TWI687217B - Compositions and methods of use of phorbol esters for the treatment of stroke - Google Patents

Compositions and methods of use of phorbol esters for the treatment of stroke Download PDF

Info

Publication number
TWI687217B
TWI687217B TW106126482A TW106126482A TWI687217B TW I687217 B TWI687217 B TW I687217B TW 106126482 A TW106126482 A TW 106126482A TW 106126482 A TW106126482 A TW 106126482A TW I687217 B TWI687217 B TW I687217B
Authority
TW
Taiwan
Prior art keywords
crotyl alcohol
crotonate
deoxycrotonol
formula
acetate
Prior art date
Application number
TW106126482A
Other languages
Chinese (zh)
Other versions
TW201735915A (en
Inventor
韓正濤
程宏峰
Original Assignee
華鴻新藥股份有限公司
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 華鴻新藥股份有限公司 filed Critical 華鴻新藥股份有限公司
Priority to TW106126482A priority Critical patent/TWI687217B/en
Publication of TW201735915A publication Critical patent/TW201735915A/en
Application granted granted Critical
Publication of TWI687217B publication Critical patent/TWI687217B/en

Links

Abstract

Methods and compositions containing a phorbol ester or a derivative of a phorbol ester are provided for the treatment and prevention of stroke and the sequelae of stroke. Additional compositions and methods are provided which employ a phorbol ester or derivative compound in combination with at least one additional agent to yield more effective treatment tools to treat or prevent stroke and the long term effects of stroke in mammalian subjects.

Description

用於中風治療之巴豆酯組成物及使用方法Crotonate composition for stroke treatment and method of use

本發明大致上關於一種巴豆酯於治療及預防中風及中風後遺症之之醫藥用途。 關於本申請書之其他揭露可於2008年1月31日由Richard D.Chang等人所申請之美國專利申請號12/023,753之「使用巴豆酯之組成物及方法(Compositions And Methods Of Use Of Phorbol Esters)」中尋得,此美國專利申請案主張於2007年1月31日所提出之美國臨時申請案號60/898,810之優先權效益,並於此全部併入以於各方面作為參考。The present invention generally relates to a medical use of crotonate for the treatment and prevention of stroke and stroke sequelae. For other disclosures related to this application, US Patent Application No. 12/023,753, "Compositions And Methods Of Use Of Phorbol", applied by Richard D. Chang et al. on January 31, 2008 Esters)”, this US patent application claims the priority benefit of US Provisional Application No. 60/898,810 filed on January 31, 2007, and is hereby incorporated by reference for all aspects.

植物歷來已作為許多醫藥用途。世界衛生組織(WHO)估計4億人,佔世界人口之80%目前在某些情況下係使用草藥作為初步之衛生保健。(世界衛生組織第134號文件,2008年12月)。然而,很難分離出植物中具有理想醫藥效果之特定化合物並依商業規格複製。另外,雖然可以從植物分離出活性化合物,植物之其他部份如礦物質、維生素、揮發油(volatile oils)、苷(glycosides)、生物鹼(alkaloids)、生物類黃酮(bioflavanoids)及其他物質亦可能涉及了活性化合物之運作或此植物已知之醫藥用途,使得使用、純化和商品化以植物為基礎的藥劑為一種挑戰。 巴豆醇是二萜(diterpenes)之萜烷類(tigliane family)之天然植物性有機化合物。其首先於1934年自原產於東南亞之大戟科(Euphorbiaceae family)灌木之巴豆(Croton tiglium)之種子作為巴豆油(croton oil)之水解產物所分離出。巴豆醇之各種酯穎具有據知包含仿甘油二酯(diacylglycerols)及活 化蛋白激酶C(protein kinase C, PKC)之能力;以及調整含有有絲分裂活化蛋白激酶(mitogen-activated protein kinase,MAPK)路徑之下游細胞訊號路徑的重要生物學特性。巴豆酯另外也被認為會結合至嵌合蛋白(chimaerins)、Ras活化子Ras GRP、及突觸囊泡啟動蛋白質(vesicle-priming protein)Munc-13(Brose N, Rosenmund C., JCell Sci;115:4399-411 (2002))。部份巴豆酯也誘導核轉錄因子‑κB(NF-κB)。最值得一提的是巴豆酯據知能夠作為腫瘤促進劑(promoter)之生理特性。(Blumberg,1988;Goel, G et al., Int,Journal of Toxicology 26,279-288(2007)) 12-O-十四烷醯巴豆醇-13-乙酸酯(TPA)為一種巴豆酯,亦稱為巴豆醇-12-肉蔻酸酯值-13-乙酸酯(phorbol-12-myristate-13-acetate, PMA),在多種細胞株及初代細胞中的致癌模式用於作為誘導分化及/或細胞凋亡。TPA亦已被報導在因化療而抑制骨髓作用之患者中,可使循環之白血球細胞及嗜中性粒細胞增加(Han Z. T. et al. Proc. Natl. Acad. Sci. 95, 5363-5365 (1998)),並抑制MT-4細胞上經HIV誘導之細胞病變的效果(Mekkawy S. et al., Phytochemistry 53, 47-464 (2000))。然而,由於包含接觸皮膚時之腐蝕反應(caustic reactions)的各種因素及對於其潛在毒性之疑慮,TPA無法展現出做為一種用於治療、管理或預防疾病之有效工具。實際上,由於巴豆酯在誘發造成發炎反應及腫瘤發展之各種細胞反應的蛋白激酶C之活化中扮演了重要的角色(Goel et al., Int, Journal of Toxicology 26, 279-288 (2007)),故巴豆酯通常將會自用於涉及發炎反應之症狀(如中風)的可能治療候選藥物中排除。 心臟疾病及中風一年造成1700萬人喪生,佔全球所有死亡率之幾乎三分之一。此預計將會變成全世界死亡及失能之首要原因,死亡的人數預測在2020年時將增加超過2000萬人,在2030年時則會增加至2400萬人。(Atlas of Heart Disease and Stroke,世界衛生組織2004) 雖然有超過300個危險因素與冠狀動脈心臟疾病和中風相關(Atlas of Heart Disease and Stroke,世界衛生組織2004),然而在已發展國家中,至少有1/3的心血管疾病係歸因於煙草使用、酒精使用、高血壓、高膽固醇和肥胖。 目前管理和預防中風的治療一般係結合如血管收縮素轉換酶抑制劑(ACE inhibitors)、阿斯匹靈(aspirin)、β受體阻滯劑(beta blockers)和降血脂藥物(lipid lowering medications)之藥物;如心臟起搏器(pacemakers)、植入式除顫器(implantable defibrillators)、冠狀動脈支架(coronary stents)、人工心臟瓣膜(prosthetic valves)、人工心臟(artificial hearts)之裝置;以及如冠狀動脈搭橋(coronary artery bypass)、球囊擴張術(balloon angioplasty)、瓣膜修復和置換(valve repair and replacement)、心臟移植(heart transplants)及人工心臟手術(artificial heart operations)的手術。「越來越傾向選擇高科技治療技術勝於較便宜但依舊有效果之治療法。」(Atlas of Heart Disease and Stroke,世界衛生組織2004),此增加了醫療保健成本之提高,且使不同個體之群組間的治療品質有著顯著的差距。 然而,即使可使用先進的技術和設施,經歷中風之60%的患者仍然會死亡或變得無法自理,且每次中風會使得復發的風險顯著地增加。「以全球來看,對大多數患者來說心血管疾病及其危險因素的治療仍然不夠。」(Atlas of Heart Disease and Stroke,世界衛生組織2004)。因此顯然需要更新且更有效的措施來預防和治療中風,以及治療或預防因中風所引起的長期影響。Plants have historically been used for many medical purposes. The World Health Organization (WHO) estimates that 400 million people, accounting for 80% of the world's population, currently use herbal medicine as the primary health care in some cases. (World Health Organization Document 134, December 2008). However, it is difficult to isolate specific compounds in plants with ideal medicinal effects and replicate them according to commercial specifications. In addition, although the active compound can be isolated from the plant, other parts of the plant such as minerals, vitamins, volatile oils (glycosides), alkaloids (alkaloids), bioflavanoids (bioflavanoids) and other substances are also possible The operation of the active compound or the known medical use of this plant makes the use, purification and commercialization of plant-based pharmaceuticals a challenge. Croton alcohol is a natural plant organic compound of the diterpenes (tigliane family). It was first isolated in 1934 from Croton tiglium seeds of Euphorbiaceae family shrubs native to Southeast Asia as hydrolysates of croton oil. Various esters of crotonol have known ability to include diacylglycerols and protein kinase C (PKC); and to adjust the pathway containing mitogen-activated protein kinase (MAPK) The important biological characteristics of the downstream cell signal path. Crotonate is also believed to bind to chimaerins, Ras activator Ras GRP, and synaptic vesicle-priming protein Munc-13 (Brose N, Rosenmund C., JCell Sci; 115 : 4399-411 (2002)). Some crotonates also induce nuclear transcription factor-κB (NF-κB). The most noteworthy is the physiological properties of crotonate known to be a tumor promoter. (Blumberg, 1988; Goel, G et al., Int, Journal of Toxicology 26, 279-288 (2007)) 12-O-tetradecane crotonol-13-acetate (TPA) is a crotonate, Also known as crotonol-12 myristate-13-acetate (phora-12-myristate-13-acetate, PMA), it is used as a carcinogenic model in various cell lines and primary cells to induce differentiation and /Or apoptosis. TPA has also been reported to increase circulating white blood cells and neutrophils in patients who suppress bone marrow due to chemotherapy (Han ZT et al. Proc. Natl. Acad. Sci. 95, 5363-5365 (1998 )) and inhibit the effect of HIV-induced cytopathy on MT-4 cells (Mekkawy S. et al., Phytochemistry 53, 47-464 (2000)). However, due to various factors including caustic reactions in contact with the skin and doubts about its potential toxicity, TPA cannot be shown as an effective tool for treating, managing, or preventing diseases. In fact, crotonate plays an important role in the activation of protein kinase C that induces various cellular responses that cause inflammation and tumor development (Goel et al., Int, Journal of Toxicology 26, 279-288 (2007)) Therefore, crotonate will usually be excluded from possible therapeutic candidates for symptoms involving inflammatory reactions, such as stroke. Heart disease and stroke kill 17 million people a year, accounting for almost one-third of all deaths worldwide. This is expected to become the leading cause of death and disability in the world. The number of deaths is predicted to increase by more than 20 million by 2020 and to 24 million by 2030. (Atlas of Heart Disease and Stroke, World Health Organization 2004) Although there are more than 300 risk factors associated with coronary heart disease and stroke (Atlas of Heart Disease and Stroke, World Health Organization 2004), in developed countries, at least One third of cardiovascular diseases are due to tobacco use, alcohol use, high blood pressure, high cholesterol and obesity. Current treatments for management and prevention of stroke generally combine treatments such as angiotensin converting enzyme inhibitors (ACE inhibitors), aspirin, beta blockers, and lipid lowering medications Drugs; such as pacemakers, implantable defibrillators, coronary stents, prosthetic valves, artificial hearts devices; and such as Coronary artery bypass, balloon angioplasty, valve repair and replacement, heart transplants, and artificial heart operations. "There is an increasing tendency to choose high-tech treatment techniques over cheaper but still effective treatments." (Atlas of Heart Disease and Stroke, World Health Organization 2004), which increases the cost of health care and makes different individuals There is a significant gap in the quality of treatment between the groups. However, even if advanced technology and facilities are available, 60% of patients who experience a stroke will still die or become unable to take care of themselves, and each stroke will significantly increase the risk of recurrence. "Globally, the treatment of cardiovascular disease and its risk factors is still insufficient for most patients." (Atlas of Heart Disease and Stroke, World Health Organization 2004). It is therefore clear that newer and more effective measures are needed to prevent and treat stroke, as well as to treat or prevent the long-term effects caused by stroke.

本發明關於一種包含巴豆酯之組成物及使用巴豆酯之方法。本文所述之組成物及方法在預防及治療中風及在控制包含急性缺血(acute ischemic events)之中風之後遺症上係相當有效。 可藉由使用本文所述之巴豆酯及巴豆酯衍生物來預防或治療中風之後果,其包括但不限於癱瘓、空間障礙、判斷障礙、忽視症(left-sided neglect)、記憶力減退、失語、協調和平衡問題、噁心、嘔吐、認知功能障礙、知覺障礙、定向障礙、單側偏盲和行為衝動。在一些實施例中,使用巴豆酯和巴豆酯衍生物可防止初始及後續中風的發生。 成功治療與否將根據常規方法來判定,如減少中風之嚴重性或後遺症、降低或消除中風後遺症、降低個體預見中風的風險因素、及/或減少包含後續中風(subsequent strokes)的中風次數或嚴重性。 在另一個實施例中,本文所述之巴豆酯及巴豆酯衍生物可用於調控細胞訊號路徑。這種調控可具有各種效果,例如,在一些實施例中,包含巴豆酯和巴豆酯衍生物之組成物之使用可能改變哺乳動物對象的Th1細胞激素的釋放。在另一實施例中,包含巴豆酯及/或巴豆酯衍生物之組成物可能改變哺乳動物對象的白介素2(interleukin 2, IL-2)的釋放。在另外的實施例中,包含巴豆酯及/或巴豆酯衍生物的組成物可能會改變哺乳動物對象的干擾素(interferon)的釋放。在又一實施例中,包含巴豆酯及/或巴豆酯衍生物之組成物可以改變ERK磷酸化的速率。 本發明藉著提供一種使用包含下式I巴豆酯之組成物之用於治療及預防中風、調節細胞訊號路徑及/或管理、治療及預防中風之後遺症之新穎且特別有效之方法及組成物來實現上述並達成另外目的及優點,

Figure 02_image001
式I 其中R1 及R2 可各獨立地為氫;羥基;
Figure 02_image004
,其中烷基包含1至15個碳原子;
Figure 02_image006
,其中低級烯基包含1至7間的碳原子;
Figure 02_image008
Figure 02_image010
Figure 02_image012
Figure 02_image014
及其經取代之衍生物。R3 可各獨立地為氫或
Figure 02_image016
及其經取代之衍生物。本發明之方法及組成物更包含任何式I之組成物之醫藥鹽、對映異構體、異構體、多晶型物、前藥、水合物及溶劑化物。 在一些實施例中,R1 及R2 之至少之一非為氫,且R3 為氫或
Figure 02_image006
及其取代之衍生物。在另一實施例中,R1 或R2 之一者為
Figure 02_image019
,而R1 或R2 之另一者為
Figure 02_image006
,其中低級烷基為1至7個碳原子之間,且R3 為氫。 在此式I化學式中之烷基、烯基、苯基、及芐基可未經鹵素取代或經鹵素取代,較佳為氯、氟或溴;可未經硝基取代或經硝基取代;可未經胺基取代或經胺基取代;及/或可未經相似之自由基取代或經相似之自由基取代。 用於此處所述之方法及組成物中之例示性巴豆酯組成物為下列式II之12-O-十四烷醯巴豆醇-13-乙酸酯(TPA)。
Figure 02_image022
式II 本發明之製劑及方法中之有用的式I巴豆酯及其相關化合物及衍生物包含但不限於其他醫藥適用之此化合物之活性鹽,以及此化合物之異構體、對映異構體、多晶型物、糖苷化衍生物、溶劑化物、水合物、及/或前藥。本發明之組成物及方法中所使用之巴豆酯之例示性形式包含但不限於巴豆醇13-丁酸酯;巴豆醇12-癸酸;巴豆醇13-癸酸;巴豆醇12,13-二乙酸酯;巴豆醇13,20-二乙酸酯;巴豆醇12,13-二苯甲酸;巴豆醇12,13-二丁酯;巴豆醇12,13-二癸酸酯;巴豆醇12,13-二己酸酯;巴豆醇12,13-二丙酸酯;巴豆醇12-肉蔻酸酯;巴豆醇13-肉蔻酸酯;巴豆醇12-肉蔻酸酯-13-乙酸酯(亦習知為TPA或PMA);巴豆醇12,13,20-三乙酸酯;12-去氧巴豆醇13-當歸酸酯;12-去氧巴豆醇13-當歸酸20-乙酸酯;12-去氧巴豆醇13-異丁酸酯;12-去氧巴豆醇13-異丁酸酯-20-乙酸酯;12-去氧巴豆醇13-苯乙酸酯;12-去氧巴豆醇13-苯乙酸酯20-乙酸酯;12-去氧巴豆醇13-肉蔻酸酯;巴豆醇12-順芷酸酯13-癸酸酯;12-去氧巴豆醇13-乙酸酯;巴豆醇12-乙酸酯;及巴豆醇13-乙酸酯。 哺乳動物對象係適用於以式I巴豆酯或式I巴豆酯衍生物來治療,尤其是TPA,根據本發明的方法包括但不限於罹患中風或對中風有風險之個體。適合以式I巴豆酯,尤其是TPA,來治療之對象則另外包括那些苦於中風影響之個體,其包括但不限於癱瘓、空間障礙、判斷力障礙、忽視症、記憶力減退、失語、協調和平衡問題、噁心、嘔吐、認知功能障礙、知覺障礙、定向障礙、單側偏盲和衝動。 這些和其它對象係對藉由對對象施用有效量的式I巴豆酯或式I巴豆酯衍生物而能有效地預防及/或治療性處置,有效量的式I巴豆酯或式I巴豆酯衍生物足以調節NF-κB的活性、增加Th1細胞激素的活性、預防或治療癱瘓、增加空間認知、減少記憶力減退、降低失語、增加協調和平衡、防止或減少中風的發病率和嚴重性、以及提高認知。 本發明治療上有效用之方法和製劑將有效地以各種形式使用式I巴豆酯或式I巴豆酯衍生物,如上所述,包括任何有活性的、醫藥上可接受之所述化合物的鹽、以及有活性的異構體、對映異構體、多晶型物、溶劑化物、水合物、前藥、及/或其組合。式II之TPA係於下文之實例中採用作為本發明的說明性實施例。 在本發明之其他態樣中,提供一種組合製劑和方法,其採用有效劑量之式I巴豆酯或式I巴豆酯衍生物來搭配與式I巴豆酯能組合配製或協調給藥之一或多個次要或輔助之活性劑,以產生針對對象之有效反應。 組合製劑和協調治療方法在中風治療上採用式I巴豆酯化合物或式I巴豆酯衍生物搭配額外之一或多個中風預防、治療或其他用途之次級或輔助治療劑。在這些實施例中用於搭配巴豆酯,例如TPA之次級或輔助治療劑可能在單獨或與巴豆酯例如TPA搭配時在中風預防或復原上 具有直接或間接之效果;可能在與如TPA搭配時會展現其他有用的輔助治療活性(例如抗凝血、抗胆甾醇血(anticholesterolemic)、舒張血管、抗高血壓、降低小動脈阻力、增加靜脈容量、減少心臟需氧量、降低心臟速率、穩定心臟速率、或保護神經);或可在單獨或與如TPA搭配時展現用於治療或預防中風或相關症狀之輔助治療活性。這樣的次級或輔助治療劑可在施用式I巴豆酯或式I巴豆酯衍生物之前、同時或之後施用。 在這些組合製劑和協調治療方法中對於預防或治療哺乳動物對象中風有用的輔助或次級治療劑包括但不限於組織血纖維蛋白溶酶原活化劑、抗凝血劑、他汀類藥物(statins)、血管收縮素II受體阻斷劑、血管收縮素轉換酶抑制劑、抗血小板藥物、ß-阻斷劑、阿斯匹靈、貝特類藥物、鈣通道阻斷劑、或利尿劑。此外,輔助治療或次級治療可使用例如但不限於外科手術介入,其包括頸動脈內膜切除術、血管成形術、球囊擴張術(balloon angioplasty)、瓣膜修復和置換、冠狀動脈搭橋術(coronary artery bypass)、支架置入術(stent placement)、開顱手術、彈簧圈栓塞術(endovascular coil emobilization)、或卵圓孔未閉封堵(patent foramen ovale closure)。 本發明之上述及其他標的、特徵、態樣及優勢將自下列詳細說明而變得顯而易知。The present invention relates to a composition containing crotonate and a method of using crotonate. The compositions and methods described herein are quite effective in preventing and treating stroke and in controlling sequelae including stroke after acute ischemic events. The consequences of stroke can be prevented or treated by using the crotonate and crotonate derivatives described herein, which include but are not limited to paralysis, spatial impairment, judgment impairment, left-sided neglect, memory loss, aphasia, Coordination and balance problems, nausea, vomiting, cognitive dysfunction, perceptual impairment, disorientation, unilateral hemianopia, and behavioral impulsivity. In some embodiments, the use of crotonate and crotonate derivatives can prevent the occurrence of initial and subsequent strokes. Successful treatment will be judged according to conventional methods, such as reducing the severity or sequelae of stroke, reducing or eliminating the sequelae of stroke, reducing the individual’s risk factors for predicting stroke, and/or reducing the number or severity of strokes including subsequent strokes Sex. In another embodiment, the crotonate and crotonate derivatives described herein can be used to regulate cellular signaling pathways. Such modulation may have various effects, for example, in some embodiments, the use of a composition comprising crotonate and crotonate derivatives may alter the release of Th1 cytokines in mammalian subjects. In another embodiment, a composition comprising crotonate and/or crotonate derivatives may alter the release of interleukin 2 (IL-2) in mammalian subjects. In other embodiments, a composition comprising crotonate and/or crotonate derivatives may alter the release of interferon in mammalian subjects. In yet another embodiment, a composition comprising crotonate and/or crotonate derivatives can change the rate of ERK phosphorylation. The present invention provides a novel and particularly effective method and composition for the treatment and prevention of stroke, the regulation of cell signal pathways and/or management, the treatment and prevention of stroke sequelae, using a composition comprising crotonate of the following formula I To achieve the above and achieve additional objectives and advantages,
Figure 02_image001
Formula I wherein R 1 and R 2 can each independently be hydrogen; hydroxyl;
Figure 02_image004
, Where the alkyl group contains 1 to 15 carbon atoms;
Figure 02_image006
, Where the lower alkenyl group contains 1 to 7 carbon atoms;
Figure 02_image008
;
Figure 02_image010
;
Figure 02_image012
;
Figure 02_image014
And their substituted derivatives. R 3 may each independently be hydrogen or
Figure 02_image016
And their substituted derivatives. The method and composition of the present invention further include pharmaceutical salts, enantiomers, isomers, polymorphs, prodrugs, hydrates and solvates of any composition of formula I. In some embodiments, at least one of R 1 and R 2 is not hydrogen, and R 3 is hydrogen or
Figure 02_image006
And their substituted derivatives. In another embodiment, one of R 1 or R 2 is
Figure 02_image019
, And the other of R 1 or R 2 is
Figure 02_image006
Where lower alkyl is between 1 and 7 carbon atoms, and R 3 is hydrogen. In this formula I, the alkyl, alkenyl, phenyl, and benzyl groups may be unsubstituted or substituted with halogen, preferably chlorine, fluorine or bromine; may be unsubstituted or substituted with nitro; May be unsubstituted or substituted with amine groups; and/or may not be substituted with similar free radicals or substituted with similar free radicals. An exemplary crotonate composition for use in the methods and compositions described herein is 12-O-tetradecane crotonol-13-acetate (TPA) of Formula II below.
Figure 02_image022
Formula II Crotonate and its related compounds and derivatives useful in the formulations and methods of the present invention include, but are not limited to, active salts of this compound for other pharmaceutical applications, as well as isomers and enantiomers of this compound , Polymorphs, glycosylated derivatives, solvates, hydrates, and/or prodrugs. Exemplary forms of crotonate used in the compositions and methods of the invention include, but are not limited to, crotonol 13-butyrate; crotonol 12-decanoic acid; crotonol 13-decanoic acid; crotonol 12,13-di Acetate; crotyl alcohol 13,20-diacetate; crotyl alcohol 12,13-dibenzoic acid; crotyl alcohol 12,13-dibutyl ester; crotyl alcohol 12,13-didecanoate; crotyl alcohol 12, 13-dihexanoate; crotyl alcohol 12,13-dipropionate; crotyl alcohol 12-myristate; crotyl alcohol 13-myristate; crotyl alcohol 12-myristate-13-acetate (Also known as TPA or PMA); crotonol 12,13,20-triacetate; 12-deoxycrotonol 13-angelica; 12-deoxycrotonol 13-angelica 20-acetate ; 12-deoxycrotonol 13-isobutyrate; 12-deoxycrotonol 13-isobutyrate-20-acetate; 12-deoxycrotonol 13-phenylacetate; 12-deoxy Crotyl alcohol 13-phenylacetate 20-acetate; 12-deoxycrotonol 13-myristate; crotonol 12-cis erythroate 13-decanoate; 12-deoxycrotonol 13-ethyl Acid ester; crotonol 12-acetate; and crotonol 13-acetate. Mammalian subjects are suitable for treatment with crotonate of formula I or derivatives of crotonate of formula I, especially TPA. The methods according to the present invention include but are not limited to individuals suffering from or at risk of stroke. Subjects suitable for treatment with crotonate of formula I, especially TPA, include those suffering from stroke, including but not limited to paralysis, spatial impairment, judgment impairment, neglect, memory loss, aphasia, coordination and balance Problems, nausea, vomiting, cognitive dysfunction, sensory disturbances, disorientation, unilateral hemianopia and impulsivity. These and other subjects are effective for preventive and/or therapeutic treatment by administering to the subject an effective amount of a crotonate of formula I or a derivative of crotonate of formula I, an effective amount of crotonate of formula I or crotonate of formula I Substances sufficient to regulate the activity of NF-κB, increase the activity of Th1 cytokines, prevent or treat paralysis, increase spatial cognition, reduce memory loss, reduce aphasia, increase coordination and balance, prevent or reduce the incidence and severity of stroke, and improve Cognition. The therapeutically effective methods and formulations of the present invention will effectively use the formula I crotonate or formula I crotonate derivative in various forms, as described above, including any active, pharmaceutically acceptable salt of the compound, And active isomers, enantiomers, polymorphs, solvates, hydrates, prodrugs, and/or combinations thereof. The TPA of Formula II is adopted in the following examples as an illustrative embodiment of the present invention. In other aspects of the present invention, a combination preparation and method is provided, which uses an effective dose of one or more of formula I crotonate or formula I crotonate derivative to be combined with formula I crotonate to formulate or coordinately administer one or more A secondary or auxiliary active agent to produce an effective response to the subject. The combined preparation and coordinated treatment method use a crotonate compound of formula I or a crotonate derivative of formula I in combination with one or more secondary or adjuvant therapeutic agents for stroke prevention, treatment, or other purposes. In these embodiments, secondary or adjuvant therapeutic agents used for crotonate, such as TPA, may have direct or indirect effects on stroke prevention or recovery when used alone or in combination with crotonate, such as TPA; may be used in combination with, for example, TPA Will display other useful adjuvant therapeutic activities (e.g. anticoagulant, anticholesterolemic, vasodilator, antihypertensive, reduce arteriolar resistance, increase venous volume, reduce cardiac oxygen demand, reduce heart rate, stabilize Heart rate, or nerve protection); or may exhibit adjuvant therapeutic activity for treating or preventing stroke or related symptoms when alone or in combination with, for example, TPA. Such secondary or adjunct therapeutic agents can be administered before, at the same time, or after administration of the formula I crotonate or formula I crotonate derivative. Auxiliary or secondary therapeutic agents useful in preventing or treating stroke in mammalian subjects in these combination formulations and coordinated treatment methods include, but are not limited to, tissue plasminogen activator, anticoagulant, statins , Angiotensin II receptor blockers, angiotensin converting enzyme inhibitors, antiplatelet drugs, ß-blockers, aspirin, fibrates, calcium channel blockers, or diuretics. In addition, adjuvant therapy or secondary therapy may use, for example, but not limited to, surgical interventions, including carotid endarterectomy, angioplasty, balloon angioplasty, valve repair and replacement, coronary artery bypass grafting ( coronary artery bypass, stent placement, craniotomy, endovascular coil emobilization, or patent foramen ovale closure. The above and other objects, features, aspects and advantages of the present invention will become apparent from the following detailed description.

