專家介紹
和水祥,醫(yī)學(xué)博士、教授、一級(jí)主任醫(yī)師,陜西省教學(xué)名師,西安交通大學(xué)第一附屬醫(yī)院消化內(nèi)科主任、陜西省消化系統(tǒng)疾?。[瘤方向)臨床醫(yī)學(xué)研究中心主任、陜西省消化內(nèi)科學(xué)??坡?lián)盟負(fù)責(zé)人、中國(guó)醫(yī)師協(xié)會(huì)消化內(nèi)鏡醫(yī)師培訓(xùn)中心(陜西)主任。先后獲得西安交通大學(xué)醫(yī)學(xué)部首批“名醫(yī)”“名師”,西安交通大學(xué)“王寬誠(chéng)育人獎(jiǎng)”“教學(xué)卓越獎(jiǎng)”“教學(xué)名師”獎(jiǎng)。入選“中國(guó)消化領(lǐng)域高價(jià)值論文TOP100”、2012—2021年度全國(guó)消化病領(lǐng)域?qū)W者學(xué)術(shù)影響力百?gòu)?qiáng)榜。接受“央廣國(guó)際在線”“央視新聞?lì)l道”專題參訪報(bào)道。現(xiàn)兼任中華醫(yī)學(xué)會(huì)消化內(nèi)鏡學(xué)分會(huì)委員兼膠囊內(nèi)鏡學(xué)組副組長(zhǎng),中國(guó)醫(yī)師協(xié)會(huì)內(nèi)鏡醫(yī)師分會(huì)常委,醫(yī)師分會(huì)、循證醫(yī)學(xué)醫(yī)師分會(huì)委員等。
摘要:胃癌是我國(guó)癌癥相關(guān)死亡的主要原因。靶向藥物治療是胃癌重要的治療方案之一。人表皮生長(zhǎng)因子受體2(HER2)是重要的胃癌治療靶點(diǎn),曲妥珠單抗是一種靶向HER2的單克隆抗體,ToGA實(shí)驗(yàn)確定曲妥珠單抗聯(lián)合化療是HER2陽(yáng)性晚期胃癌患者的標(biāo)準(zhǔn)治療方案。本文對(duì)ToGA實(shí)驗(yàn)及其進(jìn)展、曲妥珠單抗聯(lián)合其他化療藥物、曲妥珠單抗聯(lián)合其他HER2靶向藥物、曲妥珠單抗和其他藥物偶聯(lián)物及曲妥珠單抗聯(lián)合其他靶向位點(diǎn)藥物治療HER2陽(yáng)性晚期胃癌研究進(jìn)展進(jìn)行綜述。曲妥珠單抗聯(lián)合S-1及其他化療藥物表現(xiàn)出了良好的療效和安全性;帕妥珠單抗和曲妥珠單抗聯(lián)合化療未達(dá)到主要終點(diǎn);T-DM1(trastuzumab emtansine)對(duì)于HER2陽(yáng)性晚期胃癌患者在改善總生存期方面沒(méi)有優(yōu)勢(shì);KN026、T-DXd(trastuzumab deruxtecan)、曲妥珠單抗與派姆單抗聯(lián)合化療、貝伐單抗與曲妥珠單抗聯(lián)合卡培他濱和奧沙利鉑是治療HER2陽(yáng)性晚期胃癌的最前沿研究進(jìn)展。
關(guān)鍵詞:曲妥珠單抗;人表皮生長(zhǎng)因子受體2 (HER2);晚期胃癌
中圖分類(lèi)號(hào):R573.9 文獻(xiàn)標(biāo)志碼:A
DOI:10.7652/jdyxb202406001
收稿日期:2024-02-09 修回日期:2024-09-18
通信作者:和水祥.E-mail: hesx123@126.com
網(wǎng)絡(luò)出版地址:https://link.cnki.net/urlid/61.1399.r.20240927.1711.004 (2024-09-29)
Research progress in trastuzumab for treating HER2-positive advanced gastric cancer
ZHENG Yuchen, HE Shuixiang
(Department of Internal Medicine, The First Affiliated Hospital of
Xi’an Jiaotong University, Xi’an 710061, China)
ABSTRACT: Gastric cancer is the leading cause of cancer-related mortality. Targeted drug therapy is one of the most important therapeutic means for gastric cancer. Human epidermal growth factor receptor 2 (HER2) is an important therapeutic target for gastric cancer. Trastuzumab is a monoclonal antibody that targets HER2. The ToGA trial has established trastuzumab in combination with chemotherapy as the standard treatment for patients with HER2-positive advanced gastric cancer. This article reviews the progress in ToGA experiment, trastuzumab combined with other chemotherapy drugs, trastuzumab combined with other HER2-targeted drugs, trastuzumab and other drug conjugate, and trastuzumab combined with other targeted drugs in the treatment of HER2-positive advanced gastric cancer. Trastuzumab combined with S-1 and other chemotherapeutic drugs has shown good efficacy and safety; the combination of pertuzumab and trastuzumab did not meet the primary endpoint; trastuzumab emtansine (T-DM1) showed no benefit in improving overall survival in patients with HER2-positive advanced gastric cancer. KN026, trastuzumab deruxtecan (T-DXd), Trastuzumab and Pembrolizumab combined chemotherapy, as well as bevacizumab and trastuzumab combined with capecitabine and oxaliplatin are the most progress research advances in the treatment of HER2-positive advanced gastric cancer.
