示消耗起始物質並形成產物。向反應混合物中逐滴添 加水(250 mL),之後添加飽和碳酸氫鈉水溶液(250 mL)。 162775.doc -43- 201249841 將所得二相混合物劇烈攪拌2〇 min並借助於二氣甲烷沖洗 轉移至分液漏斗。分離有機相及水相且隨後用飽和氣化鈉 水溶液洗滌有機層。在相分離(緩慢)後,收集有機層,經 無水硫酸鈉乾燥,過濾並在減壓下濃縮,從而留下呂 (100%產率)褐色油狀雙-去鐵胺(中間體6),其未經純化即 用於下一步驟。丨H NMR (CDC13,300 ΜΗζ): δ 7.33 (m, 5Η), 3.97 (ddd, 4H), 3.74 (s, 2H), 3.44 (m, 2H), 2.92 (s, 6H), 2.28 (m, 1H), 1.91 (m, 1H); LCMS (m/z): 364 (M+l) 〇 在氮氣氛下向配備有機械攪拌器、溫度計、氮入口及回 流冷凝器之三頸燒瓶中裝填環丙烷甲醯胺(36 8 g , 432 mmol)。添加無水1·甲基_2_吡咯啶酮(N甲基_2吡咯啶 酮)(900 mL)並在氮下機械攪拌所得混合物,同時在冰水浴 中冷卻(内部溫度為5°C)。隨後經3〇 min—次性添加氫化鈉 (39.4 g存於礦物油中之60%分散液,98〇 mm〇1),此時溫度 在5 C與12 C之間變化《將所得混合物在冷浴液上再攪拌 10 mir^使冷’谷液升溫至環境溫度(經3〇 min),此時觀察 到輕微放熱曲線(至25C之最大溫度)及氣體釋放。將所得 混合物於環境溫度下再攪拌5〇 min (無其他氣體釋放),其 後向懸浮液中以一個批次添加雙-去鐵胺(中間體6)(143 g 393 mmol)存於無水ι_甲基_2_。比洛咬嗣(6〇〇 mL)中之溶 液。用其他無水1-甲基-2-吡咯啶酮(2x1 〇〇 mL)沖洗含有粗 製中間體6之燒瓶,向反應懸浮液中增加每次沖洗。隨後 將所得混合物於68°C下加熱3 h。加熱並在濕乙腈中令止 I62775.doc •44· 201249841 反應2 h後抽出之等份試樣的LCMS*析指示已消耗&gt;96%中 間體6且主要為期望產物。使反應混合物在氮下冷卻至環 境溫度過夜。在冰水浴冷卻中藉由添加水(約1〇 mL)中止 反應黏性溶液並用曱基第三丁基醚(1〇〇 mL)稀釋並攪拌1〇 min。隨後將混合物轉移至含有5%氯化鈉水溶液(4〇 L)及 25%乙酸乙酯/曱基第三丁基醚(3 〇 L)之分液漏斗中。攪動 所得混合物。在相分離後,取出有機層及水層。用其他 25%乙酸乙酯/曱基第三丁基醚(2x3 〇 L)萃取水相。將合併 之有機萃取物先後用水(lx3.〇 L)及飽和氣化鈉水溶液 (1 χ3.0 L)洗滌。隨後將有機相經無水硫酸鈉乾燥,過濾並 在減壓下濃縮’從而得到86 g褐色固體。藉由矽膠層析最 初用0至40%乙酸乙酯/己烷且最後用60%至ι〇〇〇/〇乙酸乙酯/ 己烷+0.5°/〇三乙胺洗脫來純化此殘餘物。濃縮所選部分, 產生56 g 6-苄基-3-環丙基羰基-3,6-二氮雜雙環[3.1.1]庚 院,HPLC純度為78%。此未經進一步純化即用於下一步 驟。Log P measurements Data were collected by potentiometric titration on a Sirius GlpKa instrument using three ratios of octanol: ionic strength adjustment (ISA) water to produce L〇g p, Log P ions and Log D values. Refine the data using ReHnement Pro software vl.O. The Log P value is predicted using ACD version 9 and Syracuse KOWWIN version 1.67 software. Example 2. Synthesis of cyclopropylcarbonyl-3,6-diazabicyclo[3.1.1]heptane (Compound A) equipped with a mechanical stirrer, two reflux condensers, a condenser, a thermometer, nitrogen The three-necked flask at the inlet and discharge outlets (directed to the aqueous sodium bisulfite/sodium hydroxide water separator) was filled with urethane (35 〇g, 2.07 m〇1), followed by bromine (160 mL, 3.12 mol). ). The resulting mixture was stirred under nitrogen and heated under slow reflux, at which time the initial temperature gradually increased from 58 ° C to 91 ° C over a period of 7 h. During the heating period and when bromine consumption/loss was observed, additional bromine was added twice (first 9 〇mL and later 12 〇mL) and then the reaction mixture was gradually cooled to ambient temperature while stirring under nitrogen. Overnight. Analysis of an aliquot of the reaction mixture in the methanol suspension reaction means incomplete conversion to dimethyl 2,4-dibromoglutarate (based on (5 (:) 8 and LCMS analysis> 98%). This 2,4-dibromopentane ruthenium chloride was used in the next step without further purification. A three-necked flask equipped with a mechanical stirrer, nitrogen inlet, addition funnel and thermometer was filled with anhydrous ether (7) L), followed by loading benzyl alcohol (493 g, 4 56 m〇1). The solution was stirred under nitrogen and cooled in an ice water bath until the internal temperature of 8t 162775.doc •39·201249841 was reached. 1.5 h was added to the stirred solution via an addition funnel (washed with anhydrous ether), during which time a maximum observed exotherm of 12 ° C occurred. After the addition, the reaction mixture was Stir in the cold bath (h (the internal temperature is observed to be 12) The reaction mixture was then stirred under a nitrogen atmosphere while gradually warming to ambient temperature overnight. LCMS-based analysis of an aliquot of the reaction mixture diluted in methanol indicated complete consumption of 2,4-dibromopentadienyl ( That is, 2,4-dibromoglutaric acid dimethyl ester was not observed. The reaction mixture was transferred to a separatory funnel (by riddle washing) and the organic solution was washed twice with water and washed with 1 aqueous sodium hydrogen sulfate solution. Two times, it was washed twice with a saturated aqueous solution of sodium hydrogencarbonate (the second washing was tested to be alkaline with a pH test paper) and finally washed once with a saturated aqueous solution of sodium sulphate. The collected organic phase was dried over anhydrous sodium sulfate and filtered. Concentration to dryness under reduced pressure afforded the title compound 2, 4- &gt; succinic acid dibasic vinegar (1.034 kg, quantitative crude yield) which was used in the next One step. NMR (CDC13, 300 MHz): δ 7.38 (s, 10 Η), 5.23 (s, 4H), 4.59-4.41 (m, 2H), 2.97-2.64 (m, 2H). , three-necked flask with nitrogen inlet, thermometer, addition funnel and reflux condenser A solution of dibenzyl 2,4-dibromoglutarate (intermediate 3) (1.034 kg, 2.199 mol) in dimethylformamide (2.93 L) at nitrogen and ambient temperature (internal temperature) The mixture was stirred for 18. The mixture was added benzylamine (707 g '720 mL, 6.60 mol) (addition time &lt; 5 min) by the addition funnel, during which time 55 〇c to (9) it was observed. The exothermic curve of the maximum temperature. LCMS analysis of the aliquot of the reaction mixture diluted in the decyl alcohol by heating the reaction mixture 162775.doc -40· 201249841 at 93 ° C to 95 ° C for 4.5 h immediately after the completion of the addition. Indicates complete consumption of intermediate 3 and formation of the product (and its trans-peer portion). Heating was stopped and the reaction mixture was gradually cooled to ambient temperature under nitrogen overnight. The reaction solution was then poured into a stirred two-phase mixture of 1:1 ethyl acetate/hexane (8 L) and water (6 L). After agitation, the organic phase and the aqueous phase are separated. The organic phase was collected by continuous washing with water, a saturated aqueous solution of sodium hydrogencarbonate (water was tested with a pH test paper), and a saturated aqueous solution of sodium sulphate. The organic layer was dried over anhydrous sodium sulphate, filtered and concentrated to dryness under reduced pressure to afford 790 g of dark oil &lt;RTI ID=0.0&gt;&gt; Thereafter, it was eluted with 15 〇 / 〇 ethyl acetate / hexane "concentrating the selected portion under reduced pressure to give 543 g of N-pyrazine azetidine 2,4-dicarboxylic acid dibasic vinegar, It has a cis (82 intermediate) of 82:17: trans stereochemistry ratio and 92% overall chemical purity. Crystallized from diethyl ether and air-dried to provide 31 〇g (3 6% overall yield from pentanedioxane) benzyl benzylazetidine-2,4-dicarboxylate as a white solid. (Intermediate 4) (based on LCMS purity &gt; 96%). H NMR (CDC13, 300 MHz): δ 7.30 (m, 10 Η), 5.07 (dd, 4H) 3.89 (s, 2H), 3.67 (dd, 2H), 2.55 (dd, 1H), 2.36 (dd, 1H) LCMS (m/z): 416 (M+l). A three-necked flask equipped with a mechanical stirrer, a thermometer and a nitrogen inlet was charged with cis-N-benzylazetidin-2,4-dicarboxylic acid dibenzyl g (173 g, 416 mmol), after which Methanol (1.14 L) and tetrahydrofuran (56 〇 mL) were charged. The resulting mixture was stirred at ambient temperature and under nitrogen until a uniform sentence was obtained. @ After cooling the solution in a dry ice/acetone bath until the internal temperature of -4 °C is 162775.doc • 41 · 201249841. Sodium borohydride (79 〇, 2 (10) was added in 1 h to 1.5 h, and a slight exothermic curve of maximum temperature was observed during this time. After the addition, the mixture was mixed under nitrogen while gradually Warming to ambient temperature overnight [Note · During heating, a second exotherm of maximum temperature of up to 27 t was observed and controlled by using an ice bath]. Aliquots were taken from the reaction mixture diluted in methanol The lcms analysis carried out indicated that the starting material had been consumed and formed the product (and benzyl alcohol) (&gt; The reaction mixture was then cooled in an ice water bath (internal temperature was 8. 〇 and water was added dropwise via a dropping funnel (100 mL)) The reaction mixture was concentrated under reduced pressure to remove most of the volatiles. The residue was partitioned between &lt;RTI ID=0.0&gt;&gt; The organic phase (upper layer) and the aqueous phase were collected. The aqueous phase was extracted again with 10% methanol/diqi methane (2 x 5 〇〇 mL; now the organic phase became the lower layer). Analysis of the aliquot of the aqueous phase at this stage showed no Product exists. Water and full The combined organic extracts were continuously washed with a gasified aqueous solution. The analysis of the aqueous phase (water plus saturated aqueous sodium chloride) showed the presence of the product (with benzyl alcohol), so it was burned with 10 〇 / 〇 methanol / chloroform (3 X) Back extraction of the aqueous phase. Combine all the decyl alcohol / dioxane extracts and dry over anhydrous sodium sulfate. After filtration and concentration under reduced pressure, 177 g of a yellow oil was obtained. The residue was concentrated to remove most of the benzyl alcohol. After a constant weight was observed, 100 g of oil remained. This material was dissolved in di- methane and concentrated under reduced pressure to give a semi-solid. Suspension in hexane twice and then concentrated under reduced pressure. The remaining solid was suspended in hexane (260 mL) and the resulting suspension was stirred 162 775.doc • 42· 201249841 overnight. The suspension was then filtered and hexanes (2χ) Washing collected solids β The LCMS of the solid sample dissolved in methanol indicated a product purity of about 90°/〇 (containing benzyl alcohol impurities). The solid was resuspended in hexane (2〇〇mL) and The suspension is stirred at 30 ° C (in The evaporator was transferred to the evaporator for 1 hour. The suspension was filtered, and the collected solid was washed again with hexane (1×). LCMS analysis of the solid sample dissolved in decyl alcohol indicated product purity &gt;96%. The solid was further air dried to Constant weight, giving 81.9 g (95% yield) of cis-N-pyryl-2,4-bis(p-fluorenyl)azetidine (interm. 5) as a white solid. iH NMR (CDC13 , 300 MHz): δ 7.28 (m, 5H), 4.24 (dd, 2H), 3.63 (s, 2H), 3.12 (dd, 4H), 3.05 (m, 2H), 2.00 (dd, 1H), 1.57 ( Dd, 1H); LCMS (m/z): 208 (M+l) » Filled with cis-N-benzyl-2,4 in a three-necked flask equipped with a mechanical stirrer, thermometer, nitrogen inlet and addition funnel - Bis(hydroxyindenyl)azetidine (81·9 g '395 mmol), followed by anhydrous dichloromethane (820 mL). The resulting solution was stirred under nitrogen, and then the whole amount of triethylamine (160 g, 220 mL, 1.58 mol) was added at once. The resulting solution was cooled in a dry ice/acetone bath until the internal temperature of -6 C was reached. An exothermic curve with a maximum observed temperature of up to 4 °C was then added during this time by adding a funnel with a concentration of 4 5 m i η by adding a funnel (109 g, 73.4 mL, 948 mmol). After the addition, the reaction mixture was gradually warmed to ambient temperature while stirring overnight under nitrogen. LCMS analysis of aliquots of the reaction mixture diluted in acetonitrile indicated consumption of starting material and formation of product. Water (250 mL) was added dropwise to the reaction mixture, followed by aqueous saturated sodium hydrogen sulfate (250 mL). 162775.doc -43- 201249841 The resulting biphasic mixture was stirred vigorously for 2 〇 min and transferred to a separatory funnel by means of a two-gas methane rinse. The organic phase and the aqueous phase were separated and then the organic layer was washed with a saturated aqueous solution of sodium sulfate. After phase separation (slow), the organic layer was taken, dried over anhydrous sodium sulfate, filtered and evaporated, then evaporated. It was used in the next step without purification.丨H NMR (CDC13,300 ΜΗζ): δ 7.33 (m, 5Η), 3.97 (ddd, 4H), 3.74 (s, 2H), 3.44 (m, 2H), 2.92 (s, 6H), 2.28 (m, 1H), 1.91 (m, 1H); LCMS (m/z): 364 (M+l) 装 Fill the ring in a three-necked flask equipped with a mechanical stirrer, thermometer, nitrogen inlet and reflux condenser under nitrogen atmosphere Propane methotrexate (36 8 g, 432 mmol). Anhydrous 1·methyl-2-pyrrolidinone (Nmethyl-2-pyrrolidone) (900 mL) was added and the resulting mixture was mechanically stirred under nitrogen while cooling in an ice water bath (internal temperature 5 ° C). Subsequently, sodium hydride (39.4 g of 60% dispersion in mineral oil, 98 〇mm〇1) was added via 3 〇 min. The temperature varied between 5 C and 12 C. The bath was stirred for another 10 mir^ to cool the cold liquid to ambient temperature (after 3 Torr), at which time a slight exotherm (to a maximum temperature of 25 C) and gas evolution were observed. The resulting mixture was stirred for an additional 5 Torr at ambient temperature (no other gas evolution), after which a mixture of bis-deferoxamine (intermediate 6) (143 g 393 mmol) was added to the suspension in one portion. _methyl_2_. Bilo bite (6〇〇 mL) in solution. The flask containing the crude intermediate 6 was rinsed with other anhydrous 1-methyl-2-pyrrolidone (2 x 1 〇〇 mL) and each rinse was added to the reaction suspension. The resulting mixture was then heated at 68 ° C for 3 h. Heating and chilling in wet acetonitrile I62775.doc •44·201249841 The LCMS* analysis of the aliquots taken after 2 h of reaction indicated that &gt;96% of intermediate 6 was consumed and was predominantly the desired product. The reaction mixture was cooled to ambient temperature under nitrogen overnight. The reaction viscous solution was stopped by adding water (about 1 〇 mL) in an ice water bath and diluted with decyl tertiary butyl ether (1 〇〇 mL) and stirred for 1 〇 min. The mixture was then transferred to a separatory funnel containing a 5% aqueous solution of sodium chloride (4 mL) and 25% ethyl acetate / decyl-t-butyl ether (3 〇L). The resulting mixture was agitated. After phase separation, the organic layer and the aqueous layer were taken out. The aqueous phase was extracted with a further 25% ethyl acetate / decyl-tert-butyl ether (2 x 3 〇 L). The combined organic extracts were washed with water (1×3. 〇 L) and saturated aqueous sodium sulfate (1 χ 3.0 L). The organic phase was dried over anhydrous sodium sulfate, filtered and evaporatedEtOAc. Purification of this residue by silica gel chromatography initially eluting with 0 to 40% ethyl acetate / hexanes and finally eluting with 60% to EtOAc / EtOAc / hexane . The selected fractions were concentrated to give &lt;RTI ID=0.0&gt;&gt;&gt;&&&&&&&&&&&&& This was used in the next step without further purification.

