• 患者服务: 与癌共舞小助手
  • 微信号: yagw_help22

QQ登录

只需一步,快速开始

开启左侧

肺腺癌

    [复制链接]
211105 309 荷花池荒岛 发表于 2014-3-24 12:31:48 |
yaoli0118  初中一年级 发表于 2014-7-29 14:14:37 | 显示全部楼层 来自: 北京
谢谢楼主的搜集汇总,非常有用的帖子。对这个坛子,有一种相见恨晚的感觉。
妈妈病情记录贴
http://www.yuaigongwu.com/forum.php?mod=viewthread&tid=14776&extra=
荷花池荒岛  硕士一年级 发表于 2014-8-9 23:52:02 | 显示全部楼层 来自: 美国
本帖最后由 荷花池荒岛 于 2015-10-14 12:11 编辑

天涯的探锁的心给hellengoodd的建议是服用环磷酰胺,http://webmail.yuaigongwu.com/thread-14080-6-1.htm

1)http://www.chinabaike.com/article/43/yao/2007/20071202696567.html
复方环磷酰胺片

用法用量       
    口服每次1片,一日3~4 次

药理作用       
    本品含环磷酰胺和人参茎叶总皂甙,环磷酰胺在体外无抗肿瘤活性进入体内后被肝脏或肿瘤细胞组织内存在的过量磷酰胺酶或磷酸酶水解,生成活化型磷酰胺氮芥,该物质对肿瘤细胞有细胞毒作用,具有抑制肿瘤生长的作用。环磷酰胺是双功能烷化剂及细胞周期非特异性药物,不干扰DNA及RNA功能,它与DNA发生交叉联结,抑制DNA合成对S期作用最明显。人参茎叶总皂甙具有明显的刺激骨髓造血机能,促使骨髓有核细胞数明显增加,从而具有提升白细胞作用,此外尚具有提高机体免疫功能,增强机体应激能力抗疲劳等作用,复方环磷酰胺药理试验表明,可明显改善单独使用环磷酰胺引起的白细胞减少,降低机体免疫功能和抗应激能力和胃肠道不良反应等毒付作用,延长化疗时间,增强抗肿瘤作用。 

适应症
    抗肿瘤药。主要用于急性白血病、慢性淋巴细胞白血病、恶性淋巴瘤、多发性骨髓瘤、肺癌,神经细胞癌。也用于乳腺癌、鼻咽癌及肾母细胞癌等。本品与环磷酰胺片相比具有保护造血功能,升高白血球,改善血象减少毒付作用,延长化疗时间的优点。
       
制剂
    每片含环磷酰胺50mg,人参茎叶总皂甙50mg。

注意事项       
    用药期间应严格检查血象及有肝病患者应慎用。

2)http://www.xinyao.com.cn/anti_tu ... 130418041308229.htm
复方环磷酰胺片的用药注意事项

。。。。。。
虽说复方环磷酰胺片具有良好的广谱抗肿瘤作用,但使用复方环磷酰胺片进行治疗时,患者可能出现一定的不良反应,其中,骨髓抑制为常见的毒性,白细胞往往在给药后10-14天,多在第21天恢复正常,血小板减少比其他烷化剂少见;常见的不良反应还有恶心、呕吐,不良反应严重程度常与复方环磷酰胺片的剂量有关。

  还有一些比较少见的不良反应有发热、过敏、皮肤及指甲色素沉着、粘膜溃疡、谷丙转氨酶升高、荨麻疹、口咽部感觉异常或视力模糊。

  要注意,复方环磷酰胺片的代谢产物可产生严重的出血性膀胱炎,大量补充液体可避免,故复方环磷酰胺片可能导致膀胱纤维化;而当大剂量复方环磷酰胺片与大量液体同时给予时,可产生水中毒,可根据实际情况给予呋塞米以防止;复方环磷酰胺片还可引起生殖系统毒性,如停经或精子缺乏,妊娠初期给药可致畸胎;而长期给予复方环磷酰胺片可产生继发性肿瘤。

  百济药师温馨提醒:常规剂量环磷酰胺不产生心脏毒性,但当高剂量时可产生心肌坏死,偶有发生肺纤维化。
。。。。。。



“人类只有经历过悲哀才能够知道什么是快乐,你只有经过死亡以后才知道重生意味着什么,上帝给人留下了四个字,那就是希望和等待。”
荷花池荒岛  硕士一年级 发表于 2014-8-9 23:52:15 | 显示全部楼层 来自: 美国
本帖最后由 荷花池荒岛 于 2014-8-10 00:43 编辑

1)咳血通过什么方法来确定咳血位置?

一般支气管镜检查可以明确出血部位,如果出血量多的,支气管动脉造影也能看得到!------ 一只白杨


2)如何止咳血 ?

服用云南白药 ------网友经验

如果咯血量大,可以考虑介入栓塞。------ 一只白杨

p.s.

介入栓塞术
指通过介入技术对血管进行栓塞的技术,多用于大出血止血、肿瘤栓塞治疗,及颅内动脉瘤栓塞治疗,栓塞材料可分为永久性栓子,和临时栓子,永久栓子不能再通,临时栓子可以溶解再通。

栓塞
在循环血液中出现的不溶于血液的异常物质,随血流运行至远处阻塞血管腔的现象称为栓塞(embolism)。阻塞血管的物质称为栓子(embolus)。栓子可以是固体(如血管壁脱落的血栓)、液体(如骨折时的脂滴)或气体(如静脉外伤时进入血流的空气)。以脱落的血栓栓子引起栓塞最常见,如肺动脉、脑动脉的栓塞。栓塞对机体的影响取决于栓塞的部位、血管的解剖特点和局部血液循环状态、栓塞后能否建立充分的侧枝循环,以及栓子的种类及来源。 常见的栓塞类型有血栓栓塞、脂肪栓塞、气体栓塞、羊水栓塞、肿瘤细胞栓塞、寄生虫栓塞和感染性栓塞。


3)肺栓塞的检测?