KEY WORDS: trastuzumab; human epidermal growth factor receptor 2 (HER2); advanced gastric cancer
胃癌是世界上最常診斷的第五大癌癥和第三大癌癥相關(guān)死亡原因,2020年全球有100多萬(wàn)例胃癌病例,導(dǎo)致超過(guò)76.8萬(wàn)人死亡。胃癌是我國(guó)癌癥相關(guān)死亡的主要原因[1]。
目前,胃癌有多種治療方案,其中靶向藥物治療是重要的治療方案之一。ToGA實(shí)驗(yàn)確定曲妥珠單抗聯(lián)合化療是人表皮生長(zhǎng)因子受體2 (human epidermal growth factor receptor 2, HER2)陽(yáng)性晚期胃癌患者的標(biāo)準(zhǔn)治療方案[1]。HER2是重要的胃癌治療靶點(diǎn),HER2與腫瘤細(xì)胞增殖、凋亡、黏附、遷移和分化相關(guān)。研究表明,HER2是胃癌發(fā)生的關(guān)鍵驅(qū)動(dòng)因素,HER2陽(yáng)性與胃癌預(yù)后不良和侵襲性相關(guān)[2]。曲妥珠單抗是一種靶向HER2的單克隆抗體,可誘導(dǎo)抗體依賴的細(xì)胞毒性,抑制HER2介導(dǎo)的信號(hào)轉(zhuǎn)導(dǎo),并防止HER2細(xì)胞外結(jié)構(gòu)域的切割[2]。
以下對(duì)ToGA實(shí)驗(yàn)及其進(jìn)展、曲妥珠單抗聯(lián)合其他化療藥物、曲妥珠單抗聯(lián)合其他HER2靶向藥物、曲妥珠單抗和其他藥物偶聯(lián)物及曲妥珠單抗聯(lián)合其他靶向位點(diǎn)藥物治療HER2陽(yáng)性晚期胃癌研究進(jìn)展進(jìn)行綜述。
1 ToGA實(shí)驗(yàn)簡(jiǎn)介及進(jìn)展
ToGA實(shí)驗(yàn)是一項(xiàng)隨機(jī)、開(kāi)放標(biāo)簽,多中心、國(guó)際、3期、隨機(jī)對(duì)照試驗(yàn)。ToGA實(shí)驗(yàn)表明,與單獨(dú)化療組相比,曲妥珠單抗聯(lián)合化療組的緩解率、進(jìn)展時(shí)間和緩解持續(xù)時(shí)間均顯著改善。根據(jù)HER2狀態(tài)的一項(xiàng)預(yù)計(jì)劃探索性分析表明,HER2高表達(dá)患者的總生存期比低表達(dá)患者的總生存期更長(zhǎng),這在ToGA實(shí)驗(yàn)中也得到了驗(yàn)證。之后的探索性事后分析也表明:與HER2低表達(dá)(免疫組織化學(xué)染色強(qiáng)度0或1+和FISH陽(yáng)性)胃癌患者相比,曲妥珠單抗加化療顯著提高了HER2高表達(dá)胃癌患者(免疫組織化學(xué)染色強(qiáng)度2+和FISH陽(yáng)性或免疫組織化學(xué)染色強(qiáng)度3+)的總生存率[2]。隨后SHITARA等[3]經(jīng)單因素分析和多因素分析研究證實(shí),曲妥珠單抗治療HER2陽(yáng)性胃癌患者的中位總生存期(24.7個(gè)月)顯著長(zhǎng)于HER2胃癌陰性患者(13.9個(gè)月)。在安全性方面,曲妥珠單抗與標(biāo)準(zhǔn)化療聯(lián)合使用不影響總體安全性[2]。在臨床實(shí)踐中,對(duì)于大部分中國(guó)患者,HER2陽(yáng)性晚期胃癌患者在化療中加入曲妥珠單抗是有效和安全的[4]。隨后,為探究對(duì)存在較差預(yù)后因素的胃癌患者,增加曲妥珠單抗的用量是否可以獲得更長(zhǎng)的總生存期,SHAH等[5]進(jìn)行了HELOISE實(shí)驗(yàn),結(jié)果表明,增加曲妥珠單抗劑量沒(méi)有獲得更長(zhǎng)的生存期,但是也沒(méi)有產(chǎn)生新的安全問(wèn)題。
目前,曲妥珠單抗聯(lián)合化療已經(jīng)作為HER2陽(yáng)性晚期胃癌患者的標(biāo)準(zhǔn)治療方案[1],標(biāo)準(zhǔn)劑量為:8 mg/kg 的負(fù)荷劑量,隨后每3 周 6 mg/kg 的維持劑量[2]。
2 曲妥珠單抗聯(lián)合其他化療藥物