一種新穎方法和組成物己證實可用於預防及/或治療包括人類之哺乳動物對象的中風及中風後遺症。 在各種實施例中,如本文所述的組成物和方法可能會增加Th1細胞激素(Th1 cytokines)的釋放,增加ERK的磷酸化,調節NF-κB的活性,預防或治療癱瘓,增加空間感,減少記憶損失,降低失語,增加協調平衡,提高認知,改進方向感,減少後續中風的發生率,以及減少衝動控制異常(impulsivity)。 在此所提供之製劑和方法採用式I巴豆酯或式I巴豆酯之衍生化合物,如於2008年1月31日所申請的美國專利申請號12/023,753所更完整地說明之內容,其係主張於2007年1月31日所提出的美國臨時專 利申請號60/898,810、以及所申請之美國臨時專利申請案之優先權效益,其全部內容各於此全部併入作為參考,

Figure 02_image001
式I 其中R1 及R2 可為氫;羥基;
Figure 02_image004
,其中烷基包含1至15個碳原子;
Figure 02_image006
,其中低級烯基包含1至7之間的碳原子;
Figure 02_image008
Figure 02_image010
Figure 02_image012
Figure 02_image014
及其經取代之衍生物。R3 可為氫或
Figure 02_image016
。 在一些實施例中,作為用於治療慢性或復發症狀之新穎組成物之R1 及R2 之至少之一非為氫,且R3 為氫或
Figure 02_image016
及其經取代之衍生物。在另一實施例中,R1 或R2 之一為
Figure 02_image019
。而R1 或R2 之另一個為
Figure 02_image006
。其中低級烯基為1至7個碳原子,且R3 為氫。 在此化學式之烷基、烯基、苯基、及芐基可未經鹵素取代或經鹵素取代,較佳為氯、氟或溴;可未經硝基取代或經硝基取代;可未經胺基取代或經胺基取代;及/或可未經相似之自由基取代或經相似之自由基取代。 在此所提供之中風治療製劑及方法採用式I之巴豆酯或式I之巴豆酯之衍生化合物,如上所述,包括所有本說明中活性藥學上可接受的化合物以及各種可能變化,且輕易地提供其複合物、鹽、溶劑化物、異構體、對映異構體、多晶型物和此些化合物之前藥及其組合作為抗中風劑。 在此提供之Th1細胞激素增加的製劑和方法採用式I巴豆酯或式I巴豆酯之衍生化合物,如上所述,包括所有本說明中活性藥學上可接受的化合物以及各種可能變化,且輕易地提供其複合物、鹽、溶劑化物、異構體、對映異構體、多晶型物和此些化合物之前藥及其組合作為新穎之Th1細胞激素增加劑,這些化合物及其組合物作為一種新穎之Th1細胞因子的增加劑。包括人類對象在內之廣泛範圍的哺乳動物對象適合使用本發明的製劑和方法來治療。這些對象包括但不限於罹患中風或有中風風險的個體。 在此所提供之ERK磷酸化增加製劑和方法採用式I巴豆酯或式I巴豆酯之衍生化合物,如上所述,包括所有本說明中活性藥學上可接受的化合物以及各種可能變化,且輕易地提供其複合物、鹽、溶劑化物、異構體、對映異構體、多晶型物和此些化合物之前藥及其組合作為新穎之ERK磷酸化增加劑。包括人類對象在內之廣泛範圍的哺乳動物對象適合使用本發明的製劑和方法來治療。這些對象包括但不限於罹患中風或有中風風險的個體。 在本發明的方法及組成物中,此處所述之一或多個式I巴豆酯化合物或式I巴豆酯之衍生化合物係有效地配製或給藥以作為用於有效地治療和預防中風或中風後遺症之藥劑。在例示性實施例中,TPA係為了說明目的而示範在醫藥上之製劑及治療方法上單獨或與一或多個輔助治療劑配合作為有效之藥劑。本揭露還另外提供醫藥上可接受的巴豆酯化合物,其係為天然或合成化合物之形式,包括此處所述之複合物、衍生物、鹽、溶劑化物、異構體、對映異構體、多晶型物和此些化合物之前藥及其組合,這些作為本發明的方法和組成物中的治療藥劑在治療和預防中風和中風的後遺症上是相當有效的。 中風為大腦血液供應中斷所造成的。此可能肇因自血管堵塞(缺血性中風(ischaemic stroke))或血管破裂(出血性中風(haemorrhagic stroke))。中風的症狀包括突然麻木或無力,特別是一側身體;突然神智不清,或說話或理解話語困難;單眼或雙眼觀看困難;突然走路困難、頭暈或失去平衡或協調性;或突然無來由的劇烈頭痛。中風之風險因素包含高血壓、血脂異常、使用煙草、缺乏運動、肥胖、壓力、糖尿病、飲酒、血液中有過多同半胱胺酸(homocystein)、發炎和凝血功能異常。亦有無法改變的危險因素,如年齡、遺傳、性別和種族。 治療中風具有三個不同的階段:預防、中風後的立刻治療、中風後的復健。本文所述的組成物和方法可用於中風治療的任何階段,可獨立地或與一或多種另外的治療方法搭配,包括其他藥劑、裝置或外科手術治療。 巴豆醇是二萜(diterpenes)之萜烷類(tigliane family)之天然植物性的多環醇。其首先於1934年自巴豆(Croton tiglium)之種子作為巴豆油(croton oil)之水解產物而分離出。巴豆醇在大部分極性有機溶劑及水中易溶。巴豆醇之酯類具有下列式I的整體結構:
Figure 02_image001
式I 其中R1 及R2 係選自氫;羥基;
Figure 02_image004
,其中烷基包含1至15個碳原子;
Figure 02_image006
,其中低級烯基包含1至7之間的碳原子;
Figure 02_image008
Figure 02_image010
Figure 02_image012
Figure 02_image014
;及其經取代之衍生物所組成之群組,而R3 可為氫或
Figure 02_image016
或其經取 代之衍生物,以及式I化合物之醫藥上可接受之鹽、對映異構體、多晶型物、前藥、溶劑化物、及水合物以及其經取代衍生物。 在此使用之詞彙「低級烷基」或「低級烯基」表示含有1至7個碳原子的基團。式I化合物中,烷基或烯基可以是直鏈或支鏈。在一些實施例中,其中R1 或R2 之一或R1 及R2 皆為一種長鏈碳基團(即式I為癸酸或肉荳蔻酸)。 此處化學式之烷基、烯基、苯基和芐基可未經鹵素取代或經鹵素取代,較佳地,氯、氟或溴;可未經硝基取代或經硝基取代;可未經胺基取代或經胺基取代;及可未經相似類型之自由基取代或經相似類型之自由基取代。 包括任何自草本來源像是巴豆(croton tiglium)之配製物或萃取物之有機及合成形式的巴豆酯用於本文之實施例中係擬作為包含巴豆酯(或巴豆酯類似物、相關化合物及/或衍生物)之有效用組成物。用於本文之實施例中之有效用之巴豆酯及/或相關化合物將通常具有如式I所示之結構,不過所屬技術領域具有通常知識者將了解此些化合物之功能相等之類似物、複合物、共軛物、及衍生物亦於本發明之範疇內。 在更詳細的實施例中,將選擇根據上述式I之說明性結構修改以提供有效用之選擇化合物,以治療及/或預防包合人類在內之哺乳動物對象之中風、肇因中風之傷害及/或管理中風之後果或後遺症,其中:R1 及R2 之至少之一非為氫,且R3 選自氫或
Figure 02_image016
及其經取代之衍生物所組成之群組。在另一實施例中,R1 或R2 之一者為
Figure 02_image038
,而R1 或R2 之另一者為
Figure 02_image006
,且R3 為氫。 在治療或預防包含人類在內之哺乳動物對象之中風、來自中風之傷害及/或管理中風之後果或後遺症而用於此治療法之式I巴豆酯化合物之例示性實施例係表示於下列式II所示之巴豆醇12-肉蔻酸酯-13-乙酸酯(亦習知為PMA或12-O-十四烷醯-巴豆醇-13-乙酸酯,TPA)。
Figure 02_image022
式II 本發明之製劑及方法中另外有用的巴豆酯及其相關化合物及衍生物包含但不限於其他醫藥適用之此化合物之活性鹽,以及此化合物之活性異構體、對映異構體、多晶型物、糖苷化衍生物、溶劑化物、水合物、及/或前藥。式I巴豆酯之衍生物可為或可非為巴豆酯本身。本發明之組成物及方法中所使用之巴豆酯之例示性形式包含但不限於巴豆醇13-丁酸酯;巴豆醇12-癸酸;巴豆醇13-癸酸;巴豆醇12,13-二乙酸酯;巴豆醇13,20-二乙酸酯;巴豆醇12,13-二苯甲酸;巴豆醇12,13-二丁酯;巴豆醇12,13-二癸酸酯;巴豆醇12,13-二己酸酯;巴豆醇12,13-二丙酸酯;巴豆醇12-肉蔻酸酯;巴豆醇13-肉蔻酸酯;巴豆醇12,13,20-三乙酸酯;12-去氧巴豆醇13-當歸酸酯;12-去氧巴豆醇13-當歸酸20-乙酸酯;12-去氧巴豆醇13-異丁酸酯;12-去氧巴豆醇13-異丁酸酯-20-乙酸酯;12-去氧巴豆醇13-苯乙酸酯;12-去氧巴豆醇13-苯乙酸酯20-乙酸酯;12-去氧巴豆醇13-肉豆蔻酸酯;巴豆醇12-順芷酸酯13-癸酸酯;12-去氧巴豆醇13-乙酸酯;巴豆醇12-乙酸酯;及巴豆醇13-乙酸酯,如表1所示。 表1 例行性巴豆酯
Figure 106126482-A0304-0001
本文所述的組成物包含包括減輕或預防中風傷害之有效量之式I化合物之巴豆酯或式I巴豆酯之衍生化合物之中風治療組成物,其對於預防及/或治療哺乳動物對象之中風或中風相關症狀或後遺症相當有效。有效量的活性化合物在「中風治療」、「抗凝血」、「抗胆甾醇血」(anticholesterolemic)」、「舒張血管」、「抗高血壓」、「降低小動脈阻力」、「增加靜脈容量」、「減少心臟需氧量」、「降低心臟速率」、「穩定心臟速率」、或「保護神經」治療上係相當有效,施以單位或多單位劑量形式超過干預治療之一特定期間以測量對象身上中風之一或多個症狀或後遺症之緩解。在例示性實施例中,本發明之組成物在預防或緩解對中風脆弱或苦於中風之人類及其他哺乳動物對象之中風症狀或中風後遺症之治療方法中係相當有效。 本發明之巴豆酯治療組成物中一般包含有效量或單位劑量的式I巴豆酯化合物或式I巴豆酯衍生化合物,其可與可加強穩定性、傳遞性、吸收性、半衰期、藥效、藥物動力學和及/或藥效學、減少不良的副作用、或提供醫藥運用上之其它優點之一或多種藥學上可接受的載劑、賦形劑、媒劑、乳化劑、穩定劑、防腐劑、緩衝劑、及/或其它添加劑配合。本技術領域具有通常知識者依據臨床和患者特定因素將能輕易地決定有效 量的式I巴豆酯化合物或其相關衍生化合物(例如,包含有效濃度/量的TPA之單位劑量,或所選擇之TPA之藥學上可接受的鹽、異構體、對映異構體、溶劑化物、多晶型物和/或前藥)。用於施予包含人類之哺乳動物對象之適合之有效量單位劑量之活性化合物可於約10μg至約1500μg、約20μg至約1000μg、約25μg至約750μg、約50μg至約500μg、約150μg至約500μg、約125μg至約500μg、約180至約500μg、約190μg至約500μg、約220μg至約500μg、約240μg至約500μg、約260μg至約500μg、約290μg至500μg之範圍內。在部份實施例中,疾病治療有效劑量之式I巴豆酯化合物或相關或衍生化合物可能選自很小的範圍,舉例來說,10至25μg、30至50μg、75至100μg、100至300μg或150至500μg。這些及其他有效單位劑量可以單一劑量、或以每日、每週或每月多次劑量之形式施用,舉例來說,包含每天、每週、或每月1至5次、或2至3次劑量施用的給藥方案。在一例示性實施例中,10μg至30μg、30μg至50μg、50μg至100μg、100μg至300μg、或300μg至500μg之劑量係以每天一次、兩次、三次、四次、或五次施用。在更詳細之實施例中,50μg至100μg、100μg至300μg、300μg至400μg、或400μg-600μg之劑量係每日施用一或兩次。在進一步的實施例中,50μg至100μg、100μg至300μg、300μg至400μg、或400μg至600μg之劑量係於每隔日施用。在另外的實施例中,劑量係基於體重來計算,且可施用量為,例如,每天約0.5μg/m2 至約300μg/m2 、約1μg/m2 至約200μg/m2 、每天約1μg/m2 至187.5μg/m2 、每天約1μg/m2 至每天約175μg/m2 、每天約1μg/m2 至約157μg/m2 、每天約1μg/m2 約125μg/m2 、每天約1μg/m2 約75μg/m2 、每天1μg/m2 約50μg/m2 、每天2μg/m2 至約50μg/m2 、每天2μg/m2 至約30μg/m2 或每天3μg/m2 至約30μg/m2 。 在其它實施例中,給藥劑量可以較少,例如,每隔天0.5μg/m2 至約300μg/m2 、約1μg/m2 至約200μg/m2 、每隔天約1μg/m2 至約187.5μg/m2 、每隔天約1μg/m2 約175μg/m2 、每天約1μg/m2 至每隔天約157μg/m2 、每隔天約1μg/m2 約125μg/m2 、每隔天約1μg/m2 至約75μg/m2 、每隔天約1μg/m2 至約50μg/m2 、每隔天2μg/m2 至約50μg/m2 、每天2μg/m2 至約30μg/m2 或每天3μg/m2 至約30μg/m2 。在另外的實施例中,劑量給藥可為3次/週、4次/週、5次/週、僅於週末、僅協同其他治療方案、數天、或依據臨床及病患特定 因素之任何適合之劑量方案。 包含有效量(選擇性地針對「中風治療」、「抗凝血」、「抗胆甾醇血」(anticholesterolemic)」、「舒張血管」、「抗高血壓」、「降低小動脈阻力」、「增加靜脈容量」、「減少心臟需氧量」、「降低心臟速率」、「穩定心臟速率」、「誘使ERK磷酸化」、「IL-2調節」及/或「保護神經」)之式I巴豆酯化合物或式I巴豆酯之衍生化合物之本發明之組成物之傳達量、時機點、及模式將基於個體基準,依據個體像是重量、年齡、性別及狀況之因素、疾病之劇烈性及/或相關症狀、給藥是預防或治療性質、以及基於包含半衰期及效率之其他習知會影響藥物傳遞、吸收及藥物代謝動力之因素而定期地調整。 本發明製劑之用於即時疾病治療(選擇性地針對「中風治療」、「抗凝血」、「抗胆甾醇血」」、「舒張血管」、「抗高血壓」、「誘使ERK磷酸化」、「降低小動脈阻力」、「增加靜脈容量」、「減少心臟需氧量」、「降低心臟速率」、「穩定心臟速率」、「減少血塊」、「保護神經」、「IL-2調節」或「NFκB調節」)之有效劑量或多劑量治療方案起初將選擇所需及足以實際預防或緩解對象身上中風之症狀之概略最小劑量方案。劑量及給藥方針將時常包含重複給藥治療數天或甚至一或多週或年之過程。有效治療方案亦可能涉及每天一次或每天多次持續數天、數週、數月或甚至數年之過程之預防性劑量。 本發明之組成物及方法在中風治療上之有效性可使用各種模式系統來表現,其包含實例9所未之暫時性中度大腦動脈阻塞、實例8所示之永久性中度大腦動脈阻塞、血管內線性中度大腦動脈阻塞(endovascular filament middle cerebral artery occlusion)、實例7所示之栓塞性中度大腦動脈阻塞、內皮素-1誘導之動脈及靜脈收縮、或大腦皮質光化學。在模式系統中使用本發明之巴豆酯組成物將展現會降低症狀或長期影響,相較於控制動物降低了0%、20%、30%、50%或甚至到75%、90%、96%以上。 本發明之組成物及方法在中風治療上之有效性可進一步展現於患有中風之個體所表現之症狀之緩減。這類的症狀包含但不限於癱瘓、空間障礙、判斷力障礙、忽視症、記憶力減退、失語、協調和平衡問題、噁心、嘔吐、認知障礙、知覺障礙、定向障礙、單側偏盲和衝動。使用本 發明之巴豆酯組成物將由起始狀態降低0%、20%、30%、50%、甚或75%至90%、96%以上來降低個體表現之症狀。 在本發明之另外態樣中,提供了組合性疾病治療(針對「中風治療」、「抗凝血」、「抗胆甾醇血」」、「舒張血管」、「抗高血壓」、「誘使ERK磷酸化」、「降低小動脈阻力」、「增加靜脈容量」、「減少心臟需氧量」、「降低心臟速率」、「穩定心臟速率」或「NFκB調節」)製劑及協調給藥方法,其採用了有效量之式I巴豆酯化合物或式I巴豆酯衍生化合物以及會與式I巴豆酯化合物結合配製或協調給藥之一或多種次級或輔助劑,以產生綜合、多活性之疾病治療組成物或協調治療方法。 預防或治療中風之例示性組合製劑及協調治療方法採用了式I巴豆酯化合物、或式I巴豆酯之衍生化合物來搭配對於治療或預防標的疾病、狀況及/或症狀有用之另外一或多種保護神經或其他指向之次級或輔助治療劑。對於大部分本發明之組合製劑及協調治療方法,式I巴豆酯化合物或其相關或衍生化合物係與一或多種次級或輔助治療劑搭配來調配或協調給藥,以產生在組合時有效或協調時有用於避免或治療中風、或中風影響之組合製劑或協調治療方法。在本內容中之例示性組合製劑及協調治療方法採用了式I巴豆酯化合物或式I巴豆酯之衍生化合物,來搭配選自由組織血纖維蛋白溶酶原活化劑、抗凝血劑、他汀類藥物、血管收縮素II受體阻斷劑、血管收縮素轉換酯抑制劑、抗血小板藥物、貝特類藥物、β-阻斷劑、鈣通道阻斷劑、或利尿劑之一或多種次級或輔助治療劑。例示性的抗凝血劑包括但不限於肝素、華法林(warfarin)、類肝素(heparinoids)、苯茚二酮(phenindione)、安托美汀(atomentin)、醋硝香豆素(acenocoumarol)、苯丙香豆素(phenprocoumon)、艾卓肝素(idraparinux)、磺達肝素(fondaparinux)以及凝血酶抑製劑。例示性的他汀類包含但不限於洛伐他汀(lovastatin)、氨氯地平(amlodipine)、阿托伐他汀(atorvastatin)、瑞舒伐他汀(rosuvastatin)、辛伐他汀(simvastatin)、氟伐他汀(fluvastatin)、匹伐他汀(pitavastatin)及普伐他汀(pravastatin)。例示性的血管收縮素II受體阻斷劑包含但不限於坎地沙坦(candesartan)、依普沙坦(eprosartan)、依貝沙坦(irbesartan)、氯沙坦(losartan)、奧美沙坦(olmesartan)、替米沙坦(telmisartan)及纈沙坦(valsartan)。血管收縮素轉換酶抑制劑包括但不限於依那澤普利(enazepril)、卡托普利(captopril)、 依那普利(enalapril)、褔辛普利(fosinopril)、賴諾普利(isinopril)、莫昔普利(moexipril)、培哚普利(perindopril)、喹那普利(quinapril)、雷米普利(ramipril)及群多普利(trandolapril)。例示性的β-阻斷劑包含但不限於心得舒(alprenolol)、布新洛爾(bucindolol)、卡替洛爾(carteolol)、卡維地洛(carvedilol)、拉貝洛爾(1abetalol)、納多洛爾(nadolol)、氧烯洛爾(oxprenolol)、噴布特羅(penbutolol)、哚洛爾(pindolol)、普萘洛爾(propranolol)、索他洛爾(sotalol)、噻嗎洛爾(timolol)、杜仲(eucommia)、醋丁洛爾(acebutolol)、阿替洛爾(atenolol)、倍他洛爾(betaxolol)、比索洛爾(bisoprolol)、塞利洛爾(celiprolol)、艾司洛爾(esmolol)、美托洛爾(metoprolol)及奈必洛爾(nebivolol)。例示性鈣通道阻斷劑包含但不限於氨氯地平(amlodipine)、氯維地平(clevidipine)、地爾硫卓(diltiazem)、非洛地平(felodipine)、伊拉地平(isradipine)、硝苯地平(nifedipine)、尼卡地平(iücardipine)、尼莫地平(nimodipine)、尼索地平(nisoldipine)及維拉帕米(verapamil)。例示性利尿劑包括但不限於氯噻嗪(chlorothiazide)、氫氯噻嗪(hydrochlorothiazide)、布美他尼(bumetanide)、依他尼酸(ethacrynic acid)、呋塞米(furosemide)、阿米洛利(amiloride)、依普利酮(eplerenone)、螺內酯(spironolactone)及氨苯蝶啶(triamterene)。例示性貝特類藥物包括但不限於苯扎貝特(benzafibrate)、環丙貝特(ciprofibrate)、氯貝丁酯(clofibrate)、吉非貝齊(gemfibrozil)或非諾貝特(fenotibrate)。例示性抗血小板藥物包括但不限於氯吡格雷(clopidogrel)及噻氯匹定(ticlopidin)。 輔助治療方法可進一步採用外科手術治療,其包含但不限於利用起博器、植入式除顫器、冠狀動脈支架、人工心臟瓣膜、冠狀動脈搭橋、球囊擴張術、瓣膜修復和置換、頸動脈內膜切除術、血管成形術、支架置入術、開顱手術、彈簧圈栓塞術、卵圓孔未閉封堵及心臟移植。 本發明一些實施例提供包含了巴豆酯及具疾病治療活性之一或多個輔助劑之組合疾病治療(「中風治療」、「抗凝血」、「抗阻甾醇血」」、「舒張血管」、「抗高血壓」、「誘使ERK磷酸化」、「降低小動脈阻力」、「增加靜脈容量」、「減少心臟需氧量」、「降低心臟速率」、「穩定心臟速率」或「NFκB調節」)製劑。在這樣的組合製劑中,式I巴豆酯及具疾病治療活性之輔助劑將以疾病治療(「中風治療」、「抗凝血」、「抗胆甾醇血」」、「舒張血管」、「抗高血壓」、「誘使ERK磷酸化」、「降低小動脈阻力」、「增加靜脈容 量」、「減少心臟需氧量」、「降低心臟速率」、「穩定心臟速率」或「NFκB調節」)有效之劑量單獨或搭配存於組合製劑中。在例示性實施例中,式I巴豆酯化合物及非巴豆酯藥劑將各以疾病治療/預防量存在(亦即,以將會針對對象之症狀獨自引發可偵測性緩解之單劑量)。此外,組合製劑可以次要治療(sub-therapeutic)之單劑量包含一或兩種式I巴豆酯化合物及非巴豆酯藥劑,其中包含兩種藥劑之組合製劑特別在於其於引導疾病、狀況或症狀緩解反應之統合效果之兩種藥劑之組合劑量。因此,一或兩種式I巴豆酯或式I巴豆酯之衍生化合物及非巴豆酯藥劑可以次要治療劑量存於製劑中,或可依搭配給藥方針來施予,而在製劑或方法中其統合性地在對象身上引發疾病症狀、中風發生率或復發率、或中風後遺症之可偵測性緩解。在又另一實施例中,組合製劑可包含一或多個神經保護劑。在另一實施例中,組合製劑可包含如本文所述之一或多種抗發炎劑或其他次級或額外治療劑。 為了實現本發明之協調給藥方法,式I巴豆酯化合物或式I巴豆酯衍生化合物可依協調治療方針與本文所擬之一或多種次級或輔助治療劑同時或依序地施用。因此,在部份實施例中化合物係與本文所擬之非巴豆酯藥劑或任何其他次級或輔助治療劑使用如上所述之個別製劑或組合製劑(亦即,包含式I巴豆酯化合物或相關或衍生化合物、以及非巴豆酯治療藥劑)一起配合給藥。此協調給藥可同時或以任何順序依序地完成,且可能存在有一段時間期間僅有一或兩者(或全部)之活性治療劑單獨及/或統合性地展現其生物活性。 在其他實施例中,舉例來說,這樣的協調治療方法可遵照或衍生自治療中風之各種方針。舉例來說,協調治療方法可包含預防或治療因中風導致之傷害之巴豆酯及/或治療法。所有這樣的協調治療方法之差別態樣在於化學式I之巴豆酯化合物或式I巴豆酯之衍生化合物展現至少部分活性,其係與互補之中風預防或治療藥劑搭配展現產生期望臨床反應,或是產生藉由次級或輔助治療藥劑所提供之臨床反應。通常,式I巴豆酯化合物或式I巴豆酯之衍生化合物與次級或輔助治療藥劑之協調給藥將在對象身上產生超越僅單獨施用式I巴豆酯化合物或式I巴豆酯之衍生化合物、或次級或輔助治療藥劑所展現之治療效果更為改善之治療或預防性效果。此條件要考量直接影響及間接影響。 在例示性實施例中,式I巴豆酯化合物或式I巴豆酯之衍生化合物將與一或多種次級中風治療化合物或其他指向或輔助治療藥劑搭調給藥(同時或依序地,組合或個別製劑),例如,組織血纖維蛋白溶酶原活化劑、抗凝血劑、他汀類藥物、血管收縮素II受體阻斷劑、血管收縮素轉化脢抑制劑、貝特類藥物、β受體阻滯劑、鈣通道阻斷劑、降血脂藥物、抗血小板藥物、或利尿劑。此外,輔助或次級療法可用於治療中風或中風後果,例如但不限於起博器、植入式除顫器、冠狀動脈支架、人工心臟瓣膜、人工心臟、冠狀動脈搭橋、球囊擴張術、瓣膜修復和置換、心臟移植、頸動脈內膜切除術、血管成形術、支架置入術、開顱手術、彈簧圈栓塞術、或卵圓孔未閉封堵。 如上所述,在所有此處所擬之本發明之各種實施例中,疾病治療方法及製劑可以任何各種形式採用式I巴豆酯化合物,其包含標的化合物之醫藥上可接受之鹽、溶劑化物、異構體、對映異構體、多晶型物、溶劑化物、水合物、及/或前藥之任何一種或其組合。在本發明之例示性實施例中,為了說明目的係於治療製劑及方法中採用TPA。 本發明之醫藥組成物可以各種達成其意旨治療或預防目的之手法來給藥。用於本發明之組成物之給藥之適合途徑包括但不限於傳統傳遞途徑、裝置和方法,其包括注射方法,例如但不限於靜脈注射、肌肉注射、腹腔注射、脊髓內注射、鞘內注射、顱內注射、動脈內注射、皮下注射及鼻腔注射途徑。 本發明之組成物可更包含適用於採用之給藥之特定模式之醫藥上可接受之載體。本發明之組成物之劑量形式包含適用於如上所述之劑量單位之製備在藥學上認可之賦形劑。這樣的賦形劑包含但非旨在限於粘合劑、填充劑、潤滑劑、乳化劑、懸浮劑、甜味劑、矯味劑、防腐劑、緩衝劑、潤濕劑、崩解劑、起泡劑和其它常規賦形劑及添加劑。 如果需要的話,本發明的組成物可藉著使用緩釋載劑以控制釋放形式給藥,像是親水性,緩釋聚合物。本文中之例示性控制釋放劑包括但不限於具有黏度於約100cps至約100,000cps之範圍內之羥丙基甲基纖維素、或其他生物相容的基質如膽固醇。 部份本發明之式I巴豆酯組成物係設計用於非消化道給藥,例如,靜脈給藥、肌肉給藥、皮下給藥或腹腔給藥,其包含水性及非水性之無菌注射液並如許多其他所擬之本發明之組成物一樣可選擇性地包含抗氧化劑、緩衝劑、抑菌劑及/或使得製劑與哺乳動物對象之血液等滲透之溶質;以及可包含懸浮劑及/或增稠劑之水性或非水性之無菌懸液。製劑可以單位劑量或多劑量容量存放。本發明之另外組成物及製劑可包含於非消化道給藥後緩釋之聚合物。非消化道製劑可為適用於這類給藥之溶液、分散液或乳液。標的藥劑亦可配製為聚合物以在非消化道給藥後緩釋。醫藥上可接受之製劑及成份通常將為無菌或能便於消毒,具有生物惰性(biologically inert)且可輕易管理。這樣的聚合材料為所屬醫藥技術領域者所習知。非消化道製劑通常包含緩衝劑及防腐劑、以及醫藥上及生理上可接受之注入之液體,像是水、生理食鹽水、均衡鹽溶液、葡萄糖水溶液、甘油等。臨時注入之溶液、乳液及懸浮液可自前文所述之種額之無菌粉末、顆粒、或藥片來製備。較佳之活性成份之單位劑量製劑為包含如本文上述之每日劑量或單位、每日分次劑量、或其適當比例之單位劑量製劑。 在更詳細的實施例中,本發明之組成物可包含封裝之式I巴豆酯化合物或式I巴豆酯衍生化合物以用例如藉由凝聚技術或藉由界面聚合所製備之微膠囊、微粒、或微球體來傳遞,例如,各別之羥甲織維素、或明膠微膠囊及聚(甲基丙烯酸甲酯)微膠囊;膠體藥物載體系統(例如,脂質體、白蛋白微球、微乳劑、奈米顆粒、及奈米膠囊);或粗乳液(macroemulsions)。 如上所述,在某些實施例中,本發明之方法及組成物可採用醫藥上可接受之鹽,例如上述之式I巴豆酯化合物及/或其相關或衍生化合物之酸加成鹽或鹼鹽。醫藥上可接受之加成鹽之示例包含無機及有機酸加成鹽。適合之酸加成鹽係由不會形成有毒鹽類之酸形成,舉例來說,氯化氫、溴化氫、碘化氫、硫酸、硫化氫、硝酸、磷酸及磷酸鹽。另外醫藥上可接受之鹽包含但不限於金屬鹽,例如納鹽、鉀鹽、銫鹽等;鹼土金屬鹽,例如鈣鹽、鎂鹽等;有機胺鹽,例如三乙胺鹽、吡啶鹽、甲基吡啶鹽、乙醇胺鹽、三乙醇胺鹽、二環己基胺鹽、N,N'-二芐基乙二胺鹽等;有機酸鹽,例如乙酸鹽、檸檬酸鹽、乳酸鹽、琥珀酸鹽、酒石酸鹽、馬來酸鹽、富馬 酸鹽、扁桃酸鹽、乙酸鹽、二氯乙酸鹽、三氟乙酸鹽、草酸鹽及甲酸鹽;磺酸鹽,例如甲磺酸鹽、苯磺酸鹽及對甲苯磺酸鹽;以及胺基酸鹽,例如精胺酸鹽、天門冬胺酸鹽、谷胺酸鹽、酒石酸鹽及葡萄糖酸鹽。適合的鹼鹽係由不會形成有毒鹽類之鹼所形成,例如,鋁鈣、鋰、鎂、鉀、鈉、鋅和二乙醇胺鹽。 在其他詳細實施例中,本發明之方法及組成物採用式I巴豆酯之前藥。前藥係考量為任何在生物體中會釋放活性原藥之共價鍵結載體。用於本發明之前藥之示例包含具羥烷基(hydroxyalkyl)或胺基烷基(aminoalkyl)作為取代基之酯類或醯胺類(amides),且這些前藥可藉由如上所述之此類化合物與酐類(anhydrides)如玻珀酸酐(succinic anhydride)反應而製備。 亦將了解於此揭露之本發明涵蓋了使用所述化合物之生物體內代謝產物(無論是在標的前驅化合物給藥後於體內產生,或直接地以其代謝產物之形式給藥)來包含式I巴豆酯之方法及組成物。這樣的產物可來自所施予之化合物主要由於酵素作用過程之例如氧化、還原、水解、醯胺化、酯化等之結果。因此,本發明包含採用由包含式I巴豆酯化合物與哺乳動物對象接觸一段足以產生其代謝產物之期間之過程所製造之化合物之本發明之方法及組成物。這樣的產物一般來說藉由準備本發明之放射性標誌化合物,以可檢測劑量非消化道性地施予像是大鼠、小鼠、天竺鼠、猴子或人之動物身上,准予足夠時間使代謝發生,並自尿液、血液或其他生物性樣本分離其轉換產物來檢驗。 亦將了解於此揭露之本發明係涵蓋用於診斷包含但不限於哺乳動物對象上之中風之風險度、存在性、嚴重度或治療取向,或其他管理疾病之診斷組成物,其包含被標誌(例如,同位素標誌、螢光標誌或其他標誌,使用常規方法得以檢測被標誌化合物)的式I巴豆酯化合物與具有或表現中風之一或多個症狀之風險之哺乳動物對象(例如細胞、組織、器官或個體)接觸,且接著使用任何習知測定(known assays)之廣測定(broad array)及標誌/偵測方法來偵測被標誌化合物之存在、位置、代謝及/或結合狀態。在例示性實施例中,式I巴豆酯化合物係藉著具有一或多個原子被具有不同原子量或質量數之原子替代而被同位素標誌。可插入所揭露之化合物之同 位素之示例包含氫、碳、氮、氧、磷、氟、及氯之同位素,例如各為2 H、3 H、13 C、14 C、15 N、18 O、17 O、31 P、32 P、35 S、18 F、及36 Cl。同位素標誌的化合物接著會給藥至個體或其他對象,且接著如上所述根據常規技術偵測、產生有用的診斷及/或治療管理資料。 實施例 下文所述之實施例中展示巴豆酯及衍生化合物作為中風治療及預防劑具新穎及有力之效用。此些及另外發現會進一步地於下列實例中補充及闡釋。 實例I TPA在週邊血液白細胞(Peripheral White Blood Cells, WBC))上之效果以及S180細胞移植小鼠中之血紅素(Hemoglobin, Hb)計數 肉瘤180(Sarcoma 180, S 180)細胞係移植入Kwen-Ming小鼠。在第三天時,小鼠由腹腔(i.p.)給予50、100或200μ g/kg/天之TPA七天。在處置完成後的第二天,為了WBC及HB分析由所處置之小鼠之尾巴取出血液樣本。處置組之WBC計數(七天的50、100或200μ g/kg/天)各別為16.1±7.4、18.7±.3.0及20.7±.3.4 x109 /L;控制組之WBC計數為13.6±1.8x109 /L。處置組之Hb為136±11、149±12及149±10g/L,而控制組之Hb為134+-15g/L。結果顯示i.p.注射TPA可依劑量依賴形式增加小鼠之外周WBC計數,而經TPA處置之小鼠之Hb水平與控制小鼠相比並無顯著地影響。 實例II 劑量範圍研究 由於TPA運用時所造成之強烈局部刺激,TPA係以靜脈輸液來給予病患。在無菌注射器內之TPA溶液係注射入200ml之無菌生理食鹽水內且充分混合以用於靜脈輸液。 不同TPA臨床給藥劑量之毒性及副作用: (1)依1mg/病患/週給予TPA: 溶液中之1mg TPA係與200ml之無菌生理食鹽水充分地混合以用於以16μg/min之速率於1小時內完成靜脈輸液。在TPA給藥1 小時過後,病患開始畏寒持續約30分鐘,接著是具輕度至重度出汗之發燒(病患的體溫到達37.5-39.5℃,持續了3至5小時然後轉為正常)。上述症狀可藉由給予病患葡萄糖皮質素而緩解。此劑量之TPA造成少數病患出血,部份病患短期間內有呼吸困難,且在尿液中檢測出Hb。然而,此些副作用相當短暫且可回復。心、肝、腎及肺功能則皆檢驗正常。 (2)依0.5mg/病患x2/週給予TPA:(一週二劑量) 溶液中之0.5mg TPA係與200ml之生理食鹽水充分地混合以用於以8μg/min之速率於1小時內完成靜脈輸液。給藥過後之反應相似於1mg之TPA劑量,但程度比1mg劑量輕。病患較輕易地承受較輕劑量。有時,Hb在病患之尿液中被檢測出。呼吸困難則未觀察到。心、肝、腎及肺功能則皆正常。 (3)依0.25mg/病患x4/週給予TPA: 溶液中之0.25mg TPA係與200ml之生理食鹽水充分地混合以用於以4μg/min之速率於1小時內完成靜脈輸液。給藥過後,亦觀察到像是畏寒及發燒之症狀,但與較高劑量相比較為輕微。在尿液中未檢測出Hb,且沒有病患有呼吸困難。心、肝、腎及肺功能則皆正常。 實例III 以TPA治療難治性/復發性腫瘤 具有組織紀錄之難治性/復發性血液系統腫瘤/骨髓疾病之病患係以TPA(Xichuan Pharmaceuticals, Nan Yang, Henan, China)、地塞米松(dexamethasone)及三水楊酸膽鹼鎂(choline magnesium trisalicylate)之組合治療。如下所述之用於展現TPA對於急性骨髓細胞性白血病(Acute Myelogenous Leukemia, AML)治療上之治療用途之比較佳方法將施用以展現TPA在治療其他癌症疾病及惡性腫瘤上之用途。除了此處之特定方針以外,成功的治療及/或緩解將使用各種任何習知之癌症檢測及評斷方法來判定不同目標之癌症及腫瘤疾病,舉例來說,判定固態腫瘤之大小縮減、病理組織學研究以評估腫瘤生長、階段、轉移潛力、組織學之腫瘤標誌物之存在/表現程度等。 AML為通常需要即時及密集治療之侵襲性疾病。AML診斷之平均病患年齡為64至68歲,且超過60歲之病患以標準化療法治療的疾病治療率<20%。在無前例血液性疾病(antecedent hematologic disorder)後或在白血病之化療/放療之治療法前發展有AML之病患具有相似的貧乏效果,因為這種病患所罹患之疾病與特定不利之細胞遺傳與臨床特徵有關。因此,大部分診斷有AML之病患具有與預後不良相關之病患及/或疾病相關之特徵。對於具復發性疾病之病患來說,沒有任何標準非移植治療法展現出治療的能力。對於這些病患,AML通常是致命的疾病。故AML治療的新方法是必要的。 採用本發明之方法及組成物,基於TPA在調節細胞訊號路徑之新穎角色,TPA發展為治療患有AML之病患之治療劑,其能力足以誘使細胞株分化及/或細胞凋亡,且表現TPA在治療包含骨髓惡性腫瘤之癌症及腫瘤疾病上之效益之臨床資料。 到目前為止TPA之臨床評估以細胞存活性(cell viability)及細胞凋亡(apoptosis assays)測量展現TPA在至少部份AML病例中表現出直接治療之細胞毒性效果。藉由西方墨點分析法(Western analysis)所分析之初代培養中,TPA於細胞培養1小時時強烈地誘發ERK磷酸化。TPA對於初代AML細胞的細胞毒性係與在體外暴露24小時後之磷酸化ERK促生存訊號(phospho-ERK pro-survival signal)之後續損失相關。此觀察與據稱在藉由MEK抑制因子對ERK訊號進行醫藥干擾後降低初代AML細胞存活率之其他研究有極佳一致性,如PD98059、U0126及PD 184352。在本研究中,ERK訊號的損失與ERK磷酸酶誘導有關。 除了蛋白激酶C及ERK活化以外,TPA在AML母細胞(AML blasts)及白血病幹細胞中為持續活耀之習知NF-κB之誘發子(inducer)、促生存轉錄因子。本實驗室的最近研究展現AML細胞NF-κB可以地塞米松(dexamethasone)+三水楊酸膽鹼鎂(choline magnesium trisalicylate, CMT)治療48h而於體內抑制。此外,發現地塞米松可誘導MKP-1 ERK磷酸酶表現且加強TPA對初代AML樣本之細胞毒性。在本內容中,選擇了下列之例示性實施例以使用地塞米松及CMT作為與TPA之輔助藥物事先於24h前治療及24h後治療。此些藥物係有極佳耐受性且預期將降低治療之發炎副作用,並藉由增加ERK磷酸酶表現及抑制NF-K B 來加強TPA細胞毒性。除此之外,因為其抗發炎性、可改善副作用、且可藉由抑制持續性NF-K B表現之抗凋亡影響以及誘導會降低訊號路徑活性之磷酸酶來加強抗白血病活性,地塞米松及CMT將用於作為輔助藥物。 招募35個病患之初TPA第1階段研究[23個具難治性/復發性AML、2個具其他骨髓惡性腫瘤(CML-芽細胞危象期(CML-blast crisis)、骨髓增生異常伴隨過多芽細胞(myelodysplasia with excess blasts))、3個具何杰金氏病(Hodgkin's disease)、3個具非何杰金氏淋巴瘤(Non-Hodgkin's Lymphoma)以及4個具固體腫瘤]。大部分的病人具有難治性/復發性AML。本臨床結果包含接受8 TPA輸液之一個AML病人病情穩定達到>5個月。在第二個病人身上,TPA給藥後發現循環芽細胞之數目明顯的下降(5倍)。在白血芽細胞(leukemic blasts)上的下降維持了4週,而病患最後死於真菌感染。最後,就算施行了高劑量化療與自體幹細胞援救仍具有難治性/復發性何杰金氏病之病患在TPA給藥後胸廓腫塊具部份緩解,在最後一批在第1天至第5天,第8天至第12天以0.188mg/m2 之劑量來治療之病患中,三分之二經歷了層級III之非血液劑量限制毒性(grade III non-hematologic dose limiting toxicities, DLT),建立出第1天至第5天,第8天至第12天之單位藥劑0.125mg/m2 /天之最大耐受TPA劑量,完備了TPA劑量累增(TPA dose escalation)。 在AML及其他血液惡性腫瘤的病例中,給予病患1mg/週x3週(第1天、第8天、第15天)之TPA起始劑量給藥與6小時之持續間歇性脈搏血氧飽和度分析(continuous/intermittent pulse oximetry)。在起始TPA治療之前24小時開始,每6小時給予病患10mg地塞米松及每8小時給予病患1500mg三水揚酸膽鹼鎂(choline magnesium trisalicylate,CMT)持續到TPA給藥完後24小時為止。在TPA起始劑量施用後,病患具有兩週休息時間,接著可再次評估。自起初TPA劑量具有藥效或穩定效果之病患係根據下列方針持續治療28天6循環。 經過兩個星期的休息時間,患者在給藥TPA前30分鐘先預藥(pre-medicated with)泰諾(Tylenol)650mg及苯海拉明(Benadryl)25至50mg(依據病患體型及年齡)。接著在經過2週休息時間後持續兩週每週五天透過中心靜脈導管給予TPA之靜脈輸液。TPA係以大於1小時依一般生 理食鹽水200ml中1mg之劑量來給藥。在起初TPA給藥24小時之前,每6小時給予病患10mg地塞米松及每8小時給予病患1500mg三水揚酸膽鹼鎂直到TPA給藥完後24小時為止。 在輸液之前及之後使用測量有機溶劑萃取差異活性之生物測定來測量血液中TPA之水平。以5ml的乙酸乙酯萃取1ml的血液兩次,重新溶解萃取殘劑於50μL的乙醇且添加等量之HL60細胞。在48小時後,測量貼壁細胞(adherent cells)。 對輸液TPA之前和之後所取得之血液樣本進行測試以檢定白血細胞、血小板、及嗜中性白血球之水平。樣本係另外對於髓性母細胞(myeloblasts)及棒狀體(Auer rods)之存在作分析。此些及持續性的實驗將會進一步闡明TPA引發之針對AML及其他癌症及腫瘤症狀中之癌細胞之治療性細胞毒性及其他效果。 實例IV 測量ERK活化之調節 在患有白血病之病患之循環惡性腫瘤細胞中以及淋巴/固體腫瘤病患之周邊血液單核球(peripheral blood mononuclear cells)中測量磷酸化ERK水平。血液樣本係從根據範例III之方針來治療之病患身上在給藥TPA之前及之後取得。 在每μL具有WBC≧1000之白血病患者上,在血液樣本上使用細胞表面特定抗原以及直接與螢光物質(BD Biosciences, San Jose, CA)結合之磷酸化ERK特定抗體來進行流式細胞儀分析。根據實例III之方針一開始治療時在第1天、第2天及第11天以及接續循環中在第1天及第11天於給藥TPA之前以及TPA輸液1小時後取得樣本。根據實例III之方針於首次循環之第1天、第8天及第15天在輸液前以及輸液1小時及4小時後在每μ L具有絕對自血病芽細胞數目≧2500之白血病患者及其他非白血病患者身上取得週邊血液(peripheral blood)樣本。樣本係亦利用西方墨點分析法對磷酸化ERK及總ERK 1/2水平來分析,以檢驗由流式細胞儀所得到之結果以及將其連結至臨床反應。 前述分析將進一步闡明TPA在治療癌症和腫瘤疾病上的角色,包括TPA對於惡性腫瘤細胞的細胞毒性影響,例如初代AML細胞、以及磷酸化-ERK促生存訊號由於TPA之相關降低情形。 實例V 測量NF-κB之調節 在先前之研究中發現患者身上之NF-κB之活性可在給藥TPA與地塞米松後受到調節。此外,地塞米松被發現會誘導MKP-1 ERK磷酸酶表現並加強TPA之細胞毒性。設計下列研究以進一步闡明患者身上之NF-Kb活性如何在經TPA加地塞米松治療下受到治療性調節。 NF-κB結合度(NF-κB binding)係以根據實例III經TPA治療之患者於基準線(baseline)以及注射之前與之後之週邊血液樣本使用抗體抗原專一性之酵素免疫方法(BD Bioscience, San Jose, USA)來測量。NF-κB水平係利用96孔細胞盤形式(96-well format)使用化學發光強度來量化以於限量之細胞萃取物中進行偵測。此外,執行電泳遷移率變動分析來測量在每μ L具有絕對自血病芽細胞數目≧2500之白血病患者及具有正常白血球細胞數量之其他非自血病患者之週邊血液樣本中之NF-κB結合度。 所述研究將進一步顯示TPA為NF-κB之誘導子;然而此些實施例表現AML細胞NF-κB可在地塞米松與三水揚酸膽鹼鎂之治療下受到抑制。 實例VI 治療患有中風之個體 患者N.C.,男性,68歲,在經TPA治療18個月前中風。在TPA治療開始時,此患者無法在沒有拐杖下走路、左手及左腳行動困難且相當疲憊及虛弱。