用於增強BAX/BCL-2表現及活性之化合物及其治療用途Compounds for enhancing the performance and activity of BAX/BCL-2 and their therapeutic uses
本發明提供增強BAX/BCL-2之表現及活性之方法及相關治療用途。The present invention provides methods for enhancing the performance and activity of BAX/BCL-2 and related therapeutic uses.
BCL-2蛋白家族決定了細胞發生凋亡(一種發育、組織恆定及免疫力所必需之古老的細胞自殺程序)之承諾。該等蛋白質管理粒腺體外膜滲透性(MOMP)且可為促細胞凋亡(尤其Bax、BAD、Bak及Bok)或抗細胞凋亡的(在各種其他蛋白質中,尤其包括固有Bcl-2、Bcl-xL及Bcl-w)。迄今為止已知之Bcl-2家族中存在總共25個基因。已發現人類BCL-2係在濾泡性淋巴瘤患者之腫瘤細胞中位於染色體18及14 (t14;18)異常連接之接合點附近之基因。此染色體易位造成正常BCL-2表現模式之誤調控而導致癌症(Tsujimoto等人,1985;Nunez等人,1989)。 各種生理死亡信號以及病理細胞傷害觸發細胞凋亡之遺傳程式化路徑(Vaux及Korsmeyer 1999)。細胞凋亡體現在死亡信號下游之兩個主要執行程式中:凋亡蛋白酶路徑及細胞器功能障礙,其中線粒體功能障礙係最明顯特徵(關於評論,參見Green及Reed 1998;Thornberry及Lazebnik 1998)。由於BCL-2家族成員駐留於不可逆細胞損傷之上游且其許多努力著重於線粒體之含量,故其在決定細胞將存活還是死亡方面起到關鍵作用。 關於線粒體參與癌症之最近研究已揭露在比較轉移性線粒體與屬未轉化細胞之線粒體時,該等細胞器之結構及功能存在大量差異。特別地,現代研究在很大程度上支持了瓦博格(Warburg)及其繼任者之新陳代謝觀察結果,同時精煉並極大地擴展了線粒體狀態及在腫瘤發展中之功能之機制知識的寬度。瓦博格效應之綜合機制尚未被孤立;然而,最近已描述多個交織病因及反應機制。癌細胞新陳代謝之指示性特徵的此認識亦直接應用於當前臨床護理,其藉助日益廣泛採用之正電子發射斷層攝影術(PET)成像使用葡萄糖類似物來鑑別以高葡萄糖攝取為特徵之癌病變(BioMed Research International 第2013卷(2013), 文章ID 612369)。 細胞凋亡可經由兩種路徑(即,外在及內在路徑)起始。然而,外在路徑係藉助細胞表面死亡受體之刺激起始,內在路徑係藉由許多不同的細胞內壓力活化且係由大多數化學療法藥劑引起。BCL-2家族蛋白主要涉及內在路徑,其中其調控粒腺體外膜滲透性(MOMP)。MOMP導致細胞凋亡觸發因子(例如細胞色素c)自線粒體膜間腔釋放至細胞質中,其中細胞凋亡觸發因子活化執行普遍蛋白水解事件之凋亡蛋白酶之級聯,導致細胞死亡(3)。MOMP標誌著死亡的承諾且通常視為細胞之不可逆點。此處,評論了最近藉由利用BCL-2家族蛋白作為抗癌療法引起MOMP之努力的合理及初步結果並討論用於藥物開發之其他相關靶標(Clin Cancer Res;21(12), 2015年6月15日)。 隨後闡述Bcl-2於前列腺癌、乳癌及結腸癌、以及神經膠質母細胞瘤中之過表現。Mcl-1 (另一抗凋亡Bcl-2相關蛋白)之過表現已在復發急性骨髓性白血病中鑑別出,且與差的預後相關。癌細胞中所鑑別之Bcl-2相關蛋白表現之其他變化包括Bax基因中之不同突變及促凋亡與抗凋亡Bcl-2蛋白比率之變化(Bettaieb等人,2003;及Oncogene (2004) 23, 2934-2949)。 Bcl-2及相關抗凋亡蛋白之過表現已證實抑制由許多刺激(包括生長因子剝奪、缺氧及氧化壓力)誘導之細胞死亡。然而,抗凋亡Bcl-2家族蛋白阻抑由細胞毒性抗癌藥物誘導之細胞死亡的能力使得該等蛋白質特別有趣的作為癌症藥物發現之潛在靶標。不管作用之主要模式如何,無論係單鏈還是雙鏈DNA斷裂,無論微管解聚還是聚集,無論核激素受體活化(醣皮質激素受體)還是抑制(雌激素及雄激素受體),基本上所有傳統抗癌藥物似乎在很大程度上依賴於Bcl-2/Bax依賴機制來殺死癌細胞(由Debatin等人,2002;Reed, 2008評論)。因此,Bcl-2係在大多數抗癌藥物所利用之保守細胞死亡路徑中之遠端點處操作,此構成與先前所鑑別涉及藥物排出、藥物新陳代謝、藥物失活之機制及相關機制不同的內在化學抗性形式。此觀察結果大概解釋了為何各種Bcl-2家族蛋白之表現顯示對於由化學療法治療之許多類型的癌症及白血病具有預後意義。 BCL-2家族之促凋亡成員之表現缺陷亦在癌症中發現,此導致該等殺手基因之腫瘤阻抑功能損失。所記錄之最佳者係BAX,其中已特定地在伴隨因缺陷性DNA錯配修復所致之微衛星不穩定性發生之癌症中鑑別出純合性缺失或失活性突變。就此而言,人類BAX基因在有義鏈中含有八個鳥嘌呤核苷殘基之同源聚物延伸,此延伸係框移突變之靶標。促凋亡BCL-2家族基因之缺陷性表現亦在p53功能損失之背景中發現。其中,p53轉錄因子之直接靶標係BAX、BID、PUMA及NOXA,由此證實p53之基因組監督與BCL-2家族之細胞死亡基因之間存在強烈關係。最近,已觀察到p53蛋白與促凋亡及抗凋亡Bcl-2家族蛋白之胞質相互作用,其直接調節p21-Bax及p26-Bcl-2蛋白之生物活性,且表明p53在轉錄及轉錄後兩個層面上調控Bcl-2家族。Bcl-2家族之其他促凋亡成員的活性亦藉助翻譯後修飾受到阻抑。舉例而言,促凋亡蛋白BAD藉由Akt (PKB)及其他已知在癌症中高度活躍之蛋白激酶磷酸化,此導致其由14-3-3隔離(Oncogene (2008) 27, 6398-6406)。 Bcl-2之過表現阻止由大多數化學治療藥物(包括烷基化劑及拓樸異構酶抑制劑)誘導之細胞凋亡。而且,在Bax及Bak二者中有缺陷之成纖維細胞對由誘發線粒體壓力之各種藥劑誘導之細胞凋亡具有抗性。然而,具有Bax或Bak之單一缺陷之成纖維細胞對於由該等藥劑誘導之細胞凋亡並無顯著缺陷,此表明Bax及Bak蛋白二者可係功能上冗餘的。此明顯冗餘可係細胞類型依賴性的,此乃因Bax缺失之HCT116細胞對星孢菌素(staurosporine)誘導之細胞凋亡具有抗性(Theodorakis等人,2002)。許多抗癌藥劑(例如砷及氯尼達明(lonidamine))至少在活體外可直接滲透化線粒體之外膜,且Bcl-2之過表現阻止砷及氯尼達明誘導之線粒體滲透化及細胞凋亡。基於Bcl-2蛋白在調控線粒體膜滲透化中之作用及其在人類癌症中通常經修改之表現,Bcl-2蛋白係藉由自身或與其他抗癌藥物組合用於誘導細胞凋亡之合理靶標(抗凋亡成員)或原型(促凋亡成員)。然而,在使用Bcl-2靶向療法時應使用分子標記,此乃因並非所有抗細胞凋亡細胞均過表現Bcl-2,如在來自NCI細胞篩選之乳癌細胞系中所證實(Nieves-Neira及Pommier, 1999;及Oncogene (2004) 23, 2934-2949)。 Bcl-2蛋白係程式化細胞死亡之阻抑劑,其與自身同源二聚且與同源蛋白Bax (細胞死亡之促進劑)形成異二聚體。Bax/Bcl-2比率可作為決定細胞對細胞凋亡之敏感性的變阻器。一些Bcl-2家族成員之次細胞定位亦似乎調控其功能。Bcl-2及Bcl-xL之一小部分可發現於線粒體外膜上。與此相比,Bax在細胞凋亡誘導之前主要存在於胞質液中。在細胞凋亡早期期間,Bax自胞質液易位至線粒體,在其中其參與線粒體分裂及細胞色素c之釋放。調節插入步驟可調控細胞凋亡。此比率之較低位準可導致人類癌細胞對細胞凋亡之抗性。因此,Bax/Bcl-2比率可影響腫瘤進展及侵襲性(Iran Biomed J. 2015年4月;19(2): 69-75)。 如同在外在路徑中一樣,靶向涉及腫瘤發生及化學抗性二者之內在路徑的媒介用於治療途徑。該等抗癌症策略試圖開發通常在癌細胞中過表現之抗細胞凋亡蛋白(例如,Bcl-2、Bcl-xL及IAP)之藥物經設計抑制劑。關於靶向Bcl-2蛋白之努力涉及開發破壞Bcl-2複合物之藥劑。BH3模擬物結合至抗凋亡蛋白之疏水性溝槽,此模擬了僅BH3之蛋白質在結合促存活蛋白中之作用,導致自複合物釋放僅BH3之蛋白質並活化BAX及BAK。到目前為止,有將近十二種BH3模擬物作為Bcl-2抑制劑在人類臨床試驗之不同階段中進行研究,例如AT-101 (R-(−)-棉子酚)、ABT-199 (維奈托克(venetoclax))、ABT-737、ABT-263 (納維托克(navitoclax),ABT-737之口服可用衍生物)、GX15-070 (奧博托克(obatoclax))及TW37。Bcl-2家族成員抑制劑之領域處於持續發展中,此強調了該等分子作為強效抗癌症藥劑之重要性。而且,靶向Bcl-2之特定BH4結構域亦正形成為抗癌療法之新穎策略。因此,Bcl-2經由其BH4結構域與若干調控涉及腫瘤進展及化學抗性之不同細胞路徑(例如缺氧及血管生成)之蛋白質合作。最近,發現小分子(即BDA-366)可作為強力且有效BH4結構域拮抗劑,其在活體外及活體內展現顯著抗癌活性,由此提供此途徑之概念驗證。抑制Bcl-2之另一創新途徑源於以下證據:人類bcl-2基因含有位於其啟動子中具有形成G四股結構之可能性之富含GC之序列且起到轉錄抑制子元件的作用。因此,G四股特異性配體可藉助四股結構之穩定化調控bcl-2之轉錄(AGING, 2016年4月, 第8卷第4期)。 Bcl-XL係Bcl-2家族中之主要抗細胞凋亡蛋白,其過表現較Bcl-2更廣泛地在人類肺癌細胞中觀察到,此表明Bcl-XL更生物上相關且因此係肺癌之更佳治療靶標。此處,使用UCSF DOCK 6.1程式套件及NCI化學文庫數據庫篩選選擇性靶向Bcl-XL之BH3結構域(aa 90-98)結合袋之小分子。針對肺癌細胞展示選擇性毒性之兩種新Bcl-XL抑制劑(BXI-61及BXI-72)與正常人類支氣管上皮細胞相當。螢光偏振分析揭露,BXI-61及BXI-72在活體外以高結合親和力優先結合至Bcl-XL蛋白而非Bcl2、Bcl-w、Bfl-1/A1或Mcl-1。利用BXI-72處理細胞導致破壞Bcl-XL/Bak或Bcl-XL/Bax相互作用,使得Bak寡聚且細胞色素c自線粒體釋放。重要地,BXI-61及BXI-72展示較ABT-737更強效之對抗人類肺癌功效,且在活體內血小板減少較少。BXI-72克服了肺癌獲得性輻射抗性。基於該等發現,開發BXI作為一類新的抗癌藥劑獲得保證且代表改良肺癌結果之新策略(Cancer Res. 2013年9月1日;73(17):5485-5496)。 在細胞凋亡之決策階段需要Bax (細胞死亡調控劑)。最近研究將Bax之絲胺酸184 (S184)鑑別為控制其促凋亡活性的關鍵功能開關。此處,使用圍繞S184之結構袋作為停泊位點以使用UCSF-DOCK程式套件篩選小分子之NCI文庫。三種化合物(即,小分子Bax激動劑SMBA1、SMBA2及SMBA3)藉由阻斷S184磷酸化誘導Bax中之構型改變,此促進Bax插入線粒體膜中並形成Bax寡聚物。後者導致細胞色素c釋放及人類肺癌細胞中之細胞凋亡,此係以Bax而非Bak依賴性方式發生。SMBA1經由細胞凋亡藉由在活體內選擇性活化Bax而強效阻抑肺腫瘤生長,而無顯著正常組織毒性。開發Bax激動劑作為一類新抗癌藥物提供治療肺癌及其他表現Bax之惡性腫瘤之策略(NATURE COMMUNICATIONS DOI: 10.1038/ncomms5935)。 BID (BH3相互作用結構域死亡受體激動劑)蛋白位於死亡受體與線粒體之間之外在凋亡信號傳導中,且充當II型細胞中線粒體外膜之滲透化的誘導子。BID之含量對於各種細胞之存活力至關重要,此乃因其沉默使得細胞抵抗由死亡受體配體(例如TNF相關之細胞凋亡誘導配體(TRAIL))誘導之細胞凋亡。而且,已證實BID之含量在若干細胞系之細胞中低於功能劑量,此乃因其可藉由BID之過表現而對TRAIL敏感。由於上述原因,已考慮將BID用於治療劑開發。然而,為界定投與BID之途徑,應解決若干重大問題。主要問題係控制遞送至細胞之BID之量。儘管全長BID已顯示參與細胞凋亡信號傳導,但細胞凋亡之有效活化需要藉由凋亡蛋白酶8特異性裂解BID並產生活性截短形式(tBID)。於細胞中表現之tBID直接誘導細胞凋亡。因此,為利用一些抗癌藥劑所展現針對癌細胞之選擇性,藉由全長BID使細胞敏感化係較佳策略。為使細胞敏感化,通常使用基於腺病毒或pcDNA載體之過表現系統。然而,其並不提供細胞中BID表現含量之嚴格控制。因此,BID在轉染細胞中之含量超過內源性蛋白質含量的數倍且在一些情形中觀察到細胞凋亡之直接活化而非細胞對細胞凋亡刺激之敏感化。根據上述,在利用表現全長BID之腺病毒載體處理之細胞中出現tBID (BMC Cancer201414:771 DOI: 10.1186/1471-2407-14-771)。 雷帕黴素(Rapamycin)及其衍生物係具有免疫阻抑劑及抗腫瘤性質二者之有前景治療劑。該等雷帕黴素的作用係藉助mTOR蛋白激酶之特定抑制來調介。已知mTOR充當響應於改變營養物含量來控制細胞週期之進化上保守之信號傳導路徑之一部分。mTOR信號傳導網絡含有許多腫瘤抑制基因(包括PTEN、LKB1、TSC1及TSC2)及許多原致癌基因(包括PI3K、Akt及eIF4E),且mTOR信號傳導在許多腫瘤類型中經組成性活化。該等觀察結果表明mTOR係抗癌藥劑之理想靶標且表明雷帕黴素係此一藥劑。實際上,早期臨床前及臨床研究指示雷帕黴素衍生物作為抗腫瘤藥劑在單獨及與其他療法模式組合時具有功效。雷帕黴素看起來藉由停止腫瘤細胞增殖、誘導腫瘤細胞凋亡及阻抑腫瘤血管生成抑制腫瘤生長。雷帕黴素免疫阻抑劑的作用係由藉助與雷帕黴素阻斷癌細胞增殖之相同機制抑制T及B細胞增殖引起。因此,可能會認為雷帕黴素誘導之免疫阻抑對於使用雷帕黴素作為抗癌藥劑將係有害的。與此相反,雷帕黴素當與廣泛使用之免疫阻抑劑環孢黴素(cyclosporine)組合時,與環孢黴素單獨使用時所觀察到之腫瘤發生率相比,在器官移植試驗中發生之腫瘤形成之頻率降低。現有證據指示,關於腫瘤生長,雷帕黴素抗癌活性顯著超過雷帕黴素免疫阻抑效應。近年來,臨床上雷帕黴素及其類似物(如CCI-779或替西羅莫司(temsirolimus)、RAD001或依維莫司(everolimus)、西羅莫司、FK-50及AP23576)已用於治療各種癌症,包括腎癌、套細胞淋巴瘤及轉移性乳癌。 雙嘧達莫(Dipyridamole)(DPM) (如阿斯匹靈(aspirin))抑制血小板黏附,且因此傾向於阻止心臟病發作及中風之血管栓塞。在1987年12月12日發行之Lancet (第1,371-4頁)中有關於歐洲中風預防研究(European Stroke Prevention Study)之報導。此報導之介紹評論利用阿斯匹靈治療在小中風TIA (暫時性腦缺血發作)中存活之患者缺乏所指示益處。在此試驗中,將雙嘧達莫300 mg/天添加至利用阿斯匹靈之治療中且結果顯著。經兩年時期,中風死亡降低50%,因心肌梗塞所致之死亡降低38%且因癌症所致之死亡降低25%。上文所指示雙嘧達莫之抗癌效應可僅由於其阻止轉移;然而,巴塞羅那大學(University of Barcelona)之Eva Bestida等人已在Cancer Research, 1985年9月發行(第4,048-4,062頁)中報告某些人類癌細胞生長受雙嘧達莫之抑制。其使大於80%之腺苷、胸苷及尿苷受到抑制。該等物質係癌細胞繁榮所需的。此可指示雙嘧達莫除預防轉移以外之抗癌效應。 已出現腫瘤壞死因子相關之細胞凋亡誘導配體(TRAIL)作為選擇性靶向癌細胞之有吸引力細胞介素;然而,其功效受到許多抗性機制之挑戰。因此,目前研究調查雙嘧達莫增強TRAIL功效之可能性及可能的潛在機制。雙嘧達莫顯著使以下p53突變型人類癌細胞系對TRAIL之抗腫瘤活性敏感化:SW480、MG63及DU145,如由使TRAIL能夠有效裂解起始者及執行者凋亡蛋白酶所證實。儘管雙嘧達莫上調DR4及DR5二者且增加其細胞表面表現,但RNA干擾揭露對DR5之優先依賴性。而且,雙嘧達莫抑制存活素表現且其重要結果已由小干擾RNA證實。機制上,雙嘧達莫誘使存活素表現之轉錄關閉,伴隨以D型週期素及cdk6之下調為特徵之G1阻滯。此外,由CCAAT/增強子結合蛋白(C/EBP)同源蛋白(CHOP)誘導驅動之轉錄機制負責由雙嘧達莫所致之DR5上調。重要地,雙嘧達莫誘導之TRAIL功效之增強及蛋白質表現之改變不依賴於蛋白激酶A或蛋白激酶G。總之,本研究之發現闡述雙嘧達莫作用之新穎機制且突出其作為TRAIL功效之潛在增強劑之有前景用途(Oncogene (2008) 27, 3435-3445)。 初步觀察結果指示雙嘧達莫(DPM)可增加人類卵巢癌細胞對順鉑(cisplatin)之敏感性。在另一研究中,Jekunen等人顯示DPM以協同方式使順鉑在順鉑敏感性2008人類卵巢癌細胞中之細胞毒性增強至4.7倍,且在順鉑抗性2008/C13*5.25亞系中增強至5.8倍。已發現,DPM以濃度依賴性方式增加順鉑之細胞攝取,而不增加台盼藍(trypan blue)或碘化丙啶攝取或改變細胞大小。其認為DPM誘導之順鉑累積增加與膜滲透性之非特異性增加不相關。在具有人類膀胱癌之裸小鼠模型中,Keane等人發現當順鉑與DPM組合時腫瘤大小減小20%。在相同模型中使用人類睪丸癌,其達成完全腫瘤消退。Barberi-Heyob等人發現DPM在MCF-7人類乳癌細胞以協同方式增加順鉑之生長抑制活性。Janice R. Perussi等人在其中報告在順鉑敏感性MDA/S人類乳癌細胞中順鉑細胞毒性因DPM增強,此表明在Pt累積與增強之順鉑細胞毒性之間存在強關聯,但在順鉑抗性MDA/R細胞中,順鉑與DPM之間之協同相互作用並不涉及順鉑之細胞累積的增加(Quím. Nova 第26卷第3期 São Paulo 2003年5月/6月)。 吉西他濱(Gemcitabine)(2’,2’-二氟去氧胞苷)係嘧啶核苷類似物。其在細胞內發揮其細胞毒性效應且具有對抗許多不同實體腫瘤(包括胰臟癌、乳癌、肺癌及膀胱癌)之活性。由於吉西他濱具有強親水性,故穿過疏水性胞漿膜脂質雙層之被動擴散較緩慢。為有效進入細胞,吉西他濱需要生理核苷轉運蛋白以跨越漿膜。該等轉運蛋白分為兩類,即平衡型轉運蛋白及濃縮型轉運蛋白。雙向人類平衡型核苷轉運蛋白(hENT)發現於大多數細胞類型中,且hENT1及hENT2二者能夠在濃度梯度方向上調介吉西他濱攝取。hENT蛋白係定位於漿膜之跨膜醣蛋白。其藉由其能力受硝基苄基巰嘌呤核糖核苷(NBMPR)(在低毫微莫耳濃度下之特異性hENT1抑制劑)及雙嘧達莫(hENT1/2抑制劑)之抑制在功能上區分,其中對吉西他濱之敏感性降低至1/39及1/1,800 (The Oncologist 2008;13:261-276)。 氟尿嘧啶(Fluorouracil)(5-FU) (尤其以商品名Adrucil出售者)係用於治療癌症之醫藥。藉由將其注射於靜脈中,其用於結腸癌、食管癌、胃癌、胰臟癌、乳癌及子宮頸癌。作為乳膏,其用於日光性角化症及基底細胞癌,且作為滴眼液用於治療眼表面鱗狀細胞瘤。 細胞凋亡之缺陷已涉及癌細胞之化學抗性。研究已發現,高含量之抗細胞凋亡Bcl-2與低含量Bax之組合與各種癌症中之高5-FU抗性相關,包括人類乳腺癌、胰臟癌、人類頭頸癌、結腸腫瘤細胞(Int J Cancer. 2002年4月1日;98(4):498-504;及Br J Cancer. 2000年11月;83(10): 1380-1386)。 雙嘧達莫已顯示在活體外加強5-FU及其他氟嘧啶之細胞毒性。此效應可能藉由「補救(salvage)」路徑與FdUMP細胞內濃度之增加及細胞外核苷攝取之抑制有關。活體外研究已顯示雙嘧達莫大概藉由改變細胞毒性劑之細胞攝取及保留可增大許多細胞毒性劑(包括依託泊苷(etoposide)、多柔比星(doxorubicin)、長春花鹼(vinblastine)及米托蒽醌(mitoxantrone))之細胞毒性(Investigational New Drugs 12: 283-287, 1994)。 癌細胞之多重耐藥性(MDR)係對各種看起來結構上且機制上不相關之抗癌藥物之抗性的同時發展。一種類型之MDR的特徵在於疏水性天然產物藥物之累積減少。在一些多重耐藥性細胞中,藥物排出係由稱為P-醣蛋白(Pgp) (MDR1基因之產物)之三磷酸腺苷(ATP)依賴性膜轉運蛋白調介(Juliano及Ling, 1976)。Pgp起到各種分子(包括某些化學治療藥物)之主動向外傳輸機制的作用。隨著關於Pgp在藥物抗性中之作用的數據累積,很明顯其他轉運蛋白可賦予對細胞毒性劑之抗性。MDR蛋白1 (MRP1)基因係自多重耐藥性肺癌細胞系選殖,且發現如Pgp一樣係轉運蛋白基因之ATP結合盒(ABC)超家族的成員(Cole等人,1992)。轉染研究指示,類似於MDR1,MRP1過表現足以賦予對各種親脂性天然產物抗腫瘤藥之抗性。MRP1係首次鑑別出之編碼多特異性有機陰離子轉運蛋白(MOAT)蛋白之基因家族的成員(Borst等人,1999)。MRP1之另外兩個同系物cMOAT/MRP2及MRP3基因編碼當轉染於藥物敏感細胞中時介導MDR之蛋白(Borst等人,1999)。所有該等膜包埋蛋白質均充當藥物排出幫浦,防止細胞毒性劑在細胞內達到致死含量。 P-醣蛋白(P-gp)係癌症中多重耐藥性表型中之關鍵角色。該蛋白藉由調介驚人的一系列抗癌藥物之ATP依賴性排出賦予抗性。其寬特異性一直係許多嘗試之主題以抑制該蛋白並恢復抗癌藥物之功效。一般策略係開發與抗癌藥物競爭傳輸或充當P-gp之直接抑制劑之化合物。儘管在活體外相當成功,但當前在臨床中沒有可用於「阻斷」P-gp介導之抗性的化合物。失敗可歸因於毒性、不利的藥物相互作用及若干藥代動力學問題。此外,多重耐藥性相關蛋白1 (MRP1)傳輸寬範圍之治療劑以及各種生理受質,且可在若干癌症(包括肺癌、乳癌及前列腺癌)以及兒童神經母細胞瘤之藥物抗性之發展中起作用。以上若干研究已顯示在活體外,雙嘧達莫可顯著增加各種化學治療劑之細胞毒性及抗腫瘤活性。此處之潛在機制係阻止核苷及核鹼基補救,及經由經抑制之P-醣蛋白及MRP1增加毒性代謝產物之細胞內累積(Clin Pharmacol Ther 2003;73: 51-60. Drug Metab Dispos. 2014年4月;42(4): 623-631. Oncogene (2003) 22, 7340-7358)。 研究已在三陰性(雌激素及黃體酮受體陰性,Her-2正常)乳癌(一種幾乎沒有有效療法之亞型)之多模型中調查雙嘧達莫作為單一藥劑在預防腫瘤發生及轉移中之潛在作用。該等發現提供在乳癌異種移植動物模型中腹膜內投與雙嘧達莫損害原發性腫瘤生長及轉移之證據。而且,數據鑑別出雙嘧達莫之新作用機制,其顯示抑制ERK1/2-MAPK、NF-kB及Wnt信號傳導路徑,且阻止腫瘤微環境中在炎症細胞中之累積。此外,雙嘧達莫在所測試化合物中係最強效BCRP抑制劑,其中IC50值為6.4 +/- 0.9 mM。因此,雙嘧達莫具有治療癌症多重耐藥性之潛力(Cancer Prev Res (Phila). 2013年5月;6(5): . doi:10.1158/1940-6207.CAPR-12-0345)。 然而,臨床投與DPM並未改善5-FU或順鉑在患有晚期結腸直腸癌、轉移性乳癌、晚期非小細胞肺癌或晚期可量測胰臟癌之患者中之抗癌活性。針對DPM所觀察到之5-FU或順鉑劑量強度增加並非臨床相關的。 奈米粒子藥物載劑進入細胞之路徑不同於習用藥物。習用藥物係藉由劑量依賴性之擴散作用進入細胞。換言之,血液中之藥物濃度越高,細胞中之藥物濃度越高,且藥物僅可進入細胞質。奈米粒子藥物載劑係藉助胞吞由細胞吸收且進入細胞後係親溶酶體的。在注射後之初始階段,奈米粒子藥物載劑之濃度係以時間依賴方式增加。 胞吞係將細胞外物質併入細胞中之過程。此過程可分類為三種類型,即胞噬、胞飲及受體介導之胞吞。胞噬僅發生於特化細胞中。該等細胞在由細胞外物質刺激時增殖並聚集並將該等物質吞沒於細胞之溶酶體中以分解。此過程發生於免疫系統之巨噬細胞及嗜中性球中。胞飲係藉助細胞膜之凹入將細胞外流體及分子內在化於其內以形成袋,然後將其掐斷(pinch off)進入細胞中以形成囊泡。囊泡然後行進至胞質液中並與其他囊泡(例如核內體及溶酶體)融合。 端視載劑之結構而定,胞飲可分為兩種類型,即流體相胞飲及吸附性胞飲。若載劑不具有與細胞相互作用之官能團,則細胞將藉由流體相胞飲吞沒藥物載劑。此過程緩慢且取決於細胞膜周圍之載劑濃度。當載劑具有疏水性基團或帶正電荷時,會發生吸附性胞飲。該等載劑將由細胞膜物理吸附並增加細胞之吞沒能力。以上兩種類型之胞吞係非特異性過程且不適於將藥物遞送至其靶標。在某些癌症組織中僅可藉助增強之滲透及滯留(EPR)達成靶向。 受體介導之胞吞係細胞藉由含有具有特定針對正吸收分子之受體位點之蛋白質之漿膜囊泡向內發芽而吸收分子(胞吞)之過程。在藥物載劑結合至細胞上之受體後,內在信號將觸發細胞膜以形成包被小窩(coated pit)。包被小窩之表面積相當於細胞膜之1%至2%。包被小窩將自細胞膜分離並進入細胞以在細胞中形成包被囊泡,隨後形成核內體並在細胞內部以跳躍運動移動。核內體係包含微管及囊泡之複雜結構。囊泡可與Golgi融合。由於質子幫浦(ATPase),核內體通常變為酸性。核內體然後將與溶酶體融合以形成次級溶酶體。 細胞膜係治療劑有效遞送至線粒體、細胞質或細胞核中之靶標位點欲克服之障壁。疏水性磷脂係阻礙治療劑傳輸之細胞膜之主要組分。因此,已開發各種遞送系統(例如脂質體、奈米粒子及病毒載體)以跨越膜轉移小分子、肽、蛋白質及寡核苷酸。藥物遞送之此種方式在本文中稱為細胞穿透藥物遞送系統。 已開發許多藥物載劑系統(脂質體、細胞穿透肽、陽離子聚合物偶聯物及聚合奈米粒子)用於治療劑之細胞內遞送。其需要適於跨越一系列膜障壁以到達細胞中之藥物作用位點。在此過程期間,在每一連續障壁處將損失重要部分之藥物分子。該等障壁包括藥物載劑之細胞締合及藉由胞吞內在化;藥物或藥物載劑之細胞內運輸及釋放至細胞質中;藥物或藥物載劑由細胞質易位至細胞核或任何其他細胞器;及細胞核/細胞器攝取。細胞含有具有特定功能之若干細胞內細胞器。治療劑細胞內靶向該等特定細胞器預計不僅顯著增強治療功效,且降低非特異性效應且因此降低毒性。因此,對使用不同載劑系統達成治療劑之細胞內靶標特異性遞送有重大興趣。 促進藥物胞吞之載劑包括奈米大小之聚合物載劑及脂質體。端視藥物之性質及製造製程,奈米大小之藥物載劑可分類為奈米粒子、奈米脂質體、奈米懸浮粒子、固體脂質奈米粒子、磁性奈米載劑及諸如此類。 除上述載劑以外,亦可使用細胞穿透肽(CPP)、生物可降解奈米粒子及病毒載體作為遞送系統用於增強藥物穿透進入細胞中。 由於細胞膜構成大的大小之親水性蛋白質、肽及寡核苷酸之細胞內遞送的主要障壁,故已開發細胞穿透肽(CPP)來克服此障壁。該等CPP可擺渡(ferry)標記至其之分子或膠體藥物載劑系統跨越細胞膜,進入細胞質中並到達細胞核。CPP之特性歸因於一段9-16個陽離子胺基酸殘基之存在;最常研究之CPP包括HIV-1反式活化之轉錄活化子(TAT)肽、HSV VP-22 (單純性皰疹病毒1型轉錄因子)肽及penetratin。已提出若干理論來確定該等CPP藉由其進入細胞之確切機制。舉例而言,TAT穿透細胞膜已顯示獨立於受體及轉運蛋白,且已建議藉由使磷脂雙層不穩定來形成反微胞而進入細胞。TAT偶聯之主要益處在於連同分子之有效遞送一起,保留偶聯分子之生物活性,且所傳輸分子之大小亦非速率限制因子。 TAT已提出不僅增強細胞內遞送,亦增強細胞核遞送,且因此已研究用於核酸遞送。偶聯至反義寡核苷酸之TAT肽已顯示將寡核苷酸遞送至細胞核。內在化之後,亦發現TAT肽與BODIPY-神經醯胺(其係高爾基體(Golgi body)之標記)一起共定位於高爾基體內部。因此,很可能自早期核內體直接運輸至高爾基體而不進入晚期核內體。可存在分泌路徑,其中肽自內質網進入胞質液。基因療法已在治療遺傳性、獲得性及神經退化性病症中證實具有重大潛力。在非病毒基因遞送方法中,正研究各種藥物遞送系統及聚合物,例如脂質體、陽離子脂質-DNA、聚合物複合物。為克服非病毒基因表現載體之相對低效之細胞攝取,一直探索將TAT肽偶聯至載體。 Kleeman等人已利用藉助聚乙二醇(PEG)間隔基共價偶聯至TAT之聚乙烯亞胺(PEI)證實肺泡基底上皮細胞中之基因表現,其證實在氣管內投與後在小鼠肺中在活體內較未偶聯PEG複合物高之轉染效率。在Rudolph等人之類似研究中,偶聯至二聚HIV-1 TAT之固體脂質粒子已證實增強的對肺之基因遞送。 CPP之胺基酸組成通常含有高相對豐度之帶正電荷胺基酸(例如離胺酸或精胺酸)或具有含有極性/帶電荷胺基酸與非極性、疏水性胺基酸之交替模式之序列。該兩種類型之結構分別稱為聚陽離子或兩親性的。第三類CPP係疏水性肽,其僅含有非極性殘基、具有低淨電荷或具有對細胞攝取至關重要之疏水性胺基酸基團。在細胞穿透肽中,富含精胺酸之細胞穿透肽的研究最廣泛。實例包括來自HIV反式活化因子蛋白TAT之TAT肽、Penetratin、來自果蠅(Drosophila)之觸角足突變蛋白(Antennapedia protein)之16胺基酸結構域、獸棚病毒(flock house virus, FHV)外套肽(序列35-49)及寡精胺酸。 生物可降解奈米粒子介導之細胞內遞送係動態過程;涉及胞吞、胞洩及分選至不同細胞內隔室。看起來NP表面及其與細胞表面之相互作用控制生物可降解奈米粒子且因此經囊封治療劑之攝取及細胞內運輸。 病毒載體係分子生物學家通常用於將遺傳物質遞送至細胞中之工具。此過程可在活生物體內部(在活體內)或在細胞培養物中(在活體外)執行。因此,病毒載體係用於細胞穿透藥物遞送系統之適用選項。 細胞穿透肽及生物可降解奈米粒子不僅用於修飾藥物,且偶聯至載劑以增強跨膜效應。 雙嘧達莫係平衡型核苷轉運蛋白(ENT)抑制劑。核苷轉運蛋白(NT)在核苷跨越細胞膜之傳輸中起重要作用。雙嘧達莫阻斷平衡型核苷轉運蛋白(ENT),此促進腺苷之跨膜擴散。雙嘧達莫將主要地在腺苷之細胞外形成增加之情形中(例如發生在缺氧或炎症期間)增加細胞外內源性腺苷濃度。然而,雙嘧達莫誘導之細胞外內源性腺苷濃度造成血管舒張,此有助於器官灌注之代謝控制。雙嘧達莫壓力心肌成像係用於診斷並評估冠狀動脈疾病之廣泛應用且成功的技術。利用IV雙嘧達莫之冠狀動脈舒張與至側枝依賴性心肌之血流顯著減少相關聯,此與患有CAD之患者之冠狀動脈竊血之一致。此外,存在其他發現雙嘧達莫在許多器官(包括腎、肺、胰臟、腦等)中之血管收縮及血管舒張效應之研究。 雙嘧達莫不僅在一些器官中造成血管收縮,亦可導致低血壓及隨後副作用,例如眩暈及由於心臟血管之擴張所致之心悸。降低血壓之效應使得雙嘧達莫不適用於治療生理上不穩定之患者,例如彼等患有(但不限於)敗血症、缺血性腦中風、出血性腦中風、急性肺損傷、急性肝損傷、心肌梗塞及心腎症候群者。而且,雙嘧達莫之血流限制效應限制其在治療涉及富含血管之器官之疾病中的應用。 由於雙嘧達莫之藥理作用主要針對細胞膜,故經設計用於膜穿透以避免與膜上之平衡型核苷轉運蛋白結合同時增強細胞內信號轉導及PPARγ調控之遞送系統可預防因增加之心血管擴張及局部血流限制所致之組織低灌注效應。因此可解除在急性及嚴重患者中由於血壓之降低導致的雙嘧達莫在臨床應用中之限制。 雙嘧達莫亦係非選擇性磷酸二酯酶抑制劑。細胞內藥物遞送之增加將增強雙嘧達莫對細胞內磷酸二酯酶(PDE)之抑制。PDE家族之成員具有獨特的細胞及組織特異性分佈。端視PDE在不同組織中於細胞膜上或細胞質中之分佈曲線,雙嘧達莫可用作抗炎症、抗氧化劑及平滑肌鬆弛劑用於治療與PDE調控相關之疾病。 PDE之獨特細胞及組織特異性分佈顯示於下表中(參見US 2012/0065165):