在ToGA實(shí)驗(yàn)基礎(chǔ)上,很多學(xué)者對(duì)化療方案進(jìn)行了改善,目前已有很多類(lèi)型的研究??傮w而言,S-1有更多的進(jìn)展。S-1是由5-氟尿嘧啶的前藥替加氟、吉美嘧啶和奧替拉西鉀以摩爾比1∶0.4∶1組合而成的氟嘧啶制劑。在日本,口服S-1+順鉑的5周方案是晚期胃癌的一線化療方案之一[6]。KUROKAWA等[7]研究了曲妥珠單抗聯(lián)合S-1和順鉑治療HER2陽(yáng)性晚期胃癌,結(jié)果表明,曲妥珠單抗聯(lián)合S-1聯(lián)合順鉑具有良好的安全性和療效。在此基礎(chǔ)上,MIURA等[8]進(jìn)一步研究的結(jié)果表明,對(duì)于HER2陽(yáng)性的晚期胃癌患者,曲妥珠單抗聯(lián)合5周S-1+順鉑治療的胃癌患者緩解率為61%,達(dá)到了主要終點(diǎn),且與單純S-1+順鉑相比,二者毒性相當(dāng)。曲妥珠單抗聯(lián)合5周S-1+順鉑具有良好的療效和可耐受性。同時(shí)也發(fā)現(xiàn)血清神經(jīng)調(diào)節(jié)蛋白1(NRG1)可能是可以預(yù)測(cè)曲妥珠單抗療效的生物標(biāo)志物,但這仍需要進(jìn)一步實(shí)驗(yàn)的驗(yàn)證。
考慮到順鉑對(duì)老年患者的副作用和毒性很大,KIMURA等[9]研究了曲妥珠單抗聯(lián)合S-1不使用順鉑的療效和安全性,結(jié)果表明,這是一種有效且安全的治療方法,可以延長(zhǎng)老年HER2陽(yáng)性晚期胃癌患者的生存期。目前聯(lián)合應(yīng)用不僅僅局限于S-1+順鉑,S-1+奧沙利鉑是日本治療HER2陰性胃癌的標(biāo)準(zhǔn)方案之一。TAKAHARI等[10]研究了曲妥珠單抗聯(lián)合S-1+奧沙利鉑治療HER2陽(yáng)性胃癌,結(jié)果顯示客觀緩解率為70.7%,達(dá)到了主要終點(diǎn),安全性方面也優(yōu)于順鉑。此研究顯示曲妥珠單抗聯(lián)合S-1聯(lián)合奧沙利鉑在HER2陽(yáng)性胃癌患者中表現(xiàn)出良好的療效和可耐受性。在另一項(xiàng)二期研究中,S-1、奧沙利鉑130 mg/m2和曲妥珠單抗聯(lián)合治療晚期和復(fù)發(fā)的HER2陽(yáng)性胃癌的客觀緩解率為82.1%,中位無(wú)進(jìn)展生存期為7.0個(gè)月,中位總生存期為27.6個(gè)月,不良事件輕微。這表明曲妥珠單抗聯(lián)合S-1和奧沙利鉑對(duì)晚期和復(fù)發(fā)的HER2陽(yáng)性胃癌患者有良好的療效,并且毒性可接受和可控[11]。KAGAWA等[12]的DASH研究證實(shí)了S-1和多西他賽聯(lián)合曲妥珠單抗化療方案具有良好的抗腫瘤效果和耐受性,但是樣本量較小,仍需進(jìn)一步研究,不過(guò)對(duì)于不適合順鉑或奧沙利鉑為基礎(chǔ)的抗HER2化療的患者提供了一種新的治療選擇方向。
曲妥珠單抗聯(lián)合其他化療藥物在非S-1藥物方面也有很多進(jìn)展,但都不完善,仍需進(jìn)一步的研究。RYU等[13]在一項(xiàng)Ⅱ期試驗(yàn)中證實(shí),曲妥珠單抗聯(lián)合卡培他濱和奧沙利鉑治療HER2陽(yáng)性胃癌患者的客觀緩解率為68%,具有良好的療效和安全性。也有研究者觀察了曲妥珠單抗加伊立替康對(duì)既往接受曲妥珠單抗治療的HER2陽(yáng)性晚期胃癌患者的療效,但由于入選患者數(shù)量只有16例,難以得到可靠的研究結(jié)果[14]。MONDACA等[15]研究了改良多西他賽、順鉑和5氟尿嘧啶聯(lián)合曲妥珠單抗治療HER2陽(yáng)性晚期胃癌的效果,中位無(wú)進(jìn)展生存期為13個(gè)月,中位總生存期為24.9個(gè)月。這表明改良多西他賽、順鉑和5氟尿嘧啶聯(lián)合曲妥珠單抗的有效性和安全性,可作為特定患者的一種治療選擇。
不僅僅局限于聯(lián)合其他藥物,也有研究者想增強(qiáng)曲妥珠單抗的效果??紤]到抗體依賴性細(xì)胞毒性(ADCC)的激活在抗HER2藥物中有重要作用, LEE等[16]在一項(xiàng)Ⅰ期實(shí)驗(yàn)中證實(shí),自體NK細(xì)胞輸注治療聯(lián)合曲妥珠單抗具有初步的抗腫瘤活性,但是這仍需進(jìn)一步的二線研究評(píng)估。
3 曲妥珠單抗聯(lián)合其他HER2靶向藥物
3.1 帕妥珠單抗
帕妥珠單抗是一種人源化單克隆HER2靶向抗體,它與曲妥珠單抗在HER2受體蛋白上的結(jié)合表位不同。VON MINCKWITZ等[17]的研究證實(shí),帕妥珠單抗加曲妥珠單抗聯(lián)合化療可顯著改善HER2陽(yáng)性乳腺癌患者生存結(jié)果。同時(shí),多名研究者進(jìn)行了帕妥珠單抗加曲妥珠單抗聯(lián)合化療對(duì)HER2陽(yáng)性晚期胃癌療效的研究。KANG等[18]在一項(xiàng)Ⅱ期研究中證實(shí),帕妥珠單抗840 mg/3周的劑量可提供更大治療收益。之后,TABERNERO等[19]進(jìn)行的JACOB研究中,帕妥珠單抗和曲妥珠單抗聯(lián)合化療的總生存期與安慰劑和曲妥珠單抗聯(lián)合化療沒(méi)有顯著性差異。據(jù)此推測(cè):與乳腺癌相比,胃癌表現(xiàn)出更多的免疫組化異質(zhì)性,且HER2表達(dá)更低,膜染色不完全。