此患者每隔天接受包含0.19mg之TPA(0.125mg/m2 )之1安瓿注射連4週,接著每隔天接受0.24mg之TPA(1.25 x 0.125mg/m2 )連2週,且再接著每隔天接受0.26mg之TPA(1.5 x 0.125mg/m2 )另外連續3週。此患者已完全恢復。 患者M.C.,男性,65歲,在開始TPA治療數年前中風。此患者每週接受0.19mg TPA(0.125mg/m2 )之注射3至4次連十週共計35次注射。此患者已恢復臉的活動度且右側之活動力已有80%的改善。 實例VII 以TPA治療栓塞性中風模式動物(Embolic Stroke Model) 使用各具有體重280至350克之雄性Sprague-Dawley大鼠(Charles River,日本)。栓塞性中風依工藤等人之方法(Kudo, et al.)(1982)之改良來誘發。要用來採集血液之大鼠在自發性呼吸下以1.0%鹵烷(FluorothaneTM ;武田,大阪,日本)來麻醉。24號(gauge) SurfloTM (Terumo Medical Products, Elkton, MD)固定於股動脈上,且0.1mL之動脈血係以用來注射之1-mL注射器(Terumo Medical Products, Elkton, MD)來採集。注射器內之動脈血係於30℃下靜置培養2天以形成血塊。在此之後,加入0.1mL生理食鹽水至注射用的注射器,並通過26號注射針(Terumo Medical Products, Elkton, MD)兩次,使血塊被壓碎。 被誘導腦栓塞性中風之大鼠在自發呼吸下以1.0%鹵烷來麻醉。大鼠頸部經正中線切開(midline incision),而頸外動脈(external carotid artery)、甲狀腺上動脈(superior thyroid artery)、枕動脈(occipital artery)和翼腭動脈(pterygopalatine artery)用雙極電燒器(bipolar coagulator)(T-45;Keisei Medical lndustrial Co. Ltd,東京,日本)燒灼。腦栓塞係藉由注射0.1mL的粉碎血塊進入頸內動脈(internal carotid)而誘發。 評估腦栓塞之形成係使用雷射杜卜勒微流儀(l aser Doppler flowmetry)(FloCl;Omegawave, Tokyo, Japan)來執行。大腦供血降低至30%或更少之程度係作為栓塞形成之陽性證據。在血塊注入之後監測大腦供血30分鐘,而監測出供血剩餘原先注射血塊之前之50%或更少。在這之後,用於施予藥物之導管(PE50)係固定於頸動脈並喚醒此動物。 成功形成腦栓塞之大鼠係分成4組。每隔天給予第1組大鼠食鹽水注射。每隔天給予第2組至第4組大鼠0.125mg/m2 之TPA注射連4週。接著犧牲第2組。每隔天給予第3組至第4組進一步之0.156 mg/m2 之TPA注射連2週,且接著犧牲第3組。每隔天給予第4組0.18775mg/m2 之TPA連3週再犧牲。 在動物犧牲後切除大腦並使用組織切片機(M cIwain tissue chopper) (Mickle Laboratory Engineering, U.K.)以1mm之間隔切成十片,並在37℃時浸於2% TTC(2,3,5-三苯基四唑氯化物(2,3,5-triphenyltetrazolium chloride);Tokyo Kasei)中20分鐘來染色。TTC染色切片之影像係使用數位相機(HC-2500;Fuji PhotoFilm)及Phatograb-2500 (Fuj i Photo Film)上傳至電腦,而腦梗死體積係使用Mac Scope (Mitani, Japan)來計算。依平均值±標準差給定腦梗死體積。至於腦梗死體積之結果之統計檢驗,評估係藉由對控制組及各TPA給藥組與控制組比較而執行丹內特測驗法(Dunnett's Test)且接著藉由對TPA給藥組執行T-檢驗來完成。 每天觀察神經症狀直到犧牲為止,且大鼠係根據三種測試來評估:(1)輕輕地由尾巴抓著大鼠,由地板懸空1公尺,再觀察前肢屈曲;(2)安置大鼠於可用爪子牢牢地抓住之軟性塑膠塗層紙上。在用手抓著尾巴時,施加輕微的側向壓力於大鼠之肩膀後直到前肢下降幾英吋;(3)允許大鼠自由地移動並觀察是否有盤旋行為。根據藉著Bederson等人(Bederson etal.)(1986)所發展之量表來進行神經症狀評分如下:0:未觀察到缺損;1:前肢屈曲;2:對橫推會降低抗力但不會盤旋;3:與第2級相同行為但會盤旋。 對控制組及各TPA給藥組與控制組比較使用Steel's test且接著藉由對TPA給藥組執行威爾卡森檢定(Wilcoxon test)來評估神經症狀。在任何檢定測試中,數值p<0.05係定義為具有統計顯著性。 實例VIII TPA在治療永久性中度大腦動脈阻塞模式動物(A Permanent Middle Cerebral Artery Occlusion Model)中風上使用之效益 在本研究中使用雄性威斯達大鼠(Wistar rats)(250至320g)。動物係以異氟醚(Isoflurane)(3%引導,1-2%維持)麻醉。麻醉度係藉著捏腳趾來監控。在此研究期間麻醉技術會用於各程序。以酒精和手術消毒液修剪和清潔手術部位,動物係安置在暖水加熱墊上以維持體溫。在脖子的 頸動脈上切出正中旁切口。組織被直接切下來以顯示頸動脈及分叉處(bifurcation)。縫合處設於近端部或總頸動脈及外頸動脈。縫合處結紮。在遠離結紮之總動脈切出一個切口,預先準備的絲線(4.0單股縫線或類似材料)設於頸動脈並置於內頸動脈中。絲線深入越過頸動脈分叉約20mm直到卡到中大腦動脈而感覺到些許阻力為止。必須很小心以在插入絲線時不會使動脈破裂。絲線於定位結紮而縫合皮膚切口。成功阻塞後使用柏德森量表(Bederson scale)(參見Bederson et al., (1986) Stroke, 17:1304-1308.)來評估醒著時的動物。每15分鐘量測體溫以維持正常體溫。經過中大腦動脈阻塞程序之動物在手術後幾小時可能會有體溫異常。動物依其體溫而決定設置於冷卻箱或加熱箱中。體溫維持於37.5℃。在中大腦動脈阻塞後監測動物6小時且接著安置於籠子中過夜。 將大鼠分為四組。每隔天給予第一組大鼠生理食鹽水注射。第2組至第4組每隔天給予0.125mg/m2 之TPA注射連4週。接著犧牲第2組。進一步每隔天給予第3組至第4組0.156mg/m2 之TPA連2週,然後犧牲第3組。每隔天給予第4組0.18775mg/m2 之TPA連3週然後犧牲。 動物被犧牲後切除其大腦且依1mm間隔使用組織切片機(Mickle Laboratory Engineering, U.K.)切片為10片,並在37℃時浸於2% TTC(2,3,5-三苯基四唑氯化物(2,3,5-triphenyltetrazolium chloride);Tokyo Kasei)中20分鐘來染色。TTC染色切片之影像係使用數位相機(HC-2500;Fuji PhotoFilm)及Phatograb-2500(Fuji Photo Film)上傳至電腦。拍下大腦切片並分析梗死面積(infarct size)、梗死體積(infarct volume)、半影(penumbra)、腫大(edema)。 每天觀察神經症狀直到犧牲為止。每天觀察神經症狀直到犧牲為止,且大鼠係根據三種測試來評估:(1)輕輕地由尾巴抓著大鼠,由地板懸空1公尺,再觀察前肢屈曲;(2)安置大鼠於可用爪子牢牢地抓住之大片軟性塑膠塗層紙上。在用手抓著尾巴時,施加輕微的側向壓力於大鼠之肩膀後直到前肢下降幾英吋;(3)允許大鼠自由地移動並觀察是否有盤旋行為。根據藉著Bederson等人(Bederson et al.)(1986)所發展之量表來進行神經症狀評分如下:0:未觀察到缺損;1:前肢屈曲;2:對橫推會降低抗力但不會 盤旋;3:與第2級相同行為但會盤旋。 對控制組及各TPA給藥組與控制組比較使用Steel's test且接著藉由對TPA給藥組執行威爾卡森檢定來評估神經症狀。在任何檢定測試中,數值p<0.05係定義為具有統計顯著性。 實例IX TPA在治療使用暫時性大腦中度動脈組塞模型之中風之效益 在本研究中使用雄性C57B16小鼠(25至30克)。小鼠係以異氟醚(3%引導,1-2%維持)麻醉。以酒精和手術消毒液修剪和清潔手術部位。在頸動脈上切出頸部正中切口,且動脈係自其分支分離。單股絲線係引入內頸動脈中並深入直到在中太腦動脈交會(lodge)為止。絲線於定位結紮而縫合切口。在阻塞後2小時將小鼠重新麻醉且絲線將自MCA移除。在手術期間及手術後藉由使用加熱墊維持體溫。動物在中大腦動脈阻塞後被監測4小時。 將小鼠分為四組。每隔天給予第一組小鼠生理食鹽水注射。第2組至第4組每隔天給予0.125mg/m2 之TPA注射連4週。接著犧牲第2組。進一步每隔天給予第3組至第4組0.156mg/m2 之TPA連2週,然後犧牲第3組。每隔天給予第4組0.18775mg/m2 之TPA連3週然後犧牲。 動物被犧牲後切除其大腦且依1mm間隔使用組織切片機(Mickle Laboratory Engineering, U.K.)切片為10片,並在37℃時浸於2% TTC(2,3,5-三苯基四唑氯化物(2,3,5-triphenyltetrazolium chloride);Tokyo Kasei)中20分鐘來染色。TTC染色切片之影像係使用數位相機(HC-2500;Fuji PhotoFilm)及Phatograb-2500(Fuji Photo Film)上傳至電腦。拍下大腦切片並分析梗死面積、梗死體積、半影、腫大。 每天觀察神經症狀直到犧牲為止,且大鼠係根據三種測試來評估:(1)輕輕地由尾巴抓著大鼠,由地板懸空1公尺,再觀察前肢屈曲。(2)安置大鼠於可用爪子牢牢地抓住之大片軟性塑膠塗層紙上。在用手抓著尾巴時,施加輕微的側向壓力於大鼠之肩膀後直到前肢下降幾英吋。(3)允許大鼠自由地移動並觀察是否有盤旋行為。根據藉著Bederson等人(1986)所 發展之量表來進行神經症狀評分如下:0:未觀察到缺損;1:前肢屈曲;2:對橫推會降低抗力但不會盤旋;3:與第2級相同行為但會盤旋。 對控制組及各TPA給藥組與控制組比較使用Steel's test且接著藉由對TPA給藥組執行威爾卡森檢定來評估神經症狀。在任何檢定測試中,數值p<0.05係定義為具有統計顯著性。 實例X 使用TPA治療中風的臨床療效 1個月內經歷中風之介於30-72歲年紀之男性和女性受招募而參與TPA之10週試驗。 受招募之個人簽署了知情同意書,並使用電腦斷層掃描(computed tomography, CT)、物理及神經測試、神經系統檢查、鎮靜程度、美國國衛院腦中風評估表(National Institute of Health Stroke Survey, NIHSS)、12導程心電圖、心電圖遙測、血氧測量、生命徵象、體重,病人背景、妊娠測試、測量尿液藥物、血液測試、凝血指數、常規臨床試驗、尿檢。臨床實驗室測試包含綜合代謝指數(鈉、鉀、氯、二氧化碳(CO2 )、谷胺酸(Gl u)、尿素氮(BUN)、鉻、鈣、總蛋白質(TP)、血蛋白(ALB)、總膽紅素(TBILI)、鹼性磷酸酶(AP)、天門冬酸胺基轉化酶(AST)、丙胺酸基轉化酶(ALT))、血液常規檢查(Hematology CBC)(血紅蛋白測定(Hgb)、血容比(Hct)、紅血球數(RBC)、自血球數(WBC)、血小板計數(Plt)、白血球分類計數(Diff))、以及所有女性之血清中人類絨毛膜促性腺激素(Serum hCG)。 每隔天給予個體0.125mg/m2 之TPA或安慰劑連4週,接著在第5週及第6週每隔天給予1.25 x0.125mg/m2 或安慰劑,而第7週至第9週則每隔天給予1.5 x 0.125mg/m2 或安慰劑。在給予TPA或安慰劑後2小時監測個體。 在第5週和第10週時,使用NIHSS(美國國衛院腦中風評估表(NIH Stroke Scale))、巴氏量表(Barthel ADL index)(Granger, 1979)、以及修改過之雷氏量表(modified Rankin scale)(Farrell, 1991)來評估對象。 藉由量測比較經TPA治療之個體與經安慰劑治療之個體而依美國國衛院腦中風評估表基準之變化來評定效益。次級效益變數 (Secondary efficacy variables)為巴氏量表及修改之雷氏量表。在試驗中收集及評估安全性測量,特別是測量由診療基準至第5週之變化。這些測量包含副作用報告(Adverse events reports)、體檢、生命徵象、體重測量、心電圖(ECGs)、臨床實驗測試結果、及生命徵象以及自殺行為及/或意念之評分。副作用為在對象接收研究用藥物時任何不好之醫療反應情形,無論其是否與研究用藥物有因果關係。因此副作用可能為暫時與研究藥物相關之任何不想要或非預期之跡象(包含例如異常實驗結果)、症狀、或疾病,而無論其是否視為與研究用藥物有關係。 若完成所有診療則視同對象完成此研究。然而若對象無法滿足納入/排除標準;苦於副作用、對治療反應不充足、撤回其同意、違反了協議、未現身或死亡則可能會從研究中退出。 參考文獻 Altuwaijri S, Lin H K, Chuang K H, Lin W J, Yeh S, Hanchett L A, Rahman M M, Kang H Y, Tsai M Y, Zhang Y, Yang L, and Chang C. Interruption of nuclear factor kappaB signaling by the androgen receptor facilitates 12-O-tetradecanoylphorbolacetate-induced apoptosis in androgen-sensitive prostate cancer LNCaP cells. Cancer Res 2003; 63: 7106-12. Ando I., Crawfor D. H. et al. Phorbol ester-induced expression and function of the interleukin 2 receptor in human B lymphocytes. Eur J Immunol. 15(4), 341-4 (1985). Aye M. T., Dunne J.V. Opposing effects of 12-O-tetradecanoylphorbol 13-acetate on human myeloid and lymphoid cell proliferation. J Cell Physiol. 114(2), 209-14 (1983). Bederson JB, Pitts LH, Tsuji M, Nishimura MC, Davis RL, Bartkowski H. Rat middle cerebral artery occlusion: evaluation of the model and development of a neurologic examination. Stroke. 1986; 17: 472-476. Boutwell R.K. Biochemical mechanism of tumor promotion, in mechanisms of tumor promotion and co-carcinogenesis. Eds. Slaga, T.J., Sivak, A. J. and Boutwell, R.K. Raven, New York, 49-58 (1978). Boutwell R.K. The function and mechanism of promoters of carcinogenesis. CRC Crit. Rev.Toxicol 2, 419-443 (1974). Brose N, Rosenmund C. Move over protein kinase C, you 've got company: alternative effectors of diacylglycerol and phorbol esters. JCell Sci; 115: 43 99-411 (2002). Cancer Chemother Pharmacol. Jun;57(6):789-95 (2006). Cui X X, Chang R L, Zheng X, Woodward D, Strair R, and Conney A H. A sensitive bioassay for measurmg blood levels of 12-O-tetradecanoylphorbol-13-acetate (TPA) in patients: preliminary pharmacokinetic studies. Oncol Res 2002; 13: 169-74. Deegan M. J., Maeda k. Differentiation of chronic lymphocytic leukemia cells after in vitro treatment with Epstein-Barr virus or phorbol ester. Immunologic and morphologic studies. Am J Hermatol. 17(4), 335-47 (1984). Falcioni F., Rautmann A. et al. Influence of TPA (12-0-tetradodecanoyl-phorbol-13-acetate) on human B lymphocte function. Clin Exp Immunol. 62(3), 163-2 (1985). Farrell B, Godwin J, Richards S, Warlow C, et al. (1991). The United Kingdom transient ischaemic attack (UK-TIA) aspirin trial: final results. J Neurol Neurosurg Psychiatry) 54 (12): 1044-1054. Forbes 1. J., Zalewski P. D.,Letarte M. Human B-lymphocyte maturation sequence revealed by TPA-induced differentiation of leukaemi cells. Immunobiology 163(1), 1-6 (1 982). Gunjan Goel, Harinder P. S. Makkar, George Francis, and Klaus Becker. Phorbol Esters: Structure, Biological Activity, and Toxicity in Animals. International Journal of Toxicology, 26:279-288, 2007. Gogusev J., Barbey S., Nezelof C. Regulation of TNF-alpha and IL-1 gene expression during TPA-induced differentiation of "Malignant histiocyosis" DEL cell line t(5:6) (q35:P21). Anticancer Res. 16(1), 455-60(1996). Granger CV, Devis LS, Peters MC, Sherwood CC, Barrett JE. Stroke rehabilitation: analysis of repeated Barthel lndex measures. Arch Phys Med Rehabil. 1979;60:14-17. Han Z. T., Tong Y. K., He L. M., Zhang玄,Sun J. Z., Wang T. Y., Zhang H., Cui Y. L., Newmark H. L., Conney A.H., Chang R. L. 12-O-Tetradecanoyl-phorbol-13-acetate (TPA) -induced increase in depressed white blood cell counts in patients treated with cytotoxic cancer chemotherapeutic drugs. Proc. Natl. Acad. Sci. 95, 5363-5365 (1998). Han Z.T., Zhu X. X., Yang R. Y., Sun J. Z., Tian G. F., Liu X. J., Cao G. S., NewMark H. L., Conney A. H., and Chang R. L. Effect of intravenous infusion of 12-O-tetradecanoyl-phorbol-13 -acetate (TPA) in patients with myelocytic leukemia: preliminary studies on therapeutic efficacy and toxicity. Pro. Natl. Acad. Sci. 95, 5357-5361 (1 998). Harada S. et al.: Tumor Promoter, TPA, Enhances Replication of HTLV-III/LAV. Virology 154, 249-258 (1986). Hecker E. In handbuch der allgemeinen patholgie, ed. Grundmann, E. (Springer-Verlag, Berlin-Heideiberg, Vol. IV 16, 651-676 (1975). Hecker E. Structure -activity relationships in deterpene esters irritant and co-carcinogenic to mouse skin, in mechanisms of tumor promotion and co-carcinogenesis. Eds. Slaga, T. J., Sevak, A. j. and Boutwell, R.K. Raven, New York, 11-49 (1978). Hofmann J. The potential for isoenzyme-selective modulation of protein kinase C. FASEB J. 11, 649-669 (1997). Huberman E., Callaham M. F. Induction of tenninal differentiation in human promyelocytic leukemia cells by tumor-promoting agents. Proc. Natl. Acad. Sci. 76, 1293-1297 (1979). Hunter T. Signaling 2000 and beyond. Cell 100, 113-117 (2000). Kassel O, Sancono A, Kratzschmar J, Kreft B, Stassen M, and Cato A C. Glucocorticoids inhibit MAP kinase via increased expression and decreased degradation of MKP-1. Embo J 2001; 20: 7108-16. Kazanietz M.G. Eyes Wide Shut: protein kinase. C isoenzymes are not the only receptors for the phorbol ester tumor promoters. Mol. Carcinog. 28, 5-12 (2000). Keoffler H. P., Bar-Eli M., Territo M. C. Phorbol ester effect on differentiation of human myeloid leukemia cells lines blocked at different stages of maturation. Cancer Res. 41, 919-926 (1981). Kim S C, Hahn J S, Min Y H, Yoo N C, Ko Y W, and Lee W J. Constitutive aetivation of extracellular signal-regulated kinase in human acute leukemias: combined role of activation of MEK, hyperexpression of extracellular signal-regulated kinase,. and downregulation of a phosphatase, PAC 1. Blood 1999; 93: 3893-9. Kiyoi H, Naoe T, Nakano Y, Yokota S, Minami S, Miyawaki S, Asou N, Kuriyama K, Jinnai I, Shimazaki C, Akiyama H, Saito K, Oh H, Motoji T, Omoto E, Saito H, Ohno R, and Ueda R. Prognostic implication of FLT3 and N-RAS gene mutations in acute myeloid leukemia. Blood 1999; 93: 3074-80. Kobayashi M., Okada N. et al. Intracellular interleukin-1 alpha production in human gingival fibroblasts is differentially regulated by various cytokines. J Dent Res. 78(4), 840-9 (1 999). Kudo M., Aoyama A., Ichimori S. and Fukunaga N. An animal model of cerebral infarction: homologous blood clot emboli in rats. Stroke 13: 505-508 (1982) Lebien T. W., Bollum F. J. et al. Phorbol ester-induced differentiation of a non-T, non-B leudemic cell line: model for human lymphoid progenitor cell development. J Immunol. 128(3), 1316-20 (1982). MD Iqbal Hossain Chowdhury et al. The Phorbol Ester TPA Strongly Inhibits HIV-1 Induced Syncytia Formation but Enhances Virus Production: possible involvement of protein kinase C pathway. Virology 176, 126-132, (1990). Meinhardt G., Roth J., Hass R. Activation of protein kinase C relays distinct signaling pathways in the same cell type: differentiation and caspase-mediated apoptosis. Cell Death Differ. 7, 795-803 (2000). Milella M, Kornblau S M, Estrov Z, Carter B Z, Lapillonne H, Harris D, Konopleva M, Zhao S, Estey E, and Andreeff M. Therapeutic targeting of the MEK/MAPK signal transduction module in acute myeloid leukemia. J Clin Invest 2001; 108: 851-9. Mochty-Rosen D., Kauvar L. M. Modulating protein kinase C signal transduction. Adv. Pharmacol. 44, 91-145 (1998). Morgan M A, Dolp O, and Reuter C W. Cell-cycle-dependent activation of mitogen-activated protein kinase kinase (MEK- 1/2) in myeloid leukemia cell lines and induction of growth inhibition and apoptosis by inhibitors of RAS signaling. Blood 2001; 97: 1823-34. Nagasawa K., Chechgik B. E. et al. Modulation of human T-cell differentiation markers by 12-O-tetradecanoylphorbal-13-acetate. Thymus. 3(4-5), 307-18, (1981). Nakao Y., Matsuda S. et al. Paradoxical anti-leukemic effects of plant-derived tumor promoters on a human thymic lymphoblast cell line. Int J Cancer 30(6), 687-95 (1982). Nakao Y., Matsuda S. et al. Phorbol ester-induced differentiation of human T-lymphoblastic cellline HPB-ALL. Cancer Res. 42(9), 33843-50 (1982). Newton A.C. Protein kinase C: structure, function and regulation. J. Biol. Chem. 270, 28495-28499 (1995). Palombella V J, Rando O J, Goldberg A L, and Maniatis T. The ubiquitin-proteasome pathway is required for processing the NF-kappa B1 precursor protein and the activation of NF-kappa B. Cell 1994; 78: 773-85. Platanias L C. Map Map kinase signaling pathways and hematologic malignancies. Blood 2003; 101: 4667-79. Rovera G., Santoli D., Damsky C. Human promyelocytic cells in culture differentiate into macrophage-like cells treated with a phorbol diester. Pro. Natl. Acad. Sci. 7, 2779-2783 (1979). YIP, Y.K. et al. Stimulation of human gamma interferon production by diterpene esters. Infection and Immunity 34(1) 131-139 (1981). Zhao J., Sharma Y., Agarwal R. Significant inhibition by the flavonoid antioxidant silymarin against 12-O-tetradecanoylphorbol 13 -acetate-caused modulation of antioxidant and inflammatory enzymes and cyclooxygenase2 and interleukin-I alpha expression in SENCAR mouse epidermis: implications in the prevention of stage 1 tumor promotion. Mol Carciriog. 26(4), 321-33 (1999).A novel method and composition has proven useful in the prevention and/or treatment of stroke and stroke sequelae in mammalian subjects including humans. In various embodiments, the compositions and methods described herein may increase the release of Th1 cytokines, increase the phosphorylation of ERK, regulate the activity of NF-κB, prevent or treat paralysis, increase the sense of space, Reduce memory loss, reduce aphasia, increase coordination and balance, improve cognition, improve sense of direction, reduce the incidence of subsequent strokes, and reduce impulsivity. The formulations and methods provided herein use formula I crotonate or a derivative compound of formula I crotonate, as described more fully in US Patent Application No. 12/023,753 filed on January 31, 2008, which is Claiming the US Provisional Patent Application No. 60/898,810 filed on January 31, 2007, and the priority benefits of the US Provisional Patent Application filed, the entire contents of which are incorporated herein by reference,
Figure 02_image001
Formula I where R 1 And R 2 Can be hydrogen; hydroxyl;
Figure 02_image004
, Where the alkyl group contains 1 to 15 carbon atoms;
Figure 02_image006
, Where the lower alkenyl group contains between 1 and 7 carbon atoms;
Figure 02_image008
;
Figure 02_image010
;
Figure 02_image012
;
Figure 02_image014
And their substituted derivatives. R 3 Can be hydrogen or
Figure 02_image016
. In some embodiments, R as a novel composition for treating chronic or recurrent symptoms 1 And R 2 At least one of them is not hydrogen, and R 3 Is hydrogen or
Figure 02_image016
And their substituted derivatives. In another embodiment, R 1 Or R 2 One is
Figure 02_image019
. And R 1 Or R 2 The other is
Figure 02_image006