在氮下向配備有機械擾拌器、溫度計、回流冷凝器及氣 入口之三頸燒瓶中裝填6-苄基-3-環丙基羰基_3,6·二氮雜雙 環[3.1.1]庚烷(56 g,78%,220 mmol)存於乙醇(200度, 670 mL)。隨後添加10〇/〇 Pd/C (33.6 g,濕),之後添加甲酸 銨(82.6 g ’ 1.31 mol)。將所得混合物在氮氣氛下劇烈攪拌 並在6(TC至66°C下逐漸加熱並保持6 h,約3.5 h後添加額 外20 g 10% Pd/C及60 g甲酸錄。隨後加熱反應物(在66°C 162775.doc •45- 201249841 與71 C之間保持6 h)並藉由LCMS週期性監測,同時以若干 次添加更多試劑(總額外甲酸銨=96 g ;總額外1〇% Pd/C=21 gp反應混合物之等份試樣之分析隨後指示消耗 起始物質。停止加熱並在氮下使反應混合物逐漸冷卻至環 境溫度過夜。隨後經由矽藻土床過濾反應混合物。隨後用 曱醇(4 X)洗滌濾餅,且在減壓下濃縮合併之濾液,從而提 供35 g淺黃色油。藉由矽膠層析用存於二氯甲烷中之〇至 60% DCMA80洗脫來純化該物質。[注意:DCMA80係二氣 甲烷、甲醇及氫氧化銨水溶液之8〇: 18:2混合物]。合併所 選部分並在減壓下濃縮,從而得到9 2 g幾乎白色固體, HPLC純度為97.7%。可藉由用曱基第三丁基醚滴定將此物 質之純度增加至98.7°/。(HPLC)。亦濃縮來自層析之較不純 淨部分’產生額外11 g物質(HPLC顯示純度為約85%)。4 NMR (D2Os 300 MHz): δ 4.02 (s, 2H), 3.73 (m, 2H), 3.64 (dd, 2H), 2.66 (m, 1H), 1.92 (m, 1H), 1.52 (d, 1H), 0.88 (m, 4H); LCMS (m/z): 167 (M+l)。 實例3 : 3-環丙基羰基-3,6-二氮雜雙環丨3.1.1]庚烷半-半乳 糖一酸鹽單水合物,6-二氣雜雙環[3.1.1】庚燒-3-基(環丙 基)甲酮半-半乳糖二酸鹽單水合物}之合成 在氮下向配備有機械攪拌器、溫度計、回流冷凝器及氮 入口之三頸圓底燒瓶中裝填3-環丙基羰基·3,6-二氮雜雙環 [3.1_1]庚烷(132 g,0.794 mol,代表多輪實例2中所示化學 法)。向反應燒瓶中添加乙醇(200度,925 mL)及水 162775.doc -46 · 201249841 mL) ’且在環境溫度下攪拌混合物直至獲得溶液為止。隨 後’添加半乳糖二酸(86.0 g,0.397 mol)並將所得混合物 於5〇C及氮下加熱2 h。隨後停止加熱,且使所得稠聚液 冷卻至環境溫度,同時在氮下授拌過夜。隨後將混合物在 冰水浴中冷卻至〇艺並抽吸過濾。簡單風乾所收集固體且 隨後將其溶解於熱水(1000 mL ; 60它)中。趁熱過渡溶液 以去除少量不溶物質且隨後濃縮至接近乾燥。向此殘餘物 中添加乙醇(200度,800 mL),且將混合物於5〇。(:下授拌 30 min且隨後在冰水浴中冷卻至〇〇c,其中將其保持45 min。過濾漿液’並在真空中乾燥所收集固體過夜。在真 空及56C下將固體進一步乾燥至怪定重量(經4 h之時段), 從而產生208 g (95%產率)白色固體狀3-環丙基羰基-3,6-二 氮雜雙環[3.1.1]庚烷半-半乳糖二酸鹽單水合物(HPLC純度 為 99%)。lH NMR (D2〇,400 ΜΗζ): δ 4.39 (m,2H),4.20 (dd, 2H), 4.09 (s, 1H), 3.88 (d, 1H), 3.79 (s, 1H), 3.74 (d, 1H), 2.93 (m, 1H), 1.77 (m, 2H), 0.80 (m, 4H); LCMS (m/z): 167 (M+l);乾燥物質之卡爾-費希爾分析指示水含 量為6.5%(對應於單水合物化學計量)。 實例4 :使用酿胺偶合程序之3_環丙基羰基-3,6_二氮雜雙 環[3.1.1】庚燒鹽酸鹽之合成 向存於25 mL圓底燒瓶中之3,6·二氮雜雙環[3.Μ]庚 烧-6-曱酸第二丁基醋(50 mg,0.25 mmol)中添加環丙烧甲 酸(26 mg,24 pL,0.30 mmol)、三乙胺(70 mL,0.50 162775.doc -47- 201249841 mmol)、二氣曱烷(5 mL)及六氟填酸Ο-苯并三唑-1-基-四曱 基脲鑌鹽(191 mg,0.50 mmol)。將反應物於環境溫度下攪 拌2 h。添加飽和氣化銨(5 mL),且將反應混合物攪拌30 min。隨後使混合物通過相萃取器,並在真空下去除溶 劑。將粗混合物溶解於3 mL乙酸乙酯中並添加濃HC1(1 mL)❶將此混合物攪拌2 h。在真空下去除溶劑,且使殘餘 物通過基於氧化石夕之陽離子交換管柱,其先後經2 mL甲醇 及2 mL二氣甲烷/甲醇(1:1)預洗滌。將殘餘物吸收於i 二氣甲烷/甲醇中並使其通過管柱’先後用3 mL二氯曱烷/ 甲醇(1:1)及甲醇/氨(7M)洗脫。濃縮洗脫物,且藉由用存 於氣仿中之CMA90 (氣仿/甲醇/氫氧化銨水溶液9〇:9:1)之 梯度洗脫㈣膠管柱層析純化殘餘物4真空下濃縮所選 部分,並將殘餘物與存於甲醇(2 mL)中之1M氣化氫合併。 經由相萃取器過滤所得混合物(用以去除任何微細粒子), 且在真空中去除溶劑1而留下油狀3,6-二氮雜雙環 ^ 27〇/〇)〇 ^ 舰(CD錢獅 MHz): s 4 49 (m,吼 4 η _ 邱 4.〇1(^«),3,3(^^3,6(111, lH),,94(mj2H);〇 9; (m,4H卜注意:儘管此物質無法結晶,但可誘導鹽酸鹽之 其他试樣結晶(在用丙酮滴定後)。所得固體具有吸濕性。 實例S:其他鹽之製備 基於所量測PKa值,選擇包括於 藥上可接受之酸: 冑擇研九中之以下醫 162775.doc •48· 201249841 酸 類别 pKa LogP MW 儲存溶液 氫氣酸37 wt% (12 Μ) 1 -6.10 - - - 36.46 1 MTHF 硫酸98% 1 -3.00 1.92 - -1.03 98.08 1 MTHF 對甲苯磺酸.Η20 2 -1.34 - - 0.93 190.22 1 MEtOH 甲烷磺酸 2 -1.20 - - -1.89 96.10 1 MTHF 草酸 2 1.27 4.27 - - - 1 MTHF 草酸 2 1.27 4.27 - - 1 MTHF L-天冬胺酸 1 1.88 3.65 -0.67 133.11 固體 馬來酸 1 1.92 6.23 -0.01 116.07 1 MTHF 磷酸 1 1.96 7.12 12.32 -2.15 98.00 1 MTHF L-麩胺酸 1 2.19 4.25 - -1.43 147.13 固體 1-羥基-2-萘甲酸 (羥萘甲酸酯) 2 2.70 13.50 - 3.29 188.17 固體 L-酒石酸 1 3.02 4.36 - -1.43 150.09 1 MTHF 富馬酸 1 3.03 4.38 - -0.01 116.07 0.5 Μ THF/MeOH (1:1) 半乳糖二酸(黏酸) 1 3.08 3.63 - -1.46 210.14 1 MDMSO 檸檬酸 1 3.13 4.76 6.40 -1.72 192.12 1 MTHF D-葡糖醛酸 1 3.18 - - -1.49 194.14 固體 蘋果酸 1 3.46 5.10 -1.26 134.09 1 MTHF 馬尿酸 1 3.55 - - 0.31 179.17 固體 D-葡萄糖酸50%, 存於水中 1 3.76 - - -3.18 196.16 固體 L -扁桃酸 3 3.85 - 1Μ,存於所 用溶劑中 L-乳酸85%水溶液 1 3.86 -0.7 90.08 1 MTHF L-抗壞血酸 1 4.17 11.57 -2.41 176.13 1 Μ水 162775.doc •49- 201249841 琥珀酸 1 4.21 5.64 -0.59 118.09 1 MTHF 乙酸 1 4.76 - - -0.29 60.05 1 MTHF 苯甲酸 2 4.19 - - 1MIPA 己二酸 1 4.44 5.44 - 0.08 146.14 固體 丙酸 2 4.87 - - 0.25 74.07 1 MTHF (+)-樟腦酸 2 4.72 5.83 - 1.47 200.23 固體 4-羥基苯甲酸 2 4.57 9.22 - 138.12 1Μ,存於所 用溶劑中 於50°C下向30.0 mg (0.165 mmol)游離鹼存於所選溶劑中 之溶液中添加相應酸(根據酸之酸性部分的數量及強度, 0.5當量或1.1當量 將混合物於50它下攪拌1 h,隨後緩慢冷卻至〇〇c過夜 (0.1 C /min) »過濾並分析固體。若未產生固體,則在環境 條件下緩慢蒸發相應溶液。 在澄清溶液、膠或油情形下,將該等物質冷卻至_20。(:, 且使其緩慢蒸發以促進結晶。其後,若油/膠不可結晶, 則添加抗溶劑(共溶劑;甲基第三丁基醚);之後用丙酮滴 定。在此增加努力下,許多鹽在放置於乙酸異丙基醋中時 產生沈澱(以下兩個表中所示之結果)。A 3-necked flask equipped with a mechanical stirrer, thermometer, reflux condenser and gas inlet was charged with 6-benzyl-3-cyclopropylcarbonyl_3,6-diazabicyclo[3.1.1] under nitrogen. Heptane (56 g, 78%, 220 mmol) was stored in ethanol (200 °, 670 mL). Then 10 〇 / 〇 Pd / C (33.6 g, wet) was added, followed by the addition of ammonium formate (82.6 g ' 1.31 mol). The resulting mixture was vigorously stirred under a nitrogen atmosphere and gradually heated at 6 (TC to 66 ° C for 6 h, after adding additional 20 g of 10% Pd/C and 60 g of formic acid for about 3.5 h. The reaction was then heated ( At 66 ° C 162775.doc •45- 201249841 and 71 C for 6 h) and periodic monitoring by LCMS, while adding more reagents in several times (total additional ammonium formate = 96 g; total additional 1% Analysis of an aliquot of the Pd/C = 21 gp reaction mixture then indicated consumption of the starting material. Heating was stopped and the reaction mixture was gradually cooled to ambient temperature overnight under nitrogen. The reaction mixture was then filtered through a bed of diatomaceous earth. The filter cake was washed with decyl alcohol (4×), and the combined filtrate was concentrated under reduced pressure to give 35 g of pale yellow oil. Purified by chromatography eluting with EtOAc to 60% DCMA. This material. [Note: DCMA80 is a mixture of dioxane methane, methanol and aqueous ammonium hydroxide: 8:18:2]. The selected fractions are combined and concentrated under reduced pressure to give 9 2 g of almost white solid, HPLC purity 97.7%. This material can be titrated by thiol tert-butyl ether. The purity was increased to 98.7 ° / (HPLC). Concentration from the less pure fraction of the chromatography 'produced an additional 11 g of material (HPLC showed a purity of about 85%). 4 NMR (D2Os 300 MHz): δ 4.02 (s, 2H), 3.73 (m, 2H), 3.64 (dd, 2H), 2.66 (m, 1H), 1.92 (m, 1H), 1.52 (d, 1H), 0.88 (m, 4H); LCMS (m/z ): 167 (M+l). Example 3: 3-cyclopropylcarbonyl-3,6-diazabicycloindole 3.1.1]heptane hemi-galactosyl monophosphate monohydrate, 6-dioxa Synthesis of bicyclo [3.1.1]heptan-3-yl (cyclopropyl)methanone hemi-galactosyl dihydrate monohydrate} equipped with a mechanical stirrer, thermometer, reflux condenser and nitrogen inlet under nitrogen The three-necked round bottom flask was charged with 3-cyclopropylcarbonyl·3,6-diazabicyclo[3.1_1]heptane (132 g, 0.794 mol, representing multiple cycles of the chemistry shown in Example 2). Add ethanol (200 °, 925 mL) and water 162775.doc -46 · 201249841 mL) to the flask and stir the mixture at ambient temperature until a solution is obtained. Then add galactosuccinic acid (86.0 g, 0.397 mol) and The resulting mixture was heated at 5 ° C under nitrogen for 2 h. Then the heating was stopped. The resulting thick polyethylene was cooled to ambient temperature, and stirred overnight under nitrogen granted. The mixture was then cooled in an ice-water bath and filtered under suction to square arts. The solid was collected by simple air drying and then dissolved in hot water (1000 mL; 60 mM). The hot transition solution was removed to remove a small amount of insoluble material and then concentrated to near dryness. To this residue was added ethanol (200 °, 800 mL) and the mixture was taken to 5 hr. (: Mix for 30 min and then cool to 〇〇c in an ice water bath, which was kept for 45 min. Filter the slurry' and dry the collected solids in vacuo overnight. Dry the solid further to blame under vacuum and 56 C Constant weight (during 4 h) to give 208 g (95% yield) of 3-cyclopropylcarbonyl-3,6-diazabicyclo[3.1.1]heptane hemi-galactose Acid salt monohydrate (HPLC purity: 99%). lH NMR (D2 〇, 400 ΜΗζ): δ 4.39 (m, 2H), 4.20 (dd, 2H), 4.09 (s, 1H), 3.88 (d, 1H) ), 3.79 (s, 1H), 3.74 (d, 1H), 2.93 (m, 1H), 1.77 (m, 2H), 0.80 (m, 4H); LCMS (m/z): 167 (M+l) The Karl-Fisher analysis of the dry material indicated a water content of 6.5% (corresponding to the monohydrate stoichiometry). Example 4: 3_cyclopropylcarbonyl-3,6-diazabicyclo ring using a brewing amine coupling procedure [3.1.1] Synthesis of Glycidate Hydrochloride to 3,6-diazabicyclo[3.Μ]heptane-6-decanoic acid second butyl vinegar (50 mg) in a 25 mL round bottom flask , 0.25 mmol) with cyclopropanecarboxylic acid (26 mg, 24 pL, 0.30 mmol), triethylamine (70 mL, 0.50 162775.doc -47- 201249841 m Mol), dioxane (5 mL) and hexafluoroantimonate-benzotriazol-1-yl-tetradecylurea salt (191 mg, 0.50 mmol). The reaction was stirred at ambient temperature 2 h. Add saturated ammonium hydride (5 mL) and stir the reaction mixture for 30 min. Then pass the mixture through a phase extractor and remove the solvent under vacuum. Dissolve the crude mixture in 3 mL of ethyl acetate and add concentrated HC1 (1 mL) ❶The mixture was stirred for 2 h. The solvent was removed in vacuo and the residue was passed through a oxidant-based cation exchange column followed by 2 mL of methanol and 2 mL of methane/methanol (1: 1) Pre-washing. The residue was taken up in i-dioxethane/methanol and passed through a column of '3 mL of dichloromethane/methanol (1:1) and methanol/ammonia (7M). Eluate, and purified by gradient elution with CMA90 (gas/methanol/ammonium hydroxide aqueous solution 9:9:1). Part and the residue was combined with 1 M hydrogenation in methanol (2 mL). The mixture was filtered through a phase extractor (to remove any fine particles) ), and remove the solvent 1 in a vacuum leaving the oily 3,6-diazabicyclo^ 27〇/〇)〇^ ship (CD lion GHz): s 4 49 (m, 吼4 η _ Qiu 4 .〇1(^«),3,3(^^3,6(111, lH),,94(mj2H);〇9; (m,4H note: although this substance cannot crystallize, it can induce hydrochloric acid The other samples of the salt were crystallized (after titration with acetone). The resulting solid is hygroscopic. Example S: Preparation of other salts based on the measured PKa values, selected to include pharmaceutically acceptable acids: 胄 研 九 九 162 162 162 775. doc • 48· 201249841 Acid Category pKa LogP MW Storage Solution Hydrogen Acid 37 wt % (12 Μ) 1 -6.10 - - - 36.46 1 MTHF Sulfuric acid 98% 1 -3.00 1.92 - -1.03 98.08 1 MTHF p-Toluenesulfonic acid. Η20 2 -1.34 - - 0.93 190.22 1 MEtOH Methanesulfonic acid 2 -1.20 - - -1.89 96.10 1 MTHF Oxalic acid 2 1.27 4.27 - - - 1 MTHF Oxalic acid 2 1.27 4.27 - - 1 MTHF L-aspartic acid 1 1.88 3.65 -0.67 133.11 Solid maleic acid 1 1.92 6.23 -0.01 116.07 1 MTHF Phosphate 1 1.96 7.12 12.32 -2.15 98.00 1 MTHF L-glutamic acid 1 2.19 4.25 - -1.43 147.13 Solid 1-hydroxy-2-naphthoic acid (hydroxynaphthoate) 2 2.70 13.50 - 3.29 188.17 Solid L-tartaric acid 1 3.02 4.36 - -1.43 150.09 1 MTHF Fumaric acid 1 3.03 4.38 - -0.01 116.07 0.5 Μ THF/MeOH (1:1) Galactosedioic acid (mucosic acid) 1 3.08 3.63 - -1.46 210.14 1 MDMSO Citric acid 1 3.13 4.76 6.40 -1.72 192.12 1 MTHF D-glucuronic acid 1 3.18 - - -1.49 194.14 Solid malic acid 1 3.46 5.10 -1.26 134.09 1 MTHF Hippuric acid 1 3.55 - - 0.31 179.17 Solid D-gluconic acid 50%, stored in water 1. 3.76 - - -3.18 196.16 Solid L-mandelic acid 3 3.85 - 1Μ, 85% aqueous solution of L-lactic acid in the solvent used 1 3.86 -0.7 90.08 1 MTHF L-ascorbic acid 1 4.17 11.57 -2.41 176.13 1 Μ水162775.doc •49- 201249841 succinic acid 1 4.21 5.64 -0.59 118.09 1 MTHF acetic acid 1 4.76 - - -0.29 60.05 1 MTHF benzoic acid 2 4.19 - - 1MIPA adipic acid 1 4.44 5.44 - 0.08 146.14 solid propionic acid 2 4.87 - - 0.25 74.07 1 MTHF (+)-camphoric acid 2 4.72 5.83 - 1.47 200.23 solid 4-hydroxybenzoic acid 2 4.57 9.22 - 138.12 1Μ, in To the solution of 30.0 mg (0.165 mmol) of the free base in the selected solvent at 50 ° C, add the corresponding acid (according to the quantity and strength of the acidic part of the acid, 0.5 equivalent or 1.1 equivalents of the mixture at 50 it Stir for 1 h, then slowly cool to 〇〇c overnight (0.1 C / min). If no solids are produced, the corresponding solution is slowly evaporated under ambient conditions. In the case of a clarified solution, glue or oil, the materials are cooled to -20. (:, and slowly evaporate to promote crystallization. Thereafter, if the oil/glue is not crystallizable, an anti-solvent (cosolvent; methyl tert-butyl ether) is added; after that, it is titrated with acetone. Many salts produced precipitation when placed in isopropyl acetate (the results shown in the two tables below).