不過(guò),一項(xiàng)中國(guó)亞群的研究表明,帕妥珠單抗聯(lián)合曲妥珠單抗和化療對(duì)HER2陽(yáng)性晚期胃癌患者有良好的療效,中位總生存期高于對(duì)照組(18.7個(gè)月 vs. 16.1個(gè)月)[20]。這與JACOB研究結(jié)果有所差異,雖然得到了總生存期數(shù)值上的改善,但是仍需要相關(guān)實(shí)驗(yàn)探究差異產(chǎn)生原因和是否能進(jìn)一步臨床應(yīng)用。類(lèi)似的,在一項(xiàng)日本亞組人群的JACOB試驗(yàn)的分析中,帕妥珠單抗組的中位無(wú)進(jìn)展生存期為12.4個(gè)月,安慰劑組為6.3個(gè)月,帕妥珠單抗組的中位總生存期為22.0個(gè)月,安慰劑組為15.6個(gè)月;與安慰劑組相比,觀察到帕妥珠單抗組有改善無(wú)進(jìn)展生存期和總生存期的趨勢(shì)。相比于JACOB的總?cè)巳海毡緛喨貉芯炕颊咧杏^察到了相對(duì)較長(zhǎng)的總生存期。對(duì)于這個(gè)結(jié)果,日本亞組研究者提出了2種可能性:日本患者的東部腫瘤合作組織評(píng)分為0的患者比例較高;日本接受癌癥治療的患者比例較高[21]。JACOB實(shí)驗(yàn)的最終分析顯示,雖然JACOB沒(méi)有達(dá)到其主要終點(diǎn),但是也得到了一些相對(duì)有限的結(jié)論:長(zhǎng)期隨訪(≥ 44.4個(gè)月)后,帕妥珠單抗加曲妥珠單抗和化療對(duì)先前未經(jīng)治療的HER2陽(yáng)性轉(zhuǎn)移性胃癌具有治療活性和可接受的安全性,并且在曲妥珠單抗和化療加入帕妥珠單抗時(shí),可以使死亡風(fēng)險(xiǎn)降低15%[22]。
XU等[23]在一項(xiàng)Ⅱ期實(shí)驗(yàn)中介紹了一種新型抗體KN026。KN026是一種雙特異性抗HER2抗體,由曲妥珠單抗和帕妥珠單抗的重鏈可變結(jié)構(gòu)域和曲妥珠單抗的片段結(jié)晶區(qū)域組成。它的獨(dú)特設(shè)計(jì)允許HER2結(jié)構(gòu)域Ⅱ(帕妥珠單抗結(jié)合位點(diǎn))和Ⅳ(曲妥珠單抗結(jié)合位點(diǎn))同時(shí)結(jié)合,并繼承了曲妥珠單抗抗體依賴性的細(xì)胞毒性和吞噬殺傷作用。在該研究中,KN026治療HER2陽(yáng)性晚期胃癌的客觀緩解率為56%,達(dá)到了主要終點(diǎn)。這對(duì)于HER2陽(yáng)性胃癌患者提供了新的治療方向,但也還需要進(jìn)一步的研究。
3.2 波奇替尼
波奇替尼是一種酪氨酸激酶抑制劑,靶點(diǎn)為人表皮生長(zhǎng)因子受體EGFR、HER2和HER4。波奇替尼在體外和體內(nèi)對(duì)HER2陽(yáng)性的胃癌細(xì)胞均表現(xiàn)出較強(qiáng)的抗腫瘤活性,與紫杉醇等化療藥物聯(lián)用時(shí)也表現(xiàn)出協(xié)同抑制作用[24]。KIM等[25]的一項(xiàng)Ⅰ/Ⅱ期研究表明,在接受了一線化療的HER2陽(yáng)性晚期胃癌患者中,曲妥珠單抗聯(lián)合波奇替尼和紫杉醇顯示出良好的臨床療效和可控的毒性。但這仍需要進(jìn)一步的臨床實(shí)驗(yàn)以證實(shí)其療效和安全性。
4 曲妥珠單抗和其他藥物偶聯(lián)物
4.1 T-DXd(trastuzumab deruxtecan)
T-DXd是一種抗體藥物偶聯(lián)物,由抗HER2抗體、可裂解的四肽接頭和細(xì)胞毒性拓?fù)洚悩?gòu)酶Ⅰ抑制劑組成。T-DXd的有效載荷具有膜滲透性,因此T-DXd對(duì) HER2 高表達(dá)的依賴性較低[26]。DOI等[27]的一項(xiàng)Ⅰ期研究表明,晚期胃癌患者對(duì)T-DXd具有良好耐受性;并且基于安全性和活性,此研究也給出了推薦的2期劑量:5.4、6.4 mg/kg。隨后,SHITARA等[28]的一項(xiàng)Ⅰ期研究證實(shí),T-DXd在HER2陽(yáng)性晚期胃癌患者中顯示出初步的抗腫瘤活性。在此基礎(chǔ)上,SHITARA等[26]繼續(xù)進(jìn)行了一項(xiàng)Ⅱ期研究,研究表明,相比于化療組(伊立替康或紫杉醇),T-DXd組獲得客觀緩解的患者比例明顯更高 (51% vs. 14%)。在安全性方面,常見(jiàn)的不良事件均可通過(guò)減少劑量和中斷治療得以改善。總之,T-DXd可顯著改善HER2陽(yáng)性胃癌患者的總生存期,且具有可控的安全性。也有研究表明,相比于其他靶向藥物,T-DXd的效率更高[29]。對(duì)于美國(guó)和歐洲的HER2陽(yáng)性胃癌患者,VANCUTSEM等[30]的最新DESTINY-Gastric02研究也支持以上結(jié)論。