做一个肺动脉CT就可以,另外看看血清D-二聚体高不高。 ------ 一只白杨


“人类只有经历过悲哀才能够知道什么是快乐,你只有经过死亡以后才知道重生意味着什么,上帝给人留下了四个字,那就是希望和等待。”
荷花池荒岛  硕士一年级 发表于 2014-8-9 23:52:22 | 显示全部楼层 来自: 美国
本帖最后由 荷花池荒岛 于 2014-8-11 05:40 编辑

可以在香港买的药:

  • 关节痛,黄道益活络油;
  • 止痒,强力无比膏;
  • 皮疹,拔毒生肌膏(也可以试特罗凯推荐的尿素维E软膏)。


点评

有突变直接上靶向,老人家做化疗对身体很伤得  发表于 2014-12-23 14:45
“人类只有经历过悲哀才能够知道什么是快乐,你只有经过死亡以后才知道重生意味着什么,上帝给人留下了四个字,那就是希望和等待。”

举报 使用道具

回复 支持 3 反对 1
荷花池荒岛  硕士一年级 发表于 2014-8-9 23:52:28 | 显示全部楼层 来自: 美国
本帖最后由 荷花池荒岛 于 2016-7-9 00:37 编辑

论坛留言摘抄(大家的经验,本人不知道对与错):
  • 放疗前几分钟喝酸奶,或者是蜂蜜水,可以搁在咽喉处,避免放射引起咽喉不适.
  • 服用靶向药期间避免葡萄柚。
  • 服用沐舒坦有可能造成恶心呕吐 (沐舒坦(盐酸氨溴索片),适应症为适用于痰液粘稠而不易咳出者。)
  • 皮疹穿宽松柔软的衣服,不能用碱性肥皂,不能用粗糙毛巾。避免日光照射。口服多西环素或米诺环素。
  • 口腔溃疡餐后用漱口水,口服VC+VB2,必要时用甲硝唑消炎。口腔局部用VC+VB2+蜂蜜涂抹 。
  • 治疗便秘:果导片、喝香油、牛黄解毒片、番泻叶(刺激性泻药)。
  • 治疗腹泻:蒙脱石散、
  • 榄香烯和康莱特注射液如果不埋管对血管伤害巨大。
  • 克里唑替尼尼最明显的副作用就是前期很容易呕吐,所以服药前半小时要先吃胃药。副作用还有恶心、视觉上有拖影
  • 吃克里唑替尼注意观察有无肺炎,如有,立刻停药。
  • 鼻腔干伽,涂红霉素眼药膏缓解;指肿痛,涂红霉素药膏,好了;腹泻,肠炎宁,已经好了几天了;角膜炎,也是用红霉素眼药膏治好的。
  • 服用299804出现皮疹:鼻翼两侧都是带有脓头的皮疹,鼻子上、下巴也是红红的,痒又疼,脸部也发红。我后来用了卤米松和夫西地酸乳膏,按照1:1混合均匀,一天涂抹2-3次,第二天就发现脓头基本没有了,今天是用此方法的第四天,脸上已经找不到几颗了;口腔溃疡 :服用299804第15天,觉得舌头疼,第二天就发现舌头的两侧及舌尖上有发红且有点溃疡的症状,影响吞咽食物,马上金银花泡水喝,再用VC和VB2各一粒碾碎,用蜂蜜拌匀,涂在疼处,一天涂N次,连续涂抹了两天,溃疡终于被我消灭在萌芽状态了。
  • 头孢外治甲沟炎:剥一粒头孢(先锋4或先锋6皆可)倒出药粉,滴上一两滴水,调成白浆,涂在红肿的趾肉与趾甲之间即可,顶多如此操作二三回,就彻底地好。
  • YL药时,用药用淀粉比乳糖好。
  • 阿西副作用主要为高血压、心率加快、尿蛋白、手足综合症、疲倦、声音沙哑。
  • 辅酶Q10要在吃饭的中途吃,它是油溶性的,空腹吃吸收很差。
  • 我们疼痛科同事处理疼痛的时候,尤其是神经痛,会联合使用加巴喷丁。这个药就是一个抗癫痫药物。
  • 凡德他尼具有胃肠道粘膜毒性。应对方法是,餐前半小时胶体果胶铋,睡前加服一次。出现胃痛的话,可以法莫替丁,睡前服用。还有就是打算再试药的时候,把凡德放到上午九、十点钟,在两餐间服用。
  • 化疗同时加些免疫药,如果有条件就上日达仙或者香菇多糖。实在不行就迈普新。榄香烯也可以用。
  • 锶89对血象的影响时间比较长。
  • 有一个体外实验显示,停药2天,阿法替尼、达可替尼的T790突变可逆,这是我家吃8天停2天的依据之一。注:992的临床试验证明,每天一次,连续服药方案的效果最好,对于病情进展的病人,不应该采用药物假期方案。
  • 凡德导致的高血压避免服用地尔硫卓、维拉帕米、尼群地平等钙离子拮抗剂。降血压的络活喜也属于钙离子拮抗剂。
  • 2992副作用主要有:腹泻、皮疹/粉刺、口腔炎,甲沟炎、食欲下降、乏力、室性早博明显。
  • 抗过敏的甲泼尼龙针剂可能造成出血。
  • EGFR抑制剂引起的腹泻被认为是由于过量的氯离子的分泌,是一种分泌型腹泻。
  • HER2是乳腺癌治疗的重要靶点。抗HER2靶向治疗药物大体可被分为4类:作用于受体分子细胞外区域的抗体(曲妥珠单抗、帕妥珠单抗)、小分子酪氨酸激酶抑制剂(拉帕替尼)、抗体-细胞毒分子耦合剂(曲妥珠单抗-微管聚合抑制剂,T-DM1)和伴侣蛋白拮抗剂。
  • 以胃、十二指肠溃疡或胃癌并发的出血,肺部疾病引起的咯血, 对膀胱癌并发血尿及出血性中风来说,出血的原因都是血管破裂,是血管因素,并不是凝血功能出毛病,所以,用安络血、止血敏、维生素K、止血环酸这几种止血针实际上是不对的。
  • 由于乳糖在机体被缓慢水解吸收,使血糖水平不至于急剧上升,这一点对糖尿病患者来说尤为有利。所以乳糖可以做糖尿病人的食物。
  • 白蛋白合成减少、球蛋白合成增多。这一个也是生活中比较常见的原因,假如有病毒性肝炎一方面会导致肝脏合成白蛋白减少,另一方面病毒的存在也会使机体产生过多的球蛋白,最使白球比例偏低。正常人很少出现白球比偏低,血液中的白蛋白与球蛋白比值不应低于1.5:1,如果低于这个值,常见于肝硬化、肝功能不全、肾病(蛋白尿)等会造成白蛋白偏低的疾病患者。
  • 水飞蓟宾40mg相当于水飞蓟素160mg ,大家买水林佳和利加隆,要注意剂量  利加隆要买140mg的不要买70mg的。
  • 肺癌按组织学分类:鳞癌、小细胞癌、大细胞癌、腺癌、肺泡癌。腺癌血管丰富,故局部浸润和血行转移较鳞癌早,常转移到肝、脑和骨,更易累及胸膜而引起胸腔积液。
  • 皮肤干燥,大量掉皮屑的问题已经解决了。老妈去菜场弄了一些猪皮,煮二次,丢弃油汤,吃肉皮,效果很好,现在皮肤很光滑。
  • 4002有利尿作用,导致肠道水分减少,大便硬结难排。晚上喝些盐水会好些。
  • 癌烧可以试试新癀片(厦门中药厂)或者萘普生。牙疼可以试试人工牛黄甲硝唑片/胶囊。
  • 医生推荐了一个对付皮疹的妙招:金银花+水,火烧,然后用冷却的金银花水洗患处。


