Panitumumab–FOLFOX4 Treatment and RAS Mutations in Colorectal Cancer
folfox4方案

Folfox4方案简介Folfox4方案是一种用于治疗结直肠癌的化疗方案。
它是通过联合应用氟尿嘧啶(5-FU)、氧铂(Oxaliplatin)和白蛋白结合型叶酸(Leucovorin)来实施的。
这种化疗方案已被证明在治疗结直肠癌方面具有显著效果,并被广泛应用于临床实践中。
药物组成Folfox4方案由以下三种药物组成:1.氟尿嘧啶(5-FU):它是一种抗代谢药物,通过干扰细胞嘌呤和嘧啶的转化来抑制癌细胞的生长。
5-FU主要通过经静脉输注的形式给予患者。
2.氧铂(Oxaliplatin):它属于铂类化合物,通过与DNA结合来干扰癌细胞的复制和分裂,从而抑制其生长。
氧铂通过经静脉输注的方式给予患者。
3.白蛋白结合型叶酸(Leucovorin):它是一种辅助药物,可以增强氟尿嘧啶的抗癌作用。
白蛋白结合型叶酸通常通过经静脉输注的方式与5-FU 联合应用。
方案步骤Folfox4方案通常分为以下几个步骤:1.预处理阶段:在开始化疗之前,医生会进行一系列的评估和检查,包括病情分析、病理学检查、肝功能检查等。
这些步骤可以确定患者是否适合接受Folfox4方案的治疗。
2.化疗准备:在开始化疗之前,医生会根据患者的身体状况确定药物剂量和使用频率。
通常情况下,氟尿嘧啶和氧铂会以静脉输注的方式给予患者,并且会同时用白蛋白结合型叶酸进行联合应用。
3.化疗周期:Folfox4方案通常为14天一个周期,其中5-FU、氧铂和白蛋白结合型叶酸会在第一天和第二天连续静脉输注,然后患者会有12天的休息期。
整个治疗过程通常会持续6个月至1年不等,具体的疗程会根据患者的病情而定。
4.治疗效果评估:在每个周期结束后,医生会进行治疗效果的评估,包括病灶缩小情况、生物学标志物的变化等。
这些评估结果会帮助医生调整下个周期的治疗方案。
5.治疗结束:在完成所有化疗周期后,医生会再次进行病情评估来确定整个化疗方案的效果。
如果病情得到控制或缓解,医生可能会考虑进一步的治疗方案,如手术切除或放疗。
folfox4化疗方案

folfox4化疗方案:治疗结直肠癌的利器结直肠癌是一种严重的癌症,近年来发病率不断上升。
据统计,全球每年有超过150万人因结直肠癌去世。
而化疗一直是治疗结直肠癌的重要手段之一。
是一种较常用的化疗方案,下面我们将详细介绍。
一、的基本介绍是一种针对结直肠癌的综合性化疗方案。
主要包括药物氟尿嘧啶、奥沙利铂和亚叶酸钙等。
该方案的具体流程为:氟尿嘧啶+亚叶酸钙:氟尿嘧啶为一种以氟代嘌呤为基础的化疗药物,可以干扰癌细胞的DNA合成,抑制癌细胞的生长及扩散。
亚叶酸钙则可以帮助氟尿嘧啶发挥最大化的作用。
氟尿嘧啶+奥沙利铂+亚叶酸钙:在基础药物的基础上,再加入奥沙利铂。
奥沙利铂是一种有效的钯族药物,可以直接对DNA结构进行作用,并善于在肝脏中代谢掉,不会对肝脏产生毒副作用。
注意事项:通常需要通过静脉输液的方式进行,持续2天。
在治疗期间应尽可能避免感染,保证饮食和休息。
二、 FOLFOX4的作用机理主要通过化学作用,破坏癌细胞内部正常的分裂与复制过程,从而抑制癌细胞的生长和繁殖。
同时,它也会影响癌细胞的细胞凋亡过程,从而促使癌细胞死亡。
FOLFOX4还可以增强患者自身免疫力,有助于防止癌细胞的复发和转移。
三、优点和不良反应作为一种较常用的化疗方案,其具有以下一些优点:1、FOLFOX4相比起其他的化疗方案,更能有效降低肿瘤的复发率和转移风险。
2、FOLFOX4的药物选择偏轻,因此治疗的副作用相对较少,负担也不会太大。
3、FOLFOX4可以比较好地控制治疗周期,并且不容易对患者的身体产生直接的影响。
而的不良反应主要表现在以下几个方面:1、术后伤口愈合问题:化疗会对患者的伤口愈合产生影响。
因此,在化疗期间,患者需要特别注意自己的术后伤口的护理。
2、恶心呕吐:化疗过程中,患者很可能会出现恶心呕吐等消化道不适症状。
此时,患者需要积极采取措施进行治疗。
3、感染控制:由于化疗期间免疫力会有所下降,容易出现感染问题。
四、结语作为一种常用的结直肠癌综合性化疗方案,可以有效地抑制肿瘤的生长和扩散,并降低肿瘤的复发率和转移风险。
folfox4化疗方案

folfox4化疗方案FOLFOX4化疗方案是一种常用于治疗结直肠癌的化疗方案。
该方案采用联合使用氟尿嘧啶、奥沙利铂和亚叶酸钙三种药物进行治疗。
本文将对FOLFOX4化疗方案的药物组合、适应症、治疗周期和可能的副作用等方面进行介绍。
一、药物组合FOLFOX4化疗方案由三种药物组成,分别是氟尿嘧啶、奥沙利铂和亚叶酸钙。
其中,氟尿嘧啶是一种抑制肿瘤细胞生长的抗代谢药物,奥沙利铂是一种铂类化合物,而亚叶酸钙则可起到增强氟尿嘧啶的药效和减轻氟尿嘧啶毒副作用的作用。
二、适应症FOLFOX4化疗方案主要适用于结直肠癌的治疗,尤其是晚期结直肠癌和转移性结直肠癌。
此外,该方案也可用于局部进展性结直肠癌的术前和术后辅助治疗,以提高手术切除的效果和减少复发率。
三、治疗周期FOLFOX4化疗方案的治疗周期通常为14天。
具体治疗方案如下:1. 第一天:奥沙利铂通过静脉滴注给药,剂量为85mg/m²,注射时间为2小时;亚叶酸钙以400 mg/m²剂量口服给药,与奥沙利铂静脉滴注同时开始。
2. 第一天至第二天:持续静脉滴注氟尿嘧啶,剂量为400 mg/m²,注射时间为2小时,随后再以1200 mg/m²剂量进行22小时的持续静脉滴注。
3. 第三天:停药。
以上三天为一个治疗周期,接下来根据病情需要,在两个周期之间有2周的休息期。
四、可能的副作用使用FOLFOX4化疗方案时,患者可能出现一些副作用。
常见的副作用包括恶心、呕吐、腹泻、口腔溃疡、皮肤变化(如手足综合征、皮疹等)和骨髓抑制(如白细胞减少、贫血、血小板减少等)等。
这些副作用通常在停药或调整剂量后会有所缓解。
除了上述常见的副作用外,FOLFOX4化疗方案还可能导致感觉异常(如手脚麻木、感觉丧失等)、过敏反应和肝功能损害等。
在使用该方案治疗的过程中,医生会根据患者的具体情况进行监测并及时处理。
