徐詣芝 張劼 楊海燕
中圖分類(lèi)號(hào) R730.51;R730.53;R969.2 文獻(xiàn)標(biāo)志碼 A 文章編號(hào) 1001-0408(2022)06-0758-06
DOI 10.6039/j.issn.1001-0408.2022.06.19
摘 要 腎素-血管緊張素系統(tǒng)(RAS)調(diào)節(jié)劑包括腎素抑制劑、血管緊張素轉(zhuǎn)化酶抑制劑、血管緊張素Ⅱ受體阻滯劑、血管緊張素Ⅱ受體激動(dòng)劑和血管緊張素1-7等。本文通過(guò)檢索1992年1月-2021年6月發(fā)表的相關(guān)文獻(xiàn),就RAS調(diào)節(jié)劑對(duì)抗腫瘤藥的輔助作用進(jìn)行匯總和分析。RAS調(diào)節(jié)劑可減輕抗腫瘤藥的心臟毒性、血液學(xué)毒性、周?chē)窠?jīng)毒性等,并具有腎臟保護(hù)作用;其聯(lián)合化療藥物可促進(jìn)藥物體內(nèi)遞送,聯(lián)合靶向藥物可抑制血管生成并改善旁路激活,聯(lián)合免疫檢查點(diǎn)抑制劑可增強(qiáng)腫瘤免疫反應(yīng)進(jìn)而提高抗腫瘤藥的療效。含RAS調(diào)節(jié)劑和抗腫瘤藥的聯(lián)合治療模式有望減少抗腫瘤藥的副作用、增強(qiáng)其療效,并改善患者預(yù)后。
關(guān)鍵詞 腎素-血管緊張素系統(tǒng);調(diào)節(jié)劑;惡性腫瘤;靶向治療;化學(xué)治療;免疫治療
Review on the adjuvant effects of renin-angiotensin system regulators on antitumor drugs
XU Yizhi1,ZHANG Jie2,YANG Haiyan2(1. Dept. of Hematology, Chongqing General Hospital, Chongqing? 400014, China; 2. Dept. of Geriatrics, Chongqing General Hospital, Chongqing 400014, China)
ABSTRACT? ?The regulators of renin-angiotensin system (RAS) include renin inhibitors, angiotensin converting enzyme inhibitors, angiotensin Ⅱ receptor blockers, angiotensin Ⅱ receptor agonists and angiotensin 1-7. This paper summarizes and analyzes the adjuvant effects of RAS regulators on antitumor drugs by searching the literature published from January 1992 to June 2021. The regulators of RAS can reduce the cardiotoxicity, hematological toxicity and peripheral neurotoxicity of antitumor drugs, and has renal protective effect; the regulators of RAS combined with other chemotherapy drugs show favorable effects on promoting chemotherapeutic drugs delivery, improving anti-angiogenesis and bypass activation of targeted drugs, enhancing tumor immune response of immune checkpoint inhibitors, so as to improve therapeutic efficacy of antitumor drugs. The combination of RAS regulators with antitumor drugs is expected to reduce the side effects of antitumor drugs, enhance its efficacy and improve the prognosis of patients.
KEYWORDS? ?renin-angiotensin system; regulator; malignant tumor; targeted treatment; chemotherapy; immunotherapy
惡性腫瘤的藥物治療,包括化療、靶向治療和免疫治療,但常因毒副作用不能耐受、療效不盡人意而導(dǎo)致腫瘤不可控。腎素-血管緊張素系統(tǒng)(renin-angiotensin system,RAS)不僅與惡性腫瘤的發(fā)生發(fā)展密切相關(guān),其調(diào)節(jié)劑還可輔助抗腫瘤藥起到減毒增效的作用[1]。RAS調(diào)節(jié)劑包括腎素抑制劑、血管緊張素轉(zhuǎn)化酶抑制劑(angiotension converting enzyme inhibitors,ACEIs)、血管緊張素Ⅱ受體阻滯劑(angiotensin Ⅱ receptor blockers,ARBs)、血管緊張素Ⅱ受體(angiotensin Ⅱ receptor,AT2R)激動(dòng)劑和血管緊張素1-7(angiotensin 1-7,Ang 1-7)等。