增加雙嘧達莫穿透膜之能力可促進特定組織中PDE3、PDE5及PDE8之抑制並賦予雙嘧達莫在與PDE3、PDE5及PDE8相關之疾病中之治療功效。在此情形中,雙嘧達莫可如同其他PDE5抑制劑一樣用於治療下泌尿道功能障礙及勃起功能障礙。而且,由於雙嘧達莫係非選擇性PDE抑制劑,故當其藉由跨膜藥物遞送系統遞送時可用於治療PDE相關疾病。 存在若干當前正使用或正在臨床試驗中測試之作用於線粒體之分子。若干臨床批准之抗癌藥物(例如紫杉醇(paclitaxel)及VP-16 (依託泊苷)及長春瑞濱(vinorelbine))以及日益增多之實驗抗癌藥物(例如神經醯胺、MKT077、CD437、氯尼達明及樺木酸)已發現直接作用於線粒體以觸發細胞凋亡。CD437能夠在活體外及在活體內在各種人類癌瘤細胞中誘導細胞凋亡。在完整細胞中,CD437依賴性凋亡蛋白酶活化之前係細胞色素C自線粒體釋放。而且,當添加至經分離線粒體時,CD437引起膜滲透化。此效應藉由線粒體滲透性轉換孔複合物(mPTPC)之抑制劑(例如環孢黴素A)阻止。因此,CD437代表經由mPTPC發揮其細胞毒性效應(即,藉由直接作用於線粒體之表面或內部)之低分子量化合物。類似地,用於治療急性前骨髓性白血病之三氧化二砷對線粒體具有多重作用。已知經由其作用於電壓依賴性陰離子通道VDAC導致誘導mPTPC形成。三氧化二砷亦已知作用於呼吸鏈且抑制呼吸鏈活性。在細胞凋亡中起主要作用之凋亡因子包括Bcl-2及Bcl-Xl。藉由結合至該等蛋白質起作用之化合物已經鑑別並研究其功效;一些實例包括色烯衍生物及棉子酚,其最近顯示作用於Bcl-2家族之蛋白質。實際上,存在如此多各種且結構上不同之化合物,故建議將其統稱為線粒體靶向抗癌藥物(mitocans)以反映其線粒體介導之抗癌效應。 靶向腫瘤線粒體之選擇性累積途徑需要兩個層面之特定累積;腫瘤中之藥物累積且然後癌細胞線粒體中之藥物累積。一般而言,藥物處置可藉由化學結構之細微修改以改變其已知決定其在各個隔室中之累積之物理-化學性質來調節。當然,該等修改必須在不會不利地影響分子靶標之作用下實施。第二途徑涉及偶聯大於簡單有機官能團之配體以改變活性分子之生物分佈。再次,只要該偶聯不會不利地影響分子之期望藥理活性,此途徑即可行。使用已知對靶標組織具有親和力之配體,該等途徑已極有效地用於改變藥物在體內之分佈並在靶標組織中達成較高累積。存在已顯示調介藥物之腫瘤特異性累積之配體,且存在已知係親粒線體之配體。然而,仍不清楚是否存在具有兩種性質達到將容許高期望累積位準之程度的配體。因此可以肯定地說,至今為止,雙重策略係最可行之途徑。此一雙重策略將需要使用一種靶向遞送途徑以達成高腫瘤累積,隨後第二途徑以確保藥物然後於線粒體中累積,在線粒體中其將發揮其作用。儘管存在許多旨在增加抗癌藥物之重量含量之組織特異性遞送之研究,但旨在亞細胞遞送之研究僅獲得了較多關注。儘管如此,存在一些暗示經改良癌症療法之希望的令人關注的線粒體遞送途徑。 醫藥奈米載劑(如脂質體、微胞及固體奈米粒子)提供可視為非化學途徑之途徑以改變藥物分子之處置。所有化學過程均可針對奈米載劑系統之組分實施,該等組分然後可加載藥物以提供靶向遞送。大多數醫藥奈米載劑可經修飾用於靶向特定組織且甚至特定細胞類型。長循環脂質體及奈米粒子能夠借助增強之滲透及滯留(EPR)效應被動靶向洩漏之脈管區域且另外可利用抗體或其他靶向配體修飾以提供細胞特異性識別。若在腫瘤細胞內既影響藥物之腫瘤特異性累積亦調介線粒體特異性累積之奈米載劑可開發用於臨床療法,則其可係線粒體靶向抗癌途徑中之終極工具。近年來已邁出了此方向中之第一步。目前奈米載劑技術達到對亞細胞遞送之需求可使用針對該等目的特定設計之奈米載劑實際得到滿足之程度。 在針對將mPTPC誘導藥物黃蜂毒素(mastoparan)遞送至細胞中之途徑中,脂質體利用運鐵蛋白及基因融合肽Chol-GALA二者修飾。運鐵蛋白修飾增強脂質體經由胞吞攝取至細胞中,此後肽促進自核內體釋放至胞質液。因此,藉由僅增加藥物之細胞內含量,遞送途徑達成藥物潛在地可與亞細胞靶標相互作用之較高濃度。微胞亦已建議用於將疏水性藥物遞送至各種亞細胞細胞器,包括線粒體。發現此研究中所用之螢光素標記之微胞係藉助若干細胞質細胞器(包括其大多數與線粒體相關聯)分佈。該等微胞之攝取並不限於單一細胞類型。而且,細胞內在化貨物(cargo)併入微胞中之程度高於游離貨物自身。 目前存在若干經特定設計以在線粒體中累積之奈米載劑之實例。可能,最早之該等係稱為DQAsome者。自親粒線體分子地喹氯銨(dequalinium chloride)製得,該等囊泡奈米載劑經開發用於線粒體特異性DNA遞送,但亦顯示能夠改變紫杉醇之亞細胞分佈以增加藥物在線粒體中之累積。線粒體特異性遞送在游離藥物不具有顯著細胞毒性效應之紫杉醇濃度下導致經改良細胞凋亡活性。亦已測試加載紫杉醇之DQAsome在裸小鼠中抑制人類結腸癌腫瘤生長之能力,且數據強烈表明紫杉醇囊封於DQAsome中導致經改良之功效。經DQAsomal囊封之紫杉醇之抗腫瘤效率亦藉由利用葉酸(FA)修飾DQAsomal表面進一步改良。葉酸受體係高親和力葉酸鹽膜結合蛋白,其在多種人類腫瘤中過表現。FA偶聯物以腫瘤細胞特異性方式藉由受體介導之胞吞內在化,導致相應藥物之毒性增加。 用於線粒體特異性奈米載劑之另一途徑係利用親粒線體配體修飾現有奈米載劑。就此而言,TPP再次在基於脂質體及聚合物之奈米載劑中充當親粒線體配體。脂質體作為遞送系統已經充分表徵且由於其生物相容性、易於表面修飾及囊封親水性或疏水性藥物之能力而係受歡迎的選擇。首次指示脂質體可藉由利用親粒線體殘基表面修飾而使得親粒線體係來自關於所謂的蛋白脂質體之報導,蛋白脂質體係藉由在預植入胚胎中將粗製線粒體膜部分併入與內源性線粒體共存之脂質體中製備。使用配體改變脂質體之亞細胞分佈之概念的進一步研究係基於硬脂醯基三苯基鏻(STPP)。STPP之硬脂醯基殘基充當脂質錨以利用TPP殘基修飾脂質體之表面並產生對線粒體具有明顯傾向性之脂質體製劑。STPP-脂質體顯示在活細胞中有效引導羅丹明(rhodamine)標記之磷脂醯乙醇胺(Rh-PE)至線粒體之累積。基於流式細胞術,STPP-脂質體展現與具有相同陽離子電荷之脂質體相同之細胞締合位準。然而,藉由共焦顯微鏡分析之後續亞細胞定位顯著不同,此指示係親粒線體配體而非表面電荷決定奈米載劑之線粒體特異性締合。亦發現,TPP配體並不改變長循環聚乙二醇化脂質體之活體內分佈及腫瘤累積。然而,STPP-脂質體確實改良神經醯胺之活體外及活體內功效二者。總之,該等數據表明長循環脂質體在實體腫瘤中被動累積(經由EPR效應)之傾向可與利用適當配體修飾所賦予之細胞器特異性向性以加強經囊封抗腫瘤藥劑之效應。 開發線粒體特異性脂質體之替代途徑集中於傾向於選擇性地與線粒體膜融合之脂質體在進入細胞時更可能與線粒體締合之概念。稱為MITO-Porter之該等脂質體利用八聚精胺酸殘基經表面修飾以促進作為完整囊泡進入細胞中(經由巨胞飲)。脂質組合物係基於在活細胞中與線粒體膜之高融合位準及其貨物至內線粒體隔室之釋放來選擇。基於共焦顯微鏡數據,MITO-porter脂質體已用於將綠色螢光蛋白以及碘化丙啶遞送至線粒體,此表明其可用於將巨分子以及小分子遞送至線粒體。 線粒體特異性奈米載劑之開發並不限於基於脂質之載劑,且亦包括使用親粒線體殘基以產生能線粒體特異性細胞內遞送生物活性分子之聚合物系統。已採用TPP修飾以產生基於親粒線體N-(2-羥基丙基)甲基丙烯醯胺(HPMA)共聚物之奈米粒子。令人感興趣地,早期研究指示儘管所表徵之聚合物展示與經分離線粒體締合,但利用卵巢癌細胞之實驗揭露聚合物之顯著溶酶體締合。然而,在最近研究中,使用螢光標記之構築體實施之微注射及培育實驗基於顯微鏡分析表明線粒體靶向能力。隨後,使用光敏劑內消旋二氫卟酚(mesochlorin) e 6 (Mce 6)合成HPMA共聚物-藥物偶聯物。HPMA共聚物結合之Mce 6之線粒體靶向與非靶向HPMA共聚物-Mce 6偶聯物相比增強細胞毒性。作者指示「可需要小的修飾以適於當前設計並允許其他治療劑之腫瘤位點特異性線粒體靶向」。因此,該等系統理論上可應用於線粒體特異性遞送一系列用於癌症療法之促細胞凋亡物質。 無機奈米粒子亦顯示能夠線粒體特異性遞送。在最近研究中,將竹紅菌素A (hypocrellin)(一種光動力學藥物)囊封於水溶性非晶形二氧化矽奈米籠(HANC)中。該等藥物加載之奈米籠據報告能夠在癌細胞之線粒體中特定累積並改良竹紅菌素A之光敏化效應。然而,尚不清楚何者介導奈米籠系統之線粒體特異性累積。然而,總而言之,目前為止所闡述之各種研究強烈支援奈米載劑確實可控制生物活性分子之亞細胞累積且因此在線粒體靶向抗癌策略之發展中代表有用工具(G.G.M. D'Souza等人,/ Biochimica et Biophysica Acta 1807 (2011) 689-696)。 細胞外腺苷破壞HuH-7細胞(Fas缺陷人類肝癌細胞系)中之線粒體膜電位,且該效應藉由腺苷轉運蛋白抑制劑雙嘧達莫或藉由過表現Bcl-XL來抑制。腺苷下調Bcl-XL及細胞凋亡蛋白抑制2 (IAP2)之mRNA及蛋白質之表現以直接抑制凋亡蛋白酶-3、-7及-9,但其以其他方式上調DIABLO (一種IAP之抑制劑)之mRNA及蛋白質表現。彼等腺苷效應藉由雙嘧達莫減弱(Cell Biol Toxicol (2010) 26:319-330)。 細胞外腺苷經由內在及外在路徑在各種癌細胞中誘導細胞凋亡。在前一路徑中,腺苷攝取於細胞中觸發細胞凋亡,且在後一路徑中,腺苷受體介導細胞凋亡。抑制腺苷之細胞攝取的雙嘧達顯著逆轉腺苷誘導之生長阻抑。The BCL-2 protein family determines the promise of cell apoptosis (an ancient cell suicide program necessary for development, tissue stability, and immunity). These proteins manage the mitochondrial outer membrane permeability (MOMP) and can be pro-apoptotic (especially Bax, BAD, Bak and Bok) or anti-apoptotic (among various other proteins, especially including intrinsic Bcl-2 , Bcl-xL and Bcl-w). There are a total of 25 genes in the Bcl-2 family known to date. It has been found that human BCL-2 is a gene located near the junction of abnormal junctions of chromosomes 18 and 14 (t14; 18) in tumor cells of patients with follicular lymphoma. This chromosomal translocation causes misregulation of the normal BCL-2 expression pattern and leads to cancer (Tsujimoto et al., 1985; Nunez et al., 1989). Various physiological death signals and pathological cell damage trigger the genetically programmed pathway of apoptosis (Vaux and Korsmeyer 1999). Apoptosis is reflected in the two main execution programs downstream of the death signal: apoptotic protease pathway and organelle dysfunction, of which mitochondrial dysfunction is the most obvious feature (for reviews, see Green and Reed 1998; Thornberry and Lazebnik 1998). Since members of the BCL-2 family reside upstream of irreversible cell damage and many of their efforts focus on the content of mitochondria, they play a key role in determining whether cells will survive or die. Recent studies on the involvement of mitochondria in cancer have revealed that when comparing metastatic mitochondria and mitochondria that are untransformed cells, there are a lot of differences in the structure and function of these organelles. In particular, modern research largely supports the observations of Warburg and his successors' metabolism, and at the same time refines and greatly expands the knowledge of the mitochondrial state and the mechanism of its function in tumor development. The comprehensive mechanism of the Warburg effect has not been isolated; however, multiple intertwined etiologies and response mechanisms have been described recently. This recognition of the indicative characteristics of cancer cell metabolism is also directly applied to current clinical care. It uses glucose analogues to identify cancerous lesions characterized by high glucose uptake through the increasingly widely adopted positron emission tomography (PET) imaging ( BioMed Research International Volume 2013 (2013), Article ID 612369). Apoptosis can be initiated through two pathways (ie, external and internal pathways). However, the external pathway is initiated by the stimulation of cell surface death receptors, and the internal pathway is activated by many different intracellular pressures and is caused by most chemotherapeutic agents. The BCL-2 family proteins are mainly involved in the intrinsic pathway, in which they regulate the mitochondrial outer membrane permeability (MOMP). MOMP causes apoptosis triggering factors (such as cytochrome c) to be released from the mitochondrial membrane space into the cytoplasm, where the apoptosis triggering factors activate the cascade of apoptotic proteases that perform common proteolytic events, leading to cell death (3). MOMP marks the promise of death and is usually regarded as the irreversible point of the cell. Here, the rational and preliminary results of recent efforts to induce MOMP by using BCL-2 family proteins as anti-cancer therapy are reviewed and other related targets for drug development are discussed (Clin Cancer Res; 21(12), June 2015 15th). Subsequently, the over-expression of Bcl-2 in prostate cancer, breast cancer, colon cancer, and glioblastoma is described. Overexpression of Mcl-1 (another anti-apoptotic Bcl-2 related protein) has been identified in relapsed acute myeloid leukemia and is associated with a poor prognosis. Other changes in the expression of Bcl-2 related proteins identified in cancer cells include different mutations in the Bax gene and changes in the ratio of pro-apoptotic and anti-apoptotic Bcl-2 proteins (Bettaieb et al., 2003; and Oncogene (2004) 23 , 2934-2949). The overexpression of Bcl-2 and related anti-apoptotic proteins has been shown to inhibit cell death induced by many stimuli including growth factor deprivation, hypoxia and oxidative stress. However, the ability of anti-apoptotic Bcl-2 family proteins to suppress cell death induced by cytotoxic anticancer drugs makes these proteins particularly interesting as potential targets for cancer drug discovery. Regardless of the main mode of action, whether it is a single-stranded or double-stranded DNA break, whether microtubules are depolymerized or aggregated, whether nuclear hormone receptor activation (glucocorticoid receptor) or inhibition (estrogen and androgen receptor), Basically all traditional anti-cancer drugs seem to rely to a large extent on the Bcl-2/Bax-dependent mechanism to kill cancer cells (reviewed by Debatin et al., 2002; Reed, 2008). Therefore, Bcl-2 operates at a remote point in the conservative cell death pathway used by most anti-cancer drugs. This composition is different from the previously identified mechanisms involving drug excretion, drug metabolism, drug inactivation, and related mechanisms. Intrinsic form of chemical resistance. This observation probably explains why the performance of various Bcl-2 family proteins has shown prognostic significance for many types of cancers and leukemias treated by chemotherapy. Defects in the expression of pro-apoptotic members of the BCL-2 family are also found in cancer, which leads to the loss of tumor suppressor function of these killer genes. The best recorded is BAX, where homozygous deletion or inactivation mutations have been specifically identified in cancers that accompany microsatellite instability due to defective DNA mismatch repair. In this regard, the human BAX gene contains a homopolymer extension of eight guanosine residues in the sense strand, and this extension is the target of frame shift mutations. The defective performance of pro-apoptotic BCL-2 family genes was also found in the background of loss of p53 function. Among them, the direct targets of p53 transcription factors are BAX, BID, PUMA, and NOXA, which confirms that there is a strong relationship between the genome supervision of p53 and the cell death genes of the BCL-2 family. Recently, it has been observed that p53 protein interacts with the cytoplasm of pro-apoptotic and anti-apoptotic Bcl-2 family proteins, which directly regulate the biological activities of p21-Bax and p26-Bcl-2 proteins, and indicate that p53 is involved in transcription and transcription. The latter two levels regulate the Bcl-2 family. The activities of other pro-apoptotic members of the Bcl-2 family are also suppressed by post-translational modifications. For example, the pro-apoptotic protein BAD is phosphorylated by Akt (PKB) and other protein kinases known to be highly active in cancer, which leads to its isolation by 14-3-3 (Oncogene (2008) 27, 6398-6406 ). The overexpression of Bcl-2 prevents apoptosis induced by most chemotherapeutic drugs (including alkylating agents and topoisomerase inhibitors). Moreover, fibroblasts defective in both Bax and Bak are resistant to apoptosis induced by various agents that induce mitochondrial stress. However, fibroblasts with a single defect of Bax or Bak do not have a significant defect in apoptosis induced by these agents, which indicates that both Bax and Bak proteins can be functionally redundant. This apparent redundancy can be cell type dependent, because HCT116 cells lacking Bax are resistant to apoptosis induced by staurosporine (Theodorakis et al., 2002). Many anti-cancer agents (such as arsenic and lonidamine) can directly permeate the outer mitochondrial membrane at least in vitro, and the overexpression of Bcl-2 prevents mitochondrial permeation and apoptosis induced by arsenic and lonidamine . Based on the role of Bcl-2 protein in regulating mitochondrial membrane permeation and its usually modified performance in human cancers, Bcl-2 protein is a reasonable target for inducing apoptosis by itself or in combination with other anti-cancer drugs (Anti-apoptotic member) or prototype (pro-apoptotic member). However, molecular markers should be used when using Bcl-2 targeted therapy. This is because not all anti-apoptotic cells overexpress Bcl-2, as confirmed in breast cancer cell lines screened by NCI cells (Nieves-Neira And Pommier, 1999; and Oncogene (2004) 23, 2934-2949). The Bcl-2 protein is an inhibitor of programmed cell death. It homodimerizes with itself and forms a heterodimer with the homologous protein Bax (a promoter of cell death). The Bax/Bcl-2 ratio can be used as a rheostat that determines the sensitivity of cells to apoptosis. The secondary cell location of some members of the Bcl-2 family also seems to regulate their functions. A small part of Bcl-2 and Bcl-xL can be found on the outer mitochondrial membrane. In contrast, Bax mainly exists in the cytoplasm before apoptosis is induced. During the early stage of apoptosis, Bax translocates from the cytoplasm to the mitochondria, where it participates in mitochondrial division and the release of cytochrome c. Regulating the insertion step can regulate cell apoptosis. The lower level of this ratio can lead to resistance of human cancer cells to apoptosis. Therefore, the Bax/Bcl-2 ratio can affect tumor progression and aggressiveness (Iran Biomed J. 2015 April; 19(2): 69-75). As in the extrinsic pathways, vehicles that target the intrinsic pathways involved in both tumorigenesis and chemoresistance are used in therapeutic pathways. These anti-cancer strategies attempt to develop drug-designed inhibitors of anti-apoptotic proteins (eg, Bcl-2, Bcl-xL, and IAP) that are commonly expressed in cancer cells. Efforts to target the Bcl-2 protein involve the development of agents that destroy the Bcl-2 complex. The BH3 mimic binds to the hydrophobic groove of the anti-apoptotic protein, which mimics the role of the BH3 only protein in binding to the pro-survival protein, resulting in the release of the BH3 only protein from the complex and activates BAX and BAK. So far, nearly twelve BH3 mimics have been studied as Bcl-2 inhibitors in different stages of human clinical trials, such as AT-101 (R-(−)-Gossypol), ABT-199 (Vitamin Venetoclax), ABT-737, ABT-263 (navitoclax, an orally available derivative of ABT-737), GX15-070 (obatoclax) and TW37. The field of Bcl-2 family member inhibitors is under continuous development, which emphasizes the importance of these molecules as potent anti-cancer agents. Moreover, targeting the specific BH4 domain of Bcl-2 is also emerging as a novel strategy for anti-cancer therapy. Therefore, Bcl-2, through its BH4 domain, cooperates with several proteins that regulate different cellular pathways (such as hypoxia and angiogenesis) involved in tumor progression and chemoresistance. Recently, it was discovered that a small molecule (ie, BDA-366) can act as a powerful and effective BH4 domain antagonist, which exhibits significant anti-cancer activity in vitro and in vivo, thus providing a proof of concept for this approach. Another innovative approach to inhibit Bcl-2 stems from the following evidence: the human bcl-2 gene contains a GC-rich sequence located in its promoter that has the possibility of forming a G four-stranded structure and functions as a transcriptional repressor element. Therefore, the G four-strand specific ligand can regulate the transcription of bcl-2 with the help of the stabilization of the four-strand structure (AGING, April 2016, Vol. 8 Issue 4). Bcl-XL is the major anti-apoptotic protein in the Bcl-2 family. Its overexpression is more widely observed in human lung cancer cells than Bcl-2, which indicates that Bcl-XL is more biologically related and therefore more important than lung cancer. The