总之,FOLFOX4化疗方案是一种常用的治疗结直肠癌的化疗方案,通过联合使用氟尿嘧啶、奥沙利铂和亚叶酸钙等药物,可以有效抑制肿瘤细胞的生长和扩散。
folfox4方案

Folfox4方案简介Folfox4方案是一种常用于治疗结直肠癌的化疗方案。
它通过联合使用多种药物,可有效控制结直肠癌的生长和扩散,提高患者的生存率。
药物组合Folfox4方案由3种药物的联合使用组成,分别是:1.氟尿嘧啶(5-FU):氟尿嘧啶是一种抗癌药物,通过干扰癌细胞的DNA合成过程来抑制癌细胞的生长。
2.奥沙利铂(Oxaliplatin):奥沙利铂是一种铂类药物,可通过交联DNA链来抑制癌细胞的增殖。
3.亮丙氨酰葡萄糖(Leucovorin):亮丙氨酰葡萄糖是一种辅助剂,可增强5-FU的作用。
这三种药物的联合使用可提高治疗效果,同时也增加了一些不良反应的风险。
治疗周期Folfox4方案的治疗周期通常为14天,包括以下几个步骤:1.第1天:奥沙利铂和亮丙氨酰葡萄糖的静脉注射。
2.第1天至第2天:5-FU通过泵式输注进入血液。
3.第2天至第14天:休息期,患者可以回家休养。
4.第15天:再次进行奥沙利铂和亮丙氨酰葡萄糖的静脉注射。
5.第15天至第16天:再次进行5-FU的泵式输注。
6.第16天至第28天:休息期。
整个治疗周期共计28天,然后根据患者的具体情况决定是否进行下一周期的治疗。
不良反应尽管Folfox4方案可以有效治疗结直肠癌,但也伴随着一些不良反应。
常见的不良反应包括:1.恶心和呕吐:由于化疗药物的毒副作用,患者可能会出现恶心和呕吐的症状。
医生通常会给予相应的抗恶心药物来缓解这些不适。
2.外周神经病变:奥沙利铂的使用可能导致患者出现外周神经病变的症状,如手脚麻木、感觉异常等。
在治疗期间,医生会密切监测患者的神经功能,必要时调整药物剂量以减少这些不良反应。
3.皮肤反应:化疗药物也可能引起一些皮肤反应,如皮疹、干燥和瘙痒等。
患者可以采取保湿护肤措施来缓解这些症状。
4.骨髓抑制:Folfox4方案可能导致骨髓抑制,使患者的血小板、白细胞和红细胞减少。
在治疗期间,医生会定期检查患者的血液指标,必要时采取相应的对策。
FOLFOX_4方案治疗晚期胃癌24例临床观察

FOLFOX_4方案治疗晚期胃癌24例临床观察陈萍;周建明;卓德斌【期刊名称】《南通医学院学报》【年(卷),期】2004(24)4【摘要】目的 :探索草酸铂 (OXA) /5 -氟尿嘧啶 (5 -Fu) /甲酰四氢叶酸钙 (CF)治疗晚期胃癌近期疗效及其毒副作用。
方法 :采用 FOIFOX4 方案 ,每 2周为 1周期 ,2周期为 1疗程 ,用足 2疗程者给予疗效评定。
结果 :全组 2 4例 ,总有效率为5 0 % ;初治组中位缓解期为 5个月 ,复治组中位缓解期为 3个月 , 、度口腔炎发生率为 2 0 .8% ,2例出现手足综合征 ,主要出现累积剂量增加而毒性也逐渐加重的外周神经毒性 5 4.1% ,血液学毒性轻微。
结论 :FOLFOX4 方案是晚期胃癌安全有效的治疗方案。
【总页数】2页(P438-438)【关键词】晚期胃癌;化学治疗;草酸铂;5-氟尿嘧啶;甲酰四氢叶酸钙【作者】陈萍;周建明;卓德斌【作者单位】江苏省南通市第三人民医院肿瘤内科;南通市肿瘤医院肿瘤内科【正文语种】中文【中图分类】R735.2【相关文献】1.CS方案和SOX方案治疗晚期胃癌的临床观察 [J], 宋红蕾;毕延智;陈慧暖;徐珍;董益忠;盛桂凤;张亚平;胡岳棣2.改良 DCF 方案与 XELOX 方案一线治疗晚期胃癌的临床观察 [J], 张从军;孙国平;熊福星;彭万仁;李笑秋;范璐璐3.PCF方案和FOLFOX4方案治疗中晚期胃癌的临床观察 [J], 刘林青;胡世莲;沈干;沈国栋;王峰;徐维平;程民;唐杨琛;徐婷娟4.mFLOT方案和mFOLFOX6方案治疗晚期或局部进展期胃癌的临床观察 [J], 刘显菊; 姬发祥5.FolFox_4方案治疗晚期胃肠道恶性肿瘤42例的临床观察 [J], 张晓东;龚振夏因版权原因,仅展示原文概要,查看原文内容请购买。
FOLFOX4方案治疗晚期胃癌的临床观察

FOLFOX4方案治疗晚期胃癌的临床观察赵泉;龚昆梅;王昆华【期刊名称】《肿瘤预防与治疗》【年(卷),期】2010(023)003【摘要】目的:观察FOLFOX4方案治疗晚期胃癌的近期疗效和毒副反应.方法:26例晚期胃癌患者,采用FOLFOX4方案,即:奥沙利铂(L-OHP) 85mg/m2静脉点滴2小时,第1天;亚叶酸钙(CF)200mg/m2静脉点滴, 2小时, 第1,2天;氟脲嘧啶 (5-Fu)400mg/m2静脉推注, 第1,2天, 5-Fu 600mg/m2微泵持续静脉滴注22小时, 第1,2天;2周重复.4个周期后评价疗效和毒性.结果:全组26例均可评价,其中完全缓解(CR)1例,部分缓解(PR)13例,无变化(NC)8例,进展(PD)4例,总有效率(CR+PR)46.1%.中位生存时间(MST)为10个月.毒副反应主要是骨髓抑制,中性粒细胞减少发生率达84.6%,其次为胃肠道反应,恶心呕吐发生率88.5%,腹泻46.1%,无Ⅲ度~Ⅳ度胃肠道反应;本组患者未见明显心功能及肝肾功能损害.结论:FOLFOX4方案治疗晚期胃癌的近期疗效较好,毒副反应可以耐受,值得进一步研究应用.【总页数】3页(P206-208)【作者】赵泉;龚昆梅;王昆华【作者单位】云南省第一人民医院,普外一科,昆明,650032;云南省第一人民医院,普外一科,昆明,650032;云南省第一人民医院,普外一科,昆明,650032【正文语种】中文【中图分类】R730.53;R735.2【相关文献】1.FOLFOX4方案治疗晚期胃癌临床观察 [J], 覃汉华2.扶正抗癌汤联合FOLFOX4方案治疗晚期胃癌临床观察 [J], 乔炳礼3.XELOX方案和FOLFOX4方案治疗晚期胃癌的疗效和毒副作用的临床观察 [J], 周江华;魏蓉4.鸦胆子油乳注射液联合FOLFOX4方案治疗晚期胃癌临床观察 [J], 吴亚丛;张迎春;戴光熙;王利军;叶进科5.参苓白术散加减联合FOLFOX4方案治疗晚期胃癌的临床观察 [J], 郭孝鸽因版权原因,仅展示原文概要,查看原文内容请购买。