另一方面,YAMAGUCHI等[31]發(fā)現(xiàn),T-DXd對(duì)HER2陰性胃癌患者有效。該研究將HER2陰性胃癌患者分為2個(gè)隊(duì)列,隊(duì)列1為HER2 IHC 2+/ISH-,隊(duì)列2為HER2 IHC 1+。結(jié)果表明,隊(duì)列1和隊(duì)列2的客觀緩解率分別為26.3%。雖然這低于接受T-DXd治療的HER2陽(yáng)性患者的客觀緩解率(43%),但表明至少T-DXd對(duì)低水平HER2表達(dá)的患者有效。SHITARA等[32]的一項(xiàng)最新研究也證實(shí)了這個(gè)發(fā)現(xiàn)。
4.2 T-DM1(trastuzumab emtansine)
T-DM1是一種抗體-藥物偶聯(lián)物,由曲妥珠單抗與小管蛋白抑制劑 T-DM1 通過(guò)穩(wěn)定連接體連接而成。GATSBY實(shí)驗(yàn)是一項(xiàng)三臂、隨機(jī)、開(kāi)放標(biāo)簽、適應(yīng)性、2/3期的全球研究,評(píng)估了T-DM1治療既往接受過(guò)治療的HER2陽(yáng)性晚期胃癌患者的療效和安全性。結(jié)果表明,T-DM1組和紫杉烷類(lèi)藥物組的中位無(wú)進(jìn)展生存期分別為2.7個(gè)月、2.9個(gè)月,提示與紫杉類(lèi)藥物相比,使用T-DM1治療晚期胃癌患者的總生存期沒(méi)有優(yōu)勢(shì)[33]。對(duì)于日本亞組分析中,日本患者的療效與總體人群一致,與紫杉烷治療相比,T-DM1組的總生存期沒(méi)有延長(zhǎng)[34]。
5 曲妥珠單抗聯(lián)合其他靶向藥物
5.1 曲妥珠單抗聯(lián)合pd-1抗體
目前最主要研究為派姆單抗。JANJIGIAN等[35]首先在一項(xiàng)Ⅱ期研究中觀察派姆單抗和曲妥珠單抗聯(lián)合氟嘧啶和順鉑化療對(duì)于HER2陽(yáng)性晚期胃癌患者的療效。結(jié)果顯示,派姆單抗和曲妥珠單抗聯(lián)合氟嘧啶和順鉑化療客觀緩解率為91%,中位總生存期為27.3個(gè)月,這表明其對(duì)HER2陽(yáng)性胃癌患者有良好效果。在此基礎(chǔ)上,JANJIGIAN等[36]進(jìn)行了Ⅲ期KEYNOTE-811研究,第一次中期分析顯示,曲妥珠單抗和派姆單抗聯(lián)合化療客觀緩解率為74.4%,相比于曲妥珠單抗聯(lián)合化療,客觀緩解率提高了22.7%,這表明曲妥珠單抗和派姆單抗聯(lián)合化療對(duì)HER2陽(yáng)性胃癌患者有良好的療效;而不良事件也與之前的Ⅱ期研究一致。第二次中期分析顯示,派姆單抗組中位無(wú)進(jìn)展生存期為10.0個(gè)月(95% CI:8.6~11.7),安慰劑組中位無(wú)進(jìn)展生存期為8.1個(gè)月(95% CI:7.0~8.5)。第三次中期分析顯示,中位無(wú)進(jìn)展生存期為10.0個(gè)月(95% CI:8.6~12.2),安慰劑組為8.1個(gè)月(95% CI:7.1~8.6)。這表明與安慰劑相比,派姆單抗與一線曲妥珠單抗和化療聯(lián)合治療HER2陽(yáng)性晚期胃癌,可顯著提高無(wú)進(jìn)展生存期[37]。也期待繼續(xù)觀察的最終分析結(jié)果,以進(jìn)一步評(píng)估其遠(yuǎn)期治療效果。類(lèi)似的,LEE等[38]在一項(xiàng)Ⅱ期研究中探究了派姆單抗和曲妥珠單抗聯(lián)合卡培他濱和順鉑治療HER2陽(yáng)性晚期胃癌,得到了與KEYNOTE-811研究相近的研究結(jié)果,派姆單抗和曲妥珠單抗聯(lián)合卡培他濱和順鉑中位無(wú)進(jìn)展生存期為8.6個(gè)月;中位總生存期為19.3個(gè)月,長(zhǎng)于ToGA試驗(yàn),安全性方面也可以耐受。總之,派姆單抗、曲妥珠單抗聯(lián)合化療是HER2陽(yáng)性晚期胃癌的一種變革性的治療方法,已經(jīng)初步證明了其效果,但仍需要進(jìn)一步研究,期待KEYNOTE-811研究的最終分析。
5.2 曲妥珠單抗聯(lián)合抗血管內(nèi)皮生長(zhǎng)因子(VEGF)抗體
雷莫盧單抗是一種VEGFR-2阻斷單克隆抗體,研究證實(shí)雷莫盧單抗聯(lián)合化療可延長(zhǎng)晚期胃癌患者的總生存期[39]。貝伐單抗是一種重組人源化抗VEGF單克隆抗體,SINGH等[40]的一項(xiàng)Ⅱ期研究貝伐單抗聯(lián)合卡培他濱、奧沙利鉑和曲妥珠單抗治療先前未經(jīng)治療的HER2陽(yáng)性局部晚期或轉(zhuǎn)移性胃癌患者的安全性和有效性。結(jié)果顯示,客觀緩解率為81%,中位無(wú)進(jìn)展生存期和總生存期分別為14.0個(gè)月和23.2個(gè)月,該研究達(dá)到了主要終點(diǎn)。這表明卡培他濱、奧沙利鉑、曲妥珠單抗和貝伐單抗在HER2陽(yáng)性胃癌中顯示出強(qiáng)大的臨床活性。該方案耐受性良好,沒(méi)有意外或嚴(yán)重毒性,期待未來(lái)進(jìn)一步的研究結(jié)果。