best therapeutic target. Here, the UCSF DOCK 6.1 program kit and NCI chemical library database are used to screen small molecules that selectively target the BH3 domain (aa 90-98) of Bcl-XL. Two new Bcl-XL inhibitors (BXI-61 and BXI-72) that exhibit selective toxicity against lung cancer cells are comparable to normal human bronchial epithelial cells. Fluorescence polarization analysis revealed that BXI-61 and BXI-72 preferentially bind to Bcl-XL protein in vitro with high binding affinity instead of Bcl2, Bcl-w, Bfl-1/A1 or Mcl-1. Treatment of cells with BXI-72 results in the destruction of Bcl-XL/Bak or Bcl-XL/Bax interactions, so that Bak is oligomerized and cytochrome c is released from mitochondria. Importantly, BXI-61 and BXI-72 show more potent anti-human lung cancer efficacy than ABT-737, and have less thrombocytopenia in vivo. BXI-72 overcomes the acquired radiation resistance of lung cancer. Based on these findings, the development of BXI as a new class of anticancer agents is guaranteed and represents a new strategy for improving lung cancer outcomes (Cancer Res. 2013, September 1, 73(17):5485-5496). Bax (a regulator of cell death) is needed in the decision-making stage of apoptosis. Recent studies have identified Bax's serine 184 (S184) as a key functional switch that controls its pro-apoptotic activity. Here, the structure bag surrounding S184 is used as a docking site to screen the NCI library of small molecules using the UCSF-DOCK program kit. Three compounds (ie, the small molecule Bax agonists SMBA1, SMBA2, and SMBA3) induce configuration changes in Bax by blocking S184 phosphorylation, which promotes the insertion of Bax into the mitochondrial membrane and the formation of Bax oligomers. The latter causes the release of cytochrome c and apoptosis in human lung cancer cells, which occurs in a Bax rather than Bak-dependent manner. SMBA1 strongly inhibits lung tumor growth by selectively activating Bax in vivo through apoptosis without significant normal tissue toxicity. Develop Bax agonists as a new class of anti-cancer drugs to provide strategies for the treatment of lung cancer and other malignant tumors with Bax (NATURE COMMUNICATIONS DOI: 10.1038/ncomms5935). BID (BH3 Interaction Domain Death Receptor Agonist) protein is located outside the death receptor and mitochondria in apoptotic signaling, and acts as an inducer of permeabilization of the outer mitochondrial membrane in type II cells. The content of BID is critical to the viability of various cells, because its silence makes cells resist apoptosis induced by death receptor ligands (such as TNF-related apoptosis-inducing ligand (TRAIL)). Moreover, it has been confirmed that the content of BID is lower than the functional dose in cells of several cell lines because it can be sensitive to TRAIL through the overexpression of BID. For the above reasons, BID has been considered for therapeutic agent development. However, in order to define the way to invest in BID, a number of major issues should be resolved. The main problem is to control the amount of BID delivered to the cells. Although full-length BID has been shown to participate in apoptosis signal transduction, effective activation of apoptosis requires specific cleavage of BID by apoptotic protease 8 and the production of an active truncated form (tBID). TBID expressed in cells directly induces apoptosis. Therefore, in order to take advantage of the selectivity of some anti-cancer agents against cancer cells, it is a better strategy to sensitize cells by full-length BID. To sensitize cells, an overexpression system based on adenovirus or pcDNA vector is usually used. However, it does not provide strict control of the expression level of BID in cells. Therefore, the content of BID in transfected cells exceeds the content of endogenous protein several times and in some cases direct activation of apoptosis is observed rather than sensitization of cells to apoptotic stimuli. According to the above, tBID appears in cells treated with adenovirus vectors expressing full-length BID (BMC Cancer201414:771 DOI: 10.1186/1471-2407-14-771). Rapamycin and its derivatives are promising therapeutic agents with both immunosuppressant and anti-tumor properties. The effects of these rapamycins are mediated by specific inhibition of mTOR protein kinase. It is known that mTOR acts as part of an evolutionarily conserved signaling pathway that controls the cell cycle in response to changes in nutrient content. The mTOR signaling network contains many tumor suppressor genes (including PTEN, LKB1, TSC1, and TSC2) and many proto-oncogenes (including PI3K, Akt, and eIF4E), and mTOR signaling is constitutively activated in many tumor types. These observations indicate that mTOR is an ideal target for anticancer agents and that rapamycin is such an agent. In fact, early preclinical and clinical studies indicate that rapamycin derivatives as anti-tumor agents have efficacy alone and in combination with other treatment modalities. Rapamycin appears to inhibit tumor growth by stopping tumor cell proliferation, inducing tumor cell apoptosis, and inhibiting tumor angiogenesis. The effect of rapamycin immunosuppressant is caused by inhibiting the proliferation of T and B cells by the same mechanism that rapamycin blocks the proliferation of cancer cells. Therefore, it may be considered that the immunosuppression induced by rapamycin will be harmful to the use of rapamycin as an anticancer agent. In contrast, when rapamycin is combined with the widely used immunosuppressant cyclosporine, compared with the tumor incidence observed when cyclosporine is used alone, in organ transplantation trials The frequency of tumor formation that occurs is reduced. Existing evidence indicates that with regard to tumor growth, the anticancer activity of rapamycin significantly exceeds the immunosuppressive effect of rapamycin. In recent years, rapamycin and its analogues (such as CCI-779 or temsirolimus, RAD001 or everolimus, sirolimus, FK-50 and AP23576) have been clinically used. It is used to treat various cancers, including kidney cancer, mantle cell lymphoma and metastatic breast cancer. Dipyridamole (DPM) (such as aspirin) inhibits platelet adhesion and therefore tends to prevent vascular embolization of heart attacks and strokes. There is a report on the European Stroke Prevention Study in Lancet (page 1, 371-4) issued on December 12, 1987. The introduction of this report commented that the use of aspirin to treat patients who survived a small stroke TIA (transient ischemic attack) lacks the indicated benefit. In this trial, 300 mg/day of dipyridamole was added to the treatment with aspirin and the results were significant. After two years, stroke deaths were reduced by 50%, deaths due to myocardial infarction were reduced by 38%, and deaths due to cancer were reduced by 25%. The anticancer effect of dipyridamole indicated above can only be due to its prevention of metastasis; however, Eva Bestida of the University of Barcelona and others have been published in Cancer Research, September 1985 (pages 4,048-4,062) It is reported that the growth of certain human cancer cells is inhibited by dipyridamole. It inhibits more than 80% of adenosine, thymidine and uridine. These substances are needed for the prosperity of cancer cells. This can indicate the anti-cancer effect of dipyridamole in addition to preventing metastasis. Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) has emerged as an attractive cytokine to selectively target cancer cells; however, its efficacy is challenged by many resistance mechanisms. Therefore, current research is investigating the possibility of dipyridamole enhancing the efficacy of TRAIL and the possible underlying mechanism. Dipyridamole significantly sensitized the following p53 mutant human cancer cell lines to the anti-tumor activity of TRAIL: SW480, MG63, and DU145, as evidenced by the ability of TRAIL to effectively cleave initiator and performer apoptotic proteases. Although dipyridamole upregulates both DR4 and DR5 and increases its cell surface expression, RNA interference reveals a preferential dependence on DR5. Moreover, dipyridamole inhibits survivin performance and its important results have been confirmed by small interfering RNA. Mechanistically, dipyridamole induces the transcriptional shutdown expressed by survivin, accompanied by G1 block characterized by D-type cyclin and cdk6 downregulation. In addition, the transcription mechanism driven by CCAAT/enhancer binding protein (C/EBP) homologous protein (CHOP) induction is responsible for the up-regulation of DR5 by dipyridamole. Importantly, dipyridamole-induced enhancement of TRAIL efficacy and changes in protein performance do not depend on protein kinase A or protein kinase G. In summary, the findings of this study illustrate the novel mechanism of dipyridamole action and highlight its promising use as a potential enhancer of TRAIL efficacy (Oncogene (2008) 27, 3435-3445). Preliminary observations indicate that dipyridamole (DPM) can increase the sensitivity of human ovarian cancer cells to cisplatin. In another study, Jekunen et al. showed that DPM enhanced the cytotoxicity of cisplatin in cisplatin-sensitive 2008 human ovarian cancer cells to 4.7 times in a synergistic manner, and in the cisplatin-resistant 2008/C13*5.25 subline Increased to 5.8 times. It has been found that DPM increases the cellular uptake of cisplatin in a concentration-dependent manner without increasing trypan blue or propidium iodide uptake or changing cell size. It is believed that the cumulative increase in cisplatin induced by DPM is not related to the non-specific increase in membrane permeability. In a nude mouse model with human bladder cancer, Keane et al. found that the tumor size was reduced by 20% when cisplatin was combined with DPM. Using human testicular cancer in the same model, it achieved complete tumor regression. Barberi-Heyob et al. found that DPM increased the growth inhibitory activity of cisplatin in MCF-7 human breast cancer cells in a synergistic manner. Janice R. Perussi et al. reported that the cytotoxicity of cisplatin in cisplatin-sensitive MDA/S human breast cancer cells was enhanced by DPM, indicating that there is a strong correlation between Pt accumulation and enhanced cisplatin cytotoxicity, but in cisplatin In platinum-resistant MDA/R cells, the synergistic interaction between cisplatin and DPM does not involve an increase in cisplatin cell accumulation (Quím. Nova Vol. 26, No. 3, São Paulo, May/June 2003). Gemcitabine (2',2'-difluorodeoxycytidine) is a pyrimidine nucleoside analogue. It exerts its cytotoxic effect in cells and has activity against many different solid tumors (including pancreatic cancer, breast cancer, lung cancer and bladder cancer). Because gemcitabine has strong hydrophilicity, passive diffusion through the lipid bilayer of the hydrophobic cytoplasmic membrane is relatively slow. In order to enter cells efficiently, gemcitabine requires a physiological nucleoside transporter to cross the serosal membrane. These transporters are divided into two types, namely balanced transporters and concentrated transporters. The bidirectional human balanced nucleoside transporter (hENT) is found in most cell types, and both hENT1 and hENT2 can mediate gemcitabine uptake in the direction of a concentration gradient. hENT protein is a transmembrane glycoprotein located in the plasma membrane. Its ability is inhibited by nitrobenzyl mercaptopurine ribonucleoside (NBMPR) (specific hENT1 inhibitor at low nanomolar concentrations) and dipyridamole (hENT1/2 inhibitor). In the upper division, the sensitivity to gemcitabine is reduced to 1/39 and 1/1,800 (The Oncologist 2008; 13:261-276). Fluorouracil (5-FU) (especially sold under the trade name Adrucil) is a medicine used to treat cancer. By injecting it into a vein, it is used for colon cancer, esophageal cancer, gastric cancer, pancreatic cancer, breast cancer, and cervical cancer. As a cream, it is used for solar keratosis and basal cell carcinoma, and as eye drops for the treatment of ocular surface squamous cell tumors. The defect of apoptosis has involved the chemoresistance of cancer cells. Studies have found that the combination of high levels of anti-apoptotic Bcl-2 and low levels of Bax is associated with high 5-FU resistance in various cancers, including human breast cancer, pancreatic cancer, human head and neck cancer, and colon tumor cells ( Int J Cancer. April 1, 2002; 98(4):498-504; and Br J Cancer. November 2000; 83(10): 1380-1386). Dipyridamole has been shown to enhance the cytotoxicity of 5-FU and other fluoropyrimidines in vitro. This effect may be related to the increase in the intracellular concentration of FdUMP and the inhibition of extracellular nucleoside uptake through the "salvage" pathway. In vitro studies have shown that dipyridamole may increase many cytotoxic agents (including etoposide, doxorubicin, and vinblastine) by changing the cellular uptake and retention of cytotoxic agents. ) And mitoxantrone (mitoxantrone)) (Investigational New Drugs 12: 283-287, 1994). Multidrug resistance (MDR) of cancer cells is the simultaneous development of resistance to various anti-cancer drugs that seem to be structurally and mechanistically unrelated. One type of MDR is characterized by a decrease in the accumulation of hydrophobic natural product drugs. In some multi-drug resistant cells, drug excretion is mediated by an adenosine triphosphate (ATP)-dependent membrane transporter called P-glycoprotein (Pgp) (product of the MDR1 gene) (Juliano and Ling, 1976). Pgp acts as an active transport mechanism for various molecules (including certain chemotherapeutic drugs). As data on the role of Pgp in drug resistance accumulates, it is clear that other transporters can confer resistance to cytotoxic agents. The MDR protein 1 (MRP1) gene was cloned from a multi-drug resistant lung cancer cell line, and it was found that, like Pgp, it is a member of the ATP-binding cassette (ABC) superfamily of the transporter gene (Cole et al., 1992). Transfection studies indicate that, similar to MDR1, MRP1 overexpression is sufficient to confer resistance to various lipophilic natural product antitumor drugs. MRP1 is the first identified member of the gene family encoding the multispecific organic anion transporter (MOAT) protein (Borst et al., 1999). The other two homologs of MRP1, cMOAT/MRP2 and MRP3, encode proteins that mediate MDR when transfected into drug-sensitive cells (Borst et al., 1999). All of these membrane-embedded proteins act as drug discharge pumps, preventing cytotoxic agents from reaching lethal levels in cells. P-glycoprotein (P-gp) is a key role in the multi-drug resistance phenotype in cancer. The protein confers resistance by mediating the ATP-dependent excretion of a surprising series of anticancer drugs. Its wide specificity has been the subject of many attempts to inhibit the protein and restore the efficacy of anticancer drugs. The general strategy is to develop compounds that compete with anticancer drugs for transmission or act as direct inhibitors of P-gp. Although quite successful in vitro, there are currently no compounds that can be used to "block" P-gp-mediated resistance in the clinic. The failure can be attributed to toxicity, adverse drug interactions, and several pharmacokinetic issues. In addition, multi-drug resistance-related protein 1 (MRP1) transmits a wide range of therapeutic agents and various physiological substrates, and can develop drug resistance in certain cancers (including lung cancer, breast cancer and prostate cancer) and childhood neuroblastoma Play a role. Several studies above have shown that in vitro, dipyridamole can significantly increase the cytotoxicity and anti-tumor activity of various chemotherapeutic agents. The underlying mechanism here is to prevent nucleoside and nucleobase salvage, and to increase the intracellular accumulation of toxic metabolites via inhibited P-glycoprotein and MRP1 (Clin Pharmacol Ther 2003; 73: 51-60. Drug Metab Dispos. 