一項(xiàng)納入了55項(xiàng)研究的薈萃分析顯示,ACEIs/ARBs的應(yīng)用使惡性腫瘤患者的總生存時(shí)間(overall survival,OS)和無(wú)進(jìn)展生存時(shí)間(progression-free survival,PFS)均顯著延長(zhǎng)[風(fēng)險(xiǎn)比(hazard ratio,HR)=0.82,95%置信區(qū)間(confidence interval,CI)(0.77,0.88),P<0.001;HR=0.74,95%CI(0.66,0.84),P<0.001];亞組分析結(jié)果顯示,與未使用ACEIs/ARBs者相比,使用ARBs者的OS顯著延長(zhǎng)[HR=0.80,95%CI(0.67,0.95),P=0.01],而使用ACEIs者的OS略有延長(zhǎng),但差異無(wú)統(tǒng)計(jì)學(xué)意義[HR=0.94,95%CI(0.85,1.04),P=0.27][2]。為了探索RAS調(diào)節(jié)劑對(duì)抗腫瘤藥的影響,筆者以“血管緊張素”“化學(xué)治療”“靶向治療”“免疫治療”“癌癥”“骨髓”“造血”“白血病”“淋巴瘤”“骨髓瘤”“angiotensin”“chemotherapy”“target therapy”“immunotherapy”“cancer”“carcinoma”“neoplasms”“marrow”“hematopoiesis”“l(fā)eukemia”“l(fā)ymphoma”“myeloma”等為關(guān)鍵詞,在中國(guó)知網(wǎng)、萬(wàn)方數(shù)據(jù)、維普網(wǎng)、PubMed、Embase等數(shù)據(jù)庫(kù)中組合查詢了1992年1月-2021年6月發(fā)表的相關(guān)文獻(xiàn)849篇,選取其中有效文獻(xiàn)49篇,對(duì)RAS調(diào)節(jié)劑對(duì)抗腫瘤藥的輔助作用進(jìn)行匯總分析,以探索腫瘤治療中新的藥物聯(lián)合治療模式,從而更好地達(dá)到減少毒副作用、提高治療效果、改善疾病預(yù)后的目的。
1 RAS調(diào)節(jié)劑對(duì)抗腫瘤藥的輔助作用
RAS調(diào)節(jié)劑對(duì)抗腫瘤藥的影響,一方面在于其對(duì)患者器官的保護(hù)作用,可減少抗腫瘤藥的毒副作用[3-4];另一方面可能在于其增加了抗腫瘤藥的療效[5],從而起到了減毒增效的作用。
1.1 減少抗腫瘤藥的毒副作用
研究發(fā)現(xiàn),RAS調(diào)節(jié)劑可減輕抗腫瘤藥的心臟毒性、血液學(xué)毒性和周?chē)窠?jīng)毒性等常見(jiàn)的毒副作用,有利于標(biāo)準(zhǔn)抗腫瘤治療方案順利、足量、按時(shí)完成,延長(zhǎng)患者OS,提高患者生活質(zhì)量,并有助于降低醫(yī)療成本[3-4]。
1.1.1 減少抗腫瘤藥的心臟毒性 局部RAS首先被發(fā)現(xiàn)存在于心肌細(xì)胞上[6]。已有文獻(xiàn)證實(shí),RAS調(diào)節(jié)劑(如ACEIs、ARBs)不僅可阻斷血管緊張素Ⅱ(angiotensin Ⅱ,Ang Ⅱ)的心肌損傷作用,還可促進(jìn)Ang 1-7的生成,從而起到心臟保護(hù)作用[6]??鼓[瘤藥心臟毒性的發(fā)生機(jī)制主要在于藥物導(dǎo)致的活性氧(reactive oxygen species,ROS)生成、拓?fù)洚悩?gòu)酶2β誘導(dǎo)的氧化應(yīng)激均可使心肌細(xì)胞受到損傷;或是影響原癌基因人類(lèi)表皮生長(zhǎng)因子受體2(human epidermal growth factor receptor-2,HER2)/HER4信號(hào)轉(zhuǎn)導(dǎo),減少了心臟保護(hù)性HER2/HER4異二聚體的形成,最終導(dǎo)致病理性心臟重塑[3-4]。誘發(fā)心臟毒性的抗腫瘤藥包括HER2抑制劑(如曲妥珠單抗、帕妥珠單抗和拉帕替尼等)和蒽環(huán)類(lèi)、紫杉烷類(lèi)、氟尿嘧啶類(lèi)化療藥物等。
RAS在心肌細(xì)胞應(yīng)激反應(yīng)中發(fā)揮著重要作用。Ang Ⅱ可促進(jìn)氧化應(yīng)激,影響HER2/HER4信號(hào)轉(zhuǎn)導(dǎo),導(dǎo)致心臟重塑、心肌細(xì)胞肥大和纖維化。RAS調(diào)節(jié)劑可通過(guò)改善Ang Ⅱ誘導(dǎo)的病理性心肌重塑來(lái)預(yù)防抗腫瘤藥的心臟毒性[3,5]。大量臨床研究發(fā)現(xiàn),在乳腺癌患者使用蒽環(huán)類(lèi)藥物和/或曲妥珠單抗治療期間聯(lián)合ACEIs/ARBs可減輕心臟毒性,減緩抗腫瘤藥介導(dǎo)的左室射血分?jǐn)?shù)下降:MANTICORE 101-breast研究為一項(xiàng)雙盲、多中心、隨機(jī)對(duì)照研究,納入了接受曲妥珠單抗治療的94例乳腺癌患者,結(jié)果發(fā)現(xiàn),與單用曲妥珠單抗治療比較,聯(lián)用培哚普利能減緩曲妥珠單抗導(dǎo)致的左室射血分?jǐn)?shù)下降[(-5±5)%vs.