“人类只有经历过悲哀才能够知道什么是快乐,你只有经过死亡以后才知道重生意味着什么,上帝给人留下了四个字,那就是希望和等待。”
荷花池荒岛  硕士一年级 发表于 2014-8-9 23:52:36 | 显示全部楼层 来自: 美国
本帖最后由 荷花池荒岛 于 2014-8-11 07:10 编辑

Going beyond EGFR
S. Zimmermann and S. Peters
http://annonc.oxfordjournals.org ... 10/x197.full#ref-31

Abstract
a substantial proportion of non-small-cell lung cancer (NSCLC), and adenocarcinoma in particular, depends on a so-called ‘driver mutation’ for their malignant phenotype. This genetic alteration induces and sustains tumorigenesis, and targeting of its protein product can result in growth inhibition, tumor response and increased patient survival. NSCLC can thus be subdivided into clinically relevant molecular subsets. Mutations in EGFR best illustrate the therapeutic relevance of molecular classification. This article reviews the scope of presently known driving molecular alterations, including ROS1, BRaF, KRaS, HER2 and PIK3Ca, with a special emphasis on aLK rearrangements, and outlines their potential therapeutic applications.

introduction
Distinct subtypes of non-small-cell lung cancer (NSCLC) are driven by a specific genetic alteration—are so-called ‘oncogene addicted’—and are thus sensitive to inhibition of the corresponding activated oncogenic pathway. This new paradigm has substantially impacted lung cancer treatment: early treatment of advanced NSCLC consisted of chemotherapy tailored for patients according to the expected toxicity and more recently according to the histologic subtype [1]. Nowadays, NSCLC can be further subdivided into clinically relevant molecular subsets, according to their driving genetic alterations affecting tumor proliferation and survival. Treatment of patients with EGFR activating and sensitizing mutation-driven NSCLC with EGFR tyrosine kinase inhibitors (TKIs) results in an unprecedented response rate (RR) of 60–80%, a median progression-free survival (PFS) of ~8–13 months, as well as an improved quality of life compared with chemotherapy [2, 3]. Tumor genotype analysis has to date identified driver alterations in ~50–80% of NSCLC patients according to demographics, and particularly ethnicity. Sequist et al. [4] carried out a multiplexed PCR-based assay to simultaneously identify >50 mutations in several key NSCLC genes on parallel to FISH analysis for EML4-aLK translocations on 552 tumors mainly from Caucasian smoker patients. Eighty-one percent were adenocarcinomas, and a genetic driver change was identified in 51% of all samples, most commonly KRaS (24%), EGFR (13%) and EML4-aLK translocation (5%). Less common mutations were also identified: TP53 (4%), PI3KCa (2%), beta-catenin (2%), BRaF (1%), NRaS (1%), HER2 (~1%) and IDH1 (~1%). a Chinese surgical series by Sun et al. [5] examined a genotyping panel of mutations in 52 resected lung adenocarcinomas from East asian never smokers and found 90% of tumors to be harboring a mutation in EGFR, KRaS, aLK or HER2. Focusing on adenocarcinoma subtype, the NCI's Lung Cancer Mutation Consortium (LCMC) tested 830 patients with lung adenocarcinoma, and detected a driver mutation in 54%: KRaS 25%, EGFR 23%, BRaF 3%, PIK3Ca 3%, HER2 1%, MEK1 0.4%, NRaS 0.2%, aLK rearrangements 6% and MET amplifications 2% [6].

Takeuchi et al. [7] identified KIF5B–aLK fusions in ~0.2% of resected adenocarcinomas, ROS1 gene rearrangements with five different fusion partners: TPM3, SDC4, SLC34a2, CD74 and E2R in 1.2% of adenocarcinomas and CCDC6-RET in an extremely small minority of adenocarcinomas. Kohno et al. [8] identified KIF5B–RET fusions in ~1.9% of adenocarcinomas (patients of Japanese ancestry). at aSCO 2012, Capelletti et al. reported on 11 patients with KIF5B-RET fusions among 643 patients [9] More recently, Togashi et al. [10] identified a KLC1–aLK fusion with an unreported incidence. all of these aLK, RET and ROS1 fusions have transforming capability.

EGFR and KRaS mutations, aLK translocations and MET amplification are found in <5% of squamous cell carcinoma, and HER2 and BRaF mutations have not yet been described in this histological subtype. Squamous cell carcinoma (SCC) of the lung is a distinct molecular subtype of lung cancer potentially amenable to distinct molecularly targeted therapies. The Cancer Genome atlas (TCGa) is conducting DNa, RNa, and miRNa sequencing along with DNa copy number profiling, quantification of mRNa expression and promoter methylation on surgically resected samples of SCC. Exome sequencing of 178 samples revealed 13 significantly mutated genes, including TP53, CDKN2a, PTEN, KEaP1, and NFE2L2. apart from the near universal loss of TP53 and CDKN2a, alterations in the NFE2L2/KEaP1 and PI3K/aKT pathways were found in 35% and 43% of tumors analyzed. Rearrangements involving several known tumor suppressors were detected including PTEN, RB1, NOTCH1, NF1 and CDKN2a. CDKN2a loss of function was observed in 72% of specimens. Potential therapeutic targets for clinical trials with currently available drugs were identified in 127 patients (75%) [11].