2014 April; 42(4): 623-631. Oncogene (2003) 22, 7340-7358). Studies have investigated the role of dipyridamole as a single agent in the prevention of tumor occurrence and metastasis in multiple models of triple-negative (estrogen and progesterone receptor negative, Her-2 normal) breast cancer (a subtype with little effective therapy) The potential role. These findings provide evidence that intraperitoneal administration of dipyridamole impairs the growth and metastasis of primary tumors in a breast cancer xenograft animal model. Moreover, the data identified a new mechanism of action of dipyridamole, which was shown to inhibit ERK1/2-MAPK, NF-kB and Wnt signaling pathways, and prevent the accumulation of inflammatory cells in the tumor microenvironment. In addition, dipyridamole is the most potent BCRP inhibitor among the tested compounds, with an IC50 value of 6.4 +/- 0.9 mM. Therefore, dipyridamole has the potential to treat multidrug resistance in cancer (Cancer Prev Res (Phila). May 2013; 6(5):. Doi:10.1158/1940-6207.CAPR-12-0345). However, clinical administration of DPM did not improve the anticancer activity of 5-FU or cisplatin in patients with advanced colorectal cancer, metastatic breast cancer, advanced non-small cell lung cancer, or advanced measurable pancreatic cancer. The increased dose intensity of 5-FU or cisplatin observed for DPM is not clinically relevant. The path for nanoparticle drug carriers to enter cells is different from conventional drugs. Conventional drugs enter cells through dose-dependent diffusion. In other words, the higher the concentration of the drug in the blood, the higher the concentration of the drug in the cells, and the drug can only enter the cytoplasm. Nanoparticle drug carriers are absorbed by cells through endocytosis and enter the cells to form lysosomes. In the initial stage after injection, the concentration of the nanoparticle drug carrier increases in a time-dependent manner. Endocytosis is the process of incorporating extracellular material into a cell. This process can be classified into three types, namely endocytosis, pinocytosis and receptor-mediated endocytosis. Cytophagy occurs only in specialized cells. These cells proliferate and aggregate when stimulated by extracellular substances and engulf the substances in the lysosomes of the cells for decomposition. This process occurs in the macrophages and neutrophils of the immune system. The pinocytosis system internalizes the extracellular fluids and molecules in the cell membrane to form a pocket by means of the recess of the cell membrane, and then pinches it off into the cell to form a vesicle. The vesicles then travel into the cytoplasm and fuse with other vesicles, such as endosomes and lysosomes. Depending on the structure of the carrier, pinocytosis can be divided into two types, namely fluid-phase pinocytosis and adsorptive pinocytosis. If the carrier does not have a functional group that interacts with the cell, the cell will engulf the drug carrier through fluid phase pinocytosis. This process is slow and depends on the concentration of the carrier around the cell membrane. When the carrier has a hydrophobic group or is positively charged, adsorptive pinocytosis will occur. These carriers will be physically adsorbed by the cell membrane and increase the engulfing capacity of the cell. The above two types of endocytosis are non-specific processes and are not suitable for delivering drugs to their targets. Targeting can only be achieved by enhanced penetration and retention (EPR) in certain cancer tissues. Receptor-mediated endocytosis is a process in which cells absorb molecules (endocytosis) by sprouting inwardly of serosal vesicles containing proteins with receptor sites specific to the uptake molecules. After the drug carrier binds to the receptor on the cell, the intrinsic signal will trigger the cell membrane to form a coated pit. The surface area of the coated pit is equivalent to 1% to 2% of the cell membrane. The coated follicles will separate from the cell membrane and enter the cell to form coated vesicles in the cell, then form endosomes and move in a jumping motion inside the cell. The intranuclear system includes a complex structure of microtubules and vesicles. Vesicles can be fused with Golgi. Due to the proton pump (ATPase), endosomes usually become acidic. Endosomes will then fuse with lysosomes to form secondary lysosomes. The cell membrane therapeutic agent is effectively delivered to the barrier to be overcome at the target site in the mitochondria, cytoplasm or nucleus. Hydrophobic phospholipids are the main components of cell membranes that hinder the delivery of therapeutic agents. Therefore, various delivery systems (such as liposomes, nanoparticles, and viral vectors) have been developed to transfer small molecules, peptides, proteins, and oligonucleotides across membranes. This method of drug delivery is referred to herein as a cell penetrating drug delivery system. Many drug carrier systems (liposomes, cell penetrating peptides, cationic polymer conjugates and polymeric nanoparticles) have been developed for intracellular delivery of therapeutic agents. It needs to be suitable for crossing a series of membrane barriers to reach the site of drug action in the cell. During this process, an important part of the drug molecule will be lost at each continuous barrier. These barriers include cell association of drug carriers and internalization by endocytosis; intracellular transport and release of drugs or drug carriers into the cytoplasm; translocation of drugs or drug carriers from the cytoplasm to the nucleus or any other organelle ; And nucleus/organelle uptake. Cells contain several intracellular organelles with specific functions. The intracellular targeting of these specific organelles of therapeutic agents is expected to not only significantly enhance the therapeutic efficacy, but also reduce non-specific effects and therefore toxicity. Therefore, there is a great interest in using different carrier systems to achieve intracellular target-specific delivery of therapeutic agents. Carriers that promote endocytosis of drugs include nano-sized polymer carriers and liposomes. Depending on the nature of the drug and the manufacturing process, nano-sized drug carriers can be classified into nanoparticle, nanoliposome, nanosuspended particle, solid lipid nanoparticle, magnetic nanocarrier and the like. In addition to the above-mentioned carriers, cell penetrating peptides (CPP), biodegradable nanoparticles, and viral vectors can also be used as delivery systems to enhance the penetration of drugs into cells. Since cell membranes constitute the main barrier for intracellular delivery of large-sized hydrophilic proteins, peptides and oligonucleotides, cell penetrating peptides (CPP) have been developed to overcome this barrier. These CPPs can ferry the molecule or colloidal drug carrier system labeled with them across the cell membrane, enter the cytoplasm and reach the nucleus. The characteristics of CPP are attributed to the existence of a segment of 9-16 cationic amino acid residues; the most commonly studied CPP includes HIV-1 transactivated transcription activator (TAT) peptide, HSV VP-22 (herpes simplex) Virus type 1 transcription factor) peptide and penetratin. Several theories have been proposed to determine the exact mechanism by which these CPP enter cells. For example, the penetration of TAT through cell membranes has been shown to be independent of receptors and transporters, and it has been suggested to enter cells by destabilizing the phospholipid bilayer to form antimicrobial cells. The main benefit of TAT coupling is that together with the effective delivery of the molecule, the biological activity of the coupled molecule is retained, and the size of the delivered molecule is not a rate limiting factor. TAT has been proposed to enhance not only intracellular delivery but also nuclear delivery, and therefore has been studied for nucleic acid delivery. TAT peptides coupled to antisense oligonucleotides have been shown to deliver the oligonucleotides to the nucleus. After internalization, it was also found that the TAT peptide and BODIPY-ceramide (which is a marker of the Golgi body) co-localized inside the Golgi body. Therefore, it is likely to be directly transported from the early endosomes to the Golgi without entering the late endosomes. There may be a secretory pathway in which the peptide enters the cytoplasm from the endoplasmic reticulum. Gene therapy has proven to have great potential in the treatment of hereditary, acquired and neurodegenerative disorders. Among non-viral gene delivery methods, various drug delivery systems and polymers are being studied, such as liposomes, cationic lipid-DNA, and polymer complexes. To overcome the relatively inefficient cellular uptake of non-viral gene expression vectors, it has been explored to couple TAT peptides to the vector. Kleeman et al. have used polyethyleneimine (PEI) covalently coupled to TAT via a polyethylene glycol (PEG) spacer to confirm gene expression in alveolar basal epithelial cells. The transfection efficiency in lung is higher than that of unconjugated PEG complex in vivo. In a similar study by Rudolph et al., solid lipid particles coupled to dimeric HIV-1 TAT have demonstrated enhanced gene delivery to the lung. The amino acid composition of CPP usually contains positively charged amino acids with high relative abundance (such as lysine or arginine) or alternates between polar/charged amino acids and non-polar, hydrophobic amino acids. The sequence of patterns. The two types of structures are called polycationic or amphiphilic, respectively. The third type of CPP is a hydrophobic peptide that contains only non-polar residues, has a low net charge, or has a hydrophobic amino acid group that is essential for cellular uptake. Among the cell penetrating peptides, arginine-rich cell penetrating peptides have been studied most extensively. Examples include the TAT peptide from HIV transactivator protein TAT, Penetratin, the 16 amino acid domain of Antennapedia protein from Drosophila, and the coat of flock house virus (FHV) Peptide (sequence 35-49) and oligoarginine. A dynamic process of intracellular delivery system mediated by biodegradable nanoparticles; involving endocytosis, exocytosis and sorting into different intracellular compartments. It appears that the NP surface and its interaction with the cell surface controls the uptake and intracellular transport of the biodegradable nanoparticles and therefore the encapsulated therapeutic agent. Viral vectors are tools commonly used by molecular biologists to deliver genetic material into cells. This process can be performed inside a living organism (in vivo) or in cell culture (in vitro). Therefore, viral vector systems are suitable options for cell-penetrating drug delivery systems. Cell penetrating peptides and biodegradable nanoparticles are not only used to modify drugs, but also coupled to carriers to enhance transmembrane effects. Dipyridamole is a balanced nucleoside transporter (ENT) inhibitor. Nucleoside transporter (NT) plays an important role in the transport of nucleosides across cell membranes. Dipyridamole blocks the balanced nucleoside transporter (ENT), which promotes the diffusion of adenosine across the membrane. Dipyridamole will increase the concentration of extracellular endogenous adenosine mainly in situations where the extracellular formation of adenosine is increased (for example, occurring during hypoxia or inflammation). However, the extracellular endogenous adenosine concentration induced by dipyridamole causes vasodilation, which contributes to the metabolic control of organ perfusion. Dipyridamole pressure myocardial imaging is a widely used and successful technique for diagnosing and evaluating coronary artery disease. Coronary artery dilation with IV dipyridamole is associated with a significant reduction in blood flow to the collateral-dependent myocardium, which is consistent with coronary blood stealing in patients with CAD. In addition, there are other studies that have discovered the vasoconstriction and vasodilation effects of dipyridamole in many organs (including kidney, lung, pancreas, brain, etc.). Dipyridamole not only causes vasoconstriction in some organs, but can also cause hypotension and subsequent side effects, such as dizziness and palpitations due to the dilation of cardiovascular blood vessels. The effect of lowering blood pressure makes dipyridamole unsuitable for the treatment of physiologically unstable patients, such as those suffering from (but not limited to) sepsis, ischemic stroke, hemorrhagic stroke, acute lung injury, acute liver injury, Patients with myocardial infarction and cardiorenal syndrome. Moreover, the blood flow restriction effect of dipyridamole limits its application in the treatment of diseases involving organs rich in blood vessels. Since the pharmacological effects of dipyridamole are mainly on the cell membrane, a delivery system designed for membrane penetration to avoid binding to the balanced nucleoside transporter on the membrane while enhancing intracellular signal transduction and PPARγ regulation can prevent the increase The effect of tissue hypoperfusion caused by cardiovascular expansion and local blood flow restriction. Therefore, the limitation of dipyridamole in clinical application due to the decrease of blood pressure in acute and severe patients can be lifted. Dipyridamole is also a non-selective phosphodiesterase inhibitor. The increase in intracellular drug delivery will enhance the inhibition of intracellular phosphodiesterase (PDE) by dipyridamole. The members of the PDE family have a unique cell and tissue specific distribution. Depending on the distribution curve of PDE on the cell membrane or in the cytoplasm in different tissues, dipyridamole can be used as an anti-inflammatory, antioxidant, and smooth muscle relaxant for the treatment of diseases related to PDE regulation. The unique cell and tissue specific distribution of PDE is shown in the following table (see US 2012/0065165):