. Where lower alkenyl is 1 to 7 carbon atoms, and R 3 For hydrogen. The alkyl, alkenyl, phenyl, and benzyl groups in this chemical formula may be unsubstituted or substituted with halogen, preferably chlorine, fluorine, or bromine; may be unsubstituted or substituted with nitro; Amino group substitution or substitution with an amine group; and/or may be unsubstituted or substituted with similar free radicals. The stroke treatment formulations and methods provided herein use crotonate of formula I or derivative compounds of crotonate of formula I, as described above, including all active pharmaceutically acceptable compounds and various possible variations in this specification, and easily Provide complexes, salts, solvates, isomers, enantiomers, polymorphs, and prodrugs of these compounds and combinations thereof as anti-stroke agents. The preparations and methods for increasing Th1 cytokines provided herein use the formula I crotonate or a derivative compound of formula I crotonate, as described above, including all active pharmaceutically acceptable compounds in this specification and various possible changes, and easily Provide their complexes, salts, solvates, isomers, enantiomers, polymorphs, and prodrugs of these compounds and their combinations as novel Th1 cytokine enhancers, these compounds and their compositions as a Novel Th1 cytokine increasing agent. A wide range of mammalian subjects, including human subjects, are suitable for treatment using the formulations and methods of the present invention. These subjects include but are not limited to individuals suffering from stroke or at risk of stroke. The ERK phosphorylation increasing preparations and methods provided herein use formula I crotonate or a derivative compound of formula I crotonate, as described above, including all active pharmaceutically acceptable compounds in this specification and various possible changes, and easily The complexes, salts, solvates, isomers, enantiomers, polymorphs, and prodrugs of these compounds and combinations thereof are provided as novel ERK phosphorylation increasing agents. A wide range of mammalian subjects, including human subjects, are suitable for treatment using the formulations and methods of the present invention. These subjects include but are not limited to individuals suffering from stroke or at risk of stroke. In the methods and compositions of the present invention, one or more of the crotonate compounds of formula I or derivative compounds of crotonate of formula I described herein are effectively formulated or administered as effective for the treatment and prevention of stroke or A medicine for the sequelae of stroke. In the exemplary embodiment, TPA is demonstrated for the purpose of illustration in pharmaceutical preparations and treatment methods alone or in combination with one or more auxiliary therapeutic agents as effective agents. The disclosure also provides pharmaceutically acceptable crotonate compounds in the form of natural or synthetic compounds, including the complexes, derivatives, salts, solvates, isomers, and enantiomers described herein , Polymorphs and prodrugs of these compounds and combinations thereof, these as therapeutic agents in the methods and compositions of the present invention are quite effective in treating and preventing stroke and sequelae of stroke. Stroke is caused by the interruption of blood supply to the brain. This may be caused by occlusion of blood vessels (ischaemic stroke) or rupture of blood vessels (haemorrhagic stroke). Symptoms of stroke include sudden numbness or weakness, especially one side of the body; sudden unconsciousness, or difficulty speaking or understanding words; difficulty viewing with one or both eyes; sudden difficulty walking, dizziness, or loss of balance or coordination; or sudden absence Severe headache caused by. Risk factors for stroke include high blood pressure, dyslipidemia, tobacco use, lack of exercise, obesity, stress, diabetes, alcohol consumption, excessive homocysteine (homocystein) in the blood, inflammation and abnormal coagulation. There are also risk factors that cannot be changed, such as age, genetics, gender, and race. There are three different stages in the treatment of stroke: prevention, immediate treatment after stroke, and rehabilitation after stroke. The compositions and methods described herein can be used at any stage of stroke treatment, and can be used alone or in combination with one or more additional treatment methods, including other agents, devices, or surgical treatments. Croton alcohol is a natural plant-based polycyclic alcohol of diterpenes (tigliane family). It was first isolated from the seeds of Croton tiglium as a hydrolysate of croton oil in 1934. Croton alcohol is soluble in most polar organic solvents and water. Crotyl alcohol esters have the following overall structure of formula I:
Figure 02_image001
Formula I where R 1 And R 2 Selected from hydrogen; hydroxyl;
Figure 02_image004
, Where the alkyl group contains 1 to 15 carbon atoms;
Figure 02_image006
, Where the lower alkenyl group contains between 1 and 7 carbon atoms;
Figure 02_image008
;
Figure 02_image010
;
Figure 02_image012
;
Figure 02_image014
; And the group of substituted derivatives, and R 3 Can be hydrogen or
Figure 02_image016
Or substituted derivatives thereof, and pharmaceutically acceptable salts, enantiomers, polymorphs, prodrugs, solvates, and hydrates of the compounds of formula I and substituted derivatives thereof. The term "lower alkyl" or "lower alkenyl" used herein means a group containing 1 to 7 carbon atoms. In the compound of formula I, the alkyl or alkenyl group may be linear or branched. In some embodiments, where R 1 Or R 2 One or R 1 And R 2 All are a long-chain carbon group (that is, formula I is capric acid or myristic acid). The alkyl, alkenyl, phenyl and benzyl groups of the chemical formula herein may be unsubstituted or substituted with halogen, preferably chlorine, fluorine or bromine; may be unsubstituted or substituted with nitro; may be unsubstituted Amino group substitution or substitution by amine group; and may not be substituted by similar type of free radical or substituted by similar type of free radical. Including organic or synthetic forms of crotonate from any herbal source like croton tiglium formulations or extracts used in the examples herein is intended to include crotonate (or crotonate analogs, related compounds and/or Or derivatives). Effectively used crotonate and/or related compounds in the examples herein will generally have the structure shown in Formula I, but those of ordinary skill in the art will understand that these compounds have functionally equivalent analogs, complexes Compounds, conjugates, and derivatives are also within the scope of the present invention. In a more detailed embodiment, the illustrative structure modified according to the above illustrative formula I will be selected to provide an effective selective compound to treat and/or prevent strokes and stroke-induced injuries in mammalian subjects including humans And/or manage the consequences or sequelae of stroke, including: R 1 And R 2 At least one of them is not hydrogen, and R 3 Selected from hydrogen or
Figure 02_image016
And its substituted derivatives. In another embodiment, R 1 Or R 2 One of them is
Figure 02_image038
While R 1 Or R 2 The other is
Figure 02_image006
, And R 3 For hydrogen. Exemplary embodiments of the crotonate compound of Formula I used in this treatment method in the treatment or prevention of stroke, injury from stroke, and/or management of stroke aftermath in mammalian subjects including humans are shown in the following formula Crotyl alcohol 12-myristate-13-acetate shown in II (also known as PMA or 12-O-tetradecanoyl-crotonol-13-acetate, TPA).
Figure 02_image022
Formula II Crotonate and other related compounds and derivatives useful in the formulations and methods of the present invention include, but are not limited to, active salts of this compound for other pharmaceutical applications, as well as active isomers, enantiomers, Polymorphs, glycosylated derivatives, solvates, hydrates, and/or prodrugs. The derivative of crotonate of formula I may or may not be crotonate itself. Exemplary forms of crotonate used in the compositions and methods of the invention include, but are not limited to, crotonol 13-butyrate; crotonol 12-decanoic acid; crotonol 13-decanoic acid; crotonol 12,13-di Acetate; crotyl alcohol 13,20-diacetate; crotyl alcohol 12,13-dibenzoic acid; crotyl alcohol 12,13-dibutyl ester; crotyl alcohol 12,13-didecanoate; crotyl alcohol 12, 13-dihexanoate; crotyl alcohol 12,13-dipropionate; crotyl alcohol 12-myristate; crotyl alcohol 13-myristate; crotyl alcohol 12,13,20-triacetate; 12 -Deoxycrotonol 13-angelica; 12-deoxycrotonol 13-angelica 20-acetate; 12-deoxycrotonol 13-isobutyrate; 12-deoxycrotonol 13-isobutyrate Ester-20-acetate; 12-deoxycrotonol 13-phenylacetate; 12-deoxycrotonol 13-phenylacetate 20-acetate; 12-deoxycrotonol 13-nutmeg Acid esters; crotyl alcohol 12-cisyl ester 13-decanoate; 12-deoxycrotonol 13-acetate; crotyl alcohol 12-acetate; and crotyl alcohol 13-acetate, as shown in Table 1 Show. Table 1 Routine crotonate
Figure 106126482-A0304-0001
The composition described herein includes an effective amount of a crotonate compound of formula I or a derivative compound of formula I crotonate including an effective amount of a stroke treatment composition for reducing or preventing stroke damage, which is useful for preventing and/or treating stroke in mammalian subjects or Stroke-related symptoms or sequelae are quite effective. Effective amounts of active compounds in "stroke treatment", "anticoagulation", "anticholesterolemic" (anticholesterolemic), "vasodilation", "antihypertension", "reducing arteriolar resistance", "increasing venous volume"","Reducing Cardiac Oxygen Demand", "Reducing Heart Rate", "Stabilizing Heart Rate", or "Protecting Nerve" are very effective treatments, given in unit or multi-unit dosage form over a specific period of intervention treatment to measure Relief of one or more symptoms or sequelae of stroke on the subject. In an exemplary embodiment, the composition of the present invention is quite effective in a method of preventing or relieving stroke symptoms or stroke sequelae in humans and other mammal subjects who are vulnerable to stroke or suffering from stroke. The crotonate therapeutic composition of the present invention generally contains an effective amount or unit dose of a crotonate compound of formula I or a crotonate-derived compound of formula I, which can enhance stability, transferability, absorbability, half-life, drug efficacy, drugs Kinetics and/or pharmacodynamics, reducing undesirable side effects, or providing one or more other advantages in pharmaceutical applications , Buffer, and/or other additives. Those with ordinary knowledge in the technical field will be able to easily determine the effective amount of the crotonate compound of formula I or its related derivative compounds based on clinical and patient-specific factors (eg, a unit dose containing an effective concentration/amount of TPA, or the selected TPA Pharmaceutically acceptable salts, isomers, enantiomers, solvates, polymorphs and/or prodrugs). A suitable effective amount of a unit dose of active compound for administration to mammalian subjects including humans may be from about 10 μg to about 1500 μg, about 20 μg to about 1000 μg, about 25 μg to about 750 μg, about 50 μg to about 500 μg, about 150 μg to about 500 μg, about 125 μg to about 500 μg, about 180 to about 500 μg, about 190 μg to about 500 μg, about 220 μg to about 500 μg, about 240 μg to about 500 μg, about 260 μg to about 500 μg, about 290 μg to 500 μg. In some embodiments, the therapeutically effective dose of a crotonate compound of formula I or a related or derived compound may be selected from a very small range, for example, 10 to 25 μg, 30 to 50 μg, 75 to 100 μg, 100 to 300 μg or 150 to 500 μg. These and other effective unit doses can be administered in a single dose, or in multiple doses daily, weekly, or monthly, for example, including 1 to 5 times, or 2 to 3 times daily, weekly, or monthly Dosing regimen. In an exemplary embodiment, the dosage of 10 μg to 30 μg, 30 μg to 50 μg, 50 μg to 100 μg, 100 μg to 300 μg, or 300 μg to 500 μg is administered once, twice, three times, four times, or five times per day. In a more detailed embodiment, a dose of 50 μg to 100 μg, 100 μg to 300 μg, 300 μg to 400 μg, or 400 μg-600 μg is administered once or twice a day. In a further embodiment, a dose of 50 μg to 100 μg, 100 μg to 300 μg, 300 μg to 400 μg, or 400 μg to 600 μg is administered every other day. In other embodiments, the dose is calculated based on body weight, and the amount that can be administered is, for example, about 0.5 μg/m per day 2 Up to about 300μg/m 2 , About 1μg/m 2 Up to about 200μg/m 2 、About 1μg/m per day 2 Up to 187.5μg/m 2 、About 1μg/m per day 2 To about 175μg/m per day 2 、About 1μg/m per day 2 Up to about 157μg/m 2 、About 1μg/m per day 2 About 125μg/m 2 、About 1μg/m per day 2 About 75μg/m 2 , 1μg/m per day 2 About 50μg/m 2 , 2μg/m per day 2 Up to about 50μg/m 2 , 2μg/m per day 2 Up to about 30μg/m 2 Or 3μg/m per day 2 Up to about 30μg/m 2 . In other embodiments, the administered dose may be less, for example, 0.5 μg/m every other day 2 Up to about 300μg/m 2 , About 1μg/m 2 Up to about 200μg/m 2 、About 1μg/m every other day 2 Up to about 187.5μg/m 2 、About 1μg/m every other day 2 About 175μg/m 2 、About 1μg/m per day 2 To about 157μg/m every other day 2 、About 1μg/m every other day 2 About 125μg/m 2 、About 1μg/m every other day 2 Up to about 75μg/m 2 、About 1μg/m every other day 2 Up to about 50μg/m 2 、2μg/m every other day 2 Up to about 50μg/m 2 , 2μg/m per day 2 Up to about 30μg/m 2 Or 3μg/m per day 2 Up to about 30μg/m 2 . In other embodiments, the dosage administration may be 3 times/week, 4 times/week, 5 times/week, only on weekends, only in conjunction with other treatment regimens, several days, or any based on clinical and patient-specific factors Suitable dosage regimen. Contains an effective amount (selectively targeting "stroke treatment", "anticoagulation", "anticholesterolemic"(anticholesterolemic)","vasodilation","antihypertensive","reducing arterial resistance", "increasing ``Venous volume'', ``Reduce cardiac oxygen demand'', ``Reduce heart rate'', ``Stabilize heart rate'', ``Induce ERK phosphorylation'', ``IL-2 regulation'' and/or ``Protect nerve'') The amount, timing, and mode of the composition of the present invention of an ester compound or a derivative compound of a crotonate of formula I will be based on individual criteria, based on factors such as weight, age, sex, and condition of the individual, the severity of the disease, and/or Or related symptoms, the preventive or therapeutic nature of the administration, and regular adjustments based on factors including half-life and efficiency that affect drug delivery, absorption, and drug metabolic motility. The preparation of the present invention is used for immediate disease treatment (selectively targeting "stroke treatment", "anticoagulation", "anticholesterol blood"", "vasodilation", "antihypertension", and "induced ERK phosphorylation"'', ``Reduce arterial resistance'', ``Increase venous volume'', ``Reduce cardiac oxygen demand'', ``Reduce heart rate'', ``Stabilize heart rate'', ``Reduce blood clots'', ``Protect nerves'', ``IL-2 regulation "Or "NFκB modulation"), the effective dose or multi-dose treatment plan will initially select the required minimum dose plan that is sufficient to actually prevent or alleviate the symptoms of stroke in the subject. Dosage and dosing guidelines will often include a process of repeated dosing for several days or even one or more weeks or years. An effective treatment regimen may also involve a prophylactic dose that lasts for several days, weeks, months, or even years once a day or multiple times a day. The effectiveness of the composition and method of the present invention in the treatment of stroke can be demonstrated using various model systems, including temporary moderate cerebral artery occlusion as shown in Example 9, permanent moderate cerebral artery occlusion as shown in Example 8, Endovascular linear middle cerebral artery occlusion, embolic moderate cerebral artery occlusion shown in Example 7, endothelin-1 induced arterial and venous contraction, or cerebral cortical photochemistry. Use of the crotonate composition of the present invention in a model system will show a reduction in symptoms or long-term effects, which is reduced by 0%, 20%, 30%, 50% or even 75%, 90%, 96% compared to control animals the above. The effectiveness of the compositions and methods of the present invention in the treatment of stroke can be further demonstrated in the relief of symptoms exhibited by individuals suffering from stroke. Symptoms of this category include, but are not limited to, paralysis, spatial impairment, impaired judgment, neglect, memory loss, aphasia, coordination and balance problems, nausea, vomiting, cognitive impairment, perceptual impairment, disorientation, unilateral blindness and impulsivity. Using the crotonate composition of the present invention will reduce the initial state by 0%, 20%, 30%, 50%, or even 75% to 90%, 96% or more to reduce the symptoms manifested by the individual. In another aspect of the present invention, combined disease treatment (for "stroke treatment", "anticoagulation", "anticholesterol blood"", "vasodilation", "antihypertension", "inducement ERK phosphorylation", "reducing arteriolar resistance", "increasing venous volume", "decreasing cardiac oxygen demand", "decreasing heart rate", "stabilizing heart rate" or "NFκB regulation") and coordinated administration methods, It uses an effective amount of a crotonate compound of formula I or a crotonate derivative compound of formula I and one or more secondary or auxiliary agents that will be formulated or coordinated with the crotonate compound of formula I to produce a comprehensive, multi-active disease Treatment composition or coordinated treatment method. Exemplary combination preparations and coordinated treatment methods for the prevention or treatment of strokes use a crotonate compound of formula I, or a derivative compound of crotonate of formula I, in combination with one or more protections useful for the treatment or prevention of the underlying disease, condition, and/or symptoms Nerve or other secondary or auxiliary therapeutic agents. For most of the combination preparations and coordinated treatment methods of the present invention, the crotonate compound of Formula I or its related or derivative compounds is formulated or coordinated with one or more secondary or adjunct therapeutic agents to produce an effective combination or In coordination, there are combination preparations or coordinated treatment methods for avoiding or treating stroke, or the effects of stroke. Exemplary combination preparations and coordinated treatment methods in this content use a crotonate compound of formula I or a derivative compound of crotonate of formula I to match with a tissue plasminogen activator, anticoagulant, statin Drugs, angiotensin II receptor blockers, angiotensin-converting ester inhibitors, antiplatelet drugs, fibrates, β-blockers, calcium channel blockers, or one or more secondary drugs Or adjuvant therapy. Exemplary anticoagulants include but are not limited to heparin, warfarin, heparinoids, phenindione, phenindione, atomin, and acenocoumarol , Phenprocoumon, phenprocoumon, idraparinux, fondaparinux, and thrombin inhibitors. Exemplary statins include but are not limited to lovastatin (lovastatin), amlodipine (amlodipine), atorvastatin (atorvastatin), rosuvastatin (rosuvastatin), simvastatin (simvastatin), fluvastatin ( fluvastatin), pitavastatin and pravastatin. Exemplary angiotensin II receptor blockers include but are not limited to candesartan (candesartan), eprosartan (eprosartan), irbesartan (irbesartan), losartan (losartan), olmesartan (olmesartan), telmisartan (telmisartan) and valsartan (valsartan). Angiotensin-converting enzyme inhibitors include but are not limited to enazepril, captopril, enalapril, fosinopril, isinopril ), moexipril (moexipril), perindopril (perindopril), quinapril (quinapril), ramipril (ramipril) and trandolapril (trandolapril). Exemplary β-blockers include but are not limited to alprenolol, bucindolol, carteolol, carvedilol, carvedilol, labetalol, Nadolol, oxprenolol, penbutolol, pindolol, propranolol, sotalol, timolol (Timolol), eucommia, acebutolol, atenolol, atenolol, betaxolol, bisoprolol, celiprolol, Ai Esmolol, metoprolol and nebivolol. Exemplary calcium channel blockers include, but are not limited to, amlodipine (amlodipine), clevidipine (clevidipine), diltiazem (diltiazem), felodipine (felodipine), isradipine, nifedipine (nifedipine) , Nicardipine (iücardipine), nimodipine (nimodipine), nisoldipine (nisoldipine) and verapamil (verapamil). Exemplary diuretics include, but are not limited to, chlorothiazide, hydrochlorothiazide, bumetanide, ethacrynic acid, furosemide, amiloride ), eplerenone (eplerenone), spironolactone (spironolactone) and triamterene (triamterene). Exemplary fibrates include, but are not limited to, bezafibrate, ciprofibrate, clofibrate, gemfibrozil, or fenotibrate. Exemplary antiplatelet drugs include but are not limited to clopidogrel and ticlopidin. The adjuvant treatment method can further adopt surgical treatment, including but not limited to the use of pacemakers, implantable defibrillators, coronary stents, artificial heart valves, coronary artery bypass, balloon dilatation, valve repair and replacement, neck Endarterectomy, angioplasty, stent implantation, craniotomy, coil embolization, oval hole closure and heart transplantation. Some embodiments of the present invention provide combined disease treatments including "crotonate" and one or more adjuvants with disease treatment activity ("stroke treatment", "anticoagulation", "anti-sterol blood"", "vasodilation" , "Antihypertensive", "Induce ERK phosphorylation", "Reduce arterial resistance", "Increase venous volume", "Decrease cardiac oxygen demand", "Decrease heart rate", "Stabilize heart rate" or "NFκB"Regulation") preparation. In such a combined preparation, the crotonate of formula I and the adjuvants with disease therapeutic activity will be treated with disease ("stroke therapy", "anticoagulation", "anticholesterolemia", "vasodilation", "anti- (Hypertension'', ``Induce ERK phosphorylation'', ``Reduce arteriolar resistance'', ``Increase venous volume'', ``Reduce cardiac oxygen demand'', ``Reduce heart rate'', ``Stabilize heart rate'' or ``NFκB regulation'') The effective dose is stored in the combination preparation alone or in combination. In an exemplary embodiment, the crotonate compound of formula I and the non-crotonate agent will each be present in a disease treatment/preventive amount (ie, in a single dose that will individually trigger detectable relief for the subject's symptoms). In addition, the combined preparation may contain one or two crotonate compounds of formula I and a non-crotonate agent in a single dose of sub-therapeutic, wherein the combined preparation containing the two agents is particularly aimed at guiding diseases, conditions or symptoms The combined dose of two agents that alleviates the combined effect of the response. Therefore, one or two crotonate formula I or derivative compounds of crotonate formula and non-crotonate medicament can be stored in the preparation at a secondary therapeutic dose, or can be administered according to the combined administration guidelines, and in the preparation or method It collectively causes detectable remission of disease symptoms, stroke incidence or recurrence rate, or stroke sequelae in the subject. In yet another embodiment, the combination formulation may include one or more neuroprotective agents. In another embodiment, the combination formulation may contain one or more anti-inflammatory agents or other secondary or additional therapeutic agents as described herein. In order to achieve the coordinated administration method of the present invention, the crotonate compound of formula I or the crotonate-derived compound of formula I may be administered simultaneously or sequentially with one or more secondary or adjuvant therapeutic agents contemplated herein in accordance with the coordinated treatment strategy. Therefore, in some embodiments, the compound is a separate formulation or combination formulation as described above (ie, contains a crotonate compound of formula I or related to the non-crotonate medicament or any other secondary or adjuvant therapeutic agent contemplated herein) Or derivative compounds, and non-crotonate therapeutic agents) are co-administered together. This coordinated administration can be completed simultaneously or sequentially in any order, and there may be a period of time when only one or both (or all) of the active therapeutic agents exhibit their biological activity individually and/or collectively. In other embodiments, for example, such coordinated treatment methods may follow or derive from various guidelines for treating stroke. For example, a coordinated treatment method may include crotonate and/or treatment to prevent or treat injury caused by stroke. The difference between all such coordinated treatment methods is that the crotonate compound of formula I or the derivative compound of crotonate of formula I exhibits at least part of the activity, which is combined with complementary stroke preventive or therapeutic agents to exhibit the desired clinical response, or Clinical response provided by secondary or adjunct therapeutic agents. Generally, the co-administration of a crotonate compound of formula I or a derivative compound of formula I with a secondary or adjuvant therapeutic agent will produce in the subject more than just a crotonate compound of formula I or a derivative compound of crotonate of formula I alone, or The therapeutic or prophylactic effect of the secondary or auxiliary therapeutic agent is more improved. This condition needs to consider the direct and indirect effects. In an exemplary embodiment, a crotonate compound of formula I or a derivative compound of crotonate of formula I will be administered in conjunction with one or more secondary stroke therapeutic compounds or other directed or adjuvant therapeutic agents (simultaneously or sequentially, combined or Individual preparations), for example, tissue plasminogen activators, anticoagulants, statins, angiotensin II receptor blockers, angiotensin conversion inhibitors, fibrates, beta receptors Body blockers, calcium channel blockers, hypolipidemic drugs, antiplatelet drugs, or diuretics. In addition, adjuvant or secondary therapy can be used to treat stroke or stroke consequences, such as but not limited to pacemakers, implantable defibrillators, coronary stents, artificial heart valves, artificial hearts, coronary artery bypass, balloon dilation, Valve repair and replacement, heart transplantation, carotid endarterectomy, angioplasty, stenting, craniotomy, coil embolization, or oval foramen occlusion. As described above, in all the various embodiments of the present invention contemplated herein, the disease treatment methods and formulations can use the crotonate compound of Formula I in any of various forms, which includes the pharmaceutically acceptable salts, solvates, Any one or combination of a conformer, enantiomer, polymorph, solvate, hydrate, and/or prodrug. In the exemplary embodiments of the present invention, TPA is used in therapeutic formulations and methods for illustrative purposes. The pharmaceutical composition of the present invention can be administered in various ways to achieve its intended therapeutic or preventive purpose. Suitable routes for administration of the compositions of the present invention include, but are not limited to, traditional delivery routes, devices, and methods, including injection methods, such as, but not limited to, intravenous injection, intramuscular injection, intraperitoneal injection, intraspinal injection, intrathecal injection , Intracranial injection, intra-arterial injection, subcutaneous injection and nasal injection. The composition of the present invention may further include a pharmaceutically acceptable carrier suitable for the specific mode of administration employed. The dosage form of the composition of the present invention contains pharmaceutically acceptable excipients suitable for the preparation of dosage units as described above. Such excipients include but are not intended to be limited to binders, fillers, lubricants, emulsifiers, suspending agents, sweeteners, flavoring agents, preservatives, buffers, wetting agents, disintegrants, foaming agents Agents and other conventional excipients and additives. If necessary, the composition of the present invention can be administered in a controlled-release form by using a sustained-release carrier, such as a hydrophilic, sustained-release polymer. Exemplary controlled release agents herein include, but are not limited to, hydroxypropyl methyl cellulose having a viscosity in the range of about 100 cps to about 100,000 cps, or other biocompatible matrix such as cholesterol. Part of the crotonate composition of formula I of the present invention is designed for non-digestive tract administration, for example, intravenous administration, intramuscular administration, subcutaneous administration, or intraperitoneal administration, which contains aqueous and non-aqueous sterile injections and Like many other proposed compositions of the present invention, it may optionally contain antioxidants, buffers, bacteriostatic agents and/or solutes that allow the preparation to penetrate the blood of mammalian subjects, etc.; and may contain suspending agents and/or Aqueous or non-aqueous sterile suspension of thickener. The formulation can be stored in unit dose or multi-dose volumes. The additional compositions and formulations of the present invention may contain polymers that are sustained-released after parenteral administration. The non-digestible preparation may be a solution, dispersion or emulsion suitable for such administration. The subject agent can also be formulated as a polymer for sustained release after administration in the non-digestive tract. Pharmaceutically acceptable preparations and ingredients will generally be sterile or easily sterilizable, biologically inert and easily manageable. Such polymeric materials are well known in the field of medical technology. Non-digestive preparations usually include buffers and preservatives, as well as medically and physiologically acceptable liquids for injection, such as water, physiological saline, balanced salt solution, aqueous glucose solution, glycerin, etc. The temporarily injected solutions, emulsions and suspensions can be prepared from sterile powders, granules, or tablets of the aforementioned amount. The preferred unit dosage formulation of the active ingredient is a unit dosage formulation comprising the daily dose or unit as described herein, a divided daily dose, or an appropriate ratio thereof. In a more detailed embodiment, the composition of the present invention may include an encapsulated crotonate compound of formula I or a crotonate derivative compound of formula I to use microcapsules, microparticles, or microcapsules prepared by coagulation techniques or by interfacial polymerization, or Microspheres to deliver, for example, individual oxymetrovine, or gelatin microcapsules and poly(methyl methacrylate) microcapsules; colloidal drug carrier systems (eg, liposomes, albumin microspheres, microemulsions, Nanoparticles, and nanocapsules); or macroemulsions. As mentioned above, in certain embodiments, the methods and compositions of the present invention may employ pharmaceutically acceptable salts, such as the acid addition salts or bases of the above-mentioned crotonate compound of formula I and/or its related or derived compounds salt. Examples of pharmaceutically acceptable addition salts include inorganic and organic acid addition salts. Suitable acid addition salts are formed from acids that do not form toxic salts, for example, hydrogen chloride, hydrogen bromide, hydrogen iodide, sulfuric acid, hydrogen sulfide, nitric acid, phosphoric acid, and phosphate. In addition, pharmaceutically acceptable salts include but are not limited to metal salts, such as sodium salt, potassium salt, cesium salt, etc.; alkaline earth metal salts, such as calcium salt, magnesium salt, etc.; organic amine salts, such as triethylamine salt, pyridine salt, Picoline salt, ethanolamine salt, triethanolamine salt, dicyclohexylamine salt, N,N'-dibenzylethylenediamine salt, etc.; organic acid salts, such as acetate, citrate, lactate, succinate , Tartrate, maleate, fumarate, mandelate, acetate, dichloroacetate, trifluoroacetate, oxalate and formate; sulfonates, such as methanesulfonate, benzene Sulfonate and p-toluenesulfonate; and amino acid salts such as spermine, aspartate, glutamate, tartrate and gluconate. Suitable alkali salts are formed from alkalis that do not form toxic salts, such as aluminum calcium, lithium, magnesium, potassium, sodium, zinc, and diethanolamine salts. In other detailed embodiments, the methods and compositions of the present invention employ a prodrug of formula I crotonate. Prodrugs are considered to be any covalently bonded carriers that will release the active drug substance in the organism. Examples of prodrugs used in the present invention include esters or amides with hydroxyalkyl or aminoalkyl as substituents, and these prodrugs can be The compounds are prepared by reacting anhydrides such as succinic anhydride. It will also be understood that the invention disclosed herein covers the use of the compound in vivo metabolites (whether it is produced in the body after administration of the target precursor compound, or is directly administered in the form of its metabolite) to include Formula I Croton ester method and composition. Such products may come from the results of the compounds administered mainly due to enzymes such as oxidation, reduction, hydrolysis, amidation, esterification, etc. Therefore, the present invention encompasses the methods and compositions of the present invention employing compounds made by a process comprising a crotonate compound of formula I in contact with a mammalian subject for a period of time sufficient to produce its metabolites. Such products are generally prepared by preparing the radiolabeled compound of the present invention in a non-digestible tract at a detectable dose to animals such as rats, mice, guinea pigs, monkeys or humans, allowing sufficient time for metabolism to occur , And separate the conversion products from urine, blood or other biological samples for testing. It will also be understood that the invention disclosed herein covers diagnostic compositions for the diagnosis of stroke, including, but not limited to, the risk, presence, severity, or treatment orientation of stroke in mammalian subjects, or other diagnostic compositions that manage diseases, including (E.g., isotope markers, fluorescent markers, or other markers, which can be detected using conventional methods) and crotonate compounds of formula I and mammalian subjects (eg, cells, tissues) who are at risk of or exhibiting one or more symptoms of stroke , Organs or individuals), and then use any broad array of known assays and labeling/detection methods to detect the presence, location, metabolism, and/or binding status of the labeled compound. In an exemplary embodiment, the crotonate compound of formula I is labeled by isotopes by having one or more atoms replaced by atoms having different atomic weights or mass numbers. Examples of isotopes that can be inserted into the disclosed compounds include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, fluorine, and chlorine, such as 2 H, 3 H, 13 C, 14 C, 15 N, 18 O, 17 O, 31 P, 32 P, 35 S, 18 F, and 36 Cl. The isotope-labeled compound is then administered to the individual or other subject, and then detected and produced useful diagnostic and/or therapeutic management data according to conventional techniques as described above. EXAMPLES The examples described below demonstrate the novel and potent effects of crotonate and derivative compounds as a treatment and prevention agent for stroke. These and other findings will be further supplemented and explained in the following examples. Example I The effect of TPA on Peripheral White Blood Cells (WBC) and the hemoglobin (Hb) count in S180 cell transplantation mice Sarcoma 180 (Sarcoma 180, S 180) cell line was transplanted into Kwen- Ming mice. On the third day, mice were given 50, 100, or 200 intraperitoneal (ip) μ Seven days for g/kg/day of TPA. On the second day after the treatment was completed, blood samples were taken from the tails of the treated mice for WBC and HB analysis. WBC count of treatment group (seven days of 50, 100 or 200 μ g/kg/day) 16.1±7.4, 18.7±.3.0 and 20.7±.3.4 x10 respectively 9 /L; WBC count of the control group is 13.6±1.8x10 9 /L. The Hb of the treatment group was 136±11, 149±12 and 149±10g/L, while the Hb of the control group was 134+-15g/L. The results showed that ip injection of TPA can increase the peripheral WBC count of mice in a dose-dependent manner, and the Hb levels of TPA-treated mice did not significantly affect the control mice. Example II Dose Range Study Due to the strong local irritation caused by the use of TPA, TPA is given to patients by intravenous infusion. The TPA solution in a sterile syringe is injected into 200 ml of sterile physiological saline and mixed thoroughly for intravenous infusion. Toxicity and side effects of different doses of clinically administered TPA: (1) 1 mg/patient/week of TPA: 1 mg of TPA in the solution is thoroughly mixed with 200 ml of sterile physiological saline for use at a rate of 16 μg/min Complete intravenous