This review focuses on aLK modification in NSCLC, but encompasses other genetic alterations that contribute to lung carcinogenesis. Many oncogene products represent targets for drug therapy, and the expanding knowledge of molecular pathways implicated in lung tumorigenesis will radically change treatment, with hope for less toxic, targeted treatment and subsequently better outcomes.

aLK rearrangements
anaplastic lymphoma kinase (aLK) is an orphan member of the insulin superfamily of receptor tyrosine kinases, whose normal function is poorly understood. Chromosomal rearrangements involving the aLK gene occur in a variety of malignancies, including anaplastic large cell lymphoma, inflammatory myofibroblastic tumors and NSCLC. To date, in NSCLC, aLK has three reported fusion partners: EML4, KLC1 and KIF5B. TFG represents a potential forth fusion partner which has however not been histopathologically proven until today [12]. Soda et al. [13] showed that a small inversion within chromosome 2p results in the formation of a fusion gene comprising portions of the echinoderm microtubule-associated protein-like 4 (EML4) gene and the aLK gene. The fusion gene results in constitutive aLK kinase activation, serving as a potent ‘oncogenic driver’ with transforming ability. The EML4–aLK fusion transcript could be detected in 6.7% of the 75 NSCLC patients (of Japanese ethnicity), and this alteration was mainly mutually exclusive with EGFR or KRaS mutations. EML4-aLK-positive tumors were mostly adenocarcinomas and tended to affect younger and more frequently never/light smokers [14]. In these tumors, aLK is the sole determinant of critical growth pathways, resulting in the activation of downstream canonical PI3K/aKT as well as MaPK/ERK pathways. Within the pivotal phase I/II clinical trial by Kwak et al. [15], treatment of 82 EML4-aLK-positive patients with the aLK TKI crizotinib resulted in a 57% RR and stable disease in 33% of the patients. The kinetics of clinical response was comparable to previous experiences with EGFR TKIs in EGFR-mutated NSCLC. The median PFS was 10 months, 1-year overall survival (OS) was 74% [95% confidence interval (CI) 63–82] and 2-year OS was 54% (95% CI 40–66), with a median OS not yet reached. a retrospective study by Shaw et al. [16] suggests that crizotinib might prolong OS in aLK-positive NSCLC, when compared with historical aLK-positive patients not exposed to crizotinib, who had a similar course to the general NSCLC population, suggesting that the aLK fusion is predictive but not prognostic. On the basis of its demonstrated efficacy and safety in phase I and II studies, crizotinib was granted accelerated approval by the Food and Drug administration (FDa) for the treatment of advanced, aLK-positive NSCLC. Phase III study results are awaited, both in the second-line setting comparing crizotinib to pemetrexed or docetaxel (NCT00932893) and in the first-line setting in comparison to cisplatin and pemetrexed or carboplatin and pemetrexed (NCT01154140).

resistance to aLK inhibitors
as is the case with EGFR TKIs, clinical benefit of crizotinib therapy is limited by the development of acquired resistance. Resistance to TKIs can be mediated either by aLK point mutation and/or gene amplification, or activation of bypass signaling. The most frequent resistance mutations consist in amino acid substitutions increasing kinase activity and/or hindering drug binding, as described, for example, for EGFR in NSCLC and BCR-aBL in chronic myeloid leukemia, respectively. In the aLK setting, at least eight point mutations conferring resistance to aLK inhibitors have already been described and most of them shown to result in cross-resistance to other aLK inhibitors [17, 18]. Mutations can involve the gatekeeper site within the kinase domain and interfere with inhibitor binding (L1196M), the solvent front—also altering crizotinib binding (G1202R, S1206Y), the aTP-binding pocket (G1269a) or amino acids C- or N-terminal to the αC-helix—affecting aTP kinase affinity (C1156Y, L1152R, F1174L, 1151Tins) [19]. More potent and irreversible inhibitors might be capable of overcoming gatekeeper resistance [20].

another mechanism of resistance is the activation of a parallel ‘side-road’ or of the downstream pathways. In this situation combination targeted approaches need to be evaluated, such as in the case of MET amplification in EGFR-mutated NSCLCs. a combination of EGFR and MET inhibitors effectively overcomes this resistance in preclinical models [21]. Two recent small series have looked into the mechanisms of resistance to crizotinib and will be summarized below.

Katayama et al. analyzed biopsies of 18 patients with NSCLC who had developed secondary resistance to crizotinib and observed multiple additional genetic changes in the aLK gene, including aLK gene amplification, and further not previously described point mutations (T1151 insertion, G1202R and S1206Y) [22]. In addition, some cells harbor both aLK amplification and gatekeeper mutations [23]. The clinically available aLK inhibitors tested (CH5424802 and aSP-3026) showed distinct selectivity profiles against the various aLK resistance mutations, with accordingly different degrees of drug sensitivity. Doebele et al. [24] also analyzed tissue from 14 patients with aLK-positive NSCLC with radiological progression while on crizotinib, and identified aLK mutations in 4 patients, including a novel mutation G1269 which induced crizotinib resistance in cell lines. Doebele et al. also found copy number gain, defined as a more than two-fold increase in the mean rearranged gene per cell in the post-treatment biopsy compared with the pre-treatment biopsy, to be the mechanism of resistance in 2 patients out of 14. These results indicate that multiple distinct mutations in the aLK kinase domain can abrogate the inhibitory capacity of crizotinib. This is in sharp contrast to the EGFR-activating mutations, where T790M essentially represents the sole resistance mutation [25].