6 總結(jié)及展望
綜上,ToGA實(shí)驗(yàn)確定了曲妥珠單抗聯(lián)合化療是HER2陽(yáng)性晚期胃癌患者的標(biāo)準(zhǔn)治療方案,隨后不斷有研究針對(duì)化療藥物進(jìn)行改善,其中S-1的研究相對(duì)較成熟,表現(xiàn)出了良好的療效和安全性,其他化療藥物的改善仍需要進(jìn)一步研究。帕妥珠單抗和曲妥珠單抗聯(lián)合化療未達(dá)到主要終點(diǎn),T-DM1對(duì)于HER2陽(yáng)性晚期胃癌患者在改善總生存期方面沒(méi)有優(yōu)勢(shì)。目前,KN026、T-DXd、曲妥珠單抗與派姆單抗聯(lián)合化療、貝伐單抗與曲妥珠單抗聯(lián)合卡培他濱和奧沙利鉑是治療HER2陽(yáng)性晚期胃癌的最前沿進(jìn)展,T-DXd對(duì)HER2陰性胃癌也表現(xiàn)出一定的療效。這四種方案都具有廣闊的前景,可能可以有效延長(zhǎng)HER2陽(yáng)性晚期胃癌患者的總生存期和無(wú)進(jìn)展生存期,這尚需未來(lái)進(jìn)一步研究。
參考文獻(xiàn):
[1]AJANI J A, D’AMICO T A, BENTREM D J, et al. Gastric cancer, version 2.2022, NCCN clinical practice guidelines in oncology[J]. J Natl Compr Canc Netw, 2022, 20(2): 167-192.
[2]BANG Y J, VAN CUTSEM E, FEYEREISLOVA A, et al. Trastuzumab in combination with chemotherapy versus chemotherapy alone for treatment of HER2-positive advanced gastric or gastro-oesophageal junction cancer (ToGA): a phase 3, open-label, randomised controlled trial[J]. Lancet, 2010, 376(9742): 687-697.
[3]SHITARA K, YATABE Y, MATSUO K, et al. Prognosis of patients with advanced gastric cancer by HER2 status and trastuzumab treatment[J]. Gastric Cancer, 2013, 16(2): 261-267.
[4]LI Q, LI H, JIANG H, et al. Predictive factors of trastuzumab-based chemotherapy in HER2 positive advanced gastric cancer: a single-center prospective observational study[J]. Clin Transl Oncol, 2018, 20(6): 695-702.
[5]SHAH M A, XU R H, BANG Y J, et al. HELOISE: phase Ⅲb randomized multicenter study comparing standard-of-care and higher-dose trastuzumab regimens combined with chemotherapy as first-line therapy in patients with human epidermal growth factor receptor 2-positive metastatic gastric or gastroesophageal junction adenocarcinoma[J]. J Clin Oncol, 2017, 35(22): 2558-2567.
[6]KOIZUMI W, NARAHARA H, HARA T, et al. S-1 plus cisplatin versus S-1 alone for first-line treatment of advanced gastric cancer (SPIRITS trial): a phase Ⅲ trial[J]. Lancet Oncol, 2008, 9(3): 215-221.