(-3±4)%,P=0.001],且培哚普利是維持患者左室射血功能的獨(dú)立預(yù)測(cè)因子[7];另一項(xiàng)納入了468例乳腺癌患者的雙盲、多中心、隨機(jī)對(duì)照研究顯示,賴(lài)諾普利降低了接受蒽環(huán)類(lèi)藥物聯(lián)合曲妥珠單抗治療的患者心臟毒性的發(fā)生率(37%vs. 47%,P=0.011),減少了曲妥珠單抗治療中斷的次數(shù)[8];PRADA研究為一項(xiàng)2×2析因、雙盲、隨機(jī)對(duì)照研究,納入了130例無(wú)嚴(yán)重并發(fā)癥的早期乳腺癌成年女性患者,給予坎地沙坦聯(lián)合曲妥珠單抗和/或蒽環(huán)類(lèi)藥物治療,抗腫瘤治療后停用坎地沙坦,結(jié)果顯示,坎地沙坦組患者的左室射血分?jǐn)?shù)下降率顯著低于安慰劑組(0.8%vs. 2.6%,P=0.026)[9];近期該研究更新了隨訪數(shù)據(jù)(中位隨訪23個(gè)月),結(jié)果顯示,抗腫瘤治療期間給予的坎地沙坦并不能阻止抗腫瘤藥治療2年后患者左室射血分?jǐn)?shù)的降低,這可能與坎地沙坦用藥時(shí)間不足有關(guān)[10];一項(xiàng)針對(duì)2001-2009年新診斷的6 542例66歲及以上患者的真實(shí)世界數(shù)據(jù)隊(duì)列研究顯示,隨訪5年內(nèi),ACEIs使用者的心臟毒性發(fā)生率[HR=0.77,95%CI(0.62,0.95),P<0.05]和全因死亡率[HR=0.79,95%CI(0.70,0.90),P<0.05]均較低[11]。
除了臨床和真實(shí)世界數(shù)據(jù)研究,臨床前實(shí)驗(yàn)也在不斷探索RAS調(diào)節(jié)劑減少抗腫瘤藥心臟毒性的劑量、組合和應(yīng)用模式:Olorundare等[12]開(kāi)展的一項(xiàng)動(dòng)物實(shí)驗(yàn)研究結(jié)果表明,單用賴(lài)諾普利、纈沙坦或者兩藥聯(lián)用均可逆轉(zhuǎn)曲妥珠單抗致心臟損傷相關(guān)實(shí)驗(yàn)室和病理指標(biāo)的變化;一項(xiàng)動(dòng)物實(shí)驗(yàn)還發(fā)現(xiàn),除了ACEIs和ARBs,腎素抑制劑阿利吉侖對(duì)他克莫司所致大鼠心臟毒性也具有對(duì)抗作用[13]。然而,腎素抑制劑雖可更高效地減少Ang Ⅱ的生成,但也可同時(shí)減少Ang 1-7的生成,故其對(duì)心臟的保護(hù)作用可能有限[13]。這方面的研究報(bào)道少,且近年無(wú)進(jìn)一步的基礎(chǔ)研究及臨床數(shù)據(jù),尚待進(jìn)一步驗(yàn)證。
1.1.2 促進(jìn)化療后骨髓抑制的恢復(fù) 在造血系統(tǒng)亦發(fā)現(xiàn)骨髓RAS的存在,其能參與并調(diào)控原始和成熟血細(xì)胞的生成[14]。有研究指出,活性肽Ang Ⅱ促進(jìn)了骨髓、臍血造血祖細(xì)胞向紅系祖細(xì)胞的分化及擴(kuò)增,外源性給予Ang Ⅱ能促進(jìn)小鼠體內(nèi)造血干細(xì)胞(hematopoietic stem cell,HSC)的增殖和分化[15]?;钚噪腁ng 1-7除了通過(guò)激活Mas受體而具有對(duì)抗調(diào)節(jié)Ang Ⅱ病理作用的能力外,還能夠模擬Ang Ⅱ而發(fā)揮促進(jìn)HSC分化的作用[16]。當(dāng)化療藥物引起骨髓抑制時(shí),HSC的Mas受體表達(dá)會(huì)明顯增高,Ang 1-7可與之結(jié)合,并通過(guò)激活Janus激酶2(Janus kinase 2,JAK2)來(lái)增加化療后骨髓中髓系細(xì)胞、巨核細(xì)胞和紅系祖細(xì)胞的濃度;且Ang 1-7與重組人粒細(xì)胞集落刺激因子(recombinant human granulocyte colony stimulating factor,rhG-CSF)、重組人促紅細(xì)胞生成素(recombinant human erythropoietin,rhEPO)等重組人造血因子有協(xié)同作用,能促進(jìn)化療后患者骨髓抑制的恢復(fù)[16]。一項(xiàng)針對(duì)卵巢癌的Ⅱb期臨床研究結(jié)果表明,患者經(jīng)吉西他濱+鉑類(lèi)雙藥化療后再皮下注射Ang 1-7 100 mg/kg,可降低化療后血小板減少的發(fā)生率及嚴(yán)重程度[17]。Nle3-Ang 1-7是用正亮氨酸替代Ang 1-7第3位纈氨酸所得的Ang 1-7類(lèi)似物,其外用時(shí)可顯示出比Ang 1-7更強(qiáng)的促進(jìn)傷口愈合的能力[18]。為了提高臨床用藥的便利性,Gaffney等[19]以吉西他濱誘導(dǎo)的骨髓抑制模型小鼠為對(duì)象,比較了由β-環(huán)糊精制成的Ang 1-7、Nle3-Ang 1-7口服制劑與Ang 1-7、Nle3-Ang 1-7皮下注射劑對(duì)小鼠骨髓抑制的恢復(fù)能力,結(jié)果顯示,兩者效果相當(dāng)。