结直肠癌KRAS和BRAF基因突变及其与临床病理的相关性
结直肠癌KRAS和BRAF基因突变及其与临床病理的相关性谢明智;李科志;利基林;蔡政民;胡榜利【摘要】目的探讨结直肠癌患者KRAS和BRAF基因突变与临床病理特征的关系.方法应用实时荧光TaqMan探针法检测322例结直肠癌患者KRAS和BRAF基因突变情况,分析患者不同年龄、性别、肿瘤大小、分化程度等不同组别的基因突变情况与临床病理特征的关系.结果 322例结直肠癌患者中,结肠癌患者182例,直肠癌患者140例,KRAS基因突变105例(32.6%),BRAF基因突变13例(4.0%).结肠癌患者和直肠癌患者在性别、年龄、肿瘤分化程度、淋巴结转移、远处转移、肿瘤分期指标比较差异无统计学意义(P>0.05);但结肠癌患者T3 +T4的比例高于直肠癌患者(P=0.004).结肠癌和直肠癌患者KRAS和BRAF基因突变情况比较差异无统计学意义(P>0.05).在105例KRAS基因突变中,G12D突变40例(38.1%),G12S突变38例(36.2%),G12A突变12例(11.4%),G12V突变9例(8.6%),Gl3A突变6例(5.7%).13例BRAF基因突变型均为V600E型.右半结肠癌的KRAS基因突变率显著高于左半结肠癌和直肠癌(P=0.010);BRAF基因突变的患者年龄高于未突变者(P =0.022),突变患者发生淋巴结转移的比例比未发生转移的比例低(P =0.020);突变患者Ⅲ+Ⅳ期的比例低于未突变患者(P =0.044).结论结直肠癌患者存在较高的KRAS基因突变率,而BRAF基因突变率较低.KRAS基因突变与肿瘤的原发部位密切相关,且与BRAF基因突变无直接相关性.【期刊名称】《广东医学》【年(卷),期】2019(040)008【总页数】4页(P1128-1131)【关键词】结直肠肿瘤;KRAS;BRAF;基因突变【作者】谢明智;李科志;利基林;蔡政民;胡榜利【作者单位】广西医科大学附属肿瘤医院实验研究部广西南宁530021;广西医科大学附属肿瘤医院实验研究部广西南宁530021;广西医科大学附属肿瘤医院实验研究部广西南宁530021;广西医科大学附属肿瘤医院实验研究部广西南宁530021;广西医科大学附属肿瘤医院实验研究部广西南宁530021【正文语种】中文【中图分类】R735.3;R394.3结直肠癌是消化系统最常见的肿瘤之一,除了手术治疗外,肿瘤靶向治疗成为结直肠癌的重要治疗方式,其中抗表皮生长因子受体(EGFR)是目前治疗转移性结直肠癌的主要靶向药物[1]。
folfox4方案化疗
1. 简介Folfox4方案是一种常用于结直肠癌化疗的治疗方案。
本文将介绍Folfox4方案的具体内容、治疗原理、适应症和不良反应等相关内容。
2. Folfox4方案的内容Folfox4方案的化疗药物包括氟尿嘧啶(5-FU)、氧铂(Oxaliplatin)和亚叶酸(Leucovorin)。
具体方案如下:•氟尿嘧啶(5-FU):根据体表面积给予静脉滴注,每周一次,剂量为400mg/m^2。
•氧铂(Oxaliplatin):根据体表面积给予静脉滴注,每隔两周一次,剂量为85mg/m^2。
•亚叶酸(Leucovorin):与氟尿嘧啶(5-FU)联合使用,剂量为200mg/m^2,与氟尿嘧啶(5-FU)一同静脉滴注。
整个方案包含12个周期,每个周期的治疗时间为2周。
3. Folfox4方案的治疗原理Folfox4方案通过联合使用氟尿嘧啶、氧铂和亚叶酸来进行化疗。
这三种药物分别发挥不同的作用:•氟尿嘧啶是一种抗代谢药物,它能干扰癌细胞的DNA合成过程,抑制癌细胞的增殖能力。
•氧铂是一种铂类药物,它通过与DNA结合,造成DNA损伤,直接杀死癌细胞。
•亚叶酸是一种维生素B9的衍生物,它能增强氟尿嘧啶的抗肿瘤活性。
三种药物的联合使用,通过不同的作用机制,可以增强化疗的效果,提高对癌细胞的杀伤力。
4. Folfox4方案的适应症Folfox4方案一般适用于结直肠癌的治疗,特别是转移性结直肠癌。
具体适应症如下:•原发性结直肠癌:Folfox4方案可用于原发性结直肠癌的术前或术后辅助化疗。
•转移性结直肠癌:Folfox4方案可用于转移性结直肠癌的一线治疗或二线治疗。
需要注意的是,对于不同患者,具体的治疗方案可能会有所不同,医生会根据患者的实际情况进行调整。
5. Folfox4方案的不良反应Folfox4方案的化疗药物具有一定的毒副作用,常见的不良反应包括:•恶心呕吐:氟尿嘧啶和氧铂可能引起恶心呕吐,但一般可以通过适当的药物控制来减轻这些症状。
转移性结直肠癌治疗的新进展_Mar_2014_
雷替曲塞的临床应用
雷替曲塞单药一线治疗转移性结直肠癌的Ⅱ期临床试验
欧洲、澳洲、南非的15个中心 N=177 转移灶未治,5%接受过辅化
雷替曲塞 3mg/m2,推注15分钟,每3周重复
Gallagher DJ, et al. Oncology. 2010;78:237-248.
mCRC: 主要的一线化疗效果
比较方案
IFL vs FOLFOX vs IROX[1] FOLFIRI vs FOLFOX4[2] XELOX (CapeOx) vs FOLFOX4[3,4] FOLFIRI vs mIFL vs CapeIRI[5]
mCRC : 分子靶点药
分子靶点药
Cetuximab (Erbitux,C225)
雷替曲赛临床应用特点 (3)
患者依从性好 雷替曲赛推荐剂量为3mg/M2,15分钟静脉 滴注,每三周一个疗程。故雷替曲赛的用 药方案明显由于5-FU/亚叶酸钙的每4周连 续5天的用药方案。给患者带来更大便利。
雷替曲赛临床应用特点 (4)
抗肿瘤谱广 雷替曲赛除对结直肠癌有效外,临床试验 中单药或与其他抗肿瘤药及化疗联用,对 其他多种肿瘤(如乳腺癌,胰腺癌,非小 细胞肺癌,难治性卵巢癌)有效。
50%病人最终死于本病。
结直肠癌 : 诊断时的分期
Stage 0 Stage IV 7% 19% Stage I 24%
Stage III 25% Stage II 25%
National Cancer Database.