162775.doc -50- 201249841162775.doc -50- 201249841

甲烷磺酸 1.1 固體 固體 - 分離後幾分鐘内 潮解 草酸 0.5 固體 固體 - 分離後幾分鐘内 潮解 草酸 1.1 固體 固體 分離後幾分鐘内 潮解 L-天冬胺酸 0.5 固體 固體 結晶 L-天冬胺酸 L-天冬胺酸 1.1 固體 固體 結晶 L-天冬胺酸 馬來酸 0.5 固體 固體 結晶 與TB-09-10相同 之 XRPD 馬來酸 1.1 固體 固體 結晶 與TB-09-09相同 之 XRPD 磷酸 1.1 固體 固體 - 分離後幾分鐘内 潮解 L-麩胺酸 0.5 固體 固體 結晶 - L-麩胺酸 1.1 固體 固體 結晶 垂 1-經基-2-蔡 曱酸(羥萘甲 酸酉旨) 1.1 固體 固體 結晶 L-酒石酸 0.5 固體 固體 - 分離後幾分鐘内 潮解 L-酒石酸 1.1 固體 固體 - 分離後幾分鐘内 潮解 富馬酸 0.5 固體 固體 結晶 - 富馬酸 1.1 固體 固體 結晶 與具有額外峰之 TB-09-17相同之 XRPD 半乳糖二酸 (黏酸) 0.5 固體 固體 結晶 與半-半乳糖二酸 鹽參照相同之 XRPD 162775.doc .51 · 201249841 半乳糖二酸 (黏酸) 1.1 固體 固體 結晶 與半-半乳糖二酸 鹽參照相同之 XRPD 檸檬酸 0.5 固體 固體 分離後幾分鐘内 潮解 檸檬酸 1.1 固體 固體 分離後幾分鐘内 潮解 D-葡糖醛酸 1.1 固體 固體 分離後幾分鐘内 潮解 蘋果酸 1.1 膠 固體 分離後幾分鐘内 潮解 馬尿酸 1.1 固體 固體 結晶 - D-葡萄糖酸 50%,存於 水中 1.1 油 油 - - L -扁桃酸 1.1 固體 固體 結晶 L-乳酸85% 水溶液 1.1 膠 膠 - - L-抗壞血酸 1.1 澄清溶液 油 - 琥珀酸 0.5 固體 固體 結晶 - 琥珀酸 1.1 澄清溶液 澄清溶液 • 乙酸 1.1 澄清溶液 澄清溶液 - 苯甲酸 1.1 澄清溶液 澄清溶液 - 己二酸 0.5 固體 固體 結晶 與TB-09-35相同 之 XRPD 己二酸 1.1 固體 固體 結晶 與TB-09-34相同 之 XRPD 丙酸 1.1 澄清溶液 澄清溶液 - - (+)-樟腦酸 1.1 固體 固體 結晶 162775.doc ·52· 201249841 酸 當量 在-20°c 下觀察 緩慢蒸發後 觀察 於TBME中 成熟化後 觀察 用丙酮滴定 後觀察 XRPD 氫氯酸 37wt% (12 Μ) 1.1 膠 膠 膠 固體 結晶 硫酸 1.1 膠 膠 膠 膠 D-葡萄糖酸 50%,存於 水中 1.1 油 膠 膠 固體 非晶形 L-乳酸85% 水溶液 1.1 膠 膠 膠 澄清溶液 - L-天冬胺酸 1.1 澄清 溶叙 膠 膠 固體 非晶形 琥珀酸 1.1 澄清 溶液 膠 膠 膠 乙酸 1.1 澄清 溶液 膠 膠 膠 苯曱酸 1.1 固體 - 結晶 丙酸 1.1 澄清 溶液 膠 膠 澄清溶液 最初,使用乙酸異丙基酯實施鹽篩選,然而,對於彼等 產生不可分離鹽之實驗而言,使用異丙醇及2-丁酮(曱基乙 基酮)實施進一步鹽篩分(參見以下兩個表)。 酸 當量 於50°C下 於o°c下 緩慢蒸發 後觀察 用丙明滴 定後觀察 XRPD 硫酸 1.1 澄清溶液 固體 - 分離時 潮解 L-天冬胺酸 0.5 澄清溶液 澄清溶液 膠 膠 L-天冬胺酸 1.1 澄清溶液 澄清溶液 膠 膠 L-麩胺酸 0.5 澄清溶液 澄清溶液 1個月後 為固體 如游離驗 之結晶+ 額外峰 L-麩胺酸 1.1 澄清溶液 澄清溶液 1個月後 為固體 - 結晶 162775.doc •53· 201249841 D-葡糖 醛酸 1.1 油 油 膠 固體 非晶形 L·乳酸 1.1 澄清溶液 澄清溶液 膠 澄清溶液 _ 丙酸 1.1 澄清溶液 澄清溶液 膠 澄清溶液 4-羥基苯 甲酸 1.1 澄清溶液 固體 - 結晶 酸 當量 於 50°C 下 於 o°c 下 於 -20°C 下 緩慢 蒸發後 觀察 抗溶劑 添加/ 成熟化 後觀察 XRPD 硫酸 1.1 油 膠 膠 膠 膠 - L-天冬胺酸 0.5 澄清 溶液 澄清 溶液 澄清 溶液 膠 膠 L-天冬胺酸 1.1 澄清 溶液 澄清 溶液 澄清 溶液 膠 膠 - L-麩胺酸 0.5 澄清 溶液 澄清 溶液 澄清 溶液 膠 固體 與 TB-776-24-05相同之 結晶 L-麩胺酸 1.1 澄清 溶液 油 油 膠 固體 與TB- 776-24-05相同之 結晶 D-葡糖 醛酸 1.1 澄清 溶液 油 油 膠 固體 非晶形 L-乳酸85% 水溶液 1.1 澄清 溶液 膠 油 膠 膠 - 丙酸 1.1 澄清 溶液 澄清 溶液 澄清 溶液 膠 膠 - 4-羥基苯 甲酸 1.1 固體 膠 膠 膠 固體 結晶 過濾所有固體並藉由XRPD分析。 162775.doc -54- 201249841 圖1繪示化合物A之半-半乳糖二酸鹽單水合物之晶體結 構。 圖2繪示採用編號順序之晶體結構。 圖3繪示化合物A之半-半乳糖二酸鹽單水合物之 TGA/DSC。 圖4繪示化合物A之半-半乳糖二酸鹽單水合物之GVS。 圖5繪示於40°C/75% RH下儲存之前及之後及GVS之後的 化合物A之半-半乳糖二酸鹽單水合物的XRPD圖案。 圖6繪示化合物A之羥萘曱酸鹽之XRPD。 圖7繪示化合物A之羥萘曱酸鹽之TGA/DSC。 圖8繪示化合物A之羥萘甲酸鹽之GVS。 圖9繪示化合物A之苯甲酸鹽之XRPD。 圖10繪示化合物A之苯曱酸鹽之TGA/DSC。 圖11繪示化合物A之苯曱酸鹽之GVS。 圖12繪示化合物A之苯曱酸鹽之高解析度VT-XRPD。 圖13繪示化合物A之馬尿酸鹽之XRPD。 圖14繪示化合物A之馬尿酸鹽之TGA/DSC。 圖15繪示化合物A之馬尿酸鹽之GVS。 圖16繪示化合物A之馬尿酸鹽之高解析度VT-XRPD。 圖17繪示用於多晶型分析之化合物A之半-半乳糖二酸鹽 的 XRPD。 圖18繪示用於多晶型分析之化合物A之苯曱酸鹽的 XRPD。 圖19繪示用於多晶型分析之化合物A之馬尿酸鹽的 162775.doc -55- 201249841 XRPD。 圖20繪示用於多晶型分析之化合物A之羥萘曱酸鹽的 XRPD。 圖21繪示化合物A (+)-樟腦酸鹽之XRPD。 圖22繪示化合物A (+)-樟腦酸鹽之TGA/DSC。 圖23繪示化合物A之甲苯磺酸鹽之XRPD。 圖24繪示化合物A之甲苯磺酸鹽之TGA/DSC。 圖25繪示使用0.5當量酸之化合物A之馬來酸鹽的 XRPD。試樣潮解,故未量測TGA及DSC。 圖26繪示使用1當量酸之化合物A之馬來酸鹽的XRPD。 圖27繪示使用1當量酸之化合物A之馬來酸鹽的 TGA/DSC。 圖28繪示化合物A之L-扁桃酸鹽之XRPD。 圖29繪示化合物A之L-扁桃酸鹽之TGA/DSC。 具有藥物研發之可接受物理特性之鹽包括鹽酸鹽、對曱 苯磺酸鹽、L-天冬胺酸鹽、馬來酸鹽、L-麩胺酸鹽、1-羥 基-2-萘曱酸鹽(羥萘甲酸鹽)、富馬酸鹽、半乳糖二酸鹽、 馬尿酸鹽、L-扁桃酸鹽、琥珀酸鹽、己二酸鹽、或(+)-樟 腦酸鹽。較佳鹽包括對曱苯磺酸鹽、馬來酸鹽、半乳糖二 酸鹽、苯甲酸鹽、馬尿酸鹽、羥萘曱酸鹽、或(+)-樟腦酸 鹽。 下文給出若干鹽之NMR譜及熔點。該等數據係自初步鹽 篩選獲得。 化合物A之對甲苯磺酸鹽:4 NMR (d6-DMS〇, 400 162775.doc •56· 201249841 MHz): δ 7.48 (d, 2H), 7.12 (d, 2H), 4.37 (m, 2H), 4.13 (dd, 2H), 3.79 (d, 1H), 3.70 (d, 1H), 2.81 (m, 1H), 2.29 (s, 3H), 1_87 (m,1H),1·75 (d, 1H),0.81 (m, 4H); mp: 172〇C。 化合物A之馬來酸鹽:NMR (d6-DMSO, 400 MHz): δ 6.08 (s, 2H), 4.42 (m, 2H), 4.20 (dd, 2H), 3.84 (d, 1H), 3.78 (d, 1H), 2.88 (m, 1H), 1.94 (m, 1H), 1.82 (d, 1H), 0.85 (m, 4H) ; mp: 163°C。 化合物 A之羥萘曱酸鹽:1111&lt;[1411((16-01^80,4001^1^): δ 8.24 (m, 1Η), 7.75 (m, 2H), 7.51 (dd, 1H), 7.41 (dd, 1H), 7.06 (d, 1H), 4.42 (m, 2H), 4.19 (m, 2H), 3.82 (dd, 2H), 2.91 (m,1H),1.93 (m, 1H),1.79 (d,1H),0.84 (m, 4H)。 化合物 A 之 L-扁桃酸鹽:NMR (d6-DMSO, 400 MHz): δ 7.41 (d, 2H), 7.31 (m5 2H), 7.23 (m, 1H), 4.72 (s, 1H), 4.10 (m, 4H), 3.69 (dd, 2H), 2.72 (m, 1H), 1.91 (m, 1H), 1.62 (d,1H), 0.82 (m,4H) ; mp: 143°C。 化合物A之馬尿酸鹽:NMR (d6-DMSO, 400 MHz): δ 7.92 (d, 2H), 7.57 (m, 3H), 4.01 (s, 2H), 3.88 (m, 4H), 3.60 (dd, 2H), 2.62 (m, 1H), 1.94 (m, 1H), 1-52 (d, 1H), 0.82 (m, 4H)。 化合物 A之苯甲酸鹽·· iH NMR (d6-DMSO, 400 MHz): δ 7.92 (d, 2H), 7.54 (t, 1H), 7.44 (m, 2H), 3.95 (m, 2H), 3.84 (m, 2H), 3.56 (dd, 2H), 2.63 (m, 1H), 1.87 (m, 1H), 1.49 (d, 1H),0.77 (m,4H)。 化合物A之(+)-樟腦酸鹽:lH NMR (d6-DMSO,400 162775.doc -57- 201249841 MHz): δ 3.94 (m, 2H), 3.70 (m, 2H), 3.56 (dd, 2H), 2.78 (t, 1H), 2.55 (m, 1H), 2.40 (m, 1H), 2.04 (m, 1H), 1.91 (m, 1H), 1.75 (η, 1H), 1.44 (m, 2H), 1.23 (s, 3H), 1.16 (s, 3H), 0.82 (m,7H); mp: 170d。 優先放大鹽篩選中鑑別之某些鹽的合成。此處報告彼等 程序、熔點及溶解性數據。 化合物A之經萘甲酸鹽:於7〇°c下將3-環丙基羰基-3,6· 二氮雜雙環[3.1.1]庚烧(500 mg,3.01 mmol)溶解於乙酸異 丙基醋(5 mL)中。於60C下將經萘甲酸(xinaf〇ic acid)(626 mg,3.31 mmol)懸浮於98:2乙酸異丙基酯/水(6·ι mL)中並 添加至游離鹼之溶液中。將所得混合物緩慢冷卻至〇它, 且藉由抽吸過濾收集懸浮固體並在真空及環境溫度下乾燥 5 h»獲得灰白色粉末(l.oi g,95%產率)。試樣展現m〇c 之炫點及2.6 mg/mL之水溶解性。 化合物A之苯甲酸鹽:於7〇。(:下將3-環丙基羰基-3,6-二 氮雜雙環[3.1.1]庚烧(500 mg, 3.01 mmol)溶解於乙酸異丙 基醋(5 mL)中。於60。(:下將苯甲酸(405 mg, 3.31 mmol)溶 解於98.2乙酸異丙基醋/水(3.1 mL)中並添加至游離驗之溶 液中。將所得混合物緩慢冷卻至〇°C,且藉由抽吸過濾收 集懸浮固體並在真空及環境溫度下乾燥5 h。獲得白色粉 末(833 mg,95%產率)。試樣展現136。(:之熔點及&gt;15 mg/mL之水溶解性。 化合物A之馬尿酸鹽:於7〇eC下將3-環丙基羰基-3,6-二 氮雜雙環[3.M]庚烷(500 mg, 3.01 mmol)溶解於乙酸異丙 162775.doc -58- 201249841 基S旨(5 mL)中。於60°C下將馬尿酸(596 mg,3.31 mmol)懸 浮於98:2乙酸異丙基酯/水(6.1 mL)中並添加至游離鹼之溶 液中。將所得混合物緩慢冷卻至0°C,且藉由抽吸過濾收 集懸浮固體並在真空及環境溫度下乾燥5 h。獲得灰白色 粉末(884 mg,85%產率)。試樣展現157°C之熔點及&gt;15 mg/mL之水溶解性。 實例6:於菸鹼乙醯膽鹼受體處相互作用之表徵 細胞系 將 SH-EP1/人類 α4β2 (Eaton等人,2003)、SH-EP1/人類 α4β4 (Gentry等人,2003)、3Η-ΕΡ1/α6β3β4α5 (Grinevich等 人,2005)、SH-EP1_ 人類(自 Paul Whiteaker,Barrow Neurological Institute, St. Joseph's Hospital and Medical Center,Phoenix,Arizona獲得)、TE671/RD及 SH-SY5Y細胞 系(自 Dr. Ron Lukas, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, Arizona獲 得)於杜貝克氏改良鷹氏培養基(Dulbecco's modified Eagle's medium)(Gibco/BRL)中維持於增殖生長期中,該培 養基具有10%馬血清(Gibco BRL)、5%胎牛血清(HyClone, Logan UT)、1 mM丙酮酸鈉、4 mM L-麩胺醯胺。為維持 穩定轉染子,α4β2及α4β4細胞培養基補充有0.25 mg/mL 26〇(^11及〇.13 11^/1111^潮黴素8。對於〇160304〇15細胞之維持 選擇利用 0.25 mg/mL zeocin、0.13 mg/mL 潮黴素 B、0.4 mg/mL遺傳黴素及0.2 mg/mL灰瘂素來進行。對於 α6/α3β2β3細胞之維持選擇利用0.3 mg/mL zeocin及0.3 162775.doc -59- 201249841 mg/mL潮黴素B及0.6 mg/mL G418硫酸鹽來進行。將HEK/ 人類 a7/RIC3 細胞(自 J. Lindstrom, U. Pennsylvania, Philadelphia, Pennsylvania獲得)於杜貝克氏改良鷹氏培養 基(Gibco/BRL)中維持於增殖生長期中,該培養基具有1 〇〇/〇 胎牛血清(HyClone, Logan UT)、1 mM 丙酮酸鈉、4 mM L-麩胺醯胺、0·4 mg/mL遺傳黴素、0_2 mg/ml潮黴素B。 受體結合分析 自大鼠組織之膜之製備:自Analytical BiologicalMethanesulfonic acid 1.1 Solid solid - deliquescent oxalic acid 0.5 solid solid after a few minutes of separation - deliquescent oxalic acid within a few minutes after separation 1.1 solid solids separated within a few minutes after deliquescence L-aspartic acid 0.5 solid solid crystal L-aspartic acid L-aspartic acid 1.1 solid solid crystal L-aspartic acid maleic acid 0.5 solid solid crystals identical to TB-09-10 XRPD maleic acid 1.1 solid solid crystals identical to TB-09-09 XRPD phosphoric acid 1.1 Solid solids - deliquescent L-glutamic acid 0.5 solid solid crystals within a few minutes after separation - L-glutamic acid 1.1 solid solid crystals 1-meryl-2-caeic acid (hydroxynaphthoic acid) 1.1 Solid solid crystal L-tartaric acid 0.5 solid solid - deliquescent L-tartaric acid in a few minutes after separation 1.1 solid solid - deliquescent fumaric acid within a few minutes after separation 0.5 solid solid crystal - fumaric acid 1.1 solid solid crystal with TB-09-17 with extra peak The same XRPD galactosedioic acid (mucosic acid) 0.5 solid solid crystal and semi-galactose diacid salt reference XRPD 162775.doc .51 · 201249841 half Sodium disaccharide (mucoacid) 1.1 Solid solid crystal and semi-galactose diphosphate refer to the same XRPD citric acid 0.5 Solid solid separated after a few minutes of decomposing citric acid 1.1 Solids decomposed D-glucan within a few minutes after separation Acid 1.1 solid solids separated within a few minutes after deliquescence of malic acid 1.1 gel solids separated within a few minutes of deliquescent hippuric acid 1.1 solid solid crystal - D-gluconic acid 50%, stored in water 1.1 oil - L - mandelic acid 1.1 solid solid Crystalline L-Lactic Acid 85% Aqueous Solution 1.1 Glue - - L-Ascorbic Acid 1.1 Clarified Solution Oil - Succinic Acid 0.5 Solid Solid Crystallization - Succinic Acid 1.1 Clear Solution Clear Solution • Acetic Acid 1.1 Clear Solution Clear Solution - Benzoic Acid 1.1 Clear Solution Clear Solution - Adipic acid 0.5 solid solid crystals identical to TB-09-35 XRPD adipic acid 1.1 solid solid crystals identical to TB-09-34 XRPD propionic acid 1.1 clear solution clear solution - - (+)-camphoric acid 1.1 solid solid Crystalline 162775.doc ·52· 201249841 Acid equivalent observed at -20 °c after slow evaporation After maturing in TBME, observe the observation of XRPD hydrochloric acid 37wt% (12 Μ) after titration with acetone 1.1 Glue rubber solid crystal sulfuric acid 1.1 Glue rubber D-gluconic acid 50%, stored in water 1.1 Oil gelatin solid non- Crystal L-Lactic acid 85% Aqueous solution 1.1 Glue gel clear solution - L-aspartic acid 1.1 Clarified lysine gel solid amorphous succinic acid 1.1 Clear solution Glue acetate acetic acid 1.1 Clear solution Glue phthalic acid 1.1 Solid - Crystalline Propionic Acid 1.1 Clarified Solution Glue Clarification Solution Initially, salt screening was performed using isopropyl acetate, however, for the experiments in which they produced inseparable salts, isopropanol and 2-butanone (mercaptoethyl) were used. Ketone) Perform further salt sieving (see the two tables below). The acid equivalent was slowly evaporated at 50 ° C under o ° c. After observation, the XRPD sulfuric acid was observed after titration with propyl acetate. 