infusion within 1 hour. After 1 hour after the administration of TPA, the patient began to chill for about 30 minutes, followed by a fever with mild to severe sweating (the patient's body temperature reached 37.5-39.5°C, which lasted 3 to 5 hours and then became normal ). The above symptoms can be alleviated by giving patients glucocorticoids. This dose of TPA caused bleeding in a few patients, some patients had difficulty breathing for a short period of time, and Hb was detected in urine. However, these side effects are quite short-lived and recoverable. Heart, liver, kidney, and lung functions were all normal. (2) TPA is given at 0.5 mg/patient x2/week: (two doses per week) 0.5 mg TPA in the solution is thoroughly mixed with 200 ml of physiological saline for completion within 1 hour at a rate of 8 μg/min Intravenous infusion. The response after administration was similar to the 1 mg TPA dose, but to a lesser extent than the 1 mg dose. Patients can withstand lighter doses more easily. Sometimes, Hb is detected in the patient's urine. Dyspnea was not observed. Heart, liver, kidney and lung functions are normal. (3) TPA is given at 0.25 mg/patient x 4/week: 0.25 mg TPA in the solution is thoroughly mixed with 200 ml of physiological saline for intravenous infusion within 4 hours at a rate of 4 μg/min. After administration, symptoms like chills and fever were also observed, but were milder than the higher dose. No Hb was detected in the urine, and no patient had trouble breathing. Heart, liver, kidney and lung functions are normal. Example III Treatment of refractory/recurrent tumors with TPA for refractory/recurrent hematological tumors/marrow diseases with TPA (Xichuan Pharmaceuticals, Nan Yang, Henan, China), dexamethasone (dexamethasone) And choline magnesium trisalicylate (choline magnesium trisalicylate) combination treatment. As described below, a better method for demonstrating the therapeutic use of TPA in the treatment of acute myelogenous leukemia (AML) will be administered to demonstrate the use of TPA in the treatment of other cancer diseases and malignant tumors. In addition to the specific guidelines here, successful treatment and/or remission will use various conventional cancer detection and assessment methods to determine cancer and tumor diseases of different targets, for example, to determine the size reduction of solid tumors, histopathology Research to assess tumor growth, stage, metastatic potential, histological presence/performance of tumor markers, etc. AML is an aggressive disease that usually requires immediate and intensive treatment. The average age of patients diagnosed with AML is 64 to 68 years old, and the treatment rate of patients over 60 years old with standardized therapy is <20%. Patients with AML who develop AML after an unprecedented hematologic disorder or before chemotherapy/radiotherapy for leukemia have similar debilitating effects because the disease is inherited from specific unfavorable cells Related to clinical features. Therefore, most patients diagnosed with AML have characteristics related to patients and/or diseases associated with poor prognosis. For patients with recurrent disease, there is no standard non-transplantation therapy that demonstrates the ability to treat. For these patients, AML is usually a fatal disease. Therefore, a new method of AML treatment is necessary. Using the method and composition of the present invention, based on the novel role of TPA in regulating cell signaling pathways, TPA has developed into a therapeutic agent for treating patients with AML, and its ability is sufficient to induce cell line differentiation and/or apoptosis, and Clinical data showing the effectiveness of TPA in the treatment of cancers and tumor diseases including bone marrow malignancies. The clinical evaluation of TPA so far has measured cell viability and apoptosis assays to show that TPA has shown direct therapeutic cytotoxicity in at least some AML cases. In the primary culture analyzed by Western analysis, TPA strongly induced ERK phosphorylation at 1 hour of cell culture. The cytotoxicity of TPA on primary AML cells is related to the subsequent loss of phosphor-ERK pro-survival signal after 24 hours of in vitro exposure. This observation is in good agreement with other studies that allegedly reduce the survival rate of primary AML cells after medical interference with ERK signals by MEK inhibitors, such as PD98059, U0126, and PD 184352. In this study, the loss of ERK signal was related to ERK phosphatase induction. In addition to protein kinase C and ERK activation, TPA is an inducer and pro-survival transcription factor of NF-κB in AML blasts and leukemia stem cells. Recent research in this laboratory shows that AML cell NF-κB can be inhibited in vivo by treatment with dexamethasone + choline magnesium trisalicylate (CMT) for 48 hours. In addition, it was found that dexamethasone can induce the expression of MKP-1 ERK phosphatase and enhance the cytotoxicity of TPA to primary AML samples. In this content, the following exemplary embodiments were selected to use dexamethasone and CMT as adjuvant drugs with TPA before treatment before 24 hours and after 24 hours. These drugs are very well tolerated and are expected to reduce the inflammatory side effects of treatment, and by increasing ERK phosphatase performance and inhibiting NF- K B to strengthen TPA cytotoxicity. In addition, because of its anti-inflammatory properties, it can improve side effects and can suppress persistent NF- K The anti-apoptotic effect of B and the induction of phosphatases that reduce the activity of the signal pathway to enhance anti-leukemia activity, dexamethasone and CMT will be used as adjuvant drugs. Recruitment of 35 patients at the beginning of the TPA phase 1 study [23 with refractory/recurrent AML, 2 with other marrow malignancies (CML-blast crisis), myelodysplastic abnormalities accompanied by excessive Bud cells (myelodysplasia with excess blasts)), 3 with Hodgkin's disease, 3 with Non-Hodgkin's Lymphoma, and 4 with solid tumors]. Most patients have refractory/recurrent AML. This clinical result includes an AML patient receiving 8 TPA infusion stable for> 5 months. In the second patient, the number of circulating bud cells was significantly decreased (5 times) after TPA administration. The decline in leukemic blasts lasted for 4 weeks, and the patient eventually died of a fungal infection. Finally, even if high-dose chemotherapy and autologous stem cell rescue were performed, patients with refractory/recurrent Hodgkin's disease had partial relief of the thoracic mass after TPA administration. 5 days, 0.188mg/m from the 8th day to the 12th day 2 Two-thirds of patients treated with different doses experienced grade III non-hematologic dose limiting toxicities (DLT), and established Day 1 to Day 5, and Day 8 to Unit pharmacy on the 12th day 0.125mg/m 2 The maximum tolerated TPA dose per day is complete with TPA dose escalation. In the case of AML and other hematological malignancies, the patient is given a 1 mg/week x 3 weeks (Day 1, Day 8, Day 15) initial dose of TPA and 6 hours of continuous intermittent pulse oximetry Degree analysis (continuous/intermittent pulse oximetry). Starting 24 hours before starting TPA treatment, giving patients 10 mg of dexamethasone every 6 hours and giving patients 1500 mg of choline magnesium trisalicylate (CMT) every 8 hours until the end of TPA administration 24 Hours. After the initial dose of TPA is administered, the patient has a two-week rest period, which can then be evaluated again. From the beginning, patients with TPA doses that had medicinal or stabilizing effects were treated continuously for 6 cycles for 28 days according to the following guidelines. After a two-week rest period, patients were pre-medicated with Tylenol 650 mg and Benadryl 25 to 50 mg 30 minutes before TPA administration (depending on patient size and age) . Then, TPA intravenous infusion was given through a central venous catheter every five days for two weeks after a 2-week rest period. TPA is administered at a dose of 1 mg in 200 ml of normal saline for more than 1 hour. Twenty-four hours before the initial TPA administration, patients were given 10 mg of dexamethasone every 6 hours and patients were given 1500 mg of choline magnesium salicylate every 8 hours until 24 hours after TPA administration. The level of TPA in the blood was measured before and after the infusion using bioassays that measure the differential activity of organic solvent extraction. Extract 1 ml of blood twice with 5 ml of ethyl acetate, re-dissolve the extraction residue in 50 μL of ethanol and add an equal amount of HL60 cells. After 48 hours, adherent cells were measured. Blood samples taken before and after TPA infusion are tested to determine the levels of white blood cells, platelets, and neutrophils. The samples were additionally analyzed for the presence of myeloblasts and Auer rods. These and ongoing experiments will further clarify the therapeutic cytotoxicity and other effects of TPA against cancer cells in AML and other cancers and tumor symptoms. Example IV Measurement of the regulation of ERK activation Phosphorylated ERK levels were measured in circulating malignant tumor cells in patients with leukemia and in peripheral blood mononuclear cells in lymphoid/solid tumor patients. Blood samples were obtained from patients treated according to the guidelines of Example III before and after administration of TPA. Flow cytometry analysis was performed on blood samples using specific antigens on the cell surface and phosphorylated ERK specific antibodies directly bound to fluorescent substances (BD Biosciences, San Jose, CA) on leukemia patients with WBC≧1000 per μL . Samples were taken at the beginning of treatment according to the guidelines of Example III on day 1, day 2 and day 11 and in successive cycles on day 1 and day 11 before TPA administration and 1 hour after TPA infusion. According to the guidelines of Example III, on the 1st, 8th, and 15th days of the first cycle before the infusion and 1 hour and 4 hours after the infusion μ L. Peripheral blood samples were obtained from leukemia patients with absolute blood cell number ≧2500 and other non-leukemia patients. The sample system was also analyzed for phosphorylated ERK and total ERK 1/2 levels using Western blot analysis to verify the results obtained by flow cytometry and to link them to clinical responses. The aforementioned analysis will further clarify the role of TPA in the treatment of cancer and tumor diseases, including the cytotoxic effect of TPA on malignant tumor cells, such as primary AML cells, and the phosphorylated-ERK pro-survival signal due to the related reduction of TPA. Example V Measuring the regulation of NF-κB In previous studies, it was found that the activity of NF-κB in patients can be regulated after administration of TPA and dexamethasone. In addition, dexamethasone was found to induce MKP-1 ERK phosphatase expression and enhance TPA cytotoxicity. The following study was designed to further clarify how NF-Kb activity in patients is therapeutically regulated by TPA plus dexamethasone. NF-κB binding (NF-κB binding) is based on the baseline and the peripheral blood samples before and after the injection of TPA-treated patients according to Example III using an antibody antigen-specific enzyme immunization method (BD Bioscience, San Jose, USA). NF-κB levels are quantified using 96-well format using chemiluminescence intensity to detect in limited cell extracts. In addition, perform electrophoretic mobility change analysis to measure μ L. NF-κB binding in peripheral blood samples of leukemia patients with absolute autologous blood cell numbers ≧2500 and other non-autologous patients with normal white blood cell count. The study will further show that TPA is an inducer of NF-κB; however, these examples show that AML cell NF-κB can be inhibited under the treatment of dexamethasone and choline magnesium trihydrate. Example VI Treatment of an individual patient with stroke, NC, male, 68 years old, who suffered a stroke 18 months before TPA treatment. At the beginning of TPA treatment, this patient was unable to walk without crutches, had difficulty moving his left hand and left foot, and was quite tired and weak. This patient receives TPA containing 0.19mg every other day (0.125mg/m 2 ) Of 1 ampoule for 4 weeks, and then receive 0.24 mg of TPA every other day (1.25 x 0.125 mg/m 2 ) For 2 weeks, and then receive 0.26mg of TPA every other day (1.5 x 0.125mg/m 2 ) 3 consecutive weeks. This patient has fully recovered. Patient MC, male, 65 years old, had a stroke a few years before starting TPA treatment. This patient receives 0.19mg TPA (0.125mg/m 2 ) For 3 to 4 injections for a total of 35 injections over ten weeks. This patient has recovered his face's mobility and his right side has improved by 80%. Example VII Treatment of Embolic Stroke Model with TPA (Embolic Stroke Model) Male Sprague-Dawley rats (Charles River, Japan) each weighing 280 to 350 grams were used. Embolism stroke is induced by the improvement of the method of Kudo et al. (Kudo, et al.) (1982). Rats to be used for blood collection were spontaneously breathed with 1.0% haloalkane (Fluorothane TM ; Takeda, Osaka, Japan) came to anesthetize. 24 (gauge) Surflo TM (Terumo Medical Products, Elkton, MD) was fixed on the femoral artery, and 0.1 mL of arterial blood was collected with a 1-mL syringe (Terumo Medical Products, Elkton, MD) for injection. The arterial blood line in the syringe was incubated at 30°C for 2 days to form a blood clot. After this, 0.1 mL of physiological saline was added to the syringe for injection and passed through a 26-gauge injection needle (Terumo Medical Products, Elkton, MD) twice to crush the blood clot. Rats with induced cerebral embolic stroke were anesthetized with 1.0% halocarbon under spontaneous breathing. The midline incision of the rat's neck, while the external carotid artery, superior thyroid artery, occipital artery and pterygopalatine artery are bipolar A bipolar coagulator (T-45; Keisei Medical lndustrial Co. Ltd, Tokyo, Japan) was cauterized. Cerebral embolism is induced by injecting 0.1 mL of crushed blood clot into the internal carotid. Evaluation of the formation of cerebral embolism was performed using a laser Doppler flowmetry (FloCl; Omegawave, Tokyo, Japan). The decrease in cerebral blood supply to 30% or less is used as positive evidence of embolism. The blood supply to the brain was monitored for 30 minutes after the blood clot was injected, and 50% or less of the remaining blood supply before the blood clot was injected was monitored. After this, the catheter (PE50) used to administer the drug is fixed to the carotid artery and awakens the animal. The rats that successfully formed cerebral embolism were divided into 4 groups. Group 1 rats were given saline injection every other day. Give rats from groups 2 to 4 0.125 mg/m every other day 2 The TPA was injected for 4 weeks. Then sacrifice group 2. Give every group 3 to group 4 a further 0.156 mg/m every other day 2 The TPA injection was continued for 2 weeks, and then the third group was sacrificed. Give the fourth group 0.18775mg/m every other day 2 The TPA was sacrificed for 3 weeks. After the animal was sacrificed, the brain was excised and cut into ten pieces at a 1 mm interval using a tissue slicer (M cIwain tissue chopper) (Mickle Laboratory Engineering, UK), and immersed in 2% TTC (2,3,5- Stain for 20 minutes in 2,3,5-triphenyltetrazolium chloride (Tokyo Kasei). The images of TTC stained slices were uploaded to a computer using a digital camera (HC-2500; Fuji PhotoFilm) and Phatograb-2500 (Fuj i Photo Film), and the cerebral infarct volume was calculated using Mac Scope (Mitani, Japan). The volume of cerebral infarction is given as the mean ± standard deviation. As for the statistical test of the result of cerebral infarction volume, the evaluation was performed by performing the Dunnett's Test (Dunnett's Test) by comparing the control group and each TPA administration group with the control group and then by performing T- Inspection to complete. Observe neurological symptoms every day until sacrifice, and the rats are evaluated according to three tests: (1) gently grasp the rat by its tail, hang from the floor for 1 meter, and then observe forelimb flexion; (2) place the rat in Can be firmly grasped on the soft plastic coated paper with claws. While holding the tail by hand, apply slight lateral pressure behind the rat's shoulder until the forelimb drops a few inches; (3) Allow the rat to move freely and observe whether there is circling behavior. The neurological symptoms were scored according to the scale developed by Bederson et al. (1986) as follows: 0: no defect was observed; 1: forelimb flexion; 2: lateral thrust reduces resistance but does not hover ; 3: Same behavior as level 2 but circling. Steel's test was used for the control group and each TPA-administered group compared with the control group and then neurological symptoms were evaluated by performing a Wilcoxon test for the TPA-administered group. In any verification test, a value of p<0.05 is defined as statistically significant. Example VIII Benefits of using TPA in the treatment of stroke in a permanent moderate cerebral artery occlusion model animal (A Permanent Middle Cerebral Artery Occlusion Model). In this study, male Wistar rats (250 to 320 g) were used. The animal system was anesthetized with Isoflurane (3% guidance, 1-2% maintenance). The degree of anesthesia is monitored by pinching toes. During this study, anesthesia techniques will be used for each procedure. Trim and clean the surgical site with alcohol and surgical disinfectant, and the animal department is placed on a warm water heating pad to maintain body temperature. Make a paramedian incision in the carotid artery of the neck. The tissue is cut directly to show the carotid artery and bifurcation. The suture is located at the proximal end or the common carotid artery and external carotid