Nevertheless, in contrast to EGFR inhibitors, resistance due to secondary mutations or amplification of the drug target does not represent the predominant mechanism of acquired resistance to aLK inhibitors. Bypass signaling, including the KIT and EGFR pathways, has been identified as potential resistance mechanisms. Doebele et al. identified 1 patient out of 14 with an EGFR-activating mutation and 2 patients with KRaS mutations. Nevertheless, combined inhibition of aLK and EGFR failed to induce apoptosis in resistant cell lines, a phenomenon that was also observed by Katayama et al. [22]. The KIT amplification-mediated resistance resembles the resistance to EGFR TKI mediated by MET amplification, with preclinical data suggesting a combination of imatinib and crizotinib may overcome this particular mechanism of resistance.

Overall, aLK mutations account for ~37% of acquired resistance, copy number gain for 18%; alternate oncogene activation with or without loss of aLK fusion gene accounts for ~36%, and unknown mechanisms for 18%. Interestingly, sometimes multiple mechanisms of resistance were observed in the same patient, e.g. an aLK resistance mutation and EGFR activation, or copy number gain and an aLK mutation [18] To support this, Doebele et al. subsequently reported the analysis of 19 aLK-positive patients that underwent rebiopsy after progression under crizotinib therapy. Rebiopsy yielded tumor in 84% of cases, 81% of which demonstrated a plausible mechanism of resistance. 50% were aLK non-dominant, with 31% aLK kinase domain mutations and 19% aLK fusion gene copy number gain. 50% were aLK-dominant, with 31% demonstrating the emergence of alternate oncogenes (EGFR or KRaS activating mutation), and 19% unknown [26]. This potential for multiple and simultaneous resistance mechanisms has several clinical implications, affecting diagnostic evaluations as well as treatment strategies. First, new aLK inhibitors might effectively inhibit some resistant aLK fusion products, with a coexistent mechanism of resistance hampering tumor responsiveness. This clearly supports the use of combination therapy to overcome resistance. Second, the question of TKI continuation upon progression remains unresolved. Third, tumor heterogeneity, both within the primary tumor and among individual metastases, will raise the question of multiple re-biopsies. From a therapeutic standpoint, the wide array of resistance alterations will make it challenging to develop strategies to overcome aLK inhibitor resistance [27].

Various aLK inhibitors are being tested in ongoing trials. a phase I dose escalation of aLK TKI LDK378 (Novartis Pharmaceuticals) is recruiting patients with aLK-positive advanced NSCLC (NCT01283516). a phase I/II study (safety, tolerability, pharmacokinetics and preliminary antitumor activity) is testing the combined aLK/EGFR inhibitor aP26113 (aRIaD Pharmaceuticals) in aLK-positive tumors including advanced NSCLC (NCT01449461).

heat shock protein 90 inhibitors in aLK-driven NSCLC
Heat shock protein 90 (Hsp90) is a molecular chaperone that plays a key role in the conformational maturation of oncogenic signaling proteins. Inhibitors bind to Hsp90 and induce the proteasomal degradation of its client proteins. Tumor cells contain Hsp90 complexes in an activated conformation that facilitates malignant progression but also exhibit high-affinity to Hsp90 inhibitors and therefore represent a unique target for cancer therapeutics [28]. Clinical data have shown the efficacy of Hsp90 inhibitors in combination with EGFR TKIs after progression on TKI therapy. Hsp90 inhibitors might abrogate the oncogenic switch that allows cancer cells to signal through alternative receptor tyrosine kinases. In fact, most oncogenic kinases, including BRaF, MET and aLK, are Hsp90 clients that are sensitive to Hsp90 inhibition [29]. Clinical evaluation of another Hsp90 inhibitor is currently in progress in unselected advanced NSCLC patients (STa 9090, Synta Pharmaceuticals Corp., NCT01031225). a single-arm phase II study is testing ganetespib (STa-9090), an Hsp90 inhibitor (Synta Pharmaceuticals Corp.) in aLK-positive NSCLC patients (NCT01562015). a phase II study is testing the Hsp90 inhibitor aUY922 (Novartis Pharmaceuticals) in patients with advanced NSCLC including EGFR-mutated or aLK-positive patients who have received at least two lines of prior chemotherapy (NCT01124864). another phase II study testing IPI-204, a novel Hsp90 inhibitor (Infinity Pharmaceuticals) tested in advanced aLK-positive NSCLC patients, has been closed due to slow accrual (NCT01228435).

ROS1 rearrangements
ROS1, like aLK, is a receptor tyrosine kinase of the insulin receptor family. It has been initially identified as a potential driver mutation in an NSCLC cell line and one NSCLC patient [12]. Translocations leading to ROS1 fusion transcripts were shown to lead to constitutive kinase activity and sensitivity to TKIs. at present, data suggest that ROS1 is inhibited by some aspecific multiple kinase inhibitors, including crizotinib. There are currently no specific ROS1 inhibitors in clinical trial. The clinical characteristics of patients with ROS1 rearrangements were described by Bergethon et al. [30], who screened 1073 NSCLC patients using an ROS1 FISH assay, mainly in the United States. approximately 2% of NSCLC harbored ROS1 rearrangements. as is the case with aLK translocations, these patients tended to be younger than the wild-type patients and were more likely to be never/light smokers. Of all never smokers screened, 6% harbored ROS1 rearrangements. In vitro, crizotinib inhibits ROS1 activity and cell proliferation. Preclinical development of ROS1-specific kinase inhibitors is ongoing [31]. In the phase I study PROFILE 1001, crizotinib demonstrated marked antitumor activity in 14 evaluable patients with advanced NSCLC harboring ROS1 gene rearrangement, with 7 patients experiencing a partial response, 1 experiencing a complete response, and a 79% disease control rate at 8 weeks [15, 32].