Increasing the ability of dipyridamole to penetrate the membrane can promote the inhibition of PDE3, PDE5 and PDE8 in specific tissues and endow dipyridamole with therapeutic effects in diseases related to PDE3, PDE5 and PDE8. In this case, dipyridamole can be used to treat lower urinary tract dysfunction and erectile dysfunction like other PDE5 inhibitors. Moreover, since dipyridamole is a non-selective PDE inhibitor, it can be used to treat PDE-related diseases when it is delivered by a transmembrane drug delivery system. There are several molecules that act on mitochondria that are currently in use or are being tested in clinical trials. Certain clinically approved anticancer drugs (e.g. paclitaxel and VP-16 (etoposide) and vinorelbine) and an increasing number of experimental anticancer drugs (e.g. ceramide, MKT077, CD437, cloni Daming and betulinic acid) have been found to act directly on mitochondria to trigger apoptosis. CD437 can induce apoptosis in various human cancer cells in vitro and in vivo. In intact cells, CD437-dependent apoptotic protease activates the release of cytochrome C from the mitochondria before activation. Moreover, when added to isolated mitochondria, CD437 caused membrane permeabilization. This effect is prevented by inhibitors of the mitochondrial permeability transition pore complex (mPTPC), such as cyclosporine A. Therefore, CD437 represents a low molecular weight compound that exerts its cytotoxic effect (ie, by directly acting on the surface or inside of mitochondria) through mPTPC. Similarly, arsenic trioxide used to treat acute promyelogenous leukemia has multiple effects on mitochondria. It is known that via its action on the voltage-dependent anion channel VDAC leads to the induction of mPTPC formation. Arsenic trioxide is also known to act on the respiratory chain and inhibit the activity of the respiratory chain. The apoptotic factors that play a major role in cell apoptosis include Bcl-2 and Bcl-Xl. Compounds that act by binding to these proteins have been identified and studied for their efficacy; some examples include chromene derivatives and gossypol, which have recently been shown to act on proteins of the Bcl-2 family. In fact, there are so many different and structurally different compounds, so it is suggested to collectively refer to them as mitochondria-targeted anticancer drugs (mitocans) to reflect their mitochondrial-mediated anticancer effects. The selective accumulation pathway targeting tumor mitochondria requires two levels of specific accumulation; drug accumulation in tumors and then drug accumulation in cancer cell mitochondria. Generally speaking, drug treatment can be adjusted by subtle modification of the chemical structure to change its known physical-chemical properties that determine its accumulation in each compartment. Of course, these modifications must be implemented without adversely affecting the molecular target. The second approach involves coupling ligands larger than simple organic functional groups to alter the biodistribution of active molecules. Again, as long as the coupling does not adversely affect the desired pharmacological activity of the molecule, this approach will work. Using ligands that are known to have affinity for target tissues, these approaches have been extremely effective in changing the distribution of drugs in the body and achieving higher accumulation in target tissues. There are ligands that have been shown to mediate the tumor-specific accumulation of drugs, and there are ligands that are known to be mitochondrial. However, it is still unclear whether there are ligands with two properties to the extent that will allow a high desired accumulation level. Therefore, it can be said with certainty that so far, the dual strategy is the most feasible way. This dual strategy will require the use of a targeted delivery route to achieve high tumor accumulation, followed by a second route to ensure that the drug then accumulates in the mitochondria where it will exert its effect. Although there are many tissue-specific delivery studies aimed at increasing the weight content of anticancer drugs, studies aimed at subcellular delivery have only received more attention. Nonetheless, there are some interesting mitochondrial delivery routes suggesting the promise of improved cancer therapy. Pharmaceutical nanocarriers (such as liposomes, micelles, and solid nanoparticles) provide a way that can be regarded as a non-chemical way to change the disposal of drug molecules. All chemical processes can be performed on the components of the nanocarrier system, which can then be loaded with drugs to provide targeted delivery. Most pharmaceutical nanocarriers can be modified to target specific tissues and even specific cell types. Long-circulating liposomes and nanoparticles can passively target leaky vascular regions with enhanced penetration and retention (EPR) effects, and can also be modified with antibodies or other targeting ligands to provide cell-specific recognition. If nano-carriers that affect both the tumor-specific accumulation of drugs and mediate the mitochondrial-specific accumulation in tumor cells can be developed for clinical therapy, it can be the ultimate tool in the mitochondrial targeted anti-cancer pathway. The first step in this direction has been taken in recent years. The current nano-carrier technology has reached the extent that the demand for subcellular delivery can be actually met by using nano-carriers specifically designed for these purposes. In the route for delivering the mPTPC-inducing drug wasptoparan (mastoparan) into cells, liposomes are modified with both transferrin and the gene fusion peptide Chol-GALA. The transferrin modification enhances the uptake of liposomes into cells via endocytosis, after which the peptide promotes release from endosomes into the cytosolic fluid. Therefore, by only increasing the intracellular content of the drug, the delivery route achieves a higher concentration of the drug that can potentially interact with subcellular targets. Micelles have also been suggested for the delivery of hydrophobic drugs to various subcellular organelles, including mitochondria. It is found that the luciferin-labeled microcell lines used in this study are distributed through several cytoplasmic organelles (including most of them associated with mitochondria). The uptake of these micelles is not limited to a single cell type. Moreover, the cellular internalization cargo (cargo) is incorporated into the micelles to a higher degree than the free cargo itself. There are currently several examples of nanocarriers specifically designed to accumulate in mitochondria. Possibly, the earliest such systems are called DQAsome. Prepared from the mitochondrial molecule dequalinium chloride. These vesicle nano-carriers have been developed for mitochondrial-specific DNA delivery, but have also been shown to change the subcellular distribution of paclitaxel to increase the drug mitochondria In the accumulation. Mitochondrial-specific delivery results in improved apoptotic activity at paclitaxel concentrations where the free drug does not have a significant cytotoxic effect. DQAsome loaded with paclitaxel has also been tested for its ability to inhibit the growth of human colon cancer tumors in nude mice, and the data strongly suggests that encapsulation of paclitaxel in DQAsome leads to improved efficacy. The anti-tumor efficiency of paclitaxel encapsulated by DQAsomal is further improved by modifying the surface of DQAsomal with folic acid (FA). Folic acid is a high-affinity folate membrane-bound protein, which has been expressed in a variety of human tumors. FA conjugates are internalized by receptor-mediated endocytosis in a tumor cell-specific manner, leading to increased toxicity of the corresponding drugs. Another approach for mitochondrial specific nanocarriers is to modify existing nanocarriers with mitochondrial ligands. In this regard, TPP once again acts as a mitochondrial ligand in liposome and polymer-based nanocarriers. Liposomes have been well characterized as delivery systems and are a popular choice due to their biocompatibility, ease of surface modification, and ability to encapsulate hydrophilic or hydrophobic drugs. It is the first to indicate that liposomes can be modified by using mitochondrial residues to make the mitochondrial system derived from reports on the so-called proteoliposomes. The proteolipid system incorporates the crude mitochondrial membrane part in the preimplanted embryo Prepared in liposomes coexisting with endogenous mitochondria. Further research on the concept of using ligands to alter the subcellular distribution of liposomes is based on stearyl triphenyl phosphonium (STPP). The stearyl residues of STPP act as lipid anchors to use TPP residues to modify the surface of liposomes and produce liposome formulations that have a clear predisposition to mitochondria. STPP-liposomes have been shown to effectively guide the accumulation of rhodamine-labeled phosphatidylethanolamine (Rh-PE) to mitochondria in living cells. Based on flow cytometry, STPP-liposomes exhibit the same level of cell association as liposomes with the same cationic charge. However, the subsequent subcellular localization by confocal microscopy analysis is significantly different, indicating that the mitochondrial specific association of the nanocarrier is determined by the mitochondrial ligand rather than the surface charge. It was also found that TPP ligand does not change the in vivo distribution of long-circulating pegylated liposomes and tumor accumulation. However, STPP-liposomes do improve both the in vitro and in vivo efficacy of Ceramide. In summary, these data indicate that the tendency of long-circulating liposomes to passively accumulate in solid tumors (via the EPR effect) can be combined with the organelle-specific tropism conferred by appropriate ligand modification to enhance the effect of encapsulated anti-tumor agents. Alternative approaches to the development of mitochondrial-specific liposomes focused on the concept that liposomes that tend to selectively fuse with mitochondrial membranes are more likely to associate with mitochondria when they enter cells. These liposomes, called MITO-Porter, utilize octaarginine residues to be surface-modified to facilitate entry into cells as intact vesicles (via giant pinocytosis). The lipid composition is selected based on the high level of fusion with the mitochondrial membrane in living cells and the release of its cargo to the inner mitochondrial compartment. Based on confocal microscopy data, MITO-porter liposomes have been used to deliver green fluorescent protein and propidium iodide to mitochondria, indicating that it can be used to deliver macromolecules and small molecules to mitochondria. The development of mitochondrial-specific nanocarriers is not limited to lipid-based carriers, and also includes the use of mitochondrial residues to generate polymer systems capable of mitochondrial-specific intracellular delivery of biologically active molecules. TPP has been modified to produce nanoparticles based on mitochondrial mitochondrial N-(2-hydroxypropyl)methacrylamide (HPMA) copolymer. Interestingly, early studies indicated that although the characterized polymer exhibits association with isolated mitochondria, experiments with ovarian cancer cells revealed significant lysosomal association of the polymer. However, in recent studies, microinjection and incubation experiments using fluorescently labeled constructs based on microscopic analysis have shown the ability of mitochondria to target. Subsequently, the photosensitizer mesochlorin e 6 (Mce 6) was used to synthesize the HPMA copolymer-drug conjugate. The mitochondrial targeting of HPMA copolymer-bound Mce 6 enhances cytotoxicity compared with non-targeting HPMA copolymer-Mce 6 conjugate. The authors indicate that "small modifications may be required to suit the current design and allow tumor site-specific mitochondrial targeting of other therapeutic agents." Therefore, these systems can theoretically be applied to mitochondria to specifically deliver a series of pro-apoptotic substances for cancer therapy. Inorganic nanoparticles have also been shown to be capable of mitochondrial specific delivery. In a recent study, hypocrellin A (a photodynamic drug) was encapsulated in a water-soluble amorphous silica nanocage (HANC). These drug-loaded nanocages are reported to specifically accumulate in the mitochondria of cancer cells and improve the photosensitizing effect of Hypocrellin A. However, it is unclear which mediates the specific accumulation of mitochondria in the nanocage system. However, all in all, the various studies described so far strongly support that nanocarriers can indeed control the subcellular accumulation of bioactive molecules and therefore represent a useful tool in the development of mitochondrial targeted anti-cancer strategies (GGM D'Souza et al., / Biochimica et Biophysica Acta 1807 (2011) 689-696). Extracellular adenosine disrupts the mitochondrial membrane potential in HuH-7 cells (Fas-deficient human liver cancer cell line), and this effect is inhibited by the adenosine transporter inhibitor dipyridamole or by overexpression of Bcl-XL. Adenosine down-regulates the expression of mRNA and protein of Bcl-XL and apoptosis protein inhibitor 2 (IAP2) to directly inhibit apoptotic proteases-3, -7 and -9, but it up-regulates DIABLO (an inhibitor of IAP) in other ways ) MRNA and protein performance. These adenosine effects are attenuated by dipyridamole (Cell Biol Toxicol (2010) 26:319-330). Extracellular adenosine induces apoptosis in various cancer cells through internal and external pathways. In the former pathway, adenosine uptake in cells triggers apoptosis, and in the latter pathway, adenosine receptors mediate apoptosis. Dipyridamole, which inhibits the cellular uptake of adenosine, significantly reverses the growth suppression induced by adenosine.