[7]KUROKAWA Y, SUGIMOTO N, MIWA H, et al. Phase Ⅱ study of trastuzumab in combination with S-1 plus cisplatin in HER2-positive gastric cancer (HERBIS-1)[J]. Br J Cancer, 2014, 110(5): 1163-1168.
[8]MIURA Y, SUKAWA Y, HIRONAKA S, et al. Five-weekly S-1 plus cisplatin therapy combined with trastuzumab therapy in HER2-positive gastric cancer: a phase Ⅱ trial and biomarker study (WJOG7212G)[J]. Gastric Cancer, 2018, 21(1): 84-95.
[9]KIMURA Y, FUJII M, MASUISHI T, et al. Multicenter phase Ⅱ study of trastuzumab plus S-1 alone in elderly patients with HER2-positive advanced gastric cancer (JACCRO GC-06)[J]. Gastric Cancer, 2018, 21(3): 421-427.
[10]TAKAHARI D, CHIN K, ISHIZUKA N, et al.Multicenter phase Ⅱ study of trastuzumab with S-1 plus oxaliplatin for chemotherapy-nave, HER2-positive advanced gastric cancer[J]. Gastric Cancer, 2019, 22(6): 1238-1246.
[11]YUKI S, SHINOZAKI K, KASHIWADA T, et al. Multicenter phase Ⅱ study of SOX plus trastuzumab for patients with HER2+ metastatic or recurrent gastric cancer: KSCC/HGCSG/CCOG/PerSeUS 1501B[J]. Cancer Chemother Pharmacol, 2020, 85(1): 217-223.
[12]KAGAWA S, MURAOKA A, KAMBARA T, et al. A multi-institution phase Ⅱ study of docetaxel and S-1 in combination with trastuzumab for HER2-positive advanced gastric cancer (DASH study)[J]. Cancer Chemother Pharmacol, 2018, 81(2): 387-392.
[13]RYU M H, YOO C, KIM J G, et al. Multicenter phase Ⅱ study of trastuzumab in combination with capecitabine and oxaliplatin for advanced gastric cancer[J]. Eur J Cancer, 2015, 51(4): 482-488.
[14]KAWAMOTO Y, YUKI S, MEGURO T, et al. Phase Ⅱ study of continued trastuzumab plus irinotecan in patients with HER2-positive gastric cancer previously treated with trastuzumab (HGCSG 1201)[J]. Oncologist, 2022, 27(5): 340-e374.
[15]MONDACA S, MARGOLIS M, SANCHEZ V F, et al. Phase Ⅱ study of trastuzumab with modified docetaxel, cisplatin, and 5 fluorouracil in metastatic HER2-positive gastric cancer[J]. Gastric Cancer, 2019, 22(2): 355-362.
[16]LEE S C, SHIMASAKI N, LIM J S J, et al. Phase Ⅰ trial of expanded, activated autologous NK-cell infusions with trastuzumab in patients with HER2-positive cancers[J]. Clin Cancer Res, 2020, 26(17): 4494-4502.
[17]VON MINCKWITZ G, PROCTER M, DE AZAMBUJA E, et al. Adjuvant pertuzumab and trastuzumab in early HER2-positive breast cancer[J]. N Engl J Med, 2017, 377(2): 122-131.
[18]KANG Y K, RHA S Y, TASSONE P, et al. A phase Ⅱa dose-finding and safety study of first-line pertuzumab in combination with trastuzumab, capecitabine and cisplatin in patients with HER2-positive advanced gastric cancer[J]. Br J Cancer, 2014, 111(4): 660-666.
[19]TABERNERO J, HOFF P M, SHEN L, et al. Pertuzumab plus trastuzumab and chemotherapy for HER2-positive metastatic gastric or gastro-oesophageal junction cancer (Jacob): final analysis of a double-blind, randomised, placebo-controlled phase 3 study[J]. Lancet Oncol, 2018, 19(10): 1372-1384.
[20]LIU T, QIN Y, LI J, et al. Pertuzumab in combination with trastuzumab and chemotherapy for Chinese patients with HER2-positive metastatic gastric or gastroesophageal junction cancer: a subpopulation analysis of the Jacob trial[J]. Cancer Commun (Lond), 2019, 39(1): 38.
[21]SHITARA K, HARA H, YOSHIKAWA T, et al. Pertuzumab plus trastuzumab and chemotherapy for Japanese patients with HER2-positive metastatic gastric or gastroesophageal junction cancer: a subgroup analysis of the Jacob trial[J]. Int J Clin Oncol, 2020, 25(2): 301-311.
[22]TABERNERO J, HOFF P M, SHEN L, et al. Pertuzumab, trastuzumab, and chemotherapy in HER2-positive gastric/gastroesophageal junction cancer: end-of-study analysis of the Jacob phase Ⅲ randomized clinical trial[J]. Gastric Cancer, 2023, 26(1): 123-131.
[23]XU J, YING J, LIU R, et al. KN026 (anti-HER2 bispecific antibody) in patients with previously treated, advanced HER2-expressing gastric or gastroesophageal junction cancer[J]. Eur J Cancer, 2023, 178: 1-12.
[24]NAM H J, KIM H P, YOON Y K, et al. Antitumor activity of HM781-36B, an irreversible Pan-HER inhibitor, alone or in combination with cytotoxic chemotherapeutic agents in gastric cancer[J]. Cancer Lett, 2011, 302(2): 155-165.