转移性结直肠癌化疗
化疗药物, 化疗方案; 首次治疗; 个体化用药策略 根据生物标记物选择; 较新靶向药物; 进展后的延续治疗 小结
folfox4化疗方案
folfox4化疗方案化疗是一种通过使用化学药物来治疗癌症的治疗方法,常用于治疗结直肠癌。
folfox4化疗方案是一种常见的化疗方案,被广泛应用于结直肠癌的治疗中。
本文将介绍folfox4化疗方案的具体内容及其疗效。
一、folfox4化疗方案的组成folfox4化疗方案由三种不同的化疗药物组成:氟尿嘧啶(5-FU)、亚叶酸(Leucovorin)和奥沙利铂(Oxaliplatin)。
1. 氟尿嘧啶(5-FU):氟尿嘧啶是一种常用的抗癌药物,通过抑制DNA合成和细胞分裂来阻止癌细胞的生长和扩散。
在folfox4方案中,氟尿嘧啶以静脉注射的形式给药。
2. 亚叶酸(Leucovorin):亚叶酸是一种葡萄糖胺,通过增加氟尿嘧啶的抗癌活性来增强治疗效果。
亚叶酸被认为是氟尿嘧啶的增效剂,在folfox4方案中与氟尿嘧啶一起使用。
3. 奥沙利铂(Oxaliplatin):奥沙利铂是一种铂类化学药物,通过干扰DNA链的结构和功能来杀死癌细胞。
奥沙利铂在folfox4方案中以静脉注射的形式给药,与氟尿嘧啶和亚叶酸一起使用。
二、folfox4化疗方案的治疗周期和剂量folfox4化疗方案通常按照14天一个周期进行,每个周期包括治疗日和休息日。
1. 治疗日:在治疗日,患者会接受化疗药物的静脉注射。
根据患者的具体情况,氟尿嘧啶、亚叶酸和奥沙利铂的剂量会有所调整。
2. 休息日:在休息日,患者不接受化疗药物治疗,给予身体恢复的时间。
治疗周期的具体次数取决于医生的建议和患者的情况,通常为6至12个周期。
三、folfox4化疗方案的疗效folfox4化疗方案在结直肠癌的治疗中显示出良好的疗效。
1. 提高患者的生存率:研究表明,与单独使用氟尿嘧啶治疗相比,folfox4化疗方案可以显著提高结直肠癌患者的生存率。
2. 缩小肿瘤大小:folfox4化疗方案可以有效地缩小肿瘤的大小,有助于手术切除肿瘤或减轻症状。
3. 防止癌症复发:folfox4化疗方案可以减少癌细胞的复发和转移,提高治疗的长期效果。
- 1、下载文档前请自行甄别文档内容的完整性,平台不提供额外的编辑、内容补充、找答案等附加服务。
- 2、"仅部分预览"的文档,不可在线预览部分如存在完整性等问题,可反馈申请退款(可完整预览的文档不适用该条件!)。
- 3、如文档侵犯您的权益,请联系客服反馈,我们会尽快为您处理(人工客服工作时间:9:00-18:30)。
T h e ne w engl a nd jour na l o f medicinen engl j med 369;11 september 12, 20131023Panitumumab–FOLFOX4 Treatment and RAS Mutations in Colorectal CancerJean-Yves Douillard, M.D., Ph.D., Kelly S. Oliner, Ph.D., Salvatore Siena, M.D.,Josep Tabernero, M.D., Ronald Burkes, M.D., Mario Barugel, M.D.,Yves Humblet, M.D., Ph.D., Gyorgy Bodoky, M.D., Ph.D.,David Cunningham, M.D., Jacek Jassem, M.D., Ph.D., Fernando Rivera, M.D., Ph.D., Ilona Kocákova, M.D., Ph.D., Paul Ruff, M.D., Maria Błasińska-Morawiec, M.D.,Martin Šmakal, M.D., Jean Luc Canon, M.D., Mark Rother, M.D.,Richard Williams, M.B., B.S., Ph.D., Alan Rong, Ph.D., Jeffrey Wiezorek, M.D.,Roger Sidhu, M.D., and Scott D. Patterson, Ph.D.From Institut de Cancérologie de l’Ouest (ICO) René Gauducheau, Nantes, France (J.-Y.D.); Amgen, Thousand Oaks, CA (K.S.O., R.W., A.R., J.W., R.S., S.D.P.); Os-pedale Niguarda Ca’ Granda, Milan (S.S.); Vall d’Hebron University Hospital, Univer-sitat Autònoma de Barcelona, Barcelona (J.T.), and Hospital Universitario Marqués de Valdecilla, Santander (F.R.) — both in Spain; Mount Sinai Hospital, Toronto (R.B.), and Credit Valley Hospital, Mississauga, ON (M.R.) — both in Canada; Hospital de Gastroenterología, Buenos Aires (M.B.); Université Catholique de Louvain, Brussels (Y.H.), and Grand Hôpital de Charleroi, Charleroi (J.L.C.) — both in Belgium; Szent László Hospital, Budapest, Hungary (G.B.); Royal Marsden National Health Service Foundation Trust, London (D.C.); Medical University of Gdansk, Gdansk (J.J.), and Copernicus Memorial Hospital, Lodz (M.B.-M.) — both in Poland; Ma-saryk Memorial Cancer Institute, Brno (I.K.), and Institute of Oncology and Reha-bilitation Pleší, Nová Ves pod Pleší (M.Š.) — both in the Czech Republic; and the University of the Witwatersrand Faculty of Health Sciences, Johannesburg (P.R.). Address reprint requests to Dr. Douillard at ICO René Gauducheau, Blvd. Jacques Monod, 44805 Saint Herblain, France, or at jean-yves.douillard@ico.unicancer.fr.Drs. Douillard and Oliner contributed equal-ly to this article.N Engl J Med 2013;369:1023-34.DOI: 10.1056/NEJMoa1305275Copyright © 2013 Massachusetts Medical Society.ABSTR ACTBackgroundPatients with metastatic colorectal cancer that harbors KRAS mutations in exon 2 do not benefit from anti–epidermal growth factor receptor (EGFR) therapy. Other activating RAS mutations may also be negative predictive biomarkers for anti-EGFR therapy.MethodsIn this prospective–retrospective analysis, we assessed the efficacy and safety of pani-tumumab plus oxaliplatin, fluorouracil, and leucovorin (FOLFOX4) as compared with FOLFOX4 alone, according to RAS (KRAS or NRAS ) or BRAF mutation status. A total of 639 patients who had metastatic colorectal cancer without KRAS mutations in exon 2 had results for at least one of the following: KRAS exon 3 or 4; NRAS exon 2, 3, or 4; or BRAF exon 15. The overall rate of ascertainment of RAS status was 90%.ResultsAmong 512 patients without RAS mutations, progression-free survival was 10.1 months with panitumumab–FOLFOX4 versus 7.9 months with FOLFOX4 alone (hazard ratio for progression or death with combination therapy, 0.72; 95% confidence interval [CI], 0.58 to 0.90; P = 0.004). Overall survival was 26.0 months in the panitumumab–FOLFOX4 group versus 20.2 months in the FOLFOX4-alone group (hazard ratio for death, 0.78; 95% CI, 0.62 to 0.99; P = 0.04). A total of 108 patients (17%) with non-mutated KRAS exon 2 had other RAS mutations. These mutations were associated with inferior progression-free survival and overall survival with panitumumab–FOLFOX4 treatment, which was consistent with the findings in patients with KRAS mutations in exon 2. BRAF mutations were a negative prognostic factor. No new safety signals were identified.ConclusionsAdditional RAS mutations predicted a lack of response in patients who received pani t umumab –FOLFOX4. In patients who had metastatic colorectal cancer with-out RAS mutations, improvements in overall survival were observed with panitum-umab–FOLFOX4 therapy. (Funded by Amgen and others; PRIME number, NCT00364013.)T h e ne w engl a nd jour na l o f medicinen engl j med 369;11 september 12, 20131024KRAS mutation is an established pre-dictive biomarker of resistance to anti–epidermal growth factor receptor (EGFR)therapy in patients with metastatic colorectal cancer.1-4 Specifically, patients with KRAS muta-tions in exon 2 do not have a response to anti-EGFR therapy and may have inferior outcomes if this therapy is combined with an oxaliplatin-con-taining chemotherapy regimen.2,5 More accurate selection of patients according to the genetic sta-tus of the tumor may improve the benefit–risk profile of anti-EGFR therapy.Activating mutations in RAS (KRAS or NRAS ) in addition to KRAS mutations in exon 2 have been suggested as negative predictive biomarkers for anti-EGFR therapy. This is biologically plausible on the basis of the existing biochemical and mutational data. KRAS and NRAS are closely re-lated RAS oncogene family members, and muta-tions in either gene at codons 12, 13, 61, 117, and 146 result in increased levels of guanosine triphosphate–bound RAS proteins.6,7 In addition, colorectal tumors harbor KRAS and NRAS muta-tions at these codons, and mutations tend to be mutually exclusive; this suggests functional redun-dancy.8 Mutations in HRAS , the third member of the RAS family, occur infrequently in colorectal cancer.8,9Clinical data have also implicated RAS genes as negative predictive biomarkers. In a randomized phase 3 study of panitumumab monotherapy 10 and other studies,11-15 most patients with metastatic colorectal-cancer tumors harboring a mutation in KRAS or NRAS did not have a response to anti-EGFR therapy.BRAF