1.1 Clarified solution solid - deliquescent L-aspartic acid 0.5 clear solution clear solution gel L-aspartate Acid 1.1 Clear solution Clear solution Glue L-glutamic acid 0.5 Clear solution After 1 month, it is a solid such as free crystals + Extra peak L-glutamic acid 1.1 Clear solution Clear solution 1 month after solid - Crystallization 162775.doc •53· 201249841 D-glucuronic acid 1.1 oil-oil gel solid amorphous L·lactic acid 1.1 clear solution clear solution gel clear solution _ propionic acid 1.1 clear solution clear solution gel clear solution 4-hydroxybenzoic acid 1.1 clear solution Solid - crystallization acid equivalent slowly evaporating at -20 ° C at 50 ° C after observation of anti-solvent addition / maturation after observation of XRPD sulfuric acid 1.1 oil gum glue - L-aspartic acid 0.5 clarification Solution clear solution clear solution glue L-aspartic acid 1.1 clear solution clear solution clear solution gel - L-glutamic acid 0.5 clear solution Clear solution clear solution gel solid same as TB-776-24-05 crystal L-glutamic acid 1.1 clear solution oil oil gel solid the same crystal as TB-776-24-05 D-glucuronic acid 1.1 clear solution oil Oil Glue Solid Amorphous L-Lactic Acid 85% Aqueous Solution 1.1 Clarified Solution Glue Rubber Glue - Propionic Acid 1.1 Clarified Solution Clarified Solution Clear Solution Glue - 4-Hydroxybenzoic Acid 1.1 Solid Glue Solid Crystals Filter all solids and use XRPD analysis. 162775.doc -54- 201249841 Figure 1 depicts the crystal structure of the semi-galactose dihydrate monohydrate of Compound A. Figure 2 illustrates the crystal structure in numerical order. Figure 3 depicts the TGA/DSC of the semi-galactosate monohydrate of Compound A. Figure 4 depicts the GVS of the semi-galactosate monohydrate of Compound A. Figure 5 depicts the XRPD pattern of the semi-galactosuccinate monohydrate of Compound A before and after storage at 40 °C / 75% RH and after GVS. Figure 6 depicts the XRPD of the hydroxynaphthoate salt of Compound A. Figure 7 depicts the TGA/DSC of the hydroxynaphthoate salt of Compound A. Figure 8 depicts the GVS of the hydroxynaphthoate salt of Compound A. Figure 9 depicts the XRPD of the benzoate salt of Compound A. Figure 10 depicts the TGA/DSC of the benzoate salt of Compound A. Figure 11 depicts the GVS of the benzoate salt of Compound A. Figure 12 depicts the high resolution VT-XRPD of the benzoate salt of Compound A. Figure 13 depicts the XRPD of the hippurate salt of Compound A. Figure 14 depicts the TGA/DSC of the hippurate of Compound A. Figure 15 depicts the GVS of the hippurate of Compound A. Figure 16 depicts the high resolution VT-XRPD of the hippurate of Compound A. Figure 17 depicts XRPD of the semi-galactosidate of Compound A for polymorphic analysis. Figure 18 depicts the XRPD of the benzoate salt of Compound A for polymorphic analysis. Figure 19 depicts 162775.doc -55 - 201249841 XRPD of the hippurate of Compound A for polymorphic analysis. Figure 20 depicts XRPD of the hydroxynaphthoate salt of Compound A for polymorphic analysis. Figure 21 depicts the XRPD of Compound A (+)-camphorate. Figure 22 depicts TGA/DSC of compound A (+)-camphorate. Figure 23 depicts the XRPD of the tosylate salt of Compound A. Figure 24 depicts the TGA/DSC of the tosylate salt of Compound A. Figure 25 depicts XRPD of the maleate salt of Compound A using 0.5 equivalents of acid. The sample was deliquescent, so TGA and DSC were not measured. Figure 26 depicts XRPD of the maleate salt of Compound A using 1 equivalent of acid. Figure 27 depicts TGA/DSC of the maleate salt of Compound A using 1 equivalent of acid. Figure 28 depicts the XRPD of L-mandelate of Compound A. Figure 29 depicts the TGA/DSC of L-mandelate of Compound A. Salts having acceptable physical properties for drug discovery include hydrochloride, p-toluenesulfonate, L-aspartate, maleate, L-glutamate, 1-hydroxy-2-naphthoquinone An acid salt (hydroxynaphthoate), a fumarate, a galactose salt, a hippurate, an L-mandelate, a succinate, an adipate, or a (+)-camphorate. Preferred salts include p-toluenesulfonate, maleate, galactate, benzoate, hippurate, hydroxynaphthoate, or (+)-camphorate. The NMR spectrum and melting point of several salts are given below. These data were obtained from preliminary salt screening. p-Toluenesulfonate of Compound A: 4 NMR (d6-DMS 〇, 400 162775.doc • 56·201249841 MHz): δ 7.48 (d, 2H), 7.12 (d, 2H), 4.37 (m, 2H), 4.13 (dd, 2H), 3.79 (d, 1H), 3.70 (d, 1H), 2.81 (m, 1H), 2.29 (s, 3H), 1_87 (m, 1H), 1·75 (d, 1H) , 0.81 (m, 4H); mp: 172〇C. Maleate of Compound A: NMR (d6-DMSO, 400 MHz): δ 6.08 (s, 2H), 4.42 (m, 2H), 4.20 (dd, 2H), 3.84 (d, 1H), 3.78 (d , 1H), 2.88 (m, 1H), 1.94 (m, 1H), 1.82 (d, 1H), 0.85 (m, 4H); mp: 163 °C. Hydroxynaphthoate of Compound A: 1111 &lt;[1411((16-01^80,4001^1^): δ 8.24 (m, 1Η), 7.75 (m, 2H), 7.51 (dd, 1H), 7.41 (dd, 1H), 7.06 (d, 1H), 4.42 (m, 2H), 4.19 (m, 2H), 3.82 (dd, 2H), 2.91 (m, 1H), 1.93 (m, 1H), 1.79 ( d,1H), 0.84 (m, 4H). L-mandelic acid salt of compound A: NMR (d6-DMSO, 400 MHz): δ 7.41 (d, 2H), 7.31 (m5 2H), 7.23 (m, 1H) ), 4.72 (s, 1H), 4.10 (m, 4H), 3.69 (dd, 2H), 2.72 (m, 1H), 1.91 (m, 1H), 1.62 (d, 1H), 0.82 (m, 4H) ; mp: 143 ° C. Horse urate of Compound A: NMR (d6-DMSO, 400 MHz): δ 7.92 (d, 2H), 7.57 (m, 3H), 4.01 (s, 2H), 3.88 (m, 4H), 3.60 (dd, 2H), 2.62 (m, 1H), 1.94 (m, 1H), 1-52 (d, 1H), 0.82 (m, 4H). Benzoate of Compound A·· iH NMR (d6-DMSO, 400 MHz): δ 7.92 (d, 2H), 7.54 (t, 1H), 7.44 (m, 2H), 3.95 (m, 2H), 3.84 (m, 2H), 3.56 (dd, 2H), 2.63 (m, 1H), 1.87 (m, 1H), 1.49 (d, 1H), 0.77 (m, 4H). (+)-camphorate of compound A: lH NMR (d6-DMSO, 400 162775.doc -57- 201249841 MHz): δ 3.94 (m, 2H), 3.70 (m, 2H), 3.56 (dd, 2H), 2.78 (t, 1 H), 2.55 (m, 1H), 2.40 (m, 1H), 2.04 (m, 1H), 1.91 (m, 1H), 1.75 (η, 1H), 1.44 (m, 2H), 1.23 (s, 3H) ), 1.16 (s, 3H), 0.82 (m, 7H); mp: 170d. The synthesis of certain salts identified in salt screening is preferentially amplified. Their procedures, melting points and solubility data are reported here. Naphthoic acid salt of compound A: 3-cyclopropylcarbonyl-3,6-diazabicyclo[3.1.1] heptane (500 mg, 3.01 mmol) dissolved in isopropyl acetate at 7 °C Base vinegar (5 mL). Nafanoic acid (xinaf〇ic acid) (626 mg, 3.31 mmol) was suspended in 98:2 isopropyl acetate/water (6.1 mL) at 60 C and added to a solution of the free base. The resulting mixture was slowly cooled to dryness, and the suspension solid was collected by suction filtration and dried under vacuum and ambient temperature for 5 h» to obtain an off-white powder (l.oi g, 95% yield). The sample exhibited a dazzling point of m〇c and a water solubility of 2.6 mg/mL. The benzoate salt of Compound A: at 7 Torr. (: 3-cyclopropylcarbonyl-3,6-diazabicyclo[3.1.1] heptane (500 mg, 3.01 mmol) was dissolved in isopropyl acetate (5 mL) at 60. : Benzoic acid (405 mg, 3.31 mmol) was dissolved in 98.2 acetic acid isopropyl vinegar / water (3.1 mL) and added to the free solution. The resulting mixture was slowly cooled to 〇 ° C, and by pumping The suspended solid was collected by suction filtration and dried under vacuum and ambient temperature for 5 h to give a white powder (833 mg, 95% yield). The sample exhibited 136. (: melting point and &gt; 15 mg/mL water solubility. Horse salt of Compound A: 3-cyclopropylcarbonyl-3,6-diazabicyclo[3.M]heptane (500 mg, 3.01 mmol) was dissolved in isopropyl acetate at 7 〇eC. -58- 201249841 Base S (5 mL). Capric acid (596 mg, 3.31 mmol) was suspended in 98:2 isopropyl acetate/water (6.1 mL) at 60 ° C and added to the free base The solution was slowly cooled to 0 ° C, and the suspended solid was collected by suction filtration and dried under vacuum and ambient temperature for 5 h to give an off-white powder (884 mg, 85% yield). 157 ° C Melting point and water solubility of &gt; 15 mg/mL. Example 6: Characterization of interaction at the nicotinic acetylcholine receptor. Cell line SH-EP1/human α4β2 (Eaton et al., 2003), SH-EP1 /Human α4β4 (Gentry et al., 2003), 3Η-ΕΡ1/α6β3β4α5 (Grinevich et al., 2005), SH-EP1_ human (obtained from Paul Whiteaker, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, Arizona) , TE671/RD and SH-SY5Y cell lines (obtained from Dr. Ron Lukas, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, Arizona) in Dulbecco's modified Eagle's medium (Gibco) /BRL) maintained in the proliferative growth phase with 10% horse serum (Gibco BRL), 5% fetal bovine serum (HyClone, Logan UT), 1 mM sodium pyruvate, 4 mM L-glutamine. In order to maintain stable transfectants, α4β2 and α4β4 cell culture media were supplemented with 0.25 mg/mL 26〇(^11 and 〇.13 11^/1111^ hygromycin 8. For the maintenance of 〇160304〇15 cells, 0.25 mg/ was used. mL zeocin, 0.13 mg/mL hygromycin B, 0.4 mg/mL geneticin and 0.2 mg/mL glucosinolate. For the maintenance of α6/α3β2β3 cells, use 0.3 mg/mL zeocin and 0.3 162775.doc -59 - 201249841 mg/mL hygromycin B and 0.6 mg/mL G418 sulfate. HEK/human a7/RIC3 cells (obtained from J. Lindstrom, U. Pennsylvania, Philadelphia, Pennsylvania) in Dubec's modified Eagle The medium (Gibco/BRL) is maintained in the proliferative growth phase with 1 〇〇/〇 fetal bovine serum (HyClone, Logan UT), 1 mM sodium pyruvate, 4 mM L-glutamine, 0.4 Mg/mL geneticin, 0_2 mg/ml hygromycin B. Receptor binding assay preparation from rat tissue membrane: from Analytical Biological