1.1.3 減少抗腫瘤藥的周?chē)窠?jīng)毒性 雖然RAS在大腦中的功能目前尚未完全清楚,但越來(lái)越多的研究表明該系統(tǒng)與神經(jīng)性疼痛有關(guān)[20]。在與疼痛傳遞、調(diào)節(jié)和感知的大腦相關(guān)區(qū)域(如丘腦和下丘腦核、藍(lán)斑、中央杏仁核和孤束核)中均發(fā)現(xiàn)了AT1R和AT2R的存在[21-22]。RAS調(diào)節(jié)劑可為接受具有周?chē)窠?jīng)毒性化療藥物治療的患者提供神經(jīng)保護(hù)作用[23]。一項(xiàng)基于紫杉醇致痛覺(jué)過(guò)敏大鼠模型的基礎(chǔ)研究顯示,氯沙坦可通過(guò)抑制背根神經(jīng)節(jié)炎癥介質(zhì)的表達(dá)來(lái)減輕化療引起的神經(jīng)性疼痛[24]。在長(zhǎng)春新堿致機(jī)械性異常性疼痛的小鼠模型研究中,坎地沙坦和AT2R激動(dòng)劑被證實(shí)均可促進(jìn)小鼠的觸覺(jué)敏感性完全恢復(fù)正常,但其中只有AT2R激動(dòng)劑顯示出針對(duì)長(zhǎng)春新堿致神經(jīng)元損傷的神經(jīng)保護(hù)作用[25]。Bessaguet等[26]的研究顯示,坎地沙坦預(yù)防功能性感覺(jué)神經(jīng)病變的作用可能來(lái)源于其減弱AT1R介導(dǎo)的腎素釋放負(fù)反饋,從而增強(qiáng)了Ang Ⅱ-AT2R的信號(hào)轉(zhuǎn)導(dǎo),進(jìn)而增強(qiáng)了AT2R激動(dòng)后的神經(jīng)保護(hù)作用。筆者推測(cè),AT2R激動(dòng)劑在減少抗腫瘤藥周?chē)窠?jīng)毒性方面的療效比其他RAS調(diào)節(jié)劑更佳,但尚需更多研究進(jìn)行驗(yàn)證。
1.1.4 具有腎臟保護(hù)作用 抗血管生成藥常有引發(fā)蛋白尿的副作用,而ACEIs/ARBs具有改善蛋白尿、保護(hù)腎臟的作用。Nihei等[27]通過(guò)回顧性分析2008-2014年在11家醫(yī)院接受抗血管生成藥貝伐珠單抗治療的非小細(xì)胞肺癌患者的臨床資料發(fā)現(xiàn),ACEIs/ARBs使用者的蛋白尿發(fā)生率顯著低于未使用者(P=0.037)。
1.2 增強(qiáng)抗腫瘤藥的療效
越來(lái)越多的臨床前研究證實(shí),RAS參與了惡性腫瘤的發(fā)生和進(jìn)展,這促使學(xué)者們就RAS調(diào)節(jié)劑對(duì)抗腫瘤藥的增效作用進(jìn)行了研究[28]。
1.2.1 RAS調(diào)節(jié)劑聯(lián)合化療藥物 Zhao等[29]回顧性分析了2010-2014年在美國(guó)馬薩諸塞州總醫(yī)院和布萊根婦女醫(yī)院接受化療的ⅢC/Ⅳ期卵巢癌患者的臨床資料,結(jié)果顯示,ACEIs/ARBs的使用與死亡風(fēng)險(xiǎn)顯著降低相關(guān)[HR=0.55,95%CI(0.36,0.95),P=0.004],且與服用其他抗高血壓藥組患者相比,ACEIs/ARBs組患者的OS延長(zhǎng)了30個(gè)月。該研究團(tuán)隊(duì)進(jìn)一步通過(guò)動(dòng)物實(shí)驗(yàn)發(fā)現(xiàn),氯沙坦通過(guò)使腫瘤基質(zhì)正?;瘉?lái)改善靜脈注射紫杉醇以及腹腔注射多柔比星的體內(nèi)遞送,進(jìn)而增強(qiáng)了化療療效,減少了卵巢癌模型動(dòng)物體內(nèi)的腹水。Murphy等[30]進(jìn)行了一項(xiàng)單臂Ⅱ期臨床試驗(yàn),納入了49例未曾治療且不可行切除術(shù)的局部晚期胰腺癌患者,其中39例接受了8周期的FOLFIRINOX方案(氟尿嘧啶+亞葉酸+奧沙利鉑+伊立替康)聯(lián)合氯沙坦治療,繼之給予放化療,這種全新輔助治療的方式起到了腫瘤降期的作用,使患者手術(shù)完全切除率達(dá)到了61%,提示新輔助治療與氯沙坦聯(lián)合應(yīng)用可提高抗腫瘤療效。Kasi等[31]回顧性分析了114例轉(zhuǎn)移性胰腺癌患者的臨床數(shù)據(jù),發(fā)現(xiàn)氯沙坦聯(lián)合化療患者與單純化療患者在OS、PFS、客觀緩解率(objective response rate,ORR)、疾病控制率(disease control rate,DCR)方面均無(wú)明顯差異;對(duì)接受FOLFIRINOX方案化療的患者進(jìn)行的亞組分析結(jié)果顯示,氯沙坦聯(lián)合化療組患者的PFS較單純化療組更長(zhǎng),但差異無(wú)統(tǒng)計(jì)學(xué)意義(350 d vs. 101 d,P=0.060 4),其原因可能是樣本量較小,故尚需進(jìn)行樣本量更大的隊(duì)列研究以驗(yàn)證氯沙坦和FOLFIRINOX方案在局部晚期惡性腫瘤新輔助治療中的益處是否也適用于轉(zhuǎn)移性惡性腫瘤。
1.2.2 RAS調(diào)節(jié)劑聯(lián)合小分子靶向藥物 鑒于RAS調(diào)節(jié)劑在血管內(nèi)皮生長(zhǎng)因子(vascular endothelial growth factor,VEGF)通路和血管生成中的獨(dú)立作用,既往臨床研究多集中在RAS調(diào)節(jié)劑聯(lián)合抗血管生成靶向藥物的作用上[32-35]。ACEIs/ARBs與抗血管生成藥(如培唑帕尼、舒尼替尼)聯(lián)用是否能改善轉(zhuǎn)移性腎癌患者的預(yù)后尚有爭(zhēng)議。Izzedine等[32]展開(kāi)的回顧性分析納入了2004-2013年接受舒尼替尼一線治療的213例轉(zhuǎn)移性腎癌患者(中位隨訪時(shí)間為3.6年),發(fā)現(xiàn)ACEIs/ARBs使用者較未使用ACEIs/ARBs者擁有更長(zhǎng)的OS[HR=0.40,95%CI(0.24,0.66),P<0.001]和PFS[HR=0.55,95%CI(0.35,0.86),P=0.009]。