artery. Ligate the suture. An incision was made in the common artery away from the ligation, and a pre-prepared silk thread (4.0 single-strand suture or similar material) was placed in the carotid artery and placed in the internal carotid artery. The silk thread penetrated the carotid bifurcation about 20mm deep until it caught the middle cerebral artery and felt a little resistance. Care must be taken not to rupture the artery when inserting the silk thread. The silk thread is ligated and the skin incision is closed. After successful occlusion, the Bederson scale (see Bederson et al., (1986) Stroke, 17: 1304-1308.) was used to evaluate the animals when they were awake. Measure body temperature every 15 minutes to maintain normal body temperature. Animals that have undergone a middle cerebral artery occlusion procedure may have abnormal body temperature a few hours after surgery. The animals decide to install in the cooling box or heating box according to their body temperature. Body temperature is maintained at 37.5°C. Animals were monitored for 6 hours after the middle cerebral artery was blocked and then placed in cages overnight. The rats were divided into four groups. The first group of rats was given saline injection every other day. Group 2 to Group 4 are given 0.125mg/m every other day 2 The TPA was injected for 4 weeks. Then sacrifice group 2. Further every day to give the third group to the fourth group 0.156mg/m 2 TPA for 2 weeks, and then sacrifice the third group. Give the fourth group 0.18775mg/m every other day 2 The TPA lasted 3 weeks and then sacrificed. After the animal was sacrificed, its brain was excised and sliced into 10 slices at 1 mm intervals using a tissue slicer (Mickle Laboratory Engineering, UK), and immersed in 2% TTC (2,3,5-triphenyltetrazolium chloride) at 37°C. Compound (2,3,5-triphenyltetrazolium chloride); Tokyo Kasei) for 20 minutes to stain. The images of TTC stained slices were uploaded to a computer using a digital camera (HC-2500; Fuji PhotoFilm) and Phatograb-2500 (Fuji Photo Film). Brain slices were taken and analyzed for infarct size, infarct volume, penumbra, and edema. Observe neurological symptoms every day until sacrifice. Observe neurological symptoms every day until sacrifice, and the rats are evaluated according to three tests: (1) gently grasp the rat by its tail, hang from the floor for 1 meter, and then observe forelimb flexion; (2) place the rat in It can be firmly grasped on a large piece of soft plastic coated paper with paws. While holding the tail by hand, apply slight lateral pressure behind the rat's shoulder until the forelimb drops a few inches; (3) Allow the rat to move freely and observe whether there is circling behavior. The neurological symptoms were scored according to the scale developed by Bederson et al. (1986) as follows: 0: no defect was observed; 1: forelimb flexion; 2: lateral thrust reduces resistance but does not Circling; 3: Same behavior as level 2 but circling. Steel's test was used for the control group and each TPA-administered group compared with the control group and then neurological symptoms were evaluated by performing the Wilkerson test for the TPA-administered group. In any verification test, a value of p<0.05 is defined as statistically significant. Example IX Benefits of TPA in the treatment of stroke using a transient moderate cerebral artery occlusion model. Male C57B16 mice (25 to 30 grams) were used in this study. The mice were anesthetized with isoflurane (3% lead, 1-2% maintenance). Trim and clean the surgical site with alcohol and surgical disinfectant. A midline neck incision was made in the carotid artery, and the artery system was separated from its branch. A single strand of silk thread is introduced into the internal carotid artery and penetrates deeper until it meets in the mid-brain artery. The incision is sutured by ligating the thread. Two hours after the blockage, the mice were re-anesthetized and the silk thread was removed from the MCA. Maintain body temperature by using a heating pad during and after surgery. Animals were monitored for 4 hours after the middle cerebral artery was blocked. The mice were divided into four groups. The first group of mice was given saline injection every other day. Group 2 to Group 4 are given 0.125mg/m every other day 2 The TPA was injected for 4 weeks. Then sacrifice group 2. Further every day to give the third group to the fourth group 0.156mg/m 2 TPA for 2 weeks, and then sacrifice the third group. Give the fourth group 0.18775mg/m every other day 2 The TPA lasted 3 weeks and then sacrificed. After the animal was sacrificed, its brain was excised and sliced into 10 slices at 1 mm intervals using a tissue slicer (Mickle Laboratory Engineering, UK), and immersed in 2% TTC (2,3,5-triphenyltetrazolium chloride) at 37°C. Compound (2,3,5-triphenyltetrazolium chloride); Tokyo Kasei) for 20 minutes to stain. The images of TTC stained slices were uploaded to a computer using a digital camera (HC-2500; Fuji PhotoFilm) and Phatograb-2500 (Fuji Photo Film). Brain slices were taken and analyzed for infarct size, infarct volume, penumbra, and swelling. The neurological symptoms were observed daily until sacrificed, and the rats were evaluated according to three tests: (1) Lightly grasp the rat by its tail, hang from the floor for 1 meter, and then observe forelimb flexion. (2) Place the rat on a large piece of soft plastic-coated paper that can be firmly grasped by paws. While holding the tail by hand, apply slight lateral pressure behind the rat's shoulder until the forelimb drops a few inches. (3) Allow the rat to move freely and observe whether there is circling behavior. The neurological symptoms were scored according to the scale developed by Bederson et al. (1986) as follows: 0: no defect was observed; 1: forelimb flexion; 2: lateral push would reduce resistance but not hover; 3: and The same behavior at level 2 but hovering. Steel's test was used for the control group and each TPA-administered group compared with the control group and then neurological symptoms were evaluated by performing the Wilkerson test for the TPA-administered group. In any verification test, a value of p<0.05 is defined as statistically significant. Example X Clinical efficacy of using TPA to treat stroke. Men and women aged 30-72 years who experienced stroke within 1 month were recruited to participate in a 10-week trial of TPA. The recruited individuals signed an informed consent form and used computed tomography (computed tomography, CT), physical and neurological tests, neurological examination, sedation, and National Institute of Health Stroke Survey. NIHSS), 12-lead electrocardiogram, electrocardiogram telemetry, blood oxygen measurement, vital signs, weight, patient background, pregnancy test, urine drug measurement, blood test, coagulation index, routine clinical trials, urine test. Clinical laboratory tests include comprehensive metabolic indexes (sodium, potassium, chlorine, carbon dioxide (CO 2 ), glutamate (Gl u), urea nitrogen (BUN), chromium, calcium, total protein (TP), blood protein (ALB), total bilirubin (TBILI), alkaline phosphatase (AP), asparagine Amino acid converting enzyme (AST), alanine converting enzyme (ALT)), routine blood test (Hematology CBC) (hemoglobin measurement (Hgb), blood volume ratio (Hct), red blood cell count (RBC), self blood cell count ( WBC), platelet count (Plt), white blood cell differential count (Diff)), and human chorionic gonadotropin (Serum hCG) in the serum of all women. Give the individual 0.125mg/m every other day 2 TPA or placebo for 4 weeks, followed by 1.25 x 0.125 mg/m every other day in weeks 5 and 6 2 Or placebo, and every week 7 to week 9 is given 1.5 x 0.125mg/m every other day 2 Or placebo. Individuals were monitored 2 hours after administration of TPA or placebo. At Weeks 5 and 10, use NIHSS (NIH Stroke Scale), Barthel ADL index (Granger, 1979), and the modified Rayleigh scale Table (modified Rankin scale) (Farrell, 1991) to evaluate the object. By measuring and comparing TPA-treated individuals with placebo-treated individuals, the benefit was assessed according to the changes in the benchmarks of the National Institutes of Health Stroke Assessment Form. Secondary efficacy variables are the Pap scale and the modified Leigh scale. Collect and evaluate safety measures during the trial, especially the changes from the baseline to the 5th week. These measurements include side effects reports (Adverse events reports), physical examination, vital signs, weight measurements, electrocardiograms (ECGs), clinical laboratory test results, and vital signs and scores for suicidal behavior and/or ideas. Side effects are any adverse medical reactions when the subject receives the study drug, regardless of whether it is causally related to the study drug. Therefore, side effects may be any unwanted or unexpected signs (including, for example, abnormal experimental results), symptoms, or disease that are temporarily related to the study drug, regardless of whether they are considered to be related to the study drug. If all treatments are completed, the subject is deemed to have completed the study. However, if the subject fails to meet the inclusion/exclusion criteria; suffering from side effects, inadequate response to treatment, withdrawing his consent, violating the agreement, not appearing, or dying may be withdrawn from the study. ReferencesAltuwaijri S, Lin HK, Chuang KH, Lin WJ, Yeh S, Hanchett LA, Rahman MM, Kang HY, Tsai MY, Zhang Y, Yang L, and Chang C. Interruption of nuclear factor kappaB signaling by the androgen receptor facilitates 12-O-tetradecanoylphorbolacetate-induced apoptosis in androgen-sensitive prostate cancer LNCaP cells. Cancer Res 2003; 63: 7106-12. Ando I., Crawfor DH et al. Phorbol ester-induced expression and function of the interleukin 2 receptor in human B lymphocytes. Eur J Immunol. 15(4), 341-4 (1985). Aye MT, Dunne JV Opposing effects of 12-O-tetradecanoylphorbol 13-acetate on human myeloid and lymphoid cell proliferation. J Cell Physiol. 114(2 ), 209-14 (1983). Bederson JB, Pitts LH, Tsuji M, Nishimura MC, Davis RL, Bartkowski H. Rat middle cerebral artery occlusion: evaluation of the model and development of a neurologic examination. Stroke. 1986; 17: 472-476. Boutwell RK Biochemical mechanism of tumor promotion, in mechanisms of tumor promotion and co-carcinogenesis. Eds. Slaga, TJ, Siv ak, AJ and Boutwell, RK Raven, New York, 49-58 (1978). Boutwell RK The function and mechanism of promoters of carcinogenesis. CRC Crit. Rev. Toxicol 2, 419-443 (1974). Brose N, Rosenmund C . Move over protein kinase C, you've got company: alternative effectors of diacylglycerol and phorbol esters. JCell Sci; 115: 43 99-411 (2002). Cancer Chemother Pharmacol. Jun;57(6):789-95 (2006 ). Cui XX, Chang RL, Zheng X, Woodward D, Strair R, and Conney A H. A sensitive bioassay for measurmg blood levels of 12-O-tetradecanoylphorbol-13-acetate (TPA) in patients: preliminary pharmacokinetic studies. Oncol Res 2002; 13: 169-74. Deegan MJ, Maeda k. Differentiation of chronic lymphocytic leukemia cells after in vitro treatment with Epstein-Barr virus or phorbol ester. Immunologic and morphologic studies. Am J Hermatol. 17(4), 335- 47 (1984). Falcioni F., Rautmann A. et al. Influence of TPA (12-0-tetradodecanoyl-phorbol-13-acetate) on human B lymphocte function. Clin Exp Immunol. 62(3), 163-2 ( 1985). Far rell B, Godwin J, Richards S, Warlow C, et al. (1991). The United Kingdom transient ischaemic attack (UK-TIA) aspirin trial: final results. J Neurol Neurosurg Psychiatry) 54 (12): 1044-1054. Forbes 1. J., Zalewski PD, Letarte M. Human B-lymphocyte maturation sequence revealed by TPA-induced differentiation of leukaemi cells. Immunobiology 163(1), 1-6 (1 982). Gunjan Goel, Harinder PS Makkar, George Francis, and Klaus Becker. Phorbol Esters: Structure, Biological Activity, and Toxicity in Animals. International Journal of Toxicology, 26:279-288, 2007. Gogusev J., Barbey S., Nezelof C. Regulation of TNF-alpha and IL -1 gene expression during TPA-induced differentiation of "Malignant histiocyosis" DEL cell line t(5:6) (q35:P21). Anticancer Res. 16(1), 455-60(1996). Granger CV, Devis LS, Peters MC, Sherwood CC, Barrett JE. Stroke rehabilitation: analysis of repeated Barthel lndex measures. Arch Phys Med Rehabil. 1979;60:14-17. Han ZT, Tong YK, He LM, Zhang Xuan, Sun JZ, Wang TY, Zhang H., Cui YL, Newmark HL, Conney AH, Chang RL 12-O-Tetradecanoyl-phorbol-13-acetate (TPA) -induced increase in depressed white blood cell counts in patients treated with cytotoxic cancer chemotherapeutic drugs. Proc. Natl. Acad. Sci. 95, 5363-5365 (1998). Han ZT, Zhu XX, Yang RY, Sun JZ, Tian GF, Liu XJ, Cao GS, NewMark HL, Conney AH, and Chang RL Effect of intravenous infusion of 12-O-tetradecanoyl-phorbol- 13 -acetate (TPA) in patients with myelocytic leukemia: Preliminary studies on therapeutic efficacy and toxicity. Pro. Natl. Acad. Sci. 95, 5357-5361 (1 998). Harada S. et al.: Tumor Promoter, TPA, Enhances Replication of HTLV-III/LAV. Virology 154, 249-258 (1986). Hecker E. In handbuch der allgemeinen patholgie, ed. Grundmann, E. (Springer-Verlag, Berlin-Heideiberg, Vol. IV 16, 651- 676 (1975). Hecker E. Structure -activity relationships in deterpene esters irritant and co-carcinogenic to mouse skin, in mechanisms of tumor promotion and co-carcinogenesis. Eds. Slaga, TJ, Sevak, A. j. and Boutwel l, RK Raven, New York, 11-49 (1978). Hofmann J. The potential for isoenzyme-selective modulation of protein kinase C. FASEB J. 11, 649-669 (1997). Huberman E., Callaham MF Induction of Tenninal differentiation in human promyelocytic leukemia cells by tumor-promoting agents. Proc. Natl. Acad. Sci. 76, 1293-1297 (1979). Hunter T. Signaling 2000 and beyond. Cell 100, 113-117 (2000). Kassel O , Sancono A, Kratzschmar J, Kreft B, Stassen M, and Cato A C. Glucocorticoids inhibit MAP kinase via increased expression and decreased degradation of MKP-1. Embo J 2001; 20: 7108-16. Kazanietz MG Eyes Wide Shut: protein kinase. C isoenzymes are not the only receptors for the phorbol ester tumor promoters. Mol. Carcinog. 28, 5-12 (2000). Keoffler HP, Bar-Eli M., Territo MC Phorbol ester effect on differentiation of human myeloid leukemia cells lines blocked at different stages of maturation. Cancer Res. 41, 919-926 (1981). Kim SC, Hahn JS, Min YH, Yoo NC, Ko YW, and Lee W J. Constitutive aetivation of extracellular signal-regulated kinase in human acute leukemias: combined role of activation of MEK, hyperexpression of extracellular signal-regulated kinase,. and downregulation of a phosphatase, PAC 1. Blood 1999; 93: 3893-9. Kiyoi H, Naoe T, Nakano Y, Yokota S, Minami S, Miyawaki S, Asou N, Kuriyama K, Jinnai I, Shimazaki C, Akiyama H, Saito K, Oh H, Motoji T, Omoto E, Saito H, Ohno R, and Ueda R. Prognostic implication of FLT3 and N-RAS gene mutations in acute myeloid leukemia. Blood 1999; 93: 3074-80. Kobayashi M., Okada N. et al. Intracellular interleukin-1 alpha production in human gingival fibroblasts is differentially regulated by various cytokines. J Dent Res. 78(4), 840-9 (1 999). Kudo M., Aoyama A., Ichimori S. and Fukunaga N. An animal model of cerebral infarction: homologous blood clot emboli in rats. Stroke 13: 505 -508 (1982) Lebien TW, Bollum FJ et al. Phorbol ester-induced differentiation of a non-T, non-B leudemic cell line: model for human lymphoid progenitor cell dev elopment. J Immunol. 128(3), 1316-20 (1982). MD Iqbal Hossain Chowdhury et al. The Phorbol Ester TPA Strongly Inhibits HIV-1 Induced Syncytia Formation but Enhances Virus Production: possible involvement of protein kinase C pathway. Virology 176, 126-132, (1990). Meinhardt G., Roth J., Hass R. Activation of protein kinase C relays distinct signaling pathways in the same cell type: differentiation and caspase-mediated apoptosis. Cell Death Differ. 7, 795 -803 (2000). Milella M, Kornblau SM, Estrov Z, Carter BZ, Lapillonne H, Harris D, Konopleva M, Zhao S, Estey E, and Andreeff M. Therapeutic targeting of the MEK/MAPK signal transduction module in acute myeloid leukemia. J Clin Invest 2001; 108: 851-9. Mochty-Rosen D., Kauvar LM Modulating protein kinase C signal transduction. Adv. Pharmacol. 44, 91-145 (1998). Morgan MA, Dolp O, and Reuter C W. Cell-cycle-dependent activation of mitogen-activated protein kinase kinase (MEK- 1/2) in myeloid leukemia cell lines and induction of growth inhi bition and apoptosis by inhibitors of RAS signaling. Blood 2001; 97: 1823-34. Nagasawa K., Chechgik BE et al. Modulation of human T-cell differentiation markers by 12-O-tetradecanoylphorbal-13-acetate. Thymus. 3( 4-5), 307-18, (1981). Nakao Y., Matsuda S. et al. Paradoxical anti-leukemic effects of plant-derived tumor promoters on a human thymic lymphoblast cell line. Int J Cancer 30(6), 687-95 (1982). Nakao Y., Matsuda S. et al. Phorbol ester-induced differentiation of human T-lymphoblastic cellline HPB-ALL. Cancer Res. 42(9), 33843-50 (1982). Newton AC Protein kinase C: structure, function and regulation. J. Biol. Chem. 270, 28495-28499 (1995). Palombella VJ, Rando OJ, Goldberg AL, and Maniatis T. The ubiquitin-proteasome pathway is required for processing the NF-kappa B1 precursor protein and the activation of NF-kappa B. Cell 1994; 78: 773-85. Platanias L C. Map Map kinase signaling pathways and hematologic malignancies. Blood 2003; 101: 4667-79. Rovera G., Santoli D. , Damsky C. Hum an promyelocytic cells in culture differentiated into macrophage-like cells treated with a phorbol diester. Pro. Natl. Acad. Sci. 7, 2779-2783 (1979). YIP, YK et al. Stimulation of human gamma interferon production by diterpene esters. Infection and Immunity 34(1) 131-139 (1981). Zhao J., Sharma Y., Agarwal R. Significant inhibition by the flavonoid antioxidant silymarin against 12-O-tetradecanoylphorbol 13 -acetate-caused modulation of antioxidant and inflammatory enzymes and cyclooxygenase2 and interleukin-I alpha expression in SENCAR mouse epidermis: implications in the prevention of stage 1 tumor promotion. Mol Carciriog. 26(4), 321-33 (1999).