mutations are typically exclusive of RAS mutations, and the clinical data suggest that the BRAF V600E mutation is prognostic of pa-tient outcome with respect to survival, but not clearly predictive of treatment effects with anti-EGFR agents, in patients with metastatic colorec-tal cancer.16-19 Although no objective responses to panitumumab or cetuximab monotherapy have been reported in patients with metastatic colorectal cancer and BRAF mutations,10,20 the low prevalence of such mutations makes it dif-ficult to evaluate them as predictive biomarkers.Previous studies of anti-EGFR therapies com-bined with oxaliplatin-containing regimens have shown negative outcomes in subgroups of pa-tients with KRAS mutations in exon 2. Identifica-tion of other biomarker-defined subgroups withsimilar outcomes would influence the choice of therapy.2,5 Here, we present the results of a pro-spective–retrospective biomarker analysis of the treatment effect of the full spectrum of cur-rently characterized RAS (KRAS and NRAS ) and BRAF mutations on progression-free survival and overall survival in a randomized phase 3 study of panitumumab plus oxaliplatin, fluorouracil, and leucovorin (FOLFOX4) as compared with FOLFOX4 alone in patients with previously un-treated metastatic colorectal cancer.MethodsStudy Design and OversightThe Panitumumab Randomized Trial in Combina-tion with Chemotherapy for Metastatic Colorectal Cancer to Determine Efficacy (PRIME) compared the efficacy and safety of panitumumab–FOLFOX4 with those of FOLFOX4 alone in the first-line treatment of patients, according to KRAS exon 2 status. The primary end point was progression-free survival. The secondary end points included overall survival and safety.2The study was designed by the sponsor, Amgen, in collaboration with the first author and the study steering committee. Clinical data were collected by the investigators, and sequencing analysis was conducted by Transgenomic under the direction of the sponsor. The sponsor performed all statis-tical analyses. All authors vouch for the accuracy of the data and analyses and for the fidelity of this report to the protocol, which is available with the full text of this article at . The preliminary draft of the manuscript was written by the second author with the assistance of a medical writer who was paid by the sponsor. Subsequent drafts were revised and reviewed by all the authors. All the authors made the deci-sion to submit the manuscript for publication.Tumor SpecimensBanked tumor specimens that were character-ized as nonmutated KRAS exon 2 on the basis of an assay for investigational use only (TheraScreen KRAS Mutation Kit, Qiagen; LightCycler, Roche) were selected for analysis.2 DNA was extracted from formalin-fixed, paraffin-embedded tumor specimens with the use of a DNA Extraction Mini Kit (Qiagen). Specimens that contained less than 50% tumor area were macrodissected. In a few cases, DNA was extracted from storedpanitumumab–folfox4 in Colorectal Cancern engl j med 369;11 september 12, 20131025slides that had been stained for immunohisto-chemical analysis.Mutational Analysis Mutations in KRAS exon 3 (at codon 61) and exon 4 (at codons 117 and 146); NRAS exon 2 (at codons 12 and 13), exon 3 (at codon 61), and exon 4 (at codons 117 and 146); and BRAF exon 15 (at codon 600) were prespecified on the basis of previous studies.7,14,21,22 Gene alterations that were not prespecified (e.g., KRAS and NRAS exon 3 [codon 59] mutations) were analyzed as explor-atory end points. Polymerase-chain-reaction (PCR) primer sequences amplified regions up to 200 bp in length to account for the fragmented nature of DNA in formalin-fixed, paraffin-embedded speci-mens. Separate data sets were generated by means of bidirectional Sanger sequencing and WAVE-based Surveyor Scan Kits (Transgenomic).23-26 Double-stranded PCR amplicons were melted and cooled to form a heteroduplex–homoduplex mixture that was treated with Surveyor nuclease. The resulting DNA fragments were analyzed with the use of high-performance liquid chroma-tography (WAVE HS System). The formation of mutant:nonmutant heteroduplexes resulted in frag-ments of various sizes. The testing plan and meth-ods were prespecified, and investigators in the testing laboratory were unaware of treatment as-signments and clinical outcomes. Bidirectional Sanger sequencing and testing with WAVE-based Surveyor Scan Kits were validated according to the Clinical Laboratory Improvement Amendments of 1988.Statistical Analysis The statistical analysis plan was prespecified be-fore the RAS and BRAF testing results became available. Two clinical data snapshots were used: the primary analysis (prespecified to be performed when >50% of patients with nonmutated KRAS exon 2 had died from any cause) and the updated analysis of overall survival (an exploratory analy-sis that was undertaken when >80% of patients in both the nonmutated and mutated KRAS exon 2 subgroups had died from any cause), which pro-vided the most up-to-date estimate of overall sur-vival in the PRIME study.The primary objective of the current prospec-tive–retrospective analysis was to evaluate the treat-ment effect of panitumumab –FOLFOX4 as com-pared with FOLFOX4 alone in patients without RAS mutations (nonmutated KRAS and NRAS exons 2, 3, and 4) and in those without RAS andBRAF mutations (nonmutated KRAS and NRASexons 2, 3, and 4, and BRAF exon 15) in the primary-analysis population of the PRIME study. Subsequent evaluation of the treatment effect on the basis of the updated overall-survival data was similarly prespecified in the statistical analysis plan, but only the results of the overall-survival end point are reported, since data collection was limited to survival information. Patients were characterized as having RAS mutations if any predefined activating mutation in KRAS or NRAS was detected, and patients were characterized as having RAS or BRAF mutations if any predefined RAS or BRAF mutation was detected.The hypothesis testing in this analysis was exploratory in nature. An overall 5% significance level was used to compare the treatment effect on progression-free survival and overall survival in subpopulations without RAS mutations and in subpopulations without RAS and BRAF mutations. To control the overall type 1 error rate, a sequential testing scheme was used for evaluation of the treatment effects of panitumumab on progression-free survival among patients with nonmutated RAS and nonmutated RAS and BRAF, followed by a test of the treatment effects on overall survival among patients in the same subgroups. No hypoth-esis testing was conducted in the subgroups with mutations. To estimate the treatment effects of panitumumab, we used Cox proportional-hazards models stratified according to randomization fac-tors, with all randomly assigned