Services, Incorporated (ABS,Wilmington,Delaware)獲得大 鼠皮質。自雌性Sprague-Dawley大鼠解剖組織,冷凍並在 乾冰上運輸。於-20°C下儲存組織直至膜製備需要為止。 彙集10只大鼠之皮質並藉由Polytron (Kinematica GmbH, Switzerland)在10體積(重量:體積)冰冷製備性緩衝液 (KC1 » 11 mM ; KH2P〇4 * 6 mM ; NaCl 137 mM ; Na2HP04 8 mM ; HEPES(游離酸)’ 20 mM ;蛾乙胺,5 mM ; £〇丁八,1.5 1111^;0.1111^1?]^?卩117.4)中均質化。於4°〇 下將所得均質物以40,000 g離心20分鐘並將所得沈澱物重 新懸浮於20體積冰冷水中。在4t下培育60分鐘後,藉由 於4 C下以40,000 g離心20分鐘收集新沈激物。將最終沈殿 物重新懸浮於製備性緩衝液中並於-20°C下儲存。在分析 當天,對組織實施解凍’以40,000 g離心2〇分鐘且隨後重 新懸浮於PBS (杜貝克氏麟酸鹽緩衝鹽水,Life Technologies,pH 7.4)中至2-3 mg蛋白/毫升之最終濃度。 使用 Pierce BCA 蛋白分析套組(Pierce Bi〇techn〇1〇gy, 162775.doc -60- 201249841The rat cortex was obtained from Services, Incorporated (ABS, Wilmington, Delaware). Tissues were dissected from female Sprague-Dawley rats, frozen and transported on dry ice. Tissues were stored at -20 °C until film preparation was required. The cortex of 10 rats was pooled and ice-cold preparative buffer (KC1 » 11 mM; KH2P〇4 * 6 mM; NaCl 137 mM; Na2HP04 8 mM) by Polytron (Kinematica GmbH, Switzerland) in 10 volumes (by weight: volume). HEPES (free acid) '20 mM; mothylamine, 5 mM; 〇 八 八, 1.5 1111^; 0.1111^1?]^?卩117.4) homogenization. The resulting homogenate was centrifuged at 40,000 g for 20 minutes at 4 ° C and the resulting precipitate was resuspended in 20 volumes of ice-cold water. After incubation for 60 minutes at 4 t, the new sink was collected by centrifugation at 40,000 g for 20 minutes at 4 C. The final sediment was resuspended in preparative buffer and stored at -20 °C. On the day of analysis, the tissue was thawed 'centrifuged at 40,000 g for 2 min and then resuspended in PBS (Dulbecker Sulfate Buffered Saline, Life Technologies, pH 7.4) to a final concentration of 2-3 mg protein/ml. . Use the Pierce BCA Protein Analysis Kit (Pierce Bi〇techn〇1〇gy, 162775.doc -60- 201249841