本發明中已發現,某些經取代嘧啶并[5,4-d
]嘧啶化合物(例如雙嘧達莫)能夠增強BAX/BCL-2之表現及活性。因此,本發明提供具有嘧啶并[5,4-d
]嘧啶主要結構之新穎類型之BAX/BCL-2調節劑及使用該等BAX/BCL-2調節劑預防或治療BAX/BCL-2相關病症或病況之方法,例如癌症、骨髓增殖性病症、前列腺上皮發育不良、淋巴血管平滑肌肉增殖症、木村病(Kimura disease)及瘢痕瘤。本發明亦係關於增加BAX/BCL-2之表現及活性之方法。 儘管研究證實雙嘧達莫展現抗癌活性,但在低劑量下之功效不顯著。雙嘧達莫係已知阻斷腺苷攝取至細胞中之ENT-1抑制劑。由於腺苷可促進癌細胞之細胞凋亡,故該阻斷降低雙嘧達莫之抗癌功效。在本發明中,雙嘧達莫可囊封於載劑中以促進細胞膜之穿透及在細胞中之累積,且由此活化細胞之內源性細胞凋亡機制。雙嘧達莫在細胞中增強之促細胞凋亡功效可藉助增加BAX/BCL-2異二聚體之形成、增加BAX同二聚體之形成及降低BCL-2同二聚體之形成來達成。在本發明中亦發現雙嘧達莫可降低BCL-XL表現,降低ENT-1之抑制並調節癌細胞之凋亡蛋白表現,由此增加其抗癌活性。 在較佳實施例中,雙嘧達莫囊封於細胞穿透載劑中以增加細胞內部雙嘧達莫之濃度並降低對ENT-1之抑制。在治療癌症之實施例中,可避免BCL-XL之增加及胞質腺苷之降低且因此抗癌功效之降低。 在實施例中,本發明係關於預防或治療BAX/BCL-2相關病症或病況之方法,其包含向需要其之個體投與治療有效量之式(I)化合物:(I) 其中R1
、R2
、R3
及R4
中之每一者獨立地選自由雜環基及二(羥烷基)胺基組成之群, 或其醫藥上可接受之鹽。 本發明亦係關於式I化合物或其醫藥上可接受之鹽之用途,其用於製造用於預防或治療BAX/BCL-2相關病症或病況之醫藥。在較佳實施例中,醫藥包含囊封於醫藥上可接受之載劑中之式I化合物或其醫藥上可接受之鹽。 本發明進一步係關於用於預防或治療BAX/BCL-2相關疾病之醫藥組合物,其包含治療有效量之囊封於醫藥上可接受之載劑中之式I化合物或其醫藥上可接受之鹽。在較佳實施例中,化合物係雙嘧達莫且載劑係脂質體。 本發明更詳細闡述於以下部分中。本發明之其他特徵、目的及優點可容易地在本發明之具體實施方式及申請專利範圍中發現。It has been found in the present invention that certain substituted pyrimido[5,4- d ]pyrimidine compounds (such as dipyridamole) can enhance the performance and activity of BAX/BCL-2. Therefore, the present invention provides a novel type of BAX/BCL-2 modulator with the main structure of pyrimido[5,4-d ]pyrimidine and the use of the BAX/BCL-2 modulator to prevent or treat BAX/BCL-2 related diseases Or methods of disease conditions, such as cancer, myeloproliferative disorders, prostatic epithelial dysplasia, lymphangiomyocytosis, Kimura disease, and keloids. The present invention also relates to methods for increasing the performance and activity of BAX/BCL-2. Although studies have confirmed that dipyridamole exhibits anti-cancer activity, its effect is not significant at low doses. Dipyridamole is an ENT-1 inhibitor known to block the uptake of adenosine into cells. Since adenosine can promote the apoptosis of cancer cells, the blockade reduces the anti-cancer efficacy of dipyridamole. In the present invention, dipyridamole can be encapsulated in a carrier to promote cell membrane penetration and accumulation in the cell, and thereby activate the cell's endogenous apoptosis mechanism. The enhanced apoptosis-promoting effect of dipyridamole in cells can be achieved by increasing the formation of BAX/BCL-2 heterodimers, increasing the formation of BAX homodimers and reducing the formation of BCL-2 homodimers . In the present invention, it was also found that dipyridamole can reduce the performance of BCL-XL, reduce the inhibition of ENT-1 and regulate the expression of apoptotic proteins of cancer cells, thereby increasing its anti-cancer activity. In a preferred embodiment, dipyridamole is encapsulated in a cell penetrating vehicle to increase the concentration of dipyridamole inside the cell and reduce the inhibition of ENT-1. In the embodiment of the treatment of cancer, the increase of BCL-XL and the decrease of cytoplasmic adenosine can be avoided and therefore the anticancer efficacy is decreased. In an embodiment, the present invention relates to a method for preventing or treating BAX/BCL-2 related disorders or conditions, which comprises administering a therapeutically effective amount of a compound of formula (I) to an individual in need thereof: (I) wherein each of R 1 , R 2 , R 3 and R 4 is independently selected from the group consisting of a heterocyclic group and a di(hydroxyalkyl)amino group, or a pharmaceutically acceptable salt thereof. The present invention also relates to the use of a compound of formula I or a pharmaceutically acceptable salt thereof for the manufacture of medicines for the prevention or treatment of BAX/BCL-2 related diseases or conditions. In a preferred embodiment, the medicine comprises a compound of formula I or a pharmaceutically acceptable salt thereof encapsulated in a pharmaceutically acceptable carrier. The present invention further relates to a pharmaceutical composition for preventing or treating BAX/BCL-2 related diseases, which comprises a therapeutically effective amount of a compound of formula I encapsulated in a pharmaceutically acceptable carrier or a pharmaceutically acceptable compound thereof salt. In a preferred embodiment, the compound is dipyridamole and the carrier is a liposome. The invention is explained in more detail in the following sections. Other features, objectives and advantages of the present invention can be easily found in the specific embodiments of the present invention and the scope of the patent application.
除非本文中另外定義,否則結合本發明所用之科學及技術術語應具有熟悉此項技術者通常理解之含義。術語之含義及範圍應係清晰的;然而,在任何潛在歧義之情形中,本文所提供之定義優先於任何字典或外在定義。 如根據本發明所利用,除非另有指示,否則以下術語應理解具有以下含義。 如本文所用之術語「BAX/BCL-2調節劑」係指可調節BAX/BCL-2同二聚體之表現或活性之藥劑。 如本文所用之術語「烷基」係指具有1至6個碳原子、尤其1至4個碳原子之飽和直鏈或具支鏈烴基團,例如甲基、乙基、丙基、1-甲基乙基、丁基、1-甲基丙基、2-甲基丙基、1,1-二甲基乙基、戊基、1-甲基丁基、2-甲基丁基、3-甲基丁基、2,2-二甲基丙基、1-乙基丙基、己基、1,1-二甲基丙基、1,2-二甲基丙基、1-甲基戊基、2-甲基戊基、3-甲基戊基、4-甲基戊基、1,1-二甲基丁基、1,2-二甲基丁基、1,3-二甲基丁基、2,2-二甲基丁基、2,3-二甲基丁基、3,3-二甲基丁基、1-乙基丁基、2-乙基丁基、1,1,2-三甲基丙基、1,2,2-三甲基丙基、1-乙基-1-甲基丙基、1-乙基-2-甲基丙基。C1
-C4
-烷基意指(例如)甲基、乙基、丙基、1-甲基乙基、丁基、1-甲基丙基、2-甲基丙基或1,1-二甲基乙基。 如本文所用之術語「雜環基」係指具有5至8個環成員之單環基,其中在每一情形中該等環成員中之1、2、3或4者係彼此獨立地選自由氧、氮或硫組成之群之雜原子。 如本文所用之術語「預防(preventing, prevention)」係指延遲易感個體之症狀發作或減少疾病之發生。 如本文所用之術語「治療(treating, treatment)」表示減少及/或改良易感個體之症狀或增加患有某些致命病症或病況之個體的存活率。 如本文所用之術語「BAX/BCL-2相關病症或病況」表示其中BAX/BCL-2之調節有益之病症或病況。舉例而言,該等病症或病況包括癌症、骨髓增殖性病症、前列腺上皮發育不良、淋巴血管平滑肌肉增殖症、木村病及瘢痕瘤。 如本文所用之術語「個體」表示動物、尤其哺乳動物。在一個較佳實施例中,術語「個體」表示人類。在另一較佳實施例中,術語「個體」表示伴侶動物,例如貓或狗。 如本文所用之術語「治療有效量」係指活性成分單獨或與用於治療PPARγ相關病症或病況之其他治療/醫藥組合使用顯示治療功效之量。 術語「載劑」或「醫藥上可接受之載劑」係指可囊封活性醫藥成分之粒子。適用於本發明之載劑實例包括非離子體(niosome)、聚合體囊胞、奈米粒子、脂質體、奈米懸浮粒子、固體脂質奈米粒子、磁性奈米載劑、微胞、巨分子偶聯物、微粒藥物載劑及諸如此類。 除非上下文另外需要,否則單數術語將包括複數形式且複數術語將包括單數形式。 本發明之發明者驚訝地發現具有嘧啶并[5,4-d
]嘧啶結構之化合物可增強BAX/BCL-2同二聚體之表現及活性,且因此可充當新穎類型之BAX/BCL-2調節劑。在較佳實施例中,嘧啶并[5,4-d
]嘧啶化合物係雙嘧達莫。 因此,本發明提供預防或治療BAX/BCL-2相關病症或病況之方法,其包含向需要其之個體投與治療有效量之式(I)化合物:(I) 其中R1
、R2
、R3
及R4
中之每一者獨立地選自由雜環基及二(羥烷基)胺基組成之群, 或其醫藥上可接受之鹽。 在實施例中,R1
及R3
係雜環基、較佳六氫吡啶基,且R2
及R4
係二(羥烷基)胺基、較佳N,N-二(羥乙基)胺基。 在較佳實施例中,化合物係雙嘧達莫。 在另一實施例中,化合物囊封於載劑中,例如非離子體、聚合體囊胞、奈米粒子、脂質體、奈米懸浮粒子、固體脂質奈米粒子、磁性奈米載劑、微胞、巨分子偶聯物或微粒藥物載劑。 在較佳實施例中,載劑係脂質體。在另一實施例中,脂質體之直徑在約50-700 nm、較佳約60-530 nm、更佳80-350 nm且最佳約130-230 nm之範圍內。 此項技術中已知當雙嘧達莫以游離形式投與時,其結合至細胞膜上之ENT受體並活化阻斷腺苷進入細胞中之信號傳導路徑。發明者發現,雙嘧達莫可調控細胞凋亡/抗細胞凋亡蛋白之表現且藉助細胞穿透藥物遞送系統進入細胞中引起細胞凋亡。癌細胞細胞凋亡路徑之活化可促進已知與異常細胞凋亡/抗細胞凋亡蛋白表現相關聯之癌症的治療。 圖1a顯示若雙嘧達莫以游離形式投與,則其主要在細胞外部起作用且將促進腺苷之累積,此將導致減少之細胞凋亡。圖1b顯示在細胞內部之雙嘧達莫可活化細胞凋亡/抗細胞凋亡蛋白之表現。在此情形中,可避免雙嘧達莫在細胞外部之不合意作用。在下文之實例中,本發明證實藉由將雙嘧達莫直接遞送至細胞中,可避免結合至細胞膜上之受體以便減少副作用,例如氧化壓力及由腺苷累積造成之血管收縮。 在實施例中,本發明之式(I)化合物囊封於載劑中以遞送至細胞中。在較佳實施例中,載體係非離子體、聚合體囊胞、奈米粒子、脂質體、奈米懸浮粒子、固體脂質奈米粒子、磁性奈米載劑、微胞、巨分子偶聯物或微粒藥物載劑。較佳地,載體係脂質體。適用於本發明之脂質體具有在約10-300 nm、較佳約80-280 nm、更佳約120-270 nm範圍內之直徑。 在本發明之另一實施例中,載劑可為直徑小於1 mm之非離子體、聚合體囊胞或聚合物。可基於表面電位、親水性/疏水性、大小、形態、形狀及/或表面曲率進行修飾。 本發明之脂質體調配物可包含各種性質(例如,單層或多層)、組成、大小及特性之囊泡,包括不同組合、pH及滲透強度之水性介質在內。在較佳實施例中,脂質體脂質層膜之主要成分係選自由天然或合成磷脂組成之群,例如以下所列示之彼等: - 1,2-二月桂醯基-sn-甘油基-3-磷酸膽鹼(DLPC) - 1,2-二肉豆蔻醯基-sn-甘油基-3-磷酸膽鹼(DMPC) - 1,2-二棕櫚醯基-sn-甘油基-3-磷酸膽鹼(DPPC) - 1,2-二硬脂醯基-sn-甘油基-3-磷酸膽鹼(DSPC) - 1,2-二油醯基-sn-甘油基-3-磷酸膽鹼(DOPC) - 1,2-二肉豆蔻醯基-sn-甘油基-3-磷酸乙醇胺(DMPE) - 1,2-二棕櫚醯基-sn-甘油基-3-磷酸乙醇胺(DPPE) - 1,2-二硬脂醯基-sn-甘油基-3-磷酸乙醇胺(DSPE) - 1,2-二油醯基-sn-甘油基-3-磷酸乙醇胺(DOPE) - 1-肉豆蔻醯基-2-棕櫚醯基-sn-甘油基-3-磷酸膽鹼(MPPC) - 1-棕櫚醯基-2-肉豆蔻醯基-sn-甘油基-3-磷酸膽鹼(PMPC) - 1-硬脂醯基-2-棕櫚醯基-sn-甘油基-3-磷酸膽鹼(SPPC) - 1-棕櫚醯基-2-硬脂醯基-sn-甘油基-3-磷酸膽鹼(PSPC) - 1,2-二肉豆蔻醯基-sn-甘油基-3-[磷酸-rac-(1-甘油)] (DMPG) - 1,2-二棕櫚醯基-sn-甘油基-3-[磷酸-rac-(1-甘油)] (DPPG) - 1,2-二硬脂醯基-sn-甘油基-3-[磷酸-rac-(1-甘油)] (DSPG) - 1,2-二油醯基-sn-甘油基-3-[磷酸-rac-(1-甘油)] (DOPG) - 1,2-二肉豆蔻醯基-sn-甘油基-3-磷酸鹽(DMPA) - 1,2-二棕櫚醯基-sn-甘油基-3-磷酸鹽(DPPA) - 1,2-二棕櫚醯基-sn-甘油基-3-[磷酸-L-絲胺酸] (DPPS) - 磷脂醯絲胺酸(PS),及 - 天然L-a-磷脂醯膽鹼(來自雞蛋,EPC;或來自大豆、SPC;及HSPC)。 較佳磷脂係長飽和磷脂,例如彼等具有多於12、較佳多於14、更佳多於16、最佳多於18個碳原子之烷基鏈者。 用於本發明之較佳脂質體組合物較佳係彼等其中脂質體係單層及/或多層者,且包含: (i) 1至100、較佳40至70 mol%較佳選自由以下組成之群之生理上可接受之磷脂:DLPC、DMPC、DPPC、DSPC、DOPC、DMPE、DPPE、DSPE、DOPE、MPPC、PMPC、SPPC、PSPC、DMPG、DPPG、DSPG、DOPG、DMPA、DPPA、DPPS、PS、EPC、SPC及HSPC; (ii) 1至100、較佳40至70 mol%神經鞘脂質、較佳神經鞘磷脂; (iii) 1至100、較佳40至70 mol%表面活性劑,其較佳以疏水性烷基醚(例如,Brij)、烷基酯、聚山梨醇酯、山梨糖醇酯及/或烷基醯胺為特徵; (iv) 5至100、較佳50至100 mol%兩親性聚合物及/或共聚物,較佳包含至少一個親水性聚合物或共聚物之嵌段(例如聚乙二醇)及至少一個疏水性聚合物或共聚物之嵌段(例如聚(丙交酯)、聚(己內酯)、聚(環氧丁烷)、聚(氧化苯乙烯)、聚(苯乙烯)、聚(乙基乙烯)或聚二甲基矽氧烷)之嵌段共聚物, (v) 0至60 mol%、較佳20至50 mol% 毒素滯留增強化合物、較佳固醇衍生物、較佳膽固醇;或 (vi) 0至30 mol%、較佳1至5 mol%空間穩定劑、較佳聚乙二醇化化合物、較佳聚乙二醇化脂質、更佳DSPE-PEG。 在較佳實施例中,脂質體樣囊泡係自聚合物製得且出於脂質在形式上並不視為脂質體而是稱為聚合體囊胞之原因,不包含脂質。然而,出於本發明之目的,聚合體囊胞意欲由如定義本發明及申請專利範圍所用之術語脂質體涵蓋。 類似地,自合成表面活性劑製得且不包含脂質之脂質體樣囊泡稱為非離子體。然而,出於本發明之目的,非離子體意欲由如定義本發明及申請專利範圍所用之術語脂質體涵蓋。 在本發明之實施例中,可使用不同高分子聚合物之聚合,其包含呈三嵌段共聚物形式之彼等(例如ABA及BAB)及呈嵌段共聚物形式之彼等(例如PLLA-PEG、PLGA-PEG、PLA-PEG、PLLA-mPEG、PLGA-mPEG及PLA-mPEG)。可設計各種形狀(例如星形及L形式),其包括PEG-(PLGA)8
、PEG-(PLLA)8
及PEG-(PDLA)8
星形嵌段共聚物。聚乙二醇化修飾可用於修飾任何媒劑(例如聚合物媒劑及脂質體)以達成降低血漿蛋白之結合速率之效應(參見Park, J.等人,(2009) 「PEGylated PLGA nanoparticles for the improved delivery of doxorubicin. Nanomedicine.」 5(4):410-418.;Lück, M.等人,(1998) 「Plasma protein adsorption on biodegradable microspheres consisting of poly(D,L-lactide-co-glycolide), poly(L-lactide) or ABA triblock copolymers containing poly(oxyethylene). Influence of production method and polymer composition.」 J. Control Release. 55(2-3):107-20.;及Sempf, K.等人,(2013) 「Adsorption of plasma proteins on uncoated PLGA nanoparticles.」 Eur. J. Pharm. Biopharm. 85(1):53-60)。 動物劑量不應藉由基於體重之簡單轉換外推至人類等效劑量(HED)。食品暨藥物管理局(Food and Drug Administration)已提出動物劑量至人類劑量之外推僅藉助正規化之BSA (其通常以mg/m2表示)正確執行。人類劑量當量可藉由使用下式更適當地計算:HED (mg/kg) = 動物劑量(mg/kg)乘以動物Km/人類Km。為將小鼠中所用之劑量轉換為基於表面積之人類劑量,將22.4 mg/kg (鮑爾小鼠(Baur’s mouse)劑量)乘以小鼠之Km因子(3)且然後除以人類之Km因子(37) (參見下表)。 基於來自FDA指南草案之數據的值
為將以mg/kg表示之劑量轉換為以表示mg/m2
之劑量,乘以Km
值。根據本發明,脂質體-雙嘧達莫在小鼠中之有效劑量係10 mg/kg-100 mg/kg,在倉鼠中6-60 mg/kg,在大鼠中5-50 mg/kg,在天竺鼠中3.75-37.5 mg/kg,在兔子中2.5-25 mg/kg,在猴中2.5-25 mg/kg,在狗中1.5-15 mg/kg,在貓中2.4-24 mg/kg,在狒狒中1.5-15 mg/kg,在兒童中1.2-12 mg/kg,且在成年人中0.81-8.1 mg/kg。考慮到物種間之藥物敏感性差異,不限制物種之最寬劑量範圍係0.4-160 mg/kg、較佳0.6-120 mg/kg、更佳0.8 mg/kg-100 mg/kg。 在實施例中,雙嘧達莫脂質體可用於治療癌症。在實施例中,癌症係耐藥性或非耐藥性的。在實施例中,癌症係乳癌及肝癌。 在實施例中,雙嘧達莫脂質體可與其他抗癌藥物組合投與,例如烷基化劑、抗代謝藥、抗生素、激素、免疫調節劑、有絲分裂抑制劑、靶標治療劑及鉑藥物。在實施例中,抗癌藥物包含化學治療藥物及靶標藥物。在實施例中,化學治療藥物包含烷基化劑(例如環磷醯胺、甲基二(氯乙基)胺(Mechlorethamine)及美法侖(Melphalan))、抗有絲分裂劑(長春花鹼、長春新鹼(Vincristine)及紫杉醇(Taxol))、DNA嵌入劑(道諾黴素(Daunorubicin)及多柔比星)、DNA斷裂劑、抗代謝劑(例如卡培他濱(Capecitabine)、克拉屈濱(Cladribine)、阿糖胞苷(Cytarabine)、磷酸氟達拉濱(Fludarabine phosphate)、5-氟尿嘧啶(5-Fluorouracil)、吉西他濱(Gemcitabine)、6-巰嘌呤、胺甲喋呤(Methotrexate) (胺甲喋呤(Amethopterin),MTX)、米托蒽醌(Mitoxantron)、培美曲塞二鈉(Pemetrexed disodium) (七水合物)及替加氟(Tegafur) (FT-207))及拓樸異構酶抑制劑(例如喜樹鹼(Camptothecin)、拓撲替康(Topotecan)、伊立替康(Irinotecan)、鬼臼毒素(Podophyllotoxin)及依託泊苷)。在實施例中,靶標藥物包含曲妥珠單抗(Trastuzumab)、拉帕替尼(Lapatinib)、吉非替尼(Genfitinib)、埃羅替尼(Erlotinib)、西妥昔單抗(Cetuximab)、貝伐珠單抗(Bevacizumab)、利妥昔單抗(Rituximab)、硼替佐米(Bortizomib)、伊馬替尼(Imatinib)、舒尼替尼(Sunitinib)、雷帕黴素、西羅莫司、替西羅莫司、依維莫司、地磷莫司(Ridaforolimus) (德福羅莫司(deforolimus))及索拉非尼(Sorafenib)。在實施例中,其他抗癌藥物係5-氟尿嘧啶(5-FU)。 現已一般性闡述本發明,藉助參考以下實例可更容易地理解本發明,該等實例提供用於產生本發明醫藥組合物及其在治療癌症中之用途的例示性方案。提供實例僅出於說明性目的,且並不意欲以任何方式限制本發明之範圍。已努力確保所用數字(例如,量、溫度等)之準確性,但當然將允許一些實驗誤差及偏差。實例 實例 1 :雙嘧達莫脂質體之製備
脂質體係利用含有正電荷及中性電荷之磷脂及膽固醇製備,其中膽固醇之莫耳百分比為5%至75%。製備小的單層囊泡。將經乾燥脂質膜利用硫酸銨水合且隨後擠出穿過一系列聚碳酸酯膜過濾器。經由跨膜pH梯度或去水-再水合將雙嘧達莫囊封於脂質體中,且經由以上製造製程,所擠出脂質體之直徑在50-602 nm之範圍內(溶於葡萄糖溶液中)。脂質體-雙嘧達莫之直徑為約50至602 nm,如表1中所示。 表1實例 2 : BCL-2 蛋白家族之細胞存活力 (MTT 分析 ) 及西方墨點分析
癌細胞系培養 用於人類乳癌之培養基係L-15培養基。為獲得完成生長培養基,將以下組分添加至基礎培養基:胎牛血清至10%之最終濃度。將培養物在37℃無CO2
下培育。所測試癌細胞系包括MDA-MB-231、PANC-1、BXPC-3、HCT-116、HT29、A375及MeWo。 MTT (溴化3-(4,5-二甲基噻唑-2-基)-2,5-二苯基-四唑鎓鹽, Sigma, St. Louis)之儲積液係藉由將5 mg MTT/ml溶於磷酸鹽緩衝鹽水(pH 7.5)中製備並藉助0.22 mm過濾器過濾。所用MTT分析基本上類似於Mosmann (1983)初始所闡述者。簡言之,將100 mL MTT溶液(0.5 mg/mL)添加至所處理癌細胞系之單層並將微板於37℃下培育3小時。將100 mL DMSO添加至各孔並充分混合以溶解藍黑色晶體。在microELISA讀取器上使用570 nm之測試波長及630 nm之參考波長讀取板。 處理後,將細胞用150 mL RIPA細胞溶解緩衝液(10mM Hepes pH=7.9、1.5mM MgCl2、10mM KCl、1.0mM DTT、0.1% Triton-X 100)洗滌,並於1200 g在4℃下離心10分鐘。然後收集含有癌細胞總蛋白之上清液並藉由西方墨點進行分析。西方墨點方法係如下執行:使用Bradford分析量測蛋白質濃度;將6X試樣緩衝液(0.8 mM Tris-HCl、10 mM EDTA、10% SDS、60%甘油、0.6 M β-巰基乙醇、0.06%溴酚藍,pH 6.8)添加至50 μg全細胞蛋白中;並將等體積之溶解緩衝液添加至試樣。於95℃下加熱10分鐘以使蛋白質變性之後,立即將試樣於冰上冷卻。 然後將試樣藉由12% SDS-PAGE電泳(100 V)分離並藉由濕墨點自SDS-PAGE凝膠轉移至PVDF膜。然後將PVDF膜用5%脫脂奶在室溫下處理60分鐘以阻斷非特異性結合。將膜與初級抗體一起於4℃下培育過夜並用PBST洗滌三次。將膜與二級抗體一起於室溫下培育60分鐘並用PBST洗滌三次。然後將膜用PBS再洗滌一次並與增強之化學發光(ECL)受質一起培育用於檢測。使用自動化化學發光及螢光成像系統(UVP Biospectrum)獲得影像照片。實例 2.1 : 在用雙嘧達莫脂質體處理之 MCF-7 細胞中之細胞存活力及 Bax/BCL-2 比率
分析中所用之細胞系係人類乳癌細胞MCF-7。將細胞用雙嘧達莫脂質體(10 mM及20 mM)或雙嘧達莫游離形式(10 mM及20 mM)處理。結果顯示於圖4a及4b中,且正規化於圖5a及5b及表2中。 表2實例 3 : 在用雙嘧達莫脂質體與雷帕黴素之組合處理之 MDA-MB-231 細胞中之細胞存活力及 Bax/BCL-2 比率
分析中所用之細胞系係人類乳癌細胞MDA-MB-231。將細胞用雷帕黴素(100 mM)、雙嘧達莫脂質體(3.125 mM及25 mM)或雙嘧達莫游離形式(3.125 mM及25 mM)與雷帕黴素(100 mM)之組合處理。結果顯示於表3中。 表3實例 4 : 在用雙嘧達莫脂質體與雷帕黴素之組合處理之 PANC-1 細胞中之細胞存活力及 Bax/BCL-2 比率
分析中所用之細胞系係人類乳癌細胞PANC-1。將細胞用雷帕黴素(10 mM)、雙嘧達莫脂質體(3.125 mM及25 mM)或雙嘧達莫游離形式(3.125 mM及25 mM)與雷帕黴素(10 mM)之組合處理。結果顯示於表4中。 表4實例 5 : 在用雙嘧達莫脂質體與 5-Fu 之組合處理之 PANC-1 、 BxPC-3 、 HCT-116 、 HT29 、 A375 及 MeWo 細胞中之細胞存活力及 Bax/BCL-2 比率
將細胞用5-Fu (10 mM及100 mM)、雙嘧達莫脂質體(3.125 mM及25 mM)或雙嘧達莫游離形式(25 mM)與5-Fu (10 mM)之組合處理。結果顯示於表5中。 表5
此實例中之結果證實藉由使用雙嘧達莫脂質體與5-Fu之組合,該等藥物對胰臟癌、肝癌、乳癌及皮膚癌細胞之細胞毒性效應藉助平衡Bcl-2與Bax比率而增加。亦可減少該等藥物之劑量。舉例而言,5-Fu之劑量可減少至少8倍。 而且,藉助統計分析,發現在所測試之8種癌細胞系中,Bax/Bcl-2比率因雙嘧達莫脂質體處理之增加與癌細胞存活率負相關(R=-0.789,p