patients in eachbiomarker subgroup included in the assessment.A log-rank test stratified according to randomiza-tion factors was used to compare the treatment effects on progression-free survival and overall survival in the panitumumab–FOLFOX4 group with the treatment effects on progression-free survival and overall survival in the FOLFOX4-alone group. Sensitivity analyses, including a multivari-ate Cox model and propensity-score analysis, were used to confirm the primary results. Interaction tests were performed to compare the treatment effects of panitumumab between the subgroup with nonmutated RAS and the subgroup with mutated RAS and between the subgroup with non-mutated RAS and the subgroup with nonmutated KRAS in exon 2 and other RAS mutations. Multi-variate Cox models were also used to explore the prognostic relevance of baseline covariates.T h e ne w engl a nd jour na l o f medicinen engl j med 369;11 september 12, 20131026R esultsPatientsOf the 1183 patients who underwent randomiza-tion, 1096 (93%) had previously been evaluated for KRAS exon 2 (656 patients without KRAS mu-tations in exon 2 [60%] and 440 patients with KRAS mutations in exon 2 [40%]) (Fig. S1 in the Supplementary Appendix, available at ).2 The status of KRAS exon 3 or 4; NRAS exon 2, 3, or 4; or BRAF exon 15 (Table 1, and Table S1 in the Supplementary Appendix) was determined in 639 of the 656 patients without KRAS mutations in exon 2. Identical results were obtained by means of bidirectional Sanger sequencing and WAVE-based Surveyor analysis.RAS status was ascertained in 1060 of the 1183 patients (90%) who underwent random-ization. Of these 1060 patients, 512 (48%) were identified as having tumors with nonmutated RAS (no KRAS or NRAS mutations in exons 2, 3, or 4) and 548 (52%) were identified as having tumors with mutated RAS (any KRAS or NRAS mutations in exon 2, 3, or 4) (Fig. S1 in the Sup-plementary Appendix). Of 620 patients with data that could be evaluated for RAS , 108 (17%) who were originally categorized as not having KRAS mutations in exon 2 had other RAS mutations. Baseline clinical and demographic characteris-tics, including race or ethnic group, age, Eastern Cooperative Oncology Group (ECOG) performance-status score (on a scale from 0 to 5, with 0 indi-cating no symptoms and full activity and higher scores indicating increasing levels of disability),27 primary tumor type, and number of metastatic lesions were generally similar between patients with nonmutated RAS and those with mutated RAS and were consistent with the reported re-sults for KRAS exon 2 in patients with meta-static colorectal-cancer tumors.2The rate of ascertainment of RAS and BRAF status was 89% (assessed in 1047 of 1183 pa-tients). Of 619 patients without KRAS mutations in exon 2 who could be evaluated for BRAF, 53 (9%) had V600E mutations. Mutations in BRAF exon 15 were mutually exclusive of KRAS and NRAS muta-tions in patients without KRAS mutations in exon 2 who could be evaluated. The proportion of pa-tients with each of the RAS or BRAF mutations (Table 1) was consistent with that reported in a recently published article.21Efficacy According to Tumor RAS StatusAt the time of the primary analysis (data-cutoff point, August 29, 2009), 54% of the patients had died.2 In patients without KRAS mutations in exon 2 who received panitumumab–FOLFOX4, as compared with those who received FOLFOX4 alone, there was a significant improvement in pro-gression-free survival (9.6 vs. 8.0 months, P = 0.02) and a 4.2-month improvement in overall survival, which was not significant (23.9 vs. 19.7 months, P = 0.07) (Table 2). In an exploratory, updated anal-ysis of overall survival (data-cutoff point, Janu-ary 24, 2013), 82% of the patients had died. On the basis of this analysis, panitumumab–FOLFOX4 was associated with a 4.4-month improvement in overall survival (23.8 months in the panitumumab–FOLFOX4 group vs. 19.4 months in the FOLFOX4-alone group, P = 0.03).These analyses were extended to evaluate the predictive value of mutations other than KRAS mutations in exon 2. In the subgroup of patients without RAS mutations (the primary-analysis pop-ulation), panitumumab –FOLFOX4, as compared with FOLFOX4 alone, was associated with a significant improvement in progression-free survival (10.1 vs. 7.9 months, P = 0.004) and a significant 5.8-month improvement in overall survival (26.0 vs. 20.2 months, P = 0.04) (Table 2 and Fig. 1 and 2A and 2B). Consistently, signifi-cant results were observed in the exploratory, updated overall-survival analysis with respect to the magnitude of improvement with panitumu-mab–FOLFOX4 as compared with FOLFOX4 alone (Table 2 and Fig. 2C).A total of 17% of patients without KRAS mu-tations in exon 2 had mutations in other RAS exons. In this subgroup of 108 patients, out-comes in the primary analysis and in the explor-atory updated analysis of overall survival showed that progression-free survival (Fig. S2A in the Supplementary Appendix) and overall survival (Fig. S2B and S2C in the Supplementary Appen-dix) were shorter in the panitumumab–FOLFOX4 group than in the FOLFOX4-alone group, though the difference was not significant. These out-comes were consistent with those observed in the subgroup of patients with KRAS mutations in exon 2; in this subgroup, progression-free sur-vival in the primary analysis was significantly shorter in the panitumumab–FOLFOX4 group than in the FOLFOX4-alone group (7.3 monthspanitumumab–folfox4 in Colorectal Cancern engl j med 369;11 september 12, 20131027* FOLFOX4 denotes oxaliplatin, fluorouracil, and leucovorin.† A total of 440 patients with KRAS mutations in exon 2 were not retested for RAS or BRAF .‡ Of 641 samples tested, 2 did not yield a result (and did not have a mutation) in at least one RAS or BRAF exon. Samples that had any RAS exon mutation, regardless of whether other RAS exons did not yield a result, were characterized as mutant RAS and thus could be evaluated for RAS mutation status.§ The total includes 440 patients with KRAS mutations in exon 2 and 620 patients with data that could be evaluated for RAS . Of 641 samples tested, 21 did not yield a result (and did not have a mutation) in at least one RAS exon.