Rockford,IL)測定蛋白濃度,利用牛血清白蛋白作為標 樣。 自純系細胞系之膜之製備:將細胞收穫於冰冷PBS (pH 7.4)中,隨後用 polytron (Brinkmann Instruments,Westbury, NY)均質化。將均質物以40,000g離心20分鐘(4°C )。將沈 澱物重新懸浮於PBS中並使用Pierce BCA蛋白分析套組 (Pierce Biotechnology, Rockford, IL)測定蛋白濃度。 膜製備中與受體之競爭結合.在膜上使用根據已公開程 序(Lippiello and Fernandes,1986 ; Davies 等人,1999)改 編之標準方法分析與菸鹼受體之結合。簡言之,自冷凍原 液(約0.2 mg蛋白)重構膜並在競爭劑化合物(0.001 nM至 100 mM)及放射性配體存在下在150 ml分析緩衝液(PBS)中 於冰上培育2 h。使用[3H]-菸鹼(L-(-)-[N-曱基-3H]-菸鹼, 69.5 Ci/mmol,Perkin-Elmer Life Sciences)進行人類 α4β2 結合研究。使用[3Η]-地棘虫圭素(52 Ci/mmol, Perkin-Elmer Life Sciences)進行其他受體亞型處之結合研究。藉由在多 歧管組織收穫器(Brandel,Gaithersburg, MD)上使用預浸於 0.33%聚乙烯亞胺(w/v)中之GF/B過濾器快速過濾終止培育 以降低非特異性結合^將過濾器洗滌3次並藉由液體閃爍 計數測定所保留放射性。 結合數據分析。結合數據表示為總對照結合百分比。取 每一點之重複的平均值並根據藥物濃度之對數繪圖。藉由 最小平方線性回歸使用GraphPad Prism軟體(GraphPAD, San Diego,C A)測定IC5Q(產生50%結合抑制之化合物濃 162775.doc -61 - 201249841 度)。使用 Cheng-Prusoff方程(Cheng及 Prusoff,1973)計算Rockford, IL) determined protein concentration using bovine serum albumin as a standard. Preparation of membranes from pure cell lines: Cells were harvested in ice-cold PBS (pH 7.4) and subsequently homogenized with a polytron (Brinkmann Instruments, Westbury, NY). The homogenate was centrifuged at 40,000 g for 20 minutes (4 ° C). The precipitate was resuspended in PBS and protein concentration was determined using a Pierce BCA protein assay kit (Pierce Biotechnology, Rockford, IL). Competitive binding to receptors in membrane preparation. Binding to nicotinic receptors was analyzed on membranes using standard methods adapted from published procedures (Lippiello and Fernandes, 1986; Davies et al., 1999). Briefly, reconstituted membranes from frozen stock (approximately 0.2 mg protein) and incubated on ice for 2 h in 150 ml assay buffer (PBS) in the presence of competitor compounds (0.001 nM to 100 mM) and radioligand . Human α4β2 binding studies were performed using [3H]-nicotine (L-(-)-[N-mercapto-3H]-nicotine, 69.5 Ci/mmol, Perkin-Elmer Life Sciences). Binding studies at other receptor subtypes were performed using [3Η]-G. serrata (52 Ci/mmol, Perkin-Elmer Life Sciences). Reducing non-specific binding by rapid filtration using a GF/B filter pre-soaked in 0.33% polyethyleneimine (w/v) on a multi-manifold tissue harvester (Brandel, Gaithersburg, MD) The filter was washed 3 times and the retained radioactivity was determined by liquid scintillation counting. Combine data analysis. Binding data is expressed as the percentage of total control binding. The average of the replicates for each point is taken and plotted against the logarithm of the drug concentration. IC5Q (concentration of compound producing 50% binding inhibition 162775.doc -61 - 201249841 degrees) was determined by least square linear regression using GraphPad Prism software (GraphPAD, San Diego, CA). Calculated using the Cheng-Prusoff equation (Cheng and Prusoff, 1973)