< 0.01)。此結果證明,雙嘧達莫與5-Fu在抑制各種類型癌細胞系之生長方面產生協同效應。而且,包含雙嘧達莫及5-Fu之醫藥組合物可增強5-Fu在癌細胞、尤其對5-Fu治療具有抗性之癌症中之細胞毒性效應(圖2)。實例 6 : 用雙嘧達莫脂質體與吉西他濱之組合處理之 A375 及 MeWo 細胞的細胞存活力
在A375及MeWo癌細胞系中,發現雙嘧達莫脂質體展現膜穿透效應。藉由此膜穿透效應,雙嘧達莫對吉西他濱之拮抗有效降低20-100%。此結果證明雙嘧達莫脂質體增加藥物穿透進入細胞中。結果顯示於表6中。細胞外雙嘧達莫阻斷ENT-1且阻止吉西他濱進入癌細胞中以產生其細胞毒性效應。此實例中之結果證實雙嘧達莫脂質體使此現象降低20-40%,此指示雙嘧達莫脂質體增強進入細胞之藥物的比率。 表6
其他化學治療藥物(例如圖3中所示之雷帕黴素類似物(rapamycin analogs, Rapalogs))可與本發明本發明之雙嘧達莫脂質體組合使用。 自以上數據,已發現本發明之雙嘧達莫脂質體當與化學治療藥物一起投與時在抑制癌細胞增殖方面產生協同效應,其中10-40%以上的細胞死亡(圖4)。癌細胞死亡之協同效應並非簡單地起因於增加Bax或降低Bcl-2,而是增加Bax/Bcl-2比率(圖5及6)。根據實例,雙嘧達莫在進入細胞中後使Bax/Bcl-2比率增加25-200%,且對癌細胞增殖之抑制增加20-50%。當與5-Fu組合時,雙嘧達莫藉助進入細胞中使5-50%之癌細胞增殖受到抑制,且使Bax/Bcl-2比率增加15-50%。雙嘧達莫與雷帕黴素之組合藉助進入細胞中使10-30%之癌細胞增殖受到抑制,且使Bax/Bcl-2比率增加10-200%。 對於熟悉此項技術者而言,預計將存在以上說明性實例中所闡述本發明之各種修改及變化。因此,僅將隨附申請專利範圍中出現之該等限制應用於本發明。Unless otherwise defined herein, the scientific and technical terms used in conjunction with the present invention shall have the meanings commonly understood by those familiar with the art. The meaning and scope of the terms should be clear; however, in the case of any potential ambiguity, the definitions provided in this article take precedence over any dictionary or external definitions. As utilized in accordance with the present invention, unless otherwise indicated, the following terms should be understood to have the following meanings. The term "BAX/BCL-2 modulator" as used herein refers to an agent that can modulate the performance or activity of the BAX/BCL-2 homodimer. The term "alkyl" as used herein refers to a saturated linear or branched hydrocarbon group having 1 to 6 carbon atoms, especially 1 to 4 carbon atoms, such as methyl, ethyl, propyl, 1-methyl Base ethyl, butyl, 1-methylpropyl, 2-methylpropyl, 1,1-dimethylethyl, pentyl, 1-methylbutyl, 2-methylbutyl, 3- Methylbutyl, 2,2-dimethylpropyl, 1-ethylpropyl, hexyl, 1,1-dimethylpropyl, 1,2-dimethylpropyl, 1-methylpentyl , 2-methylpentyl, 3-methylpentyl, 4-methylpentyl, 1,1-dimethylbutyl, 1,2-dimethylbutyl, 1,3-dimethylbutyl Base, 2,2-dimethylbutyl, 2,3-dimethylbutyl, 3,3-dimethylbutyl, 1-ethylbutyl, 2-ethylbutyl, 1,1, 2-trimethylpropyl, 1,2,2-trimethylpropyl, 1-ethyl-1-methylpropyl, 1-ethyl-2-methylpropyl. C 1 -C 4 -Alkyl means (for example) methyl, ethyl, propyl, 1-methylethyl, butyl, 1-methylpropyl, 2-methylpropyl or 1,1- Dimethyl ethyl. The term "heterocyclyl" as used herein refers to a monocyclic group having 5 to 8 ring members, wherein in each case 1, 2, 3, or 4 of the ring members are independently selected from each other Heteroatoms of the group consisting of oxygen, nitrogen or sulfur. The term "preventing, prevention" as used herein refers to delaying the onset of symptoms in susceptible individuals or reducing the occurrence of diseases. The term "treating, treatment" as used herein means reducing and/or improving the symptoms of susceptible individuals or increasing the survival rate of individuals suffering from certain fatal diseases or conditions. The term "BAX/BCL-2 related disorder or condition" as used herein means a disorder or condition in which the regulation of BAX/BCL-2 is beneficial. For example, these disorders or conditions include cancer, myeloproliferative disorders, prostatic epithelial dysplasia, lymphangiomyocytosis, Kimura disease, and keloids. The term "individual" as used herein means an animal, especially a mammal. In a preferred embodiment, the term "individual" means a human being. In another preferred embodiment, the term "individual" refers to a companion animal, such as a cat or a dog. The term "therapeutically effective amount" as used herein refers to the amount of the active ingredient used alone or in combination with other treatments/medicines for the treatment of PPARγ-related disorders or conditions to show therapeutic efficacy. The term "carrier" or "pharmaceutically acceptable carrier" refers to particles that can encapsulate active pharmaceutical ingredients. Examples of carriers suitable for the present invention include non-ionic bodies (niosome), polymeric vesicles, nanoparticles, liposomes, nano-suspended particles, solid lipid nanoparticles, magnetic nano-carriers, micelles, and macromolecules Conjugates, particulate drug carriers, and the like. Unless the context requires otherwise, singular terms shall include plural forms and plural terms shall include the singular. The inventors of the present invention surprisingly found that compounds with a pyrimido[5,4-d ]pyrimidine structure can enhance the performance and activity of BAX/BCL-2 homodimers, and therefore can serve as a novel type of BAX/BCL-2 Modifier. In a preferred embodiment, the pyrimido[5,4- d ]pyrimidine compound is dipyridamole. Therefore, the present invention provides a method for preventing or treating BAX/BCL-2 related disorders or conditions, which comprises administering to an individual in need thereof a therapeutically effective amount of a compound of formula (I): (I) wherein each of R 1 , R 2 , R 3 and R 4 is independently selected from the group consisting of a heterocyclic group and a di(hydroxyalkyl)amino group, or a pharmaceutically acceptable salt thereof. In the embodiment, R 1 and R 3 are heterocyclic groups, preferably hexahydropyridyl, and R 2 and R 4 are bis(hydroxyalkyl)amino groups, preferably N,N-bis(hydroxyethyl) Amine group. In a preferred embodiment, the compound is dipyridamole. In another embodiment, the compound is encapsulated in a carrier, such as nonionic body, polymeric vesicle, nanoparticle, liposome, nanosuspended particle, solid lipid nanoparticle, magnetic nanocarrier, micro Cells, macromolecular conjugates or microparticle drug carriers. In a preferred embodiment, the carrier is a liposome. In another embodiment, the diameter of the liposome is in the range of about 50-700 nm, preferably about 60-530 nm, more preferably 80-350 nm, and most preferably about 130-230 nm. It is known in the art that when dipyridamole is administered in a free form, it binds to the ENT receptor on the cell membrane and activates the signal transduction pathway that blocks the entry of adenosine into the cell. The inventors found that dipyridamole can regulate the expression of apoptosis/anti-apoptotic proteins and enter the cell via a cell penetrating drug delivery system to cause apoptosis. Activation of the apoptotic pathway of cancer cells can promote the treatment of cancers that are known to be associated with abnormal apoptosis/anti-apoptotic protein expression. Figure 1a shows that if dipyridamole is administered in a free form, it mainly acts on the outside of the cell and will promote the accumulation of adenosine, which will result in reduced apoptosis. Figure 1b shows the expression of dipyridamole activating apoptosis/anti-apoptotic protein inside the cell. In this case, the undesirable effect of dipyridamole on the outside of the cell can be avoided. In the following examples, the present invention demonstrates that by directly delivering dipyridamole to cells, binding to receptors on the cell membrane can be avoided to reduce side effects such as oxidative stress and vasoconstriction caused by adenosine accumulation. In the examples, the compound of formula (I) of the present invention is encapsulated in a carrier for delivery to cells. In a preferred embodiment, the carrier system is nonionic, polymeric vesicles, nanoparticles, liposomes, nano-suspended particles, solid lipid nanoparticles, magnetic nano-carriers, micelles, macromolecular conjugates Or particulate drug carriers. Preferably, the carrier system liposomes. Liposomes suitable for use in the present invention have a diameter in the range of about 10-300 nm, preferably about 80-280 nm, and more preferably about 120-270 nm. In another embodiment of the present invention, the carrier may be a non-ionic body, polymeric vesicle or polymer with a diameter of less than 1 mm. It can be modified based on surface potential, hydrophilicity/hydrophobicity, size, morphology, shape, and/or surface curvature. The liposome formulation of the present invention may contain vesicles of various properties (for example, unilamellar or multilamellar), composition, size and characteristics, including aqueous media of different combinations, pH and osmotic strength. In a preferred embodiment, the main component of the liposome lipid layer membrane is selected from the group consisting of natural or synthetic phospholipids, such as those listed below:-1,2-Dilauroyl-sn-glyceryl- Choline 3-phosphoric acid (DLPC)-1,2-Dimyristyl-sn-glyceryl-3-phosphocholine (DMPC)-1,2-Dipalmitoyl-sn-glyceryl-3-phosphate Choline (DPPC)-1,2-Distearyl-sn-glyceryl-3-phosphocholine (DSPC)-1,2-Dioleyl-sn-glyceryl-3-phosphocholine ( DOPC)-1,2-Dimyristyl-sn-glyceryl-3-phosphoethanolamine (DMPE)-1,2-Dipalmitoyl-sn-glyceryl-3-phosphoethanolamine (DPPE)-1, 2-Distearyl-sn-glyceryl-3-phosphoethanolamine (DSPE)-1,2-Dioleyl-sn-glyceryl-3-phosphoethanolamine (DOPE)-1-myristyl- 2-palmitoyl-sn-glyceryl-3-phosphocholine (MPPC)-1-palmitoyl-2-myristyl-sn-glyceryl-3-phosphocholine (PMPC)-1-hard Tallow-2-palmitoyl-sn-glyceryl-3-phosphocholine (SPPC)-1-palmitoyl-2-stearyl-sn-glyceryl-3-phosphocholine (PSPC) -1,2-Dimyristyl-sn-glyceryl-3-[phosphate-rac-(1-glyceryl)] (DMPG)-1,2-Dipalmitoyl-sn-glyceryl-3-[ Phosphoric acid-rac-(1-glycerol)] (DPPG)-1,2-Distearyl-sn-glyceryl-3-[phosphate-rac-(1-glycerol)] (DSPG)-1,2- Dioleyl-sn-glyceryl-3-[phosphate-rac-(1-glyceryl)] (DOPG)-1,2-Dimyristyl-sn-glyceryl-3-phosphate (DMPA)- 1,2-Dipalmitoyl-sn-glyceryl-3-phosphate (DPPA)-1,2-Dipalmitoyl-sn-glyceryl-3-[phospho-L-serine] (DPPS) -Phospholipid serine (PS), and-Natural La-phospholipid choline (from eggs, EPC; or from soybeans, SPC; and HSPC). Preferred phospholipids are long saturated phospholipids, for example, those having alkyl chains of more than 12, preferably more than 14, more preferably more than 16, and most preferably more than 18 carbon atoms. The preferred liposome compositions used in the present invention are preferably those in which the lipid system is unilamellar and/or multilamellar, and contains: (i) 1 to 100, preferably 40 to 70 mol%, preferably selected from the following composition Physiologically acceptable phospholipids of the group: DLPC, DMPC, DPPC, DSPC, DOPC, DMPE, DPPE, DSPE, DOPE, MPPC, PMPC, SPPC, PSPC, DMPG, DPPG, DSPG, DOPG, DMPA, DPPA, DPPS, PS, EPC, SPC and HSPC; (ii) 1 to 100, preferably 40 to 70 mol% sphingolipid, preferably sphingomyelin; (iii) 1 to 100, preferably 40 to 70 mol% surfactant, It is preferably characterized by hydrophobic alkyl ethers (for example, Brij), alkyl esters, polysorbates, sorbitol esters and/or alkyl amides; (iv) 5 to 100, preferably 50 to 100 mol% amphiphilic polymer and/or copolymer, preferably comprising at least one block of hydrophilic polymer or copolymer (e.g. polyethylene glycol) and at least one block of hydrophobic polymer or copolymer (e.g. Poly(lactide), poly(caprolactone), poly(butylene oxide), poly(styrene oxide), poly(styrene), poly(ethylethylene) or polydimethylsiloxane) (V) 0 to 60 mol%, preferably 20 to 50 mol%, a toxin retention enhancing compound, preferably a sterol derivative, preferably cholesterol; or (vi) 0 to 30 mol%, preferably 1 to 5 mol% steric stabilizer, preferably pegylated compound, preferably pegylated lipid, more preferably DSPE-PEG. In a preferred embodiment, liposome-like vesicles are made from polymers and do not contain lipids for the reason that lipids are not considered liposomes in form but are called polymeric vesicles. However, for the purposes of the present invention, polymeric vesicles are intended to be covered by the term liposome as used to define the scope of the present invention and patent applications. Similarly, liposome-like vesicles made from synthetic surfactants that do not contain lipids are called nonionic bodies. However, for the purposes of the present invention, nonionic bodies are intended to be covered by the term liposomes as used to define the scope of the present invention and patent applications. In the embodiment of the present invention, the polymerization of different high molecular polymers can be used, including those in the form of triblock copolymers (such as ABA and BAB) and those in the form of block copolymers (such as PLLA- PEG, PLGA-PEG, PLA-PEG, PLLA-mPEG, PLGA-mPEG and PLA-mPEG). Various shapes (such as star and L forms) can be designed, including PEG-(PLGA) 8 , PEG-(PLLA) 8 and PEG-(PDLA) 8 star block copolymers. PEGylation modification can be used to modify any vehicle (such as polymer vehicles and liposomes) to achieve the effect of reducing the binding rate of plasma proteins (see Park, J. et al., (2009) "PEGylated PLGA nanoparticles for the improved" delivery of doxorubicin. Nanomedicine."5(4):410-418.; Lück, M. et al., (1998) "Plasma protein adsorption on biodegradable microspheres consisting of poly(D,L-lactide-co-glycolide), poly (L-lactide) or ABA triblock copolymers containing poly(oxyethylene). Influence of production method and polymer composition." J. Control Release. 