¶ Of 1060 samples with data that could be evaluated for RAS, an additional 13 did not yield a BRAF result.T h e ne w engl a nd jour na l o f medicinen engl j med 369;11 september 12, 20131028vs. 8.8 months, P = 0.02) (Table 2). In the pri-mary analysis, interaction testing between the subgroups that did not have RAS mutations and the subgroups that did not have KRAS mutations in exon 2 but did have other RAS mutations was significant for progression-free survival (P = 0.04) but not for overall survival (P = 0.07). In the up-dated analysis of overall survival, which was based on a larger number of deaths from any cause, the results of interaction testing were sig-nificant (P = 0.01). These results indicate that treatment effects differed between the subgroups of patients without RAS mutations and those without KRASmutations in exon 2 but with other* The interaction test is for the comparison with nonmutated RAS .panitumumab–folfox4 in Colorectal Cancern engl j med 369;11 september 12, 20131029RAS mutations, suggesting that RAS mutations, in addition to KRAS mutations in exon 2, were negative predictive factors (Table 2).In the expanded subgroup of patients with mutated RAS tumors, progression-free survival (Fig. S3A in the Supplementary Appendix) and overall survival (Fig. S3B and S3C in the Supple-mentary Appendix) were significantly shorter in the panitumumab–FOLFOX4 group than in the FOLFOX4-alone group in the primary analy-sis and in the exploratory, updated analysis of overall survival. Interaction testing for progres-sion-free survival and overall survival was sig-nificant in all data sets, further suggesting that RAS mutations had a negative predictive value (Table 2). Additional efficacy results for patients with data that could not be evaluated for RAS are shown in Table S2 in the Supplementary Appendix.The treatment effect on progression-free sur-vival and overall survival in favor of panitumu m ab–FOLFOX4 in patients with nonmutated RAS was observed across subpopulations predefined accord-ing to baseline covariates, except an ECOG perfor-mance-status score of 2 (Fig. S4 in the Supplemen-tary Appendix).Subsequent to the prespecified analysis, pre-viously reported mutations in KRAS and NRAS at codon 59 (A59G and A59T)9,28-30 were identified in seven patients. In an exploratory analysis in-volving this small patient population, exclusion of these mutated alleles slightly improved pro-gression-free survival and overall survival (Table S3 and Fig. S5 in the Supplementary Appendix).Efficacy According to Tumor BRAF StatusIn the nonmutated RAS and nonmutated BRAF subgroup, panitumumab–FOLFOX4 was associ-ated with a 1.6-month improvement in progres-sion-free survival and a 7.4-month improvement in overall survival, as compared with FOLFOX4 alone (Table 3). The minor differences between FOLFOX4–panitumumab and FOLFOX4 alone in the subgroup of patients without RAS mutations but with BRAF mutations were not significant (Fig. S6 in the Supplementary Appendix).T h e ne w engl a nd jour na l o f medicinen engl j med 369;11 september 12, 20131030panitumumab–folfox4 in Colorectal Cancern engl j med 369;11 september 12, 20131031Prognostic Effects of RAS and BRAF Mutation StatusThe prognostic effects of RAS and BRAF mutation status were evaluated by comparing the hazard ratios for death from any cause with no mutation versus mutation within each treatment group and across groups (Fig. S7 in the Supplementary Appendix). Most hazard ratios favored nonmu-tated status in the panitumumab–FOLFOX4 group and were neutral in the FOLFOX4-alone group. BRAF mutations were associated with re-duced overall survival among patients withoutKRAS mutations in exon 2 and among those with NRAS mutations in exon 3.Safety The incidence rates, types, and severity of adverse events among patients with nonmutated RAS in the panitumumab–FOLFOX4 group (Table 4) were similar to those previously reported in the group of patients with nonmutated KRAS exon 2 who were treated with panitumumab–FOLFOX4.2 Treat-ment exposure, disease-control rates, and the pro-portion of patients who discontinued any study drug due to an adverse event were also similar to those previously reported.2 The safety profile for patients with RAS mutations was similar to that re-ported for patients with KRAS mutations in exon 2. No new safety signals were identified.DiscussionTesting for KRAS exon 2 tumor mutations is cur-rently recommended to help guide decisions re-garding eligibility for anti-EGFR therapy in pa-tients with metastatic colorectal cancer. Although KRAS testing has facilitated the selection of pa-tients who are most likely to have a response to anti-EGFR therapy, a substantial fraction of pa-tients do not benefit from treatment. It is hoped* NE denotes not evaluated.T h e ne w engl a nd jour na l o f medicinen engl j med 369;11 september 12, 20131032that further refinement of tumor-specific genetic markers will allow more accurate selection of pa-tients who are likely to have a response to a par-ticular treatment and prevent toxic effects in those who are unlikely to benefit.Biomarker exploration has been broadened to include EGFR pathway mutations, in addition to those in KRAS exon 2. In a retrospective bio-marker analysis of a randomized phase 3 study of panitumumab monotherapy, EGFR signaling–pathway genes were assessed for their predictive ability.10 From this study, a hypothesis was gener-ated that activating mutations in KRAS or NRAS would be predictive of nonresponse to panitum-umab therapy. The current analysis, which was based on biologic plausibility and exploratory bio-marker data, further assesses the hypothesis that additional activating RAS mutations predict unre-sponsiveness to panitumumab treatment.7,10,21,28Negative treatment effects of panitumumab–FOLFOX4 on progression-free survival and over-all survival were observed among patients with tumors that did not have KRAS mutations in exon 2 but that did have other RAS mutations . In an interaction test, treatment effects were sig-nificantly worse than those in the group of pa-tients with nonmutated RAS ; this suggests that mutations in RAS, in addition to KRAS mutations in exon 2, are predictive of adverse outcomes for panitumumab–FOLFOX4 treatment. The magni-tude of the treatment effect in the patients with mutated RAS was similar to that previously ob-served in patients with mutated KRAS exon 22 and further indicated that patients with tumors that harbored any activating RAS mutations did not benefit from and may have been harmed by panitumumab–FOLFOX4 treatment.Among patients in the primary-analysis popu-lation who did not have RAS mutations, an in-crease in overall survival of 5.8 months was noted with the addition of panitumumab to FOLFOX4 as compared with FOLFOX4 alone. The results of the exploratory, updated analysis of overall sur-vival were consistent with these findings. The ob-served incidence, types, and severity of adverse events associated with panitumumab–FOLFOX4 in the nonmutated RAS and mutated RAS sub-groups were similar to the previously reported safety findings for KRAS in PRIME,2 and no new safety signals were identified.In the subgroup of patients without RAS and BRAF mutations, a 7.4-month increase in overall survival was observed in the panitumumab–FOLFOX4 group. As suggested previously,17 BRAF V600E mutations appeared to confer a poor prog-nosis, regardless of the treatment