McKay等[33]對(duì)4 736例Ⅱ、Ⅲ期腎癌患者進(jìn)行了匯總分析,其中1 487例患者接受了ACEIs/ARBs治療,其OS相較于未使用ACEIs/ARBs者有明顯獲益(31.12個(gè)月vs. 21.94個(gè)月,P<0.000 1)。但Sorich等[34]在對(duì)兩項(xiàng)共計(jì)1 545例接受培唑帕尼或舒尼替尼治療的Ⅲ期轉(zhuǎn)移性腎癌患者的隨機(jī)對(duì)照試驗(yàn)的匯總分析中并未觀察到基線ACEIs/ARBs使用與OS[HR=0.97,95%CI(0.80,1.18),P=0.80]或PFS[HR=0.88,95%CI(0.73,1.06),P=0.17]相關(guān)。一項(xiàng)回顧性分析研究了343例一線接受舒尼替尼或帕唑帕尼治療的腎癌患者的臨床數(shù)據(jù),發(fā)現(xiàn)ACEIs/ARBs的使用未能顯著改善患者的OS或PFS[35]。此外,RAS調(diào)節(jié)劑對(duì)鼻咽癌、肝癌、肺癌等惡性腫瘤均有抑制作用,其機(jī)制涉及對(duì)促分裂原活化的蛋白激酶(mitogenactivated protein kinase,MAPK)、磷脂酰肌醇-3-激酶(phosphoinositide 3-kinase,PI3K)/蛋白激酶B(protein kinase B,PKB)等多條信號(hào)通路的抑制作用[36-39]。而MAPK、PI3K/PKB等旁路激活是小分子靶向藥物耐藥的重要機(jī)制之一[40-42]。可見(jiàn),RAS調(diào)節(jié)劑聯(lián)合小分子靶向藥物可能通過(guò)抑制多通路的旁路激活而降低小分子靶向藥物的耐藥性。
1.2.3 RAS調(diào)節(jié)劑聯(lián)合免疫檢查點(diǎn)抑制劑 免疫檢查點(diǎn)抑制劑包括細(xì)胞毒性T淋巴細(xì)胞相關(guān)抗原4(cytoto- xic T lymphocyte-associated antigen-4,CTLA-4)單抗和程序性死亡蛋白1(programmed death-1,PD-1)單抗/程序性死亡蛋白1配體(programmed death ligand-1,PD-L1)單抗等,可通過(guò)恢復(fù)或激活T細(xì)胞功能來(lái)發(fā)揮抗腫瘤的作用[43]。不同瘤種的腫瘤細(xì)胞對(duì)免疫檢查點(diǎn)抑制劑的反應(yīng)不一,如何通過(guò)聯(lián)合治療來(lái)增強(qiáng)免疫反應(yīng)性是目前的研究熱點(diǎn)[43]。大量研究證實(shí),組織局部的Ang Ⅱ具有促進(jìn)成纖維細(xì)胞增殖的作用[44-46]。成纖維細(xì)胞是一種重要的基質(zhì)細(xì)胞,在腫瘤微環(huán)境中可被激活形成癌相關(guān)成纖維細(xì)胞(cancer-associated fibroblasts,CAFs),后者有助于腫瘤細(xì)胞免疫抑制微環(huán)境的形成[45]。外源性給予Ang 1-7可抑制骨髓間充質(zhì)干細(xì)胞向成纖維細(xì)胞等基質(zhì)細(xì)胞分化[47]。RAS調(diào)節(jié)劑可通過(guò)減少CAFs和細(xì)胞外基質(zhì)來(lái)調(diào)節(jié)免疫細(xì)胞并改善細(xì)胞缺氧狀態(tài),從而增強(qiáng)腫瘤免疫反應(yīng)[48-49]。一項(xiàng)納入了299例胰腺導(dǎo)管腺癌術(shù)后患者的回顧性研究結(jié)果顯示,長(zhǎng)期使用ACEIs/ARBs者與未使用者相比具有更長(zhǎng)的OS,而這種作用與ACEIs/ARBs影響腫瘤細(xì)胞的免疫微環(huán)境有關(guān)[50]。Xie等[51]使用以4T1乳腺癌細(xì)胞構(gòu)建的小鼠腫瘤模型進(jìn)行研究,發(fā)現(xiàn)坎地沙坦可通過(guò)阻斷Ang Ⅱ信號(hào)轉(zhuǎn)導(dǎo)來(lái)提高PD-1/CTLA-4單抗的療效;坎地沙坦聯(lián)合PD-1/CTLA-4單抗在同源BALB/c小鼠的CT26結(jié)腸腫瘤模型中也取得了類(lèi)似效果,使得模型小鼠的OS明顯延長(zhǎng)。RAS調(diào)節(jié)劑聯(lián)合免疫檢查點(diǎn)抑制劑可能是一種潛在有效的治療模式,但尚需進(jìn)一步的臨床試驗(yàn)證實(shí)。
2 結(jié)語(yǔ)
綜上所述,RAS調(diào)節(jié)劑聯(lián)合抗腫瘤藥能起到減毒增效的作用,可能是一種潛在有效的抗腫瘤治療模式。RAS調(diào)節(jié)劑能減輕抗腫瘤藥的心臟毒性、血液學(xué)毒性、周?chē)窠?jīng)毒性等,并具有腎臟保護(hù)作用,其機(jī)制可能在于其可減輕氧化應(yīng)激、抑制炎癥介質(zhì)從而保護(hù)心肌、減輕神經(jīng)性疼痛,并能激活JAK2通路從而刺激骨髓造血恢復(fù)等。下一步研究可從機(jī)制出發(fā),比較不同RAS調(diào)節(jié)劑對(duì)不同器官保護(hù)效力的差異,優(yōu)化化療藥物與RAS調(diào)節(jié)劑的不同聯(lián)合方案,從而針對(duì)性減少某些化療藥物特異的毒副作用。從理論上分析,RAS調(diào)節(jié)劑可通過(guò)使腫瘤基質(zhì)正常化來(lái)促進(jìn)化療藥物體內(nèi)遞送、增強(qiáng)靶向藥物的抗血管生成作用、提高腫瘤的免疫原性,從而增強(qiáng)化療藥物、靶向藥物和免疫檢查點(diǎn)抑制劑的療效;但目前RAS調(diào)節(jié)劑聯(lián)合靶向藥物是否增效尚有爭(zhēng)議,聯(lián)合免疫檢查點(diǎn)抑制劑的益處尚停留在基礎(chǔ)研究水平,還需大樣本的臨床研究進(jìn)一步證實(shí)。