BRaF mutations
BRaF is a kinase that links RaS GTPase to downstream proteins of the MaPK pathway. BRaF lies downstream of KRaS and directly phosphorylates MEK. BRaF mutations cause increased kinase activity and constitutive activation of MaPK2 and MaPK3. BRaF mutations are found in ~1–5% of NSCLC, almost exclusively adenocarcinomas [33–36]. Paik et al. [37] found 18 out of 697 screened lung adenocarcinomas to harbor a BRaF mutation [36]. Remarkably, all patients were current or former smokers, and there seems to be a paucity of BRaF mutations in non-white populations, with Sasaki et al. [35] reporting 1 out of 97 Japanese patients with lung adenocarcinoma harboring a BRaF mutation. Marchetti et al. screened 1046 NSCLC patients and found BRaF mutations in 4.9% of adenocarcinomas and 0.3% of squamous NSCLC. The mutations found in NSCLC are distinct from the melanoma setting: whereas BRaF-mutated melanoma harbors a V600E amino acid substitution in more than 80% of cases, NSCLC harbors non-V600E mutations, distributed in exons 11 and 15, in ~40%–50% of cases. In Marchetti's publication, all non-V600E mutations (2%) detected in adenocarcinomas were found in smokers and V600E mutations (2.8%) were substantially more frequent in females and in never smokers. In this series, follow-up data were available for the 331 resected lung carcinomas, showing that patients with V600E BRaF mutations had more aggressive tumor histotype, characterized by micropapillary features and associated with shorter median disease-free survival and OS, while no prognostic impact was found for the non-V600E mutations [36]. Interestingly, mutations in BRaF were mutually exclusive with EGFR and KRaS mutations, and aLK rearrangements. Due to the large predominance of V600E mutations in melanoma, current drugs targeting BRaF such as vemurafenib have been tailored to have specific activity against V600E mutant kinase. Their activity against other BRaF mutations found in NSCLC, such as G469a (39%) and D594G (11%) is unknown. Preclinical data suggest that non-V600E mutated BRaF kinases are resistant to vemurafenib [38]. Conversely, the CRaF inhibitor sorafenib was found to be ineffective against the V600E mutant isoform, but might have increased activity against other BRaF mutants that have increased CRaF activity [39]. In addition, there are preclinical data, suggesting that BRaF mutations might predict sensitivity of NSCLC cells to MEK inhibitors [40].

Clinical data on efficacy and resistance to BRaF pathway inhibitors are not yet available. according to preclinical data, amplification of BRaF or activation of downstream pathway components such as activating MEK mutations, or signaling through other RaF family members, might be among the main mechanisms of resistance [41]. It has been observed that colon cancer patients harboring V600E BRaF mutations show only a very limited response to vemurafenib [42]. In this setting, BRaF inhibition leads to rapid feedback activation of EGFR, and blockade of EGFR with cetuximab, gefitinib or erlotinib shows strong synergy with BRaF V600E inhibition in vitro [43].

a phase I study testing the multiple RaF kinase inhibitor (including CRaF, BRaF and the activated mutant BRaF V600E) XL281 (BMS-908662, Bristol-Myer Squibb) has been completed (NCT01086267) and results are awaited. aZD6244, a novel and highly selective MEK inhibitor is in phase II clinical trial for patients with solid tumors harboring a BRaF mutation (NCT00888134), and a phase II randomized trial versus pemetrexed in patients with advanced NSCLC in the second or third line has been completed, with results pending (NCT00372788). For the same compound, data are awaited from a phase II trial comparing its combination with docetaxel versus docetaxel alone in patients with advanced NSCLC with mutated KRaS (NCT00372788). The BaTTLE-2 Program, a biomarker-integrated targeted therapy study in previously treated patients with advanced NSCLC, also tests this compound in combination with MK2206 (Merck), an aKT inhibitor (NCT01248247). and finally, an ongoing phase II study evaluates the selective BRaF kinase inhibitor GSK2118436 (GlaxoSmithKline) in patients with advanced NSCLC harboring BRaF mutations (NCT01336634).

KRaS mutations
activating mutations in codons 12 and 13 of the KRaS oncogene occur in ~24% of lung adenocarcinomas and are mutually exclusive to EGFR mutations, HER2 mutations, aLK rearrangements and BRaF mutations [4]. KRaS mutations seem to occur early in the development of smoking-related carcinomas [44]. Cappuzzo and colleagues carried out a prospective molecular marker analysis of EGFR and KRaS in a large sample of patients randomly assigned to placebo or erlotinib maintenance therapy after first-line chemotherapy. KRaS mutations seemed to be prognostic for reduced PFS, regardless of treatment received [45]. There is currently no drug available capable of inhibiting KRaS directly, and current strategies focus on potential targets downstream of KRaS in the RaS/RaF/MEK pathway. Sorafenib, a weak RaF inhibitor, showed efficacy in KRaS mutant NSCLC according to a brief report by Smit et al. [46], with a partial response in 3 out of 10 patients and stable disease in 6 out of 10 patients, with a median PFS of 3 months. Sorafenib also showed efficacy in the BaTTLE trial, a phase II adaptive randomized trial, where KRaS mutant NSCLC on sorafenib showed a lower progression rate at 8 weeks when compared with the whole study population of 244 patients [47]. an ongoing phase II study is testing the MEK inhibitor GSK1120212 (GlaxoSmithKline) versus docetaxel in the second-line setting in advanced NSCLC patients with specific mutations in the KRaS signaling pathway (including KRaS) (NCT01362296).

HER2 mutations
HER2 (or ERBB-2) is a member of the EGFR family of receptor tyrosine kinases. It does not have a known ligand and is activated by homodimerization or heterodimerization with other members of the HER family. HER2 activates the PI3K/aKT/mTOR (mammalian target of rapamycine) pathway. HER2 overexpression or gene amplification is associated with sensitivity to trastuzumab and lapatinib in breast cancer. In a meta-analysis of 40 published studies, HER2 overexpression assessed by immunohistochemistry (IHC) was shown to be a marker of poor prognosis in NSCLC, with a hazard ratio of 1.48 (95% CI 1.22–1.80) and 1.95 (95% CI 1.56–2.43) in adenocarcinomas specifically. No prognostic value was found in squamous cell carcinomas. HER2 amplification determined by FISH was not related to prognosis [48]. In the lung cancer setting, amplification was found in 2–23% of the patients, while HER2 mutations were found in 2% of lung adenocarcinomas [49]. HER2 mutations consist of insertions in exon 20, leading to constitutive activation of the receptor, with downstream activation of aKT and MEK [50]. In vitro, cells harboring these mutations are sensitive to TKIs targeting HER2 and EGFR such as lapatinib [51] but are resistant to TKIs targeting EGFR alone. This is the case irrespective of the EGFR mutational status (i.e. including the small number of tumors harboring both EGFR and HER2 mutations), the activating signals being executed through the HER2 kinase [49].