55(2-3):107-20.; and Sempf, K. et al., ( 2013) "Adsorption of plasma proteins on uncoated PLGA nanoparticles." Eur. J. Pharm. Biopharm. 85(1):53-60). Animal doses should not be extrapolated to human equivalent doses (HED) by simple conversion based on body weight. The Food and Drug Administration (Food and Drug Administration) has proposed that the extrapolation of animal doses to human doses can only be carried out correctly with the help of normalized BSA (which is usually expressed in mg/m2). The human dose equivalent can be calculated more appropriately by using the following formula: HED (mg/kg) = animal dose (mg/kg) multiplied by animal Km/human Km. To convert the dose used in mice to a human dose based on surface area, 22.4 mg/kg (Baur's mouse dose) was multiplied by the mouse Km factor (3) and then divided by the human Km factor (37) (See the table below). The value based on the data from the draft FDA guideline is to convert the dose expressed in mg/kg to the dose expressed in mg/m 2 and multiply it by the K m value. According to the present invention, the effective dose of liposome-dipyridamole is 10 mg/kg-100 mg/kg in mice, 6-60 mg/kg in hamsters, and 5-50 mg/kg in rats. 3.75-37.5 mg/kg in guinea pigs, 2.5-25 mg/kg in rabbits, 2.5-25 mg/kg in monkeys, 1.5-15 mg/kg in dogs, 2.4-24 mg/kg in cats, 1.5-15 mg/kg in baboons, 1.2-12 mg/kg in children, and 0.81-8.1 mg/kg in adults. Taking into account the differences in drug sensitivity between species, the widest dose range without limiting species is 0.4-160 mg/kg, preferably 0.6-120 mg/kg, more preferably 0.8 mg/kg-100 mg/kg. In an embodiment, dipyridamole liposomes can be used to treat cancer. In an embodiment, the cancer is drug-resistant or non-drug-resistant. In the examples, the cancer is breast cancer and liver cancer. In an embodiment, dipyridamole liposomes can be administered in combination with other anticancer drugs, such as alkylating agents, antimetabolites, antibiotics, hormones, immunomodulators, mitotic inhibitors, target therapeutic agents, and platinum drugs. In an embodiment, the anti-cancer drugs include chemotherapeutic drugs and target drugs. In an embodiment, the chemotherapeutic drugs include alkylating agents (such as cyclophosphamide, methyl bis(chloroethyl) amine (Mechlorethamine) and Melphalan), anti-mitotic agents (vinblastine, vinblastine) Vincristine and Taxol), DNA intercalators (Daunorubicin and Doxorubicin), DNA fragmentation agents, antimetabolites (e.g. Capecitabine, Cladribine) (Cladribine), Cytarabine, Fludarabine phosphate, 5-Fluorouracil, Gemcitabine, 6-mercaptopurine, Methotrexate (amine Methotrexate (Amethopterin, MTX), Mitoxantron, Pemetrexed disodium (heptahydrate) and Tegafur (FT-207)) and Topology Structure enzyme inhibitors (such as Camptothecin, Topotecan, Irinotecan, Podophyllotoxin and Etoposide). In an embodiment, the target drug includes Trastuzumab (Trastuzumab), Lapatinib (Lapatinib), Gefitinib (Genfitinib), Erlotinib (Erlotinib), Cetuximab (Cetuximab), Bevacizumab (Bevacizumab), Rituximab (Rituximab), Bortizomib (Bortizomib), Imatinib (Imatinib), Sunitinib (Sunitinib), Rapamycin, Sirolimus, Tesirolimus, Everolimus, Ridaforolimus (deforolimus) and Sorafenib. In the embodiment, the other anticancer drug is 5-fluorouracil (5-FU). Now that the present invention has been described in general, the present invention can be more easily understood by referring to the following examples, which provide exemplary solutions for producing the pharmaceutical composition of the present invention and its use in the treatment of cancer. The examples are provided for illustrative purposes only, and are not intended to limit the scope of the present invention in any way. Efforts have been made to ensure the accuracy of the numbers used (for example, quantity, temperature, etc.), but of course some experimental errors and deviations will be allowed. Examples Example 1 : Preparation of dipyridamole liposomes The lipid system is prepared using phospholipids and cholesterol containing positive and neutral charges, wherein the molar percentage of cholesterol is 5% to 75%. Prepare small unilamellar vesicles. The dried lipid membrane was hydrated with ammonium sulfate and then extruded through a series of polycarbonate membrane filters. Dipyridamole is encapsulated in liposomes through transmembrane pH gradient or dehydration-rehydration, and through the above manufacturing process, the diameter of the extruded liposomes is in the range of 50-602 nm (dissolved in glucose solution) ). The diameter of liposome-dipyridamole is about 50 to 602 nm, as shown in Table 1. Table 1 Example 2 : Cell viability of the BCL-2 protein family (MTT analysis ) and Western blot analysis The culture medium used for human breast cancer is L-15 medium. To obtain a complete growth medium, the following components are added to the basal medium: Fetal Bovine Serum to a final concentration of 10%. The culture was incubated at 37°C without CO 2 . The cancer cell lines tested included MDA-MB-231, PANC-1, BXPC-3, HCT-116, HT29, A375 and MeWo. MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-tetrazolium bromide, Sigma, St.Louis) storage solution is obtained by adding 5 mg MTT /ml was prepared in phosphate buffered saline (pH 7.5) and filtered through a 0.22 mm filter. The MTT analysis used is basically similar to that originally described by Mosmann (1983). Briefly, 100 mL of MTT solution (0.5 mg/mL) was added to the monolayer of the treated cancer cell line and the microplate was incubated at 37°C for 3 hours. Add 100 mL of DMSO to each well and mix well to dissolve the blue-black crystals. Use the test wavelength of 570 nm and the reference wavelength of 630 nm to read the plate on the microELISA reader. After treatment, the cells were washed with 150 mL RIPA cell lysis buffer (10mM Hepes pH=7.9, 1.5mM MgCl2, 10mM KCl, 1.0mM DTT, 0.1% Triton-X 100), and centrifuged at 1200 g at 4°C for 10 minute. Then the supernatant containing the total protein of cancer cells was collected and analyzed by western blotting. The Western blot method is implemented as follows: use Bradford analysis to measure protein concentration; mix 6X sample buffer (0.8 mM Tris-HCl, 10 mM EDTA, 10% SDS, 60% glycerol, 0.6 M β-mercaptoethanol, 0.06% Bromophenol blue, pH 6.8) was added to 50 μg of whole cell protein; and an equal volume of lysis buffer was added to the sample. After heating at 95°C for 10 minutes to denature the protein, immediately cool the sample on ice. The sample was then separated by 12% SDS-PAGE electrophoresis (100 V) and transferred from the SDS-PAGE gel to the PVDF membrane by wet ink dots. The PVDF membrane was then treated with 5% skim milk at room temperature for 60 minutes to block non-specific binding. The membrane was incubated with the primary antibody overnight at 4°C and washed three times with PBST. The membrane was incubated with the secondary antibody for 60 minutes at room temperature and washed three times with PBST. The membrane was then washed again with PBS and incubated with enhanced chemiluminescence (ECL) substrate for detection. Use automated chemiluminescence and fluorescence imaging system (UVP Biospectrum) to obtain image photos. Example 2.1 : The cell line used in the analysis of cell viability and Bax/BCL-2 ratio in MCF-7 cells treated with dipyridamole liposomes is the human breast cancer cell line MCF-7. The cells were treated with dipyridamole liposomes (10 mM and 20 mM) or the free form of dipyridamole (10 mM and 20 mM). The results are shown in Figures 4a and 4b, and normalized in Figures 5a and 5b and Table 2. Table 2 Example 3 : Cell line human breast cancer cells used in the analysis of cell viability and Bax/BCL-2 ratio in MDA-MB-231 cells treated with the combination of dipyridamole liposome and rapamycin MDA-MB-231. Combine cells with rapamycin (100 mM), liposomes of dipyridamole (3.125 mM and 25 mM) or the free form of dipyridamole (3.125 mM and 25 mM) and rapamycin (100 mM) deal with. The results are shown in Table 3. Table 3 Example 4 : The cell line used in the analysis of cell viability and Bax/BCL-2 ratio in PANC-1 cells treated with the combination of dipyridamole liposome and rapamycin, human breast cancer cell line PANC- 1. Combine cells with rapamycin (10 mM), liposomes of dipyridamole (3.125 mM and 25 mM) or the free form of dipyridamole (3.125 mM and 25 mM) and rapamycin (10 mM) deal with. The results are shown in Table 4. Table 4 Example 5 : Cell viability and Bax/BCL-2 in PANC-1 , BxPC-3 , HCT-116 , HT29 , A375 and MeWo cells treated with the combination of dipyridamole liposomes and 5-Fu the ratio of cells with 5-Fu (10 mM and 100 mM), dipyridamole liposomes (3.125 mM and 25 mM) in free form or dipyridamole (25 mM) and 5-Fu (10 mM) of combined treatment . The results are shown in Table 5. Table 5 The results in this example confirm that by using the combination of dipyridamole liposomes and 5-Fu, the cytotoxic effects of these drugs on pancreatic cancer, liver cancer, breast cancer and skin cancer cells balance Bcl-2 and Bax The ratio increases. The dosage of these drugs can also be reduced. For example, the dose of 5-Fu can be reduced by at least 8 times. Moreover, with the help of statistical analysis, it was found that among the 8 cancer cell lines tested, the increase in the ratio of Bax/Bcl-2 due to dipyridamole liposome treatment was negatively correlated with the survival rate of cancer cells (R=-0.789, p <0.01 ). This result proves that dipyridamole and 5-Fu have a synergistic effect in inhibiting the growth of various types of cancer cell lines. Moreover, the pharmaceutical composition containing dipyridamole and 5-Fu can enhance the cytotoxic effect of 5-Fu in cancer cells, especially cancers resistant to 5-Fu treatment (Figure 2). Example 6 : Cell viability of A375 and MeWo cells treated with a combination of dipyridamole liposomes and gemcitabine In A375 and MeWo cancer cell lines, it was found that dipyridamole liposomes exhibited a membrane penetration effect. With this membrane penetration effect, the antagonism of dipyridamole to gemcitabine is effectively reduced by 20-100%. This result proves that dipyridamole liposomes increase drug penetration into cells. The results are shown in Table 6. Extracellular dipyridamole blocks ENT-1 and prevents gemcitabine from entering cancer cells to produce its cytotoxic effect. The results in this example confirm that dipyridamole liposomes reduce this phenomenon by 20-40%, which indicates that dipyridamole liposomes enhance the rate of drug entering cells. Table 6 Other chemotherapeutic drugs (such as rapamycin analogs (Rapalogs) shown in Figure 3) can be used in combination with the dipyridamole liposomes of the present invention. From the above data, it has been found that the dipyridamole liposome of the present invention produces a synergistic effect in inhibiting the proliferation of cancer cells when administered with chemotherapeutic drugs, in which more than 10-40% of the cells die (Figure 4). The synergistic effect of cancer cell death is not simply caused by increasing Bax or decreasing Bcl-2, but increasing the Bax/Bcl-2 ratio (Figures 5 and 6). According to an example, dipyridamole increases the Bax/Bcl-2 ratio by 25-200% after entering the cell, and increases the inhibition of cancer cell proliferation by 20-50%. When combined with 5-Fu, Dipyridamole inhibits the proliferation of 5-50% of cancer cells by entering the cell and increases the Bax/Bcl-2 ratio by 15-50%. The combination of dipyridamole and rapamycin inhibits the proliferation of 10-30% of cancer cells by entering the cell and increases the ratio of Bax/Bcl-2 by 10-200%. For those familiar with the art, it is expected that there will be various modifications and changes of the present invention described in the above illustrative examples. Therefore, only the limitations appearing in the scope of the attached patent application are applied to the present invention.
圖1a及1b係顯示雙嘧達莫(a)在細胞外部及(b)細胞內部對癌細胞凋亡之不同作用的示意圖。 圖2證實細胞存活力與Bax/Bcl-2比率之間之關係(Y軸:細胞存活力;X軸:Bax/Bcl-2比率)。 圖3顯示代表性雷帕黴素類似物(Rapalogs)之結構。 圖4a及4b顯示在有/沒有5-FU下藉由雙嘧達莫處理之三陰性乳癌細胞系MDA-MB-231的細胞存活力。 圖5a及5b顯示在有/沒有5-FU下藉由雙嘧達莫處理之癌細胞之Bax、Bcl-2及Bcl-xL表現。 圖6a及6b顯示在有/沒有5-FU下藉由雙嘧達莫處理之細胞中Bax/Bcl-2比率。Figures 1a and 1b are schematic diagrams showing the different effects of dipyridamole (a) outside the cell and (b) inside the cell on the apoptosis of cancer cells. Figure 2 confirms the relationship between cell viability and Bax/Bcl-2 ratio (Y-axis: cell viability; X-axis: Bax/Bcl-2 ratio). Figure 3 shows the structure of representative rapamycin analogs (Rapalogs). Figures 4a and 4b show the cell viability of the triple-negative breast cancer cell line MDA-MB-231 treated with dipyridamole with or without 5-FU. Figures 5a and 5b show the performance of Bax, Bcl-2 and Bcl-xL in cancer cells treated with dipyridamole with or without 5-FU. Figures 6a and 6b show the ratio of Bax/Bcl-2 in cells treated with dipyridamole with or without 5-FU.