參考文獻(xiàn)
[ 1 ] JIANG H R,TAI Z G,CHEN Z J,et al. Clinical applicability of renin-angiotensin system inhibitors in cancer treatment[J]. Am J Cancer Res,2021,11(2):318-336.
[ 2 ] SUN H,LI T,ZHUANG R Y,et al. Do renin-angiotensin system inhibitors influence the recurrence,metastasis,and survival in cancer patients:evidence from a meta-analysis including 55 studies[J]. Medicine(Baltimore),2017,96(13):e6394.
[ 3 ] PINTER M,KWANTEN W J,JAIN R K. Renin-angiotensin system inhibitors to mitigate cancer treatment-related adverse events[J]. Clin Cancer Res,2018,24(16):3803- 3812.
[ 4 ] TRAPANI D,ZAGAMI P,NICOLò E,et al. Management of cardiac toxicity induced by chemotherapy[J]. J Clin Med,2020,9(9):E2885.
[ 5 ] RADIN D P,KREBS A,MAQSUDLU A,et al. Our ACE in the HOLE:justifying the use of angiotensin-converting enzyme inhibitors as adjuvants to standard chemothe-? ? ?rapy[J]. Anticancer Res,2018,38(1):45-49.
[ 6 ] PACKER M,MCMURRAY J J V. Importance of endogenous compensatory vasoactive peptides in broadening the effects of inhibitors of the renin-angiotensin system for the treatment of heart failure[J]. Lancet,2017,389(10081):1831-1840.
[ 7 ] PITUSKIN E,MACKEY J R,KOSHMAN S,et al. Multidisciplinary approach to novel therapies in cardio-onco- logy research(MANTICORE 101-breast):arandomized trial for the prevention of trastuzumab-associated cardioto- xicity[J]. J Clin Oncol,2017,35(8):870-877.
[ 8 ] GUGLIN M,KRISCHER J,TAMURA R,et al. Rando- mized trial of lisinopril versus carvedilol to prevent trastuzumab cardiotoxicity in patients with breast cancer[J]. J Am Coll Cardiol,2019,73(22):2859-2868.
[ 9 ] GULATI G,HECK S L,REE A H,et al. Prevention of cardiac dysfunction during adjuvant breast cancer therapy(PRADA):a 2×2 factorial,randomized,placebo-controlled,double-blind clinical trial of candesartan and metoprolol[J]. Eur Heart J,2016,37(21):1671-1680.
[10] HECKSL,MECINAJ A,REE AH,et al. Prevention of cardiac dysfunction during adjuvant breast cancer therapy(PRADA):extended follow-up of a 2×2 factorial,randomized,placebo-controlled,double-blind clinical trial of candesartan and metoprolol[J]. Circulation,2021,143(25):2431-2440.
[11] WITTAYANUKORN S,QIAN JJ,WESTRICK S C,et al. Prevention of trastuzumab and anthracycline-induced cardiotoxicity using angiotensin-converting enzyme inhibitors or β-blockers in older adults with breast cancer[J]. Am J Clin Oncol,2018,41(9):909-918.