In preclinical models of NSCLC trastuzumab has additive or synergistic antitumor activity in combination with various cytotoxic agents [52]. Trastuzumab has been tested in advanced NSCLC patients overexpressing HER2 in a phase II trial, in combination with cisplatin and gemcitabine (NCT00016367), and failed to show survival benefit in all HER2 IHC-positive lung cancers. Nevertheless, 80% of the patients with IHC 3+ disease on study treatment were still alive at 6-month follow-up, compared with 64% of the overall population, and an RR of 83% and a median PFS of 8.5 months were observed in the six trastuzumab-treated patients with HER2 3+ or FISH-positive NSCLC [53]. In HER2-amplified NSCLC, there seems to be no benefit from lapatinib [54]. a case report from 2006 describes a female non-smoker with metastatic adenocarcinoma resistant to cisplatin, taxane and EGFR TKI therapy, with a tumor carrying an exon 20 mutation (G776L) and HER2 amplification responding to trastuzumab given weekly together with paclitaxel [55]. a single-arm trial of the EGFR/HER2 dual inhibitor BIBW 2992 (afatinib) showed responses in 3/3 assessable patients (out of 5 identified) with adenocarcinoma harboring HER2 activating mutations, even in the context of resistance to other EGFR- or HER2-targeted compounds [56].

Trastuzumab is currently being tested alone, in IHC-positive or, respectively, HER2-mutated or -amplified NSCLC (NCT00004883 and NCT00758134) and in combination with carboplatin and paclitaxel. Results are pending. Lapatinib has been tested in molecularly unselected advanced NSCLC patients, including one trial that has been stopped for futility after interim analysis (NCT00073008). Pertuzumab has been tested in a phase II trial in advanced NSCLC patients with HER2 activation (NCT00063154). Results are pending. More trials investigating afatinib in other advanced NSCLC patients, including in combination with EGFR TKIs, are ongoing.

PIK3Ca mutations
PIK3Ca mutations regenerate phosphatidylinositol-3-phosphate, which is a key mediator between growth factor receptors and downstream signaling pathways. In NSCLC, PIK3Ca mutations affect most frequently the catalytic domain encoded in exon 9 and are found in ~2% of NSCLC, as frequently in adenocarcinoma as in squamous cell carcinoma [57]. PIK3Ca mutations induce oncogenic cellular transformation [58]. amplification of PIK3Ca has also been observed in NSCLC, particularly in squamous cell carcinoma, but the oncogenic potential of PIK3Ca amplification alone has not yet been shown [59]. Chaft et al. reported 23 out of 1125 patients harboring PIK3Ca mutations, 16 (70%) of which had coexisting mutations in other oncogenes: 10 KRaS, 1 BRaF, 1 aLK rearrangement and 3 EGFR exon 19 deletions [60]. This is in sharp contrast to the mutual exclusiveness of driver oncogene mutations found in lung adenocarcinomas harboring EGFR, KRaS or aLK transformations. In their sample, the presence of coexisting oncogene mutations was associated with an inferior outcome, with only one patient having received an experimental agent targeting PIK3Ca. Cell lines with PIK3Ca mutations are sensitive to downstream inhibitors such as mTOR inhibitors, but this sensitivity is abrogated by coexistent KRaS mutation [61]. Preclinical data actually suggest that coexisting KRaS and PIK3Ca mutations are associated with resistance to PI3K/aKT/mTOR inhibitors.

The oral PI3K inhibitor BKM120 (Novartis Pharmaceuticals) is being tested in a phase II trial in pretreated advanced NSCLC patients with activated PI3K pathway (NVT01297491). The same compound is also being tested in combination with erlotinib in the setting of resistance to EGFR TKIs (NCT01487265). another oral specific PI3K inhibitor, GDC0941 (Genentech), is being tested in a phase Ib trial in combination with carboplatine/paclitaxel ± bevacizumab or cisplatine/pemetrexed/bevacizumab in unselected patients with advanced NSCLC (NCT00974584).

MET amplification and point mutations
The MET oncogene encodes hepatocyte growth factor (HGF) receptor, a transmembrane receptor with tyrosine kinase activity. Its amplification has been reported in 1.4% of lung adenocarcinomas in a Japanese population and 21% of NSCLC in a European population including squamous cell carcinomas [62, 63]. MET amplification has transforming capacity, being sufficient to drive the proliferation of cancer cells and development of metastasis in a mouse melanoma model [64]. MET amplification is observed as a mechanism of resistance in ~20% of the patients with activating EGFR mutations progressing under EGFR TKI [65]. Point mutations in the kinase domain of MET are rare in NSCLC [66, 67]. While their prevalence is low, their potential for causing disease progression is significant [68], and when used to replace endogenous MET in the mouse germline, these mutations cause a variety of tumors including carcinomas of various tissues of origin [69]. In NSCLC, most of MET mutations are located in the extracellular sema domain and the juxtamembrane domain of MET, with a preclinical demonstrated potential to affect ligand binding, receptor activation and receptor turnover.

The MET pathway can be inhibited by monoclonal antibodies against HGF or its receptor or by MET TKIs [70]. aMG102 (amgen), a human monoclonal antibody that binds and neutralizes HGF/scatter factor (SF), is being tested with erlotinib in a phase I/II trial in pretreated patients with advanced NSCLC (NCT01233687).

Interestingly, two randomized phase II trials of a MET monoclonal antibody (onartuzumab, Genentech) and a MET-specific TKI tivantinib together with erlotinib versus erlotinib alone showed promising results in unselected pretreated NSCLC patients and are now being tested in a phase III trial [71]. Onartuzumab is being tested in various settings including this randomized phase III trial which is testing onartuzumab or placebo in combination with erlotinib in pretreated patients with advanced MET IHC-positive NSCLC (NCT01456325) ; another randomized phase III trial is testing onartuzumab or placebo in combination with carboplatin/cisplatin and paclitaxel in untreated patients with advanced squamous cell carcinoma (NCT01519804); a randomized phase II trial is testing onartuzumab or placebo in combination with bevacizumab/carboplatine/paclitaxel or cisplatin/pemetrexed (NCT01496742). Tivantinib (aRQ197, Daiichi Sankyo) is tested in a phase III trial versus placebo in combination with erlotinib in advanced non-squamous NSCLC (NCT01244191) [72]. Besides crizotinib, a potent MET inhibitor, various other small-molecule MET inhibitors are being tested in NSCLC, including GSK13693089 (foretinib, a MET/VEGFR2 inhibitor, GlaxoSmithKline) and XL184 (cabozantinib, a MET/VEGFR2 inhibitor, Exelixis), even if, to date, a predictive biomarker for MET inhibitor sensitivity remains to be defined.