group.This analysis was retrospective and explorato-ry in nature and therefore subject to limitations. The alpha error for hypothesis testing was previ-ously allocated to the primary analysis,2 and RAS and BRAF status may not be representative of the intention-to-treat population from theoriginalpanitumumab–folfox4 in Colorectal Cancern engl j med 369;11 september 12, 20131033randomization. However, methodologic aspects of the analysis provided a rigorous framework for evaluating RAS and BRAF as biomarkers. Tissue samples were collected with appropriate informed consent before randomization. The biomarker hypothesis was restricted to RAS and BRAF, and the statistical analysis plan was finalized before RAS and BRAF status became available. In addi-tion, the analysis was conducted with data from a large, randomized, controlled trial.2 The high rate of ascertainment of RAS status (90%) mini-mized the potential for ascertainment bias. Two laboratory-developed tests, validated according to the Clinical Laboratory Improvement Amend-ments of 1988, provided mutual confirmation.Two interaction tests showed a clear separa-tion of panitumumab treatment effects between nonmutated RAS and mutated RAS as well as between nonmutated RAS and nonmutated KRAS exon 2 with other RAS mutations; the latter find-ing indicates the predictive value of RAS muta-tions other than KRAS mutations in exon 2. These results were observed across all meaningful end points and in all relevant subgroups.In conclusion, RAS mutations, in addition to KRAS exon 2 mutations, predict a lack of re-sponse to anti-EGFR therapy in patients with metastatic colorectal cancer. Panitumumab plus oxaliplatin-containing regimens have no value in patients with metastatic colorectal cancer and mutated RAS . The benefit–risk profile of panitumumab–FOLFOX4 was improved by ex-cluding patients with mutated RAS metastatic colorectal-cancer tumors. Pooled trials or meta-analyses of anti-EGFR therapy are needed to confirm these findings.Supported by Amgen and by grants from the Royal Marsden Hospital and the Institute of Cancer Research, National Institute for Health Research, Biomedical Research Centre (to Dr. Cunning-ham), and Oncologia Ca’ Granda Onlus Fondazione (to Dr. Siena).Disclosure forms provided by the authors are available with the full text of this article at .We thank Shawn Lee of Amgen for writing assistance and Mark Ekdahl of Amgen for operational assistance.References1. Amado RG, Wolf M, Peeters M, et al. Wild-type KRAS is required for panitum-umab efficacy in patients with metastatic colorectal cancer. J Clin Oncol 2008;26: 1626-34.2. Douillard JY, Siena S, Cassidy J, et al. Randomized, phase III trial of panitum u -mab with infusional fluorouracil, leuco-vorin, and oxaliplatin (FOLFOX4) versus FOLFOX4 alone as first-line treatment in patients with previously untreated meta-static colorectal cancer: the PRIME study. J Clin Oncol 2010;28:4697-705.3. Peeters M, Price TJ, Cervantes A, et al. Randomized phase III study of pani-tum u mab with fluorouracil, leucovorin, and irinotecan (FOLFIRI) compared with FOLFIRI alone as second-line treatment in patients with metastatic colorectal cancer. J Clin Oncol 2010;28:4706-13.4. Van Cutsem E, Kohne CH, Hitre E, et al. Cetuximab and chemotherapy as ini-tial treatment for metastatic colorectal cancer. N Engl J Med 2009;360:1408-17.5. Bokemeyer C, Bondarenko I, Hart-mann JT, et al. Efficacy according to bio-marker status of cetuximab plus FOLFOX-4 as first-line treatment for metastatic colo-rectal cancer: the OPUS study. Ann Oncol 2011;22:1535-46.6. Karnoub AE, Weinberg RA. Ras on-cogenes: split personalities. Nat Rev Mol Cell Biol 2008;9:517-31.7. Janakiraman M, Vakiani E, Zeng Z, et al. Genomic and biological characteriza-tion of exon 4 KRAS mutations in human cancer. Cancer Res 2010;70:5901-11.8. Fernández-Medarde A, Santos E. Ras in cancer and developmental diseases. Genes Cancer 2011;2:344-58.9. Forbes SA, Bindal N, Bamford S, et al. COSMIC: mining complete cancer genomes in the Catalogue of Somatic Mutations in Cancer. Nucleic Acids Res 2011;39:D945-D950.10. Peeters M, Oliner K, Parker A, et al. Massively parallel tumor multigene se-quencing to evaluate response to pani t um-umab in a randomized phase 3 study of metastatic colorectal cancer. Clin Cancer Res 2013;19:1902-12.11. De Roock W, Jonker DJ, Di Nicolantonio F, et al. Association of KRAS p.G13D muta-tion with outcome in patients with chemo-therapy-refractory metastatic colorectal cancer treated with cetuximab. JAMA 2010; 304:1812-20.12. Peeters M, Douillard J-Y, Van Cutsem E, et al. Mutant KRAS codon 12 and 13 alleles in patients with metastatic colorectal can-cer: assessment as prognostic and predic-tive biomarkers of response to pani t um-umab. J Clin Oncol 2013;31:759-65.13. André T, Blons H, Mabro M, et al. Pan i tumumab combined with irinotecan for patients with KRAS wild-type meta-static colorectal cancer refractory to standard chemotherapy: a GERCOR effi-cacy, tolerance, and translational molec-ular study. Ann Oncol 2013;24:412-9.14. Loupakis F, Ruzzo A, Cremolini C, et al. KRAS codon 61, 146 and BRAF mutations predict resistance to cetuximab plus irino-tecan in KRAS codon 12 and 13 wild-type metastatic colorectal cancer. Br J Cancer 2009;101:715-21.15. De Roock W, Claes B, Bernasconi D, et al. Effects of KRAS, BRAF, NRAS , and PIK3CA mutations on the efficacy of cetux-i m ab plus chemotherapy in chemotherapy-refractory metastatic colorectal cancer: a retrospective consortium analysis. Lancet Oncol 2010;11:753-62.16. Phipps AI, Buchanan DD, Makar KW, et al. BRAF mutation status and survival after colorectal cancer diagnosis accord-ing to patient and tumor characteristics. Cancer Epidemiol Biomarkers Prev 2012; 21:1792-8.17. Van Cutsem E, Köhne CH, Láng I, et al. Cetuximab plus irinotecan, fluoroura-cil, and leucovorin as first-line treatment for metastatic colorectal cancer: updated analysis of overall survival according totumor KRAS and BRAF mutation status. J Clin Oncol 2011;29:2011-9.18. Fariña-Sarasqueta A, van Lijnschoten G, Moerland E, et al. The BRAF V600E muta-tion is an independent prognostic factor for survival in stage II and stage III colon can-cer patients. Ann Oncol 2010;21:2396-402.19. Bokemeyer C, Van Cutsem E, Rougier P, et al. Addition of cetuximab to chemo-therapy as first-line treatment for KRAS wild-type metastatic colorectal cancer: pooled analysis of the CRYSTAL and OPUS randomised clinical trials. Eur J Cancer2012;48:1466-75.20. Di Nicolantonio F, Martini M, Moli-nari F, et al. Wild-type BRAF is required for response to panitumumab or cetux-The New England Journal of MedicineDownloaded from on December 31, 2017. For personal use only. No other uses without permission.Copyright © 2013 Massachusetts Medical Society. All rights reserved.。