[12] OLORUNDARE O E,ADENEYE A A,AKINSOLA A O,et al. Therapeutic potentials of selected antihypertensive agents and their fixed-dose combinations against trastuzumab-mediated cardiotoxicity[J]. Front Pharmacol,2020,11:610331.
[13] AL-HARBI N O,IMAM F,NADEEM A,et al. Protection against tacrolimus-induced cardiotoxicity in rats by olme- sartan and aliskiren[J]. Toxicol Mech Methods,2014,24(9):697-702.
[14] RODGERS KE,DIZEREGA G S. Contribution of the local RAS to hematopoietic function:anovel therapeutic target[J]. Front Endocrinol(Lausanne),2013,4:157.
[15] KIM S,ZINGLER M,HARRISON J K,et al. Angiotensin Ⅱ regulation of proliferation,differentiation,and engraftment of hematopoietic stem cells[J]. Hypertension,2016,67(3):574-584.
[16] RODGERS K E,ESPINOZA T B,RODA N,et al. Angiotensin-(1-7) synergizes with colony-stimulating factors in hematopoietic recovery[J]. Cancer Chemother Pharmacol,2013,72(6):1235-1245.
[17] PHAM H,SCHWARTZ B M,DELMORE J E,et al. Pharmacodynamic stimulation of thrombogenesis by angiotensin(1-7)in recurrent ovarian cancer patients receiving gemcitabine and platinum-based chemotherapy[J]. Cancer Chemother Pharmacol,2013,71(4):965-972.
[18] RODGERS K E,ESPINOZA T,F(xiàn)ELIX J,et al. Acceleration of healing,reduction of fibrotic scar,and normalization of tissue architecture by an angiotensin analogue,NorLeu3-A(1-7)[J]. PlastReconstr Surg,2003,111(3):1195-1206.
[19] GAFFNEY K,WEINBERG M,SOTO M,et al. Development of angiotensin Ⅱ(1-7) analog as an oral therapeutic for the treatment of chemotherapy-induced myelosuppression[J]. Haematologica,2018,103(12):e567-e570.
[20] BALOGH M,AGUILAR C,NGUYEN N T,et al. Angiotensin receptors and neuropathic pain[J]. Pain Rep,2021,6(1):e869.
[21] PREMER C,LAMONDIN C,MITZEY A,et al. Immunohistochemical localization of AT1a,AT1b,and AT2 angiotensin Ⅱ receptor subtypes in therat adrenal,pituitary,and brain with a perspective commentary[J]. Int J Hypertens,2013,2013:175428.
[22] DE KLOET A D,WANG L,LUDIN J A,et al. Reporter mouse strain provides a novel look at angiotensin type-2 receptor distribution in the central nervous system[J]. Brain Struct Funct,2016,221(2):891-912.
[23] ROLDAN C J,SONG J,ENGLE M P,et al. Angiotensin-converting enzyme inhibitors and angiotensin receptor blockers modulate the function of myelinated fibers after chemotherapy:aquantitative sensory testing study[J]. Pain Physician,2017,20(4):281-292.
[24] KIM E,HWANG S H,KIM H K,et al. Losartan,an angiotensin Ⅱ type 1 receptor antagonist,alleviates mechanical hyperalgesia in a rat model of chemotherapy-induced neuropathicpain by inhibiting inflammatory cytokines in the dorsal root Ganglia[J]. Mol Neurobiol,2019,56(11):7408-7419.
[25] BESSAGUET F,DANIGO A,BOUCHENAKI H,et al. Neuroprotective effect of angiotensin Ⅱ type 2 receptor stimulation in vincristine-induced mechanical allody- nia[J]. Pain,2018,159(12):2538-2546.
[26] BESSAGUET F,DANIGO A,MAGY L,et al. Candesartan prevents resiniferatoxin-induced sensory small-fiber neuropathy in mice by promoting angiotensin Ⅱ-mediated AT2 receptor stimulation[J]. Neuropharmacology,2017,126:142-150.
[27] NIHEI S,SATO J,HARADA T,et al. Antiproteinuric effects of renin-angiotensin inhibitors in lung cancer patients receiving bevacizumab[J]. Cancer Chemother Pharmacol,2018,81(6):1051-1059.
[28] YANG J,YANG X,GAO L,et al. The role of the renin-angiotensin system inhibitors in malignancy:a review[J]. Am J Cancer Res,2021,11(3):884-897.
[29] ZHAO Y X,CAO J H,MELAMED A,et al. Losartan treatment enhances chemotherapy efficacy and reduces ascites in ovarian cancer models by normalizing the tumor stroma[J]. Proc Natl Acad Sci USA,2019,116(6):2210-2219.
[30] MURPHY J E,WO J Y,RYAN D P,et al. Total neoadjuvant therapy with FOLFIRINOX in combination with losartan followed by chemoradiotherapy for locally advanced pancreatic cancer:a phase 2 clinical trial[J]. JAMA Oncol,2019,5(7):1020-1027.
[31] KASI A,ALLEN J,MEHTA K,et al. Association of losartan with outcomes in metastatic pancreatic cancer patients treated with chemotherapy[J]. J Clin Transl Res,2021,7(2):257-262.