Figure 106126482-A0304-11-02
Figure 106126482-A0304-11-02

Claims (29)

一種式I之巴豆酯或其醫藥上可接受之鹽、異構體或對映異構體之用途,
Figure 106126482-A0305-02-0055-2
其中 R1及R2選自氫、羥基、
Figure 106126482-A0305-02-0055-6
Figure 106126482-A0305-02-0055-32
Figure 106126482-A0305-02-0055-8
Figure 106126482-A0305-02-0055-9
組成之群組;及 R3為氫或
Figure 106126482-A0305-02-0055-5
,其中該烷基、烯基、苯基及苄基係未經 取代或經鹵素、硝基或胺基取代,其係用於製造用以於哺乳動物個體中降低一或多種中風影響發生或復發之藥物;其中該式I之巴豆酯係以有效量存在;及其中該一或多種中風影響為癱瘓、空間障礙、判斷力降低、忽視症、記憶力減退、失語、協調及平衡困難、噁心、嘔吐、認知功能障礙、知覺障礙、定向障礙、單側偏盲、行動障礙或衝動控制異常。
A use of crotonate of formula I or a pharmaceutically acceptable salt, isomer or enantiomer thereof,
Figure 106126482-A0305-02-0055-2
Where R 1 and R 2 are selected from hydrogen, hydroxyl,
Figure 106126482-A0305-02-0055-6
,
Figure 106126482-A0305-02-0055-32
,
Figure 106126482-A0305-02-0055-8
and
Figure 106126482-A0305-02-0055-9
A group consisting of; and R 3 is hydrogen or
Figure 106126482-A0305-02-0055-5
, Where the alkyl, alkenyl, phenyl, and benzyl groups are unsubstituted or substituted with halogen, nitro, or amine groups, which are used to manufacture to reduce the occurrence or recurrence of one or more stroke effects in mammalian individuals Drugs; wherein the crotonate of formula I is present in an effective amount; and one or more of the stroke effects are paralysis, spatial impairment, decreased judgment, neglect, memory loss, aphasia, coordination and balance difficulties, nausea, vomiting , Cognitive dysfunction, perceptual impairment, disorientation, unilateral hemianopia, impaired mobility, or impulse control abnormalities.
如請求項1之用途,其中R1或R2
Figure 106126482-A0305-02-0055-10
,R1或R2之另一 者則為
Figure 106126482-A0305-02-0056-12
,且R3為氫。
For the purpose of claim 1, where R 1 or R 2 is
Figure 106126482-A0305-02-0055-10
, The other of R 1 or R 2 is
Figure 106126482-A0305-02-0056-12
And R 3 is hydrogen.
如請求項1之用途,其中該式I之巴豆酯為巴豆醇13-丁酸酯、巴豆醇12-癸酸、巴豆醇13-癸酸、巴豆醇12,13-二乙酸酯、巴豆醇13,20-二乙酸酯、巴豆醇12,13-二苯甲酸、巴豆醇12,13-二丁酯、巴豆醇12,13-二癸酸酯、巴豆醇12,13-二己酸酯、巴豆醇12,13-二丙酸酯、巴豆醇12-肉蔻酸酯、巴豆醇13-肉蔻酸酯、巴豆醇12,13,20-三乙酸酯、12-去氧巴豆醇13-當歸酸酯、12-去氧巴豆醇13-當歸酸20-乙酸酯、12-去氧巴豆醇13-異丁酸酯、12-去氧巴豆醇13-異丁酸酯-20-乙酸酯、12-去氧巴豆醇13-苯乙酸酯、12-去氧巴豆醇13-苯乙酸酯20-乙酸酯、12-去氧巴豆醇13-肉豆蔻酸酯、巴豆醇12-順芷酸酯13-癸酸酯、12-去氧巴豆醇13-乙酸酯、巴豆醇12-乙酸酯或巴豆醇13-乙酸酯。 The use as claimed in claim 1, wherein the crotonate of formula I is crotyl alcohol 13-butyrate, crotyl alcohol 12-decanoic acid, crotyl alcohol 13-decanoic acid, crotyl alcohol 12,13-diacetate, crotyl alcohol 13,20-diacetate, crotyl alcohol 12,13-dibenzoic acid, crotyl alcohol 12,13-dibutyl ester, crotyl alcohol 12,13-didecanoate, crotyl alcohol 12,13-dihexanoate , Crotyl alcohol 12,13-dipropionate, crotyl alcohol 12-myristate, crotyl alcohol 13-myristate, crotyl alcohol 12,13,20-triacetate, 12-deoxycrotonol 13 -Angelic acid ester, 12-deoxycrotonol 13-angelic acid 20-acetate, 12-deoxycrotonol 13-isobutyrate, 12-deoxycrotonol 13-isobutyrate-20-B Ester, 12-deoxycrotonol 13-phenylacetate, 12-deoxycrotonol 13-phenylacetate 20-acetate, 12-deoxycrotonol 13-myristate, crotyl alcohol 12 -Cisyl ester 13-decanoate, 12-deoxycrotonol 13-acetate, crotyl alcohol 12-acetate or crotyl alcohol 13-acetate. 如請求項1之用途,其中該式I之巴豆酯為12-O-十四烷醯巴豆醇-13-乙酸酯。 The use according to claim 1, wherein the crotonate of formula I is 12-O-tetradecane crotonol-13-acetate. 如請求項1之用途,其中該藥物係與至少一種次級或輔助治療劑共同投與。 The use as claimed in claim 1, wherein the drug is co-administered with at least one secondary or auxiliary therapeutic agent. 如請求項5之用途,其中在協調給藥方針中,該至少一種次級或輔助治療劑係在投與該式I之巴豆酯至該哺乳動物個體之同時、之前或之後投與至該個體。 The use according to claim 5, wherein in the coordinated administration strategy, the at least one secondary or adjuvant therapeutic agent is administered to the mammal at the same time, before or after the administration of the crotonate of formula I to the mammalian subject . 如請求項5之用途,其中該至少一種次級或輔助治療劑為組織血纖維蛋白溶酶原活化劑、抗凝血劑、他汀類(statin)藥物、貝特類藥物、血管收縮素II受體阻斷劑、血管收縮素轉換酶抑制劑、β-阻斷劑、抗血小板藥物、鈣通道阻斷劑或利尿劑。 The use as claimed in claim 5, wherein the at least one secondary or auxiliary therapeutic agent is a tissue plasminogen activator, an anticoagulant, a statin drug, a fibrate drug, angiotensin II receptor Blockers, angiotensin converting enzyme inhibitors, β-blockers, antiplatelet drugs, calcium channel blockers, or diuretics. 如請求項1之用途,其中該藥物係結合外科手術介入。 The use as claimed in claim 1, wherein the drug is combined with surgical intervention. 如請求項8之用途,其中該外科手術介入為頸動脈內膜切除術、血管成形術、支架置入術、開顱于術、置入起博器、植入除顫器、瓣膜置換、冠狀動脈搭橋術、心臟移植、彈簧圈栓塞術或卵圓孔未閉封堵。 The use according to claim 8, wherein the surgical intervention is carotid endarterectomy, angioplasty, stenting, craniotomy, pacemaker implantation, defibrillator implantation, valve replacement, coronary Arterial bypass surgery, heart transplantation, coil embolization, or oval foramen occlusion. 一種式I之巴豆酯或其醫藥上可接受之鹽、異構體或對映異構體之用途,
Figure 106126482-A0305-02-0057-13
其中 R1及R2選自氫、羥基、
Figure 106126482-A0305-02-0057-33
Figure 106126482-A0305-02-0057-34
Figure 106126482-A0305-02-0057-15
Figure 106126482-A0305-02-0057-16
組成之群組;及 R3為氫或
Figure 106126482-A0305-02-0057-20
,其中該烷基、烯基、苯基及苄基係未經 取代或經鹵素、硝基或胺基取代,其係用於製造用以於哺乳動物個體中降低一或多種中風症狀之發生或復發之藥物;其中該式I之巴豆酯係以有效量存在;及其中該包含一或多種中風症狀包括突然麻木、無力、突然神智不清、說話或理解話語困難、單眼或雙眼觀看困難、走路困難、頭暈、失去平衡或協調性、行動障礙或突然頭痛。
A use of crotonate of formula I or a pharmaceutically acceptable salt, isomer or enantiomer thereof,
Figure 106126482-A0305-02-0057-13
Where R 1 and R 2 are selected from hydrogen, hydroxyl,
Figure 106126482-A0305-02-0057-33
,
Figure 106126482-A0305-02-0057-34
,
Figure 106126482-A0305-02-0057-15
and
Figure 106126482-A0305-02-0057-16
A group consisting of; and R 3 is hydrogen or
Figure 106126482-A0305-02-0057-20
, Wherein the alkyl, alkenyl, phenyl and benzyl groups are unsubstituted or substituted with halogen, nitro or amine groups, which are used for manufacturing to reduce the occurrence of one or more stroke symptoms in mammals or Relapsed drugs; wherein the crotonate of formula I is present in an effective amount; and which contains one or more symptoms of stroke including sudden numbness, weakness, sudden unconsciousness, difficulty speaking or understanding words, difficulty viewing with one or both eyes, Difficulty walking, dizziness, loss of balance or coordination, impaired mobility, or sudden headache.
如請求項10之用途,其中R1或R2
Figure 106126482-A0305-02-0058-22
,R1或R2之另 一者則為
Figure 106126482-A0305-02-0058-21
,且R3為氫。
For the purpose of claim 10, where R 1 or R 2 is
Figure 106126482-A0305-02-0058-22
, The other of R 1 or R 2 is
Figure 106126482-A0305-02-0058-21
And R 3 is hydrogen.
如請求項10之用途,其中該式I之巴豆酯為巴豆醇13-丁酸酯、巴豆醇12-癸酸、巴豆醇13-癸酸、巴豆醇12,13-二乙酸酯、巴豆醇13,20-二乙酸酯、巴豆醇12,13-二苯甲酸、巴豆醇12,13-二丁酯、巴豆醇12,13-二癸酸酯、巴豆醇12,13-二己酸酯、巴豆醇12,13-二丙酸酯、巴豆醇12-肉蔻酸酯、巴豆醇13-肉蔻酸酯、巴豆醇12,13,20-三乙酸酯、12-去氧巴豆醇13-當歸酸酯、12-去氧巴豆醇13-當歸酸20-乙酸酯、12-去氧巴豆醇13-異丁酸酯、12-去氧巴豆醇13-異丁酸酯-20-乙酸酯、12-去氧巴豆醇13-苯乙酸酯、12-去氧巴豆醇13-苯乙酸酯20-乙酸酯、12-去氧巴豆醇13-肉豆蔻酸酯、巴豆醇12-順芷酸酯13-癸酸酯、12-去氧巴豆醇13-乙酸酯、巴豆醇12-乙酸酯或巴豆醇13-乙酸酯。 The use according to claim 10, wherein the crotonate of formula I is crotyl alcohol 13-butyrate, crotyl alcohol 12-decanoic acid, crotyl alcohol 13-decanoic acid, crotyl alcohol 12,13-diacetate, crotyl alcohol 13,20-diacetate, crotyl alcohol 12,13-dibenzoic acid, crotyl alcohol 12,13-dibutyl ester, crotyl alcohol 12,13-didecanoate, crotyl alcohol 12,13-dihexanoate , Crotyl alcohol 12,13-dipropionate, crotyl alcohol 12-myristate, crotyl alcohol 13-myristate, crotyl alcohol 12,13,20-triacetate, 12-deoxycrotonol 13 -Angelic acid ester, 12-deoxycrotonol 13-angelic acid 20-acetate, 12-deoxycrotonol 13-isobutyrate, 12-deoxycrotonol 13-isobutyrate-20-B Ester, 12-deoxycrotonol 13-phenylacetate, 12-deoxycrotonol 13-phenylacetate 20-acetate, 12-deoxycrotonol 13-myristate, crotyl alcohol 12 -Cisyl ester 13-decanoate, 12-deoxycrotonol 13-acetate, crotyl alcohol 12-acetate or crotyl alcohol 13-acetate. 如請求項10之用途,其中該式I之巴豆酯為12-O-十四烷醯巴豆醇-13- 乙酸酯。 The use according to claim 10, wherein the crotonate of formula I is 12-O-tetradecane crotonol-13- Acetate. 如請求項10之用途,其中該藥物係與至少一種次級或輔助治療劑共同投與。 The use as claimed in claim 10, wherein the drug is co-administered with at least one secondary or auxiliary therapeutic agent. 如請求項10之用途,其中該至少一種次級或輔助治療劑係在投與該式I之巴豆酯至該哺乳動物個體之同時、之前或之後投與。 Use according to claim 10, wherein the at least one secondary or adjunct therapeutic agent is administered at the same time, before or after the administration of the crotonate of formula I to the mammalian subject. 如請求項14之用途,其中該至少一種次級或輔助治療劑為抗凝血劑、他汀類藥物、貝特類藥物、血管收縮素II受體阻斷劑、抗血小板藥物、血管收縮素轉換酶抑制劑、β-阻斷劑、鈣通道阻斷劑或利尿劑。 The use according to claim 14, wherein the at least one secondary or adjuvant therapeutic agent is an anticoagulant, statin, fibrate, angiotensin II receptor blocker, antiplatelet drug, angiotensin conversion Enzyme inhibitors, β-blockers, calcium channel blockers or diuretics. 如請求項10之用途,其中該藥物係結合外科手術介入。 The use according to claim 10, wherein the drug is combined with surgical intervention. 如請求項17之用途,其中該外科手術介入為頸動脈內膜切除術、血管成形術、支架置入術、開顱于術、置入起博器、植入除顫器、瓣膜置換、冠狀動脈搭橋術、心臟移植、彈簧圈栓塞術或卵圓孔未閉封堵。 The use as claimed in claim 17, wherein the surgical intervention is carotid endarterectomy, angioplasty, stenting, craniotomy, pacemaker implantation, defibrillator implantation, valve replacement, coronary Arterial bypass surgery, heart transplantation, coil embolization, or oval foramen occlusion. 一種式I之巴豆酯或其醫藥上可接受之鹽、異構體或對映異構體之用途,
Figure 106126482-A0305-02-0060-14
其中 R1及R2選自氫、羥基、
Figure 106126482-A0305-02-0060-28
Figure 106126482-A0305-02-0060-35
Figure 106126482-A0305-02-0060-24
Figure 106126482-A0305-02-0060-26
組成之群組;及 R3為氫或
Figure 106126482-A0305-02-0060-25
,其中該烷基、烯基、苯基及苄基係未經 取代或經鹵素、硝基或胺基取代,其係用於製造用以於哺乳動物個體中治療中風後遺症之藥物;其中該式I之巴豆酯係以有效量存在。
A use of crotonate of formula I or a pharmaceutically acceptable salt, isomer or enantiomer thereof,
Figure 106126482-A0305-02-0060-14
Where R 1 and R 2 are selected from hydrogen, hydroxyl,
Figure 106126482-A0305-02-0060-28
,
Figure 106126482-A0305-02-0060-35
,
Figure 106126482-A0305-02-0060-24
and
Figure 106126482-A0305-02-0060-26
A group consisting of; and R 3 is hydrogen or
Figure 106126482-A0305-02-0060-25
, Wherein the alkyl, alkenyl, phenyl, and benzyl groups are unsubstituted or substituted with halogen, nitro, or amine groups, which are used in the manufacture of a medicament for treating stroke sequelae in mammals; wherein the formula The crotonate of I is present in an effective amount.
如請求項19之用途,其中R1或R2
Figure 106126482-A0305-02-0060-30
,R1或R2之另 一者則為
Figure 106126482-A0305-02-0060-31
,且R3為氫。
For the purpose of claim 19, where R 1 or R 2 is
Figure 106126482-A0305-02-0060-30
, The other of R 1 or R 2 is
Figure 106126482-A0305-02-0060-31
And R 3 is hydrogen.
如請求項19之用途,其中該式I之巴豆酯為巴豆醇13-丁酸酯、巴豆醇12-癸酸、巴豆醇13-癸酸、巴豆醇12,13-二乙酸酯、巴豆醇13,20-二乙酸酯、巴豆醇12,13-二苯甲酸、巴豆醇12,13-二丁酯、巴豆醇12,13-二癸酸酯、巴豆醇12,13-二己酸酯、巴豆醇12,13-二丙酸酯、巴豆醇12-肉蔻酸酯、巴豆醇13-肉蔻酸酯、巴豆醇12,13,20-三乙酸酯、12-去氧巴豆醇13-當歸酸酯、12-去氧巴豆醇13-當歸酸20-乙酸酯、12-去氧巴豆醇13-異丁 酸酯、12-去氧巴豆醇13-異丁酸酯-20-乙酸酯、12-去氧巴豆醇13-苯乙酸酯、12-去氧巴豆醇13-苯乙酸酯20-乙酸酯、12-去氧巴豆醇13-肉豆蔻酸酯、巴豆醇12-順芷酸酯13-癸酸酯、12-去氧巴豆醇13-乙酸酯、巴豆醇12-乙酸酯或巴豆醇13-乙酸酯。 The use according to claim 19, wherein the crotonate of formula I is crotyl alcohol 13-butyrate, crotyl alcohol 12-decanoic acid, crotyl alcohol 13-decanoic acid, crotyl alcohol 12,13-diacetate, crotyl alcohol 13,20-diacetate, crotyl alcohol 12,13-dibenzoic acid, crotyl alcohol 12,13-dibutyl ester, crotyl alcohol 12,13-didecanoate, crotyl alcohol 12,13-dihexanoate , Crotyl alcohol 12,13-dipropionate, crotyl alcohol 12-myristate, crotyl alcohol 13-myristate, crotyl alcohol 12,13,20-triacetate, 12-deoxycrotonol 13 -Angelic acid ester, 12-deoxycrotonol 13-Angelic acid 20-acetate, 12-deoxycrotonol 13-isobutyl Ester, 12-deoxycrotonol 13-isobutyrate-20-acetate, 12-deoxycrotonol 13-phenylacetate, 12-deoxycrotonol 13-phenylacetate 20-B Ester, 12-deoxycrotonol 13-myristate, crotyl alcohol 12-cis-enanthate 13-decanoate, 12-deoxycrotonol 13-acetate, crotyl alcohol 12-acetate or Crotyl alcohol 13-acetate. 如請求項19之用途,其中該式I之巴豆酯為12-O-十四烷醯巴豆醇-13-乙酸酯。 The use according to claim 19, wherein the crotonate of formula I is 12-O-tetradecane crotonol-13-acetate. 如請求項19之用途,其中該藥物係與至少一種次級或輔助治療劑共同投與。 The use according to claim 19, wherein the drug is co-administered with at least one secondary or auxiliary therapeutic agent. 如請求項23之用途,其中在協調給藥方針中,該至少一種次級或輔助治療劑係在投與該式I之巴豆酯至該哺乳動物個體之同時、之前或之後投與至該個體。 The use according to claim 23, wherein in the coordinated dosing regimen, the at least one secondary or adjunct therapeutic agent is administered to the mammal at the same time, before, or after the administration of the crotonate of formula I to the mammalian subject . 如請求項24之用途,其中該至少一種次級或輔助治療劑為組織血纖維蛋白溶酶原活化劑、抗凝血劑、他汀類(statin)藥物、貝特類藥物、血管收縮素II受體阻斷劑、血管收縮素轉換酶抑制劑、β-阻斷劑、抗血小板藥物、鈣通道阻斷劑或利尿劑。 The use according to claim 24, wherein the at least one secondary or auxiliary therapeutic agent is a tissue plasminogen activator, an anticoagulant, a statin drug, a fibrate drug, angiotensin II receptor Blockers, angiotensin converting enzyme inhibitors, β-blockers, antiplatelet drugs, calcium channel blockers, or diuretics. 如請求項19之用途,其中該藥物係結合外科手術介入。 The use as claimed in claim 19, wherein the drug is combined with surgical intervention. 如請求項26之用途,其中該外科手術介入為頸動脈內膜切除術、血 管成形術、支架置入術、開顱于術、置入起博器、植入除顫器、瓣膜置換、冠狀動脈搭橋術、心臟移植、彈簧圈栓塞術或卵圓孔未閉封堵。 The use according to claim 26, wherein the surgical intervention is carotid endarterectomy, blood Tube angioplasty, stenting, craniotomy, pacemaker implantation, defibrillator implantation, valve replacement, coronary artery bypass grafting, heart transplantation, coil embolization, or oval foramen occlusion. 如請求項1、10或19之用途,其中該式I之巴豆酯之有效量係約10至約1500μg。 The use according to claim 1, 10 or 19, wherein the effective amount of the crotonate of formula I is about 10 to about 1500 μg. 如請求項28之用途,其中該藥物係每日投與。 For the use of claim 28, wherein the drug is administered daily.
TW106126482A 2014-01-10 2014-01-10 Compositions and methods of use of phorbol esters for the treatment of stroke TWI687217B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
TW106126482A TWI687217B (en) 2014-01-10 2014-01-10 Compositions and methods of use of phorbol esters for the treatment of stroke

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
TW106126482A TWI687217B (en) 2014-01-10 2014-01-10 Compositions and methods of use of phorbol esters for the treatment of stroke

Publications (2)

Publication Number Publication Date
TW201735915A TW201735915A (en) 2017-10-16
TWI687217B true TWI687217B (en) 2020-03-11

Family

ID=61021702

Family Applications (1)

Application Number Title Priority Date Filing Date
TW106126482A TWI687217B (en) 2014-01-10 2014-01-10 Compositions and methods of use of phorbol esters for the treatment of stroke

Country Status (1)

Country Link
TW (1) TWI687217B (en)

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050250719A1 (en) * 2002-09-24 2005-11-10 Phenos Gmbh Inhibition of protein kinase c alpha for treatment of diabetes mellitus and cardiovascular diseases
TW201817421A (en) * 2012-01-18 2018-05-16 華鴻新藥股份有限公司 Compositions and methods of use of phorbol esters for the treatment of stroke

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050250719A1 (en) * 2002-09-24 2005-11-10 Phenos Gmbh Inhibition of protein kinase c alpha for treatment of diabetes mellitus and cardiovascular diseases
TW201817421A (en) * 2012-01-18 2018-05-16 華鴻新藥股份有限公司 Compositions and methods of use of phorbol esters for the treatment of stroke

Also Published As

Publication number Publication date
TW201735915A (en) 2017-10-16

Similar Documents

Publication Publication Date Title
TWI666015B (en) Compositions and methods of use of phorbol esters for the treatment of stroke
TWI687217B (en) Compositions and methods of use of phorbol esters for the treatment of stroke
TWI750481B (en) Compositions and methods of use of phorbol esters for the treatment of stroke
US10849871B2 (en) Compositions and methods of use of phorbol esters for the treatment of stroke
US10143672B2 (en) Compositions and methods of use of phorbol esters for the treatment of stroke
US10369222B2 (en) Compositions and methods of use of phorbol esters for the treatment of stroke
TWI619494B (en) Compositions and methods of use of phorbol esters
RU2775972C2 (en) Compositions and methods for using phorbol ethers for treatment of stroke