Ki。 表1 結構 CO. &lt;s GO. n ca 言 賧 &lt; ¥ i 140 20 3.0 2.3 620 實例7:藥理學概述 活體外藥理學: 化合物A係對α6β2*及α4β2* NNR具有高親和性之配體, 且證實對ct7 NNR具有較低親和性。化合物Α係所有三種亞 型之完全激動劑且展示與肌肉型及神經節型菸鹼受體以及 非乾標受體足夠功能分離。在&gt;60個乾標之Novascreen組 中,化合物A並不與任何非NNR受體相互作用。化合物A 亦呈現已知存於紋狀體中之三種α4β2* NNR亞型穩健脫 敏。具體而言,化合物Α於(α4)2(β2)3-「高敏感性」、 (α4)3(β2)2-「低敏感性」、α5(α4)2(β2)2受體下分別具有24 nm、68 nm及42 nm之DC5〇,在每一情況下完全抑制(在投 與10 μιη乙醯膽驗30 min之前時,&gt;98% Imax)。脫敏潛在地 與治療LID之功效相關(參見Bordia等人,]\?1^1'111.£\卩· Ther. 327(1): 239-247 (2008))。 I62775.doc •62- 201249841 如WO 2011/071758中初步報告,化合物A產生抵抗大鼠 多巴胺神經元之原代培養物中的MPP+毒性之極其有效神 經保護效應《在MPP+暴露之前預培育24或48小時時,化 合物A在寬劑量範圍(1 pM至i μΜ)内有保護性。在使用24 小時預培育時’最小有效劑量係1〇 ρΜ,使得化合物a有 效’且比此分析中之終驗更有效。與終驗相比,化合物A 之更大受體選擇性及更大神經保護效能可在用以帕金森氏 病之疾病修飾中引起更好長期投與耐受性。 活體内藥理學: 大鼠研究:在重複投與左旋多巴之6-OHDA-誘導之(半-) 帕金森病大鼠中測定化合物A減輕AIM之能力。使用交又 設計在左旋多巴未曾治療(模製預防或延遲LID發作)及左 旋多巴預處理(模製治療現有LID)之帕金森病大鼠二者中 測試化合物A。 在交又之第一期中,在左旋多巴投與之前經由皮下微型 幫浦以0.75 mg/kg/天投與化合物達兩週,隨後與左旋多 巴同時投與額外3週。化合物A以0.75 mg/kg/天在左旋多 巴未曾治療大鼠中預防LID完全發作(AIM減輕34%,參見 圖30A)且在劑量降至(K3 mg/kg/天達額外數週時維持效 應。 隨後交叉投藥組,以使已接受媒劑與左旋多巴左旋多巴 (左旋多巴預處理)組合之動物暴露於化合物A。化合物A以 0.3 mg/kg/天經由微型幫浦投與2週後產生確立之LID之較 小但顯著減輕(13%)。在劑量增加至0.7 mg/kg/天達額外2 162775.doc 63· 201249841 週時’減輕增加至21% (參見圖30B)。在根據損傷之嚴重 性分析數據時,化合物A將中度損傷動物之確立AIM減輕 36%。 由於減輕AIM之藥物可使帕金森症惡化,故亦使用肢體 不對稱使用測試或圓筒測試(其係單側多巴胺損失程度之 敏感量度)針對帕金森病大鼠之運動功能測試化合物A之效 應。結果顯示化合物A不會使基線運動功能(不服用左旋多 巴)惡化且亦不會使利用左旋多巴治療之增強肢體使用惡 化(參見圖30C)。重要的是’在功效實驗過程期間,化合 物A不產生可檢測到之副作用(即,對於體重、身體溫度、 整飾、排尿、排便、分泌)。 非人類靈長類研究:為進一步分析化合物A緩解lid而 不降低左旋多巴之抗帕金森病效應的能力,採用帕金森氏 病之MPTP損傷食蟹猴模型。在此研究中,利用針對每一 動物逆轉帕金森病症狀但誘導顯著至嚴重運動障礙優化之 可變劑量之左旋多巴(20-35 mg/kg/天,經口,每天早上一 次)治療MPTP損傷食蟹猴達兩週。接下來六週動物繼續接 受左旋多巴,但亦接受(藉由經口管飼)媒劑(每天一次)或 化合物A (每天2次,間隔8小時,早上治療與左旋多巴治 療並行)。每一組中有7只動物(治療及媒劑治療動物接 受增加劑量之化合物A :兩週為〇.〇3 mg/kg,之後兩週為 0.10 mg/kg ;之後兩週為0.30 mg/kg ; —曰兩次。在第7 ' 13、21、28、35、42、49及59天中之每一天,將動物評定 為「差品質」啟動期(on-time)(顯著至嚴重運動障礙)及 162775.doc -64- 201249841 「良好品質」啟動期(無運動障礙、輕微或中度運動障 礙)。此評定時段開始於早上治療且持續6小時β在第一小 時結束與第三小時結束之間之兩小時時段期間,評定運動 障礙嚴重性及帕金森病失能二者。 如圖31a中所示,化合物Α顯著減輕1„)。此效應在〇1〇 mg/kg劑量下尤其強。如圖31b及31c中所示,化合物a不顯 著改變總啟動期(在觀察到左旋多巴之抗帕金森病效應時 之總時間)或帕金森病失能(左旋多巴治療之效率)。如圖32 中所示,化合物A將差品質啟動期(由令人煩惱之運動障礙 表徵之時間)之百分比自50-55%顯著降低至約30%且相應 地將良好品質啟動期(無令人煩惱之運動障礙的時間)之百 分比自45-50°/。增加至約70%。總運動障礙、以及舞蹈症及 肌張力障礙終點顯著降低。此研究中化合物A耐受性極 好,不會負面地影響動物之一般運動活動,且不會抵消左 旋多巴對帕金森症之效應。 所觀察到之具體醫藥反應可根據並端視所選特定活性化 合物或是否存在醫藥載劑以及所用調配物類型及投與方式 而定’且根據本發明之實踐,此等預期結果差別或差異亦 涵蓋於本發明中。 儘管本文詳細闌釋並闡述本發明之具體實施例,但本發 明並不限於此。以上詳細說明提供為本發明之實例且不應 理解為構成本發明之任何限制。彼等熟習此項技術者將易 知各修改’且不背離本發明精神之所有修改均意欲包括於 隨附申請專利範圍之範_内。 162775.doc •65· 201249841 【圖式簡單說明】 圖1繪不化合物A之半-半乳糖二酸鹽單水合物之晶體結 構。 圖2繪示採用編號順序之晶體結構。 圖3繪示化合物A之半-半乳糖二酸鹽單水合物之 TGA/DSC。 圖4繪示化合物A之半·半乳糖二酸鹽單水合物之GVS。 圖5繪示於40°C/75% RH下儲存之前及之後及GVS之後的 化合物A之半-半乳糖二酸鹽單水合物的xrpd圖案。 圖6繪示化合物A之經萘甲酸鹽之xrpd。 圖7繪示化合物A之羥萘甲酸鹽之tgA/DSC。 圖8繪示化合物A之經萘甲酸鹽之GVS。 圖9繪示化合物A之苯曱酸鹽之xrpd。 圖10繪示化合物A之笨曱酸鹽之tga/DSC。 圖11繪示化合物A之笨甲酸鹽之gvs。 圖12繪示化合物A之笨曱酸鹽之高解析度ντ-XRPD。 圖13繪示化合物A之馬尿酸鹽之xrpd。 圖14繪示化合物A之馬尿酸鹽之tga/dsc。 圖15繪示化合物A之馬尿酸鹽之GVS。 圖16繪示化合物A之馬尿酸鹽之高解析度ντ-XRPD » 圖17繪示用於多晶型分析之化合物a之半-半乳糖二酸鹽 的XRPD » 圖18繪示用於多晶型分析之化合物a之苯曱酸鹽的 XRPD。 162775.doc • 66 - 201249841 圖19繪示用於多晶型分析之化合物A之馬尿酸鹽的 XRPD。 圖20繪示用於多晶型分析之化合物A之羥萘甲酸鹽的 XRPD。 圖21繪示化合物A (+)-樟腦酸鹽之XRPD。 圖22繪示化合物A (+)-樟腦酸鹽之TGA/DSC。 圖23繪示化合物A之曱苯磺酸鹽之XRPD。 圖24繪示化合物A之曱苯磺酸鹽之TGA/DSC。 圖25繪示使用0.5當量酸之化合物A之馬來酸鹽的 XRPD。試樣潮解,故未量測TGA及DSC。 圖26繪示使用1當量酸之化合物A之馬來酸鹽的XRPD。 圖27繪示使用1當量酸之化合物A之馬來酸鹽的 TGA/DSC。 圖28繪示化合物A之L-扁桃酸鹽之XRPD。 圖29繪示化合物A之L-扁桃酸鹽之TGA/DSC。 圖30繪示化合物A減少帕金森病大鼠中之AIM的能力。 圖31繪示化合物A減少非人類靈長類中之LID的能力。 圖32繪示化合物A在左旋多巴治療期間增加良好品質啟 動期的能力。 162775.doc 67-Ki. Table 1 Structure CO. &lt;s GO. n ca 賧&lt; ¥ i 140 20 3.0 2.3 620 Example 7: Pharmacological overview In vitro pharmacology: Compound A has a high affinity for α6β2* and α4β2* NNR , and confirmed to have a lower affinity for ct7 NNR. The compound lanthanide is a full agonist of all three subtypes and exhibits sufficient functional separation from muscle and gangliotype nicotinic receptors as well as non-dry standard receptors. In the &60; dry standard Novascreen group, Compound A did not interact with any non-NNR receptors. Compound A also exhibits robust desensitization of the three α4β2* NNR subtypes known to be present in the striatum. Specifically, the compound is ((α4)2(β2)3-“high sensitivity”, (α4)3(β2)2-“low sensitivity”, and α5(α4)2(β2)2 receptor, respectively DC5〇 with 24 nm, 68 nm, and 42 nm was completely inhibited in each case (&gt;98% Imax before administration of 10 μιη 醯 醯 30 30 min). Desensitization is potentially related to the efficacy of the therapeutic LID (see Bordia et al., &quot;1^1'111. £\卩· Ther. 327(1): 239-247 (2008)). I62775.doc •62- 201249841 As reported initially in WO 2011/071758, Compound A produces an extremely potent neuroprotective effect against MPP+ toxicity in primary cultures of rat dopamine neurons. Pre-incubation 24 or 48 prior to MPP+ exposure. Compound A is protective over a wide dose range (1 pM to i μΜ) at hours. The minimum effective dose is 1 〇 ρ 在 when used for 24 hours of pre-incubation, making Compound a effective' and more effective than the final test in this analysis. The greater receptor selectivity and greater neuroprotective efficacy of Compound A may result in better long-term administration tolerance in disease modification with Parkinson's disease compared to the final assay. In vivo pharmacology: Rat study: The ability of Compound A to attenuate AIM was determined in 6-OHDA-induced (semi-) Parkinson's disease rats repeatedly administered with levodopa. Compound A was tested in both Parkinson's disease rats that had not been treated with levodopa (molar prevention or delayed LID episodes) and levodopa pretreatment (modeled treatment of existing LID). In the first phase of the reunion, the compound was administered to the subcutaneous mini-pump at 0.75 mg/kg/day for two weeks prior to the administration of levodopa, followed by an additional 3 weeks with levodopa. Compound A prevented complete onset of LID (34% reduction in AIM, see Figure 30A) at 0.75 mg/kg/day in levodopa-untreated rats and maintained at dose reduction (K3 mg/kg/day for additional weeks) Effect. The drug was then cross-administered to expose animals that had received vehicle combination with levodopa levodopa (levodopa pretreatment) to Compound A. Compound A was administered at 0.3 mg/kg/day via a micro pump. A small but significant reduction (13%) in established LID after 2 weeks. Increased to 21% at doses increased to 0.7 mg/kg/day for an additional 2 162775.doc 63·201249841 weeks (see Figure 30B) Compound A reduced the established AIM of moderately injured animals by 36% when analyzing data based on the severity of the injury. Since AIM-relieving drugs can worsen Parkinson's disease, limb asymmetry test or cylinder test is also used ( It is a sensitive measure of the degree of unilateral dopamine loss). It is a test for the motor function test compound A in Parkinson's disease rats. The results show that Compound A does not deteriorate the baseline motor function (without taking levodopa) and does not make use of it. Levodopa treatment Enhance the deterioration of limb use (see Figure 30C). It is important that Compound A does not produce detectable side effects during the efficacy test (ie, for body weight, body temperature, finishing, urination, defecation, secretion). Primate studies: To further analyze the ability of Compound A to ameliorate lid without reducing the anti-Parkinson's effect of levodopa, the MPTP-infected cynomolgus monkey model of Parkinson's disease was used. In this study, the use of each animal was used. A variable dose of levodopa (20-35 mg/kg/day, orally, once a day) that reverses the symptoms of Parkinson's disease but induces significant to severe dyskinesia is treated for MPTP-injured cynomolgus monkeys for two weeks. Six-week animals continued to receive levodopa, but also received (by oral gavage) vehicle (once a day) or Compound A (2 times a day, 8 hours apart, morning treatment in parallel with levodopa treatment). There were 7 animals in the group (therapeutic and vehicle-treated animals received an increased dose of Compound A: two weeks for 〇.〇3 mg/kg, followed by 0.10 mg/kg for the next two weeks; and 0.30 mg/kg for the next two weeks;曰On each of the 7th '13, 21, 28, 35, 42, 49, and 59 days, the animals were rated as "poor quality" on-time (significant to severe dyskinesia) and 162,775. Doc -64- 201249841 "Good quality" start-up period (no dyskinesia, mild or moderate dyskinesia). This assessment period begins in the morning and lasts for 6 hours. β is between the end of the first hour and the end of the third hour. During the hourly period, both the severity of dyskinesia and the disability of Parkinson's disease were assessed. As shown in Figure 31a, the compound Α significantly reduced 1 „). This effect is particularly strong at doses of 〇1〇 mg/kg. As shown in Figures 31b and 31c, Compound a did not significantly alter the total initiation period (the total time when levodopa was resistant to Parkinson's disease) or Parkinson's disease disability (efficiency of levodopa treatment). As shown in Figure 32, Compound A significantly reduced the percentage of poor quality initiation period (time characterized by annoying dyskinesia) from 50-55% to approximately 30% and correspondingly a good quality initiation period (no order) The percentage of time when people are troubled with movement disorders is from 45-50°/. Increase to about 70%. Total dyskinesia, as well as chorea and dystonia endpoints were significantly reduced. Compound A was well tolerated in this study and did not negatively affect the general motor activity of the animal and did not offset the effect of levodopa on Parkinson's disease. The specific pharmaceutical response observed can be based on the particular active compound selected or the presence or absence of a pharmaceutical carrier, and the type of formulation employed, and the manner in which it is administered, and in accordance with the practice of the present invention, such differences or differences in expected results are also It is encompassed by the present invention. Although specific embodiments of the invention have been described and illustrated in detail herein, the invention is not limited thereto. The above detailed description is provided as an example of the invention and should not be construed as limiting the invention. Those skilled in the art will appreciate that modifications may be made without departing from the spirit of the invention, and are intended to be included within the scope of the appended claims. 162775.doc •65· 201249841 [Simple description of the diagram] Fig. 1 depicts the crystal structure of the semi-galactose dihydrate monohydrate of the compound A. Figure 2 illustrates the crystal structure in numerical order. Figure 3 depicts the TGA/DSC of the semi-galactosate monohydrate of Compound A. Figure 4 depicts the GVS of the semi-galactosate monohydrate of Compound A. Figure 5 depicts the xrpd pattern of the semi-galactosuccinate monohydrate of Compound A before and after storage at 40 °C / 75% RH and after GVS. Figure 6 depicts the xrpd of naphthoate salt of Compound A. Figure 7 depicts the tgA/DSC of the hydroxynaphthoate salt of Compound A. Figure 8 depicts the GVS of the naphthoate salt of Compound A. Figure 9 depicts the xrpd of the benzoate salt of Compound A. Figure 10 depicts the tga/DSC of the abbreviated acid salt of Compound A. Figure 11 depicts the gvs of the stearate of Compound A. Figure 12 depicts the high resolution ντ-XRPD of the abbreviated acid salt of Compound A. Figure 13 depicts the xrpd of the hippurate of Compound A. Figure 14 depicts the tga/dsc of the hippurate salt of Compound A. Figure 15 depicts the GVS of the hippurate of Compound A. Figure 16 is a graph showing the high resolution ντ-XRPD of the hippurate of Compound A. Figure 17 is a graph showing the XRPD of the semi-galactose diphosphate of Compound a for polymorph analysis. Figure 18 is a graph showing XRPD of the benzoate of Compound a. 162775.doc • 66 - 201249841 Figure 19 depicts XRPD of the hippurate of Compound A for polymorphic analysis. Figure 20 depicts XRPD of the hydroxynaphthoate salt of Compound A for polymorphic analysis. Figure 21 depicts the XRPD of Compound A (+)-camphorate. Figure 22 depicts TGA/DSC of compound A (+)-camphorate. Figure 23 depicts the XRPD of the tosylate salt of Compound A. Figure 24 depicts the TGA/DSC of the tosylate salt of Compound A. Figure 25 depicts XRPD of the maleate salt of Compound A using 0.5 equivalents of acid. The sample was deliquescent, so TGA and DSC were not measured. Figure 26 depicts XRPD of the maleate salt of Compound A using 1 equivalent of acid. Figure 27 depicts TGA/DSC of the maleate salt of Compound A using 1 equivalent of acid. Figure 28 depicts the XRPD of L-mandelate of Compound A. Figure 29 depicts the TGA/DSC of L-mandelate of Compound A. Figure 30 depicts Compound A's ability to reduce AIM in Parkinson's disease rats. Figure 31 depicts the ability of Compound A to reduce LID in non-human primates. Figure 32 depicts the ability of Compound A to increase the good quality initiation period during levodopa treatment. 162775.doc 67-