[32] IZZEDINE H,DEROSA L,LE TEUFF G,et al. Hypertension and angiotensin system inhibitors:impact on outcome in sunitinib-treated patients for metastatic renal cell carcinoma[J]. Ann Oncol,2015,26(6):1128-1133.
[33] MCKAY R R,RODRIGUEZ G E,LIN X,et al. Angiotensin system inhibitors and survival outcomes in patients with metastatic renal cell carcinoma[J]. Clin Cancer Res,2015,21(11):2471-2479.
[34] SORICH M J,KICHENADASSE G,ROWLAND A,et al. Angiotensin system inhibitors and survival in patients with metastatic renal cell carcinoma treated with VEGF- targeted therapy:a pooled secondary analysis of clinical trials[J]. Int J Cancer,2016,138(9):2293-2299.
[35] FIALA O,OSTA?OV P,ROZSYPALOVá A,et al. Impact of concomitant cardiovascular medication on survival of metastatic renal cell carcinoma patients treated with sunitinib or pazopanib in the first line[J]. Target Oncol,2021,16(5):643-652.
[36] LIN Y T,WANG H C,TSAI M H,et al. Angiotensin Ⅱ receptor blockers valsartan and losartan improve survival rate clinically and suppress tumor growth via apoptosis related to PI3K/AKT signaling in nasopharyngeal carci-? ?noma[J]. Cancer,2021,127(10):1606-1619.
[37] LIN Y T,WANG H C,CHUANG H C,et al. Pre-treatment with angiotensin-(1-7) inhibits tumor growth via autophagy by downregulating PI3K/Akt/mTOR signaling in human nasopharyngeal carcinoma xenografts[J]. J Mol Med(Berl),2018,96(12):1407-1418.
[38] QI R,LEI C G,BAI Y X,et al. The AT1/Raf/ERK1/2 signaling pathway is involved in angiotensin Ⅱ-enhanced proliferation of hepatic carcinoma cells[J]. Neoplasma,2019,66(1):83-91.
[39] NI L,F(xiàn)ENG Y,WAN H Y,et al. Angiotensin-(1-7) in- hibits the migration and invasion of A549 human lung? ? adenocarcinoma cells through inactivation of the PI3K/Akt and MAPK signaling pathways[J]. Oncol Rep,2012,27(3):783-790.
[40] DUTTA A,HUTCHISON R E,MOHI G. Hmga2 promotes the development of myelofibrosis in Jak2V617F knockin mice by enhancing TGF-β1 and Cxcl12 pathways[J]. Blood,2017,130(7):920-932.
[41] STIVALA S,CODILUPI T,BRKIC S,et al. Targeting compensatory MEK/ERK activation increases JAK inhibitor efficacy in myeloproliferative neoplasms[J]. J Clin Invest,2019,129(4):1596-1611.
[42] MEYER S C. Mechanisms of resistance to JAK2 inhibitors in myeloproliferative neoplasms[J]. Hematol Oncol Clin North Am,2017,31(4):627-642.
[43] SALMANINEJAD A,VALILOU S F,SHABGAH A G,et al. PD-1/PD-L1 pathway:basic biology and role in cancer immunotherapy[J]. J Cell Physiol,2019,234(10):16824- 16837.
[44] SYSOEVA V Y,AGEEVA L V,TYURIN-KUZMIN P A,et al. Local angiotensin Ⅱ promotes adipogenic differentia- tion of human adipose tissue mesenchymal stem cells through type 2 angiotensin receptor[J]. Stem Cell Res,2017,25:115-122.
[45] GASIūNIEN? M,PETKUS G,MATUZEVI?IUS D,et al. Angiotensin Ⅱ and TGF-β1 induce alterations in human amniotic fluid-derived mesenchymal stem cells leading to cardiomyogenic differentiation initiation[J]. Int J Stem Cells,2019,12(2):251-264.
[46] JIANG X,WU F,XU Y,et al. A novel role of angiotensin Ⅱ in epidermal cell lineage determination:angiotensin Ⅱpromotes the differentiation of mesenchymal stem cells into keratinocytes through the p38 MAPK,JNK and JAK2 signalling pathways[J]. Exp Dermatol,2019,28(1):59- 65.
[47] PAPINSKA A M,MORDWINKIN N M,MEEKS C J,et al. Angiotensin-(1-7)administration benefits cardiac,renal and progenitor cell function in db/db mice[J]. Br J Pharmacol,2015,172(18):4443-4453.
[48] PINTER M,JAIN R K. Targeting the renin-angiotensin system to improve cancer treatment:implications for immunotherapy[J]. Sci Transl Med,2017,9(410):eaan5616.
[49] VALLEJO-ARDILA D L,F(xiàn)IFIS T,BURRELL L M,et al. Renin-angiotensin inhibitors reprogram tumor immune microenvironment:a comprehensive view of the influences on anti-tumor immunity[J]. Oncotarget,2018,9(84):35500-35511.
[50] LIU H,NAXEROVA K,PINTER M,et al. Use of angiotensin system inhibitors is associated with immune activation and longer survival in nonmetastatic pancreatic ductal adenocarcinoma[J]. Clin Cancer Res,2017,23(19):5959-5969.
[51] XIE G Z,CHENG T,LIN J,et al. Local angiotensin Ⅱ contributes to tumor resistance to checkpoint immunothe- rapy[J]. J Immunother Cancer,2018,6(1):88.
(收稿日期:2021-08-19 修回日期:2022-02-28)
(編輯:胡曉霖)