[25]KIM T Y, HAN H S, LEE K W, et al. A phase Ⅰ/Ⅱ study of poziotinib combined with paclitaxel and trastuzumab in patients with HER2-positive advanced gastric cancer[J]. Gastric Cancer, 2019, 22(6): 1206-1214.
[26]SHITARA K, BANG Y J, IWASA S, et al. Trastuzumab deruxtecan in previously treated HER2-positive gastric cancer[J]. N Engl J Med, 2020, 382(25): 2419-2430.
[27]DOI T, SHITARA K, NAITO Y, et al. Safety, pharmacokinetics, and antitumour activity of trastuzumab deruxtecan (DS-8201), a HER2-targeting antibody-drug conjugate, in patients with advanced breast and gastric or gastro-oesophageal tumours: a phase 1 dose-escalation study[J]. Lancet Oncol, 2017, 18(11): 1512-1522.
[28]SHITARA K, IWATA H, TAKAHASHI S, et al. Trastuzumab deruxtecan (DS-8201a) in patients with advanced HER2-positive gastric cancer: a dose-expansion, phase 1 study[J]. Lancet Oncol, 2019, 20(6): 827-836.
[29]XU H, ZHANG H, GUO W, et al. Safety and efficacy profile of Trastuzumab deruxtecan in solid cancer: pooled reanalysis based on clinical trials[J]. BMC Cancer, 2022, 22(1): 923.
[30]VAN CUTSEM E, DI BARTOLOMEO M, SMYTH E, et al. Trastuzumab deruxtecan in patients in the USA and Europe with HER2-positive advanced gastric or gastroesophageal junction cancer with disease progression on or after a trastuzumab-containing regimen (DESTINY-Gastric02): primary and updated analyses from a single-arm, phase 2 study[J]. Lancet Oncol, 2023, 24(7): 744-756.
[31]YAMAGUCHI K, BANG Y J, IWASA S, et al. Trastuzumab deruxtecan in anti-human epidermal growth factor receptor 2 treatment-naive patients with human epidermal growth factor receptor 2-low gastric or gastroesophageal junction adenocarcinoma: exploratory cohort results in a phase Ⅱ trial[J]. J Clin Oncol, 2023, 41(4): 816-825.
[32]SHITARA K, BANG Y J, IWASA S, et al. Trastuzumab deruxtecan in HER2-positive advanced gastric cancer: exploratory biomarker analysis of the randomized, phase 2 DESTINY-Gastric01 trial[J]. Nat Med, 2024, 30(7): 1933-1942.
[33]THUSS-PATIENCE P C, SHAH M A, OHTSU A, et al. Trastuzumab emtansine versus taxane use for previously treated HER2-positive locally advanced or metastatic gastric or gastro-oesophageal junction adenocarcinoma (Gatsby): an international randomised, open-label, adaptive, phase 2/3 study[J]. Lancet Oncol, 2017, 18(5): 640-653.
[34]SHITARA K, HONMA Y, OMURO Y, et al. Efficacy of trastuzumab emtansine in Japanese patients with previously treated HER2-positive locally advanced or metastatic gastric or gastroesophageal junction adenocarcinoma: a subgroup analysis of the Gatsby study[J]. Asia Pac J Clin Oncol, 2020, 16(1): 5-13.
[35]JANJIGIAN Y Y, MARON S B, CHATILA W K, et al. First-line pembrolizumab and trastuzumab in HER2-positive oesophageal, gastric, or gastro-oesophageal junction cancer: an open-label, single-arm, phase 2 trial[J]. Lancet Oncol, 2020, 21(6): 821-831.
[36]JANJIGIAN Y Y, KAWAZOE A, YAEZ P, et al. The KEYNOTE-811 trial of dual PD-1 and HER2 blockade in HER2-positive gastric cancer[J]. Nature, 2021, 600(7890): 727-730.
[37]JANJIGIAN Y Y, KAWAZOE A, BAI Y, et al. Pembrolizumab plus trastuzumab and chemotherapy for HER2-positive gastric or gastro-oesophageal junction adenocarcinoma: interim analyses from the phase 3 KEYNOTE-811 randomised placebo-controlled trial[J]. Lancet, 2023, 402(10418): 2197-2208.
[38]LEE C K, RHA S Y, KIM H S, et al. A single arm phase Ⅰb/Ⅱ trial of first-line pembrolizumab, trastuzumab and chemotherapy for advanced HER2-positive gastric cancer[J]. Nat Commun, 2022, 13(1): 6002.
[39]FUCHS C S, SHITARA K, DI BARTOLOMEO M, et al. Ramucirumab with cisplatin and fluoropyrimidine as first-line therapy in patients with metastatic gastric or junctional adenocarcinoma (RAINFALL): a double-blind, randomised, placebo-controlled, phase 3 trial[J]. Lancet Oncol, 2019, 20(3): 420-435.
[40]SINGH H, LOWDER K E, KAPNER K, et al. Clinical outcomes and ctDNA correlates for CAPOX BETR: a phase Ⅱ trial of capecitabine, oxaliplatin, bevacizumab, trastuzumab in previously untreated advanced HER2+ gastroesophageal adenocarcinoma[J]. Nat Commun, 2024, 15(1): 6833.
(編輯 國(guó) 榮)