Claims (1)

201249841 七、申請專利範圍: 1· 一種3-環丙基羰基-3,6-二氮雜雙環[3. M]庚烷之酸加成 。 2. 一種3-環丙基羰基-3,6-二氮雜雙環[3丨丨]庚烷之酸加成 鹽,其中該酸係選自氫氣酸、對甲笨磺酸、L_天冬胺 酸、馬來酸、L-麵胺酸、1 -經基-2-蔡甲酸、富馬酸、半 乳糖二酸、馬尿酸、L-扁桃酸、號绍酸、己二酸或(+)_ 樟腦酸。 3 ·如請求項2之酸加成鹽’其中該鹽係對甲苯續酸鹽、馬 來酸鹽、半乳糖二酸鹽、苯曱酸鹽、馬尿酸鹽、羥萘甲 酸鹽(xinafoate)或(+)·樟腦酸鹽。 4. 如請求項3之酸加成鹽’其中該鹽係半乳糖二酸鹽、苯 曱酸鹽、馬尿酸鹽或羥萘甲酸鹽。 5. 一種3_環丙基羰基-3,6-二氮雜雙環[3丨丨]庚烷半—半乳糖 二酸鹽(hemigalactarate)單水合物。 6. 如請求項1至6中任一項之酸加成鹽,其中該鹽係結晶。 7· —種醫藥組合物,其包含如請求項1至6中任一項之化合 物及一或多種醫藥上可接受之載劑、稀釋劑或賦形劑。 8. —種治療或預防由神經元菸鹼受體介導之疾病或病況之 方法,其包括投與如請求項丨至6中任一項之化合物。 9. 一種如請求項1至ό中任一項之化合物的用途,其用於製 備用於治療或預防由神經元菸鹼受體介導之疾病或病況 的藥劑。 10. 如請求項1至6之化合物,其用作活性治療物質。 162775.doc 201249841 11.如請求項1至6之化合物, 驗受體介導之疾病或病況 其用於治療或預防由神經元菸 12. 如:求項之方法、用途或化合物*中 元菸鹼性受體係α4β2*亞型。 、也 13. 如請求項之方法、用途或化合物其中 元菸鹼性受體係α6β2*亞型。 經 Η.如請求項…以方法、用途或化合物,其中該等 元菸鹼性受體係α4β2*及α6β2*亞型二者。 15.如請求項之方法、用途或化合物,其中該疾 病況係CNS病症。 或 16.如請求項8至^之方法、用途或化合物,其中該疾 病況係異常不自主運動。 5 17·如請求項8至U之方法、用途或化合物,其中該疾病或 病況係左旋多巴(L-dopa)誘導之運動障礙。 18·如請求項8至11之方法、用途或化合物,其中該疾病艺 病況係運動障礙。 — 19. 如請求項8至11之方法、用途或化合物,其中該疾病或 病況係帕金森氏病(Parkinson’s Disease)。 20. 如請求項19之方法、用途或化合物,其中該投藥法不會 妨礙任何現有療法對運動缺陷之效應。 21. 如請求項8至11之方法、用途或化合物,其中該疾病或 病況係帕金森症(Parkinsonism)。 22. 如請求項8至11之方法、用途或化合物,其中該方法、 用途或化合物進一步包括正在投與之左旋多巴。 I62775.doc 201249841 23·如請求項22之方法、 在已存在的療程期間 時間之過程提供。 用途或化合物,其中該左旋多巴係 以—或多個較低劑量或以延遲投藥 24. 一種保護神經之方法,甘6 ^ 其包括投與3-環丙基羰基-3,6-二 氮雜雙環[3.1.1]庚烧或其醫藥上可接受之鹽。 25·如請求項7之醫藥组人你 +χ , 甲、見〇物’其中該組合物經調配用於經 皮、經鼻内、經頰或舌下投與。 26· —種合成3-環丙基羰基·36_二氮雜雙環[3丨^庚烷或其 醫藥上可接爻之鹽之方法,其包括使環丙基曱醯胺親核 劑與氮雜%: 丁基雙-親電子劑反應,以形成六氫。比咬環。 27. —種化合物 Bn— N^^OMs OMs 28.如請求項27之化合物,其呈相應鹵化物形式。 162775.doc201249841 VII. Patent application scope: 1. An acid addition of 3-cyclopropylcarbonyl-3,6-diazabicyclo[3. M]heptane. 2. An acid addition salt of 3-cyclopropylcarbonyl-3,6-diazabicyclo[3丨丨]heptane, wherein the acid is selected from the group consisting of hydrogen acid, p-toluenesulfonic acid, and L_aspartate Aminic acid, maleic acid, L- faceted acid, 1-carbo-2-carotic acid, fumaric acid, galactosuccinic acid, hippuric acid, L-mandelic acid, succinic acid, adipic acid or (+ )_ camphoric acid. 3. The acid addition salt of claim 2 wherein the salt is p-toluene, maleate, galactate, benzoate, hippurate, xinafoate Or (+)·camphorate. 4. The acid addition salt of claim 3 wherein the salt is a galactose salt, a benzoate, a hippurate or a hydroxynaphthoate. 5. A 3_cyclopropylcarbonyl-3,6-diazabicyclo[3丨丨]heptane hemi-galactose dihydrate (hemigalactarate) monohydrate. 6. The acid addition salt of any one of claims 1 to 6, wherein the salt is crystalline. A pharmaceutical composition comprising a compound according to any one of claims 1 to 6 and one or more pharmaceutically acceptable carriers, diluents or excipients. 8. A method of treating or preventing a disease or condition mediated by a neuronal nicotinic receptor, comprising administering a compound according to any one of claims 6 to 6. 9. Use of a compound according to any one of claims 1 to 3 for the preparation of a medicament for the treatment or prevention of a disease or condition mediated by a neuronal nicotinic receptor. 10. A compound according to claims 1 to 6 for use as an active therapeutic substance. 162775.doc 201249841 11. A compound according to claims 1 to 6, which is a receptor-mediated disease or condition which is used for the treatment or prevention of a tobacco by a neuron smoke 12. For example, a method, use or compound* Alkaline is affected by the system α4β2* subtype. And 13. The method, use or compound of the claim, wherein the elemental nicotinic acid is affected by the system α6β2* subtype. By the method of claim, the method, the use, or the compound, wherein the metabasin is affected by both the system α4β2* and the α6β2* subtype. 15. The method, use or compound of claim, wherein the condition is a CNS disorder. Or 16. The method, use or compound of claim 8 to ^ wherein the condition is abnormally involuntary. The method, use or compound of claim 8 to U, wherein the disease or condition is levodopa-induced dyskinesia. 18. The method, use or compound of claim 8 to 11, wherein the disease condition is dyskinesia. 19. The method, use or compound of claim 8 to 11, wherein the disease or condition is Parkinson&apos;s Disease. 20. The method, use or compound of claim 19, wherein the administration does not interfere with the effects of any existing therapies on motor deficits. 21. The method, use or compound of claim 8 to 11, wherein the disease or condition is Parkinsonism. 22. The method, use or compound of claim 8 to 11, wherein the method, use or compound further comprises levodopa being administered. I62775.doc 201249841 23. The method of claim 22, provided during the course of an existing treatment period. Use or compound, wherein the levodopa is administered at a lower dose or at a lower dose. 24. A method of protecting nerves, which comprises administering 3-cyclopropylcarbonyl-3,6-diaza Heterobicyclo[3.1.1] heptane or a pharmaceutically acceptable salt thereof. 25. The medical group of claim 7 you +χ, A, see 〇物' wherein the composition is formulated for transdermal, intranasal, buccal or sublingual administration. 26. A method for synthesizing 3-cyclopropylcarbonyl·36-diazabicyclo[3丨^heptane or a pharmaceutically acceptable salt thereof, which comprises reacting a cyclopropylguanamine nucleophile with nitrogen Hetero%: butyl bis-electrophile reacts to form hexahydrogen. Than the bite ring. 27. A compound Bn-N^^OMs OMs 28. The compound of claim 27 which is in the form of the corresponding halide. 162775.doc
TW101108527A 2011-03-14 2012-03-13 Novel salt forms of 3-cyclopropylcarbonyl-3,6-diazabicyclo[3.1.1]heptane TW201249841A (en)

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
US201161452214P 2011-03-14 2011-03-14

Publications (1)

Publication Number Publication Date
TW201249841A true TW201249841A (en) 2012-12-16

Family

ID=45888490

Family Applications (1)

Application Number Title Priority Date Filing Date
TW101108527A TW201249841A (en) 2011-03-14 2012-03-13 Novel salt forms of 3-cyclopropylcarbonyl-3,6-diazabicyclo[3.1.1]heptane

Country Status (3)

Country Link
AR (1) AR085676A1 (en)
TW (1) TW201249841A (en)
WO (1) WO2012125518A1 (en)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10231970B2 (en) 2014-09-30 2019-03-19 NV Heterocycles Methods of producing heteropolycycles via bis-epoxidation

Family Cites Families (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
IL107184A (en) 1992-10-09 1997-08-14 Abbott Lab Heterocyclic ether compounds that enhance cognitive function
US5852041A (en) 1993-04-07 1998-12-22 Sibia Neurosciences, Inc. Substituted pyridines useful as modulators of acethylcholine receptors
US5493026A (en) 1993-10-25 1996-02-20 Organix, Inc. Substituted 2-carboxyalkyl-3-(fluorophenyl)-8-(3-halopropen-2-yl) nortropanes and their use as imaging for agents for neurodegenerative disorders
US5597919A (en) 1995-01-06 1997-01-28 Dull; Gary M. Pyrimidinyl or Pyridinyl alkenyl amine compounds
US5604231A (en) 1995-01-06 1997-02-18 Smith; Carr J. Pharmaceutical compositions for prevention and treatment of ulcerative colitis
US5585388A (en) 1995-04-07 1996-12-17 Sibia Neurosciences, Inc. Substituted pyridines useful as modulators of acetylcholine receptors
US5583140A (en) 1995-05-17 1996-12-10 Bencherif; Merouane Pharmaceutical compositions for the treatment of central nervous system disorders
IL118279A (en) 1995-06-07 2006-10-05 Abbott Lab 3 - pyridyloxy (or thio) alkyl heterocyclic compounds, pharmaceutical compositions containing them and their uses in the preparation of medicaments for controlling chemical synaptic transmission
US5726189A (en) 1996-05-03 1998-03-10 The United States Of America, Represented By The Secretary, Department Of Health And Human Services Method for imaging nicotinic acetylcholinergic receptors in the brain using radiolabeled pyridyl-7-azabicyclo 2.2.1!heptanes
ZA9711092B (en) 1996-12-11 1999-07-22 Smithkline Beecham Corp Novel compounds.
US5952339A (en) 1998-04-02 1999-09-14 Bencherif; Merouane Pharmaceutical compositions and methods of using nicotinic antagonists for treating a condition or disorder characterized by alteration in normal neurotransmitter release
US6310043B1 (en) 1998-08-07 2001-10-30 Governors Of The University Of Alberta Treatment of bacterial infections
GB9821503D0 (en) 1998-10-02 1998-11-25 Novartis Ag Organic compounds
ITMI20040954A1 (en) 2004-05-12 2004-08-12 Univ Degli Studi Milano DERIVATIVES OF 3,6-DIAZABICICLO 3.1.I. HEPTANE WITH ANALGESIC ACTIVITY
AU2010328419A1 (en) 2009-12-07 2012-06-21 Targacept, Inc. 3,6-diazabicyclo[3.1.1]heptanes as neuronal nicotinic acetylcholine receptor ligands

Also Published As

Publication number Publication date
AR085676A1 (en) 2013-10-16
WO2012125518A1 (en) 2012-09-20

Similar Documents

Publication Publication Date Title
EP2465501B1 (en) Method for the preparation of (2S,3R)-N-(2-((3-pyridinyl)methyl)-1-azabicyclo[2.2.2]oct-3-yl)benzofuran-2-carboxamide
KR101737924B1 (en) Preparation and therapeutic applications of (2S,3R)-N-2-((3-pyridinyl)methyl)-1-azabicyclo[2.2.2]oct-3-yl)-3,5-difluorobenzamide
US10919879B2 (en) Synthesis and novel salt forms of (R)-5-((E)-2-pyrrolidin-3-ylvinyl)pyrimidine
WO2011071758A1 (en) 3,6-diazabicyclo[3.1.1]heptanes as neuronal nicotinic acetylcholine receptor ligands
EP2364307B1 (en) Synthesis and novel salt forms of (r)-5-((e)-2-(pyrrolidin-3-ylvinyl)pyrimidine
CA2744191A1 (en) A process for the preparation of tert-butyl (r)-3-vinylpyrrolidine-1-carboxylate and intermediates thereof
TW201249841A (en) Novel salt forms of 3-cyclopropylcarbonyl-3,6-diazabicyclo[3.1.1]heptane
TW202334120A (en) Crystal of substituted piperidine compound, salts of substituted piperidine compound, and crystals thereof
JP2011522016A (en) 4-Bromophenyl 1,4-diazabicyclo [3.2.2] nonane-4-carboxylate fumarate, its crystalline form, preparation and therapeutic use