The discovery of EGFR mutations has opened the era of targeted therapy in NSCLC, shifting treatment from platinum-based chemotherapy for all fit patients to molecularly personalized therapy. The greatest improvements in outcome are obtained by targeting the ‘driver genetic alteration’ of each specific molecularly defined subset, rather than targeted therapy of unselected patients. The identification of the key molecular abnormality will thus become crucial, even if numerous very small subsets of tumors will have to be identified.

acquired resistance has emerged as a major hurdle preventing targeted therapy from having a substantial long-term impact on outcome beyond their initial benefit. The understanding of these mechanisms will hopefully allow a better sequencing and optimal combination of targeted agents in each biologically defined setting. Resistance mutations may be overcome with more potent and/or irreversible inhibitors capable of blocking mutated targets. Combination therapies will most likely be the key to overcome resistance mediated by activation of parallel or downstream pathways. Several other mechanisms of drug resistance, such as drug efflux by antiporter efflux pumps, as well as anti-apoptotic mechanisms have the potential to limit drug efficacy and need to be further explored.

Beyond oncogenic activation, other genetic alterations not encoded by the DNa sequences, referred to as epigenetic changes, also represent potential therapeutic targets. as opposed to genetic lesions, the epigenetic changes are potentially reversible by a number of small molecules such as histone deacetylases, which are, however, beyond the scope of this review [73].

In conclusion, targeted therapies hold promise for improved outcome in advanced NSCLC patients, even after the development of acquired resistance. This, however, will demand the incorporation of broad genotyping of NSCLC into the clinic as a standard of care, as well as successive repeated and potentially multisite biopsies.



“人类只有经历过悲哀才能够知道什么是快乐,你只有经过死亡以后才知道重生意味着什么,上帝给人留下了四个字,那就是希望和等待。”
荷花池荒岛  硕士一年级 发表于 2014-8-9 23:52:42 | 显示全部楼层 来自: 美国
本帖最后由 荷花池荒岛 于 2014-8-11 10:34 编辑

Resistance to ROS1 Inhibition Mediated by EGFR Pathway Activation in Non-Small Cell Lung Cancer
http://connection.ebscohost.com/ ... ll-cell-lung-cancer

The targeting of oncogenic driver kinases with small molecule inhibitors has proven to be a highly effective therapeutic strategy in selected non-small cell lung cancer (NSCLC) patients. However, acquired resistance to targeted therapies invariably arises and is a major limitation to patient care. ROS1 fusion proteins are a recently described class of oncogenic driver, and NSCLC patients that express these fusions generally respond well to ROS1-targeted therapy. In this study, we sought to determine mechanisms of acquired resistance to ROS1 inhibition. To accomplish this, we analyzed tumor samples from a patient who initially responded to the ROS1 inhibitor crizotinib but eventually developed acquired resistance. In addition, we generated a ROS1 inhibition-resistant derivative of the initially sensitive NSCLC cell line HCC78. Previously described mechanisms of acquired resistance to tyrosine kinase inhibitors including target kinase-domain mutation, target copy number gain, epithelial-mesenchymal transition, and conversion to small cell lung cancer histology were found to not underlie resistance in the patient sample or resistant cell line. However, we did observe a switch in the control of growth and survival signaling pathways from ROS1 to EGFR in the resistant cell line. As a result of this switch, ROS1 inhibition-resistant HCC78 cells became sensitive to EGFR inhibition, an effect that was enhanced by co-treatent with a ROS1 inhibitor. Our results suggest that co-inhibition of ROS1 and EGFR may be an effective strategy to combat resistance to targeted therapy in some ROS1 fusion-positive NSCLC patients.
“人类只有经历过悲哀才能够知道什么是快乐,你只有经过死亡以后才知道重生意味着什么,上帝给人留下了四个字,那就是希望和等待。”
荷花池荒岛  硕士一年级 发表于 2014-8-9 23:52:50 | 显示全部楼层 来自: 美国
本帖最后由 荷花池荒岛 于 2014-8-11 10:46 编辑

Overcoming erlotinib resistance with tailored treatment regimen in patient-derived xenografts from na&#239;ve Asian NSCLC patients.
http://www.ncbi.nlm.nih.gov/pubmed/22948846

Abstract
Overall benefits of EGFR-TKIs are limited because these treatments are largely only for adenocarcinoma (ADC) with EGFR activating mutation. The treatments also usually lead to development of resistances. We have established a panel of patient-derived xenografts (PDXs) from treatment na&#239;ve Asian NSCLC patients, including those containing "classic" EGFR activating mutations. Some of these EGFR-mutated PDXs do not respond to erlotinib: LU1868 containing L858R/T790M mutations, and LU0858 having L858R mutation as well as c-MET gene amplification, both squamous cell carcinoma (SCC). Treatment of LU0858 with crizotinib, a small molecule inhibitor for ALK and c-MET, inhibited tumor growth and c-MET activity. Combination of erlotinib and crizotinib caused complete response, indicating the activation of both EGFR and c-MET promote its growth/survival. LU2503 and LU1901, both with wild-type EGFR and c-MET gene amplification, showed complete response to crizotinib alone, suggesting that c-MET gene amplification, not EGFR signaling, is the main oncogenic driver. Interestingly, LU1868 with the EGFR L858R/T790M, but without c-met amplification, had a complete response to cetuximab. Our data offer novel practical approaches to overcome the two most common resistances to EGFR-TKIs seen in the clinic using marketed target therapies.
“人类只有经历过悲哀才能够知道什么是快乐,你只有经过死亡以后才知道重生意味着什么,上帝给人留下了四个字,那就是希望和等待。”
荷花池荒岛  硕士一年级 发表于 2014-8-9 23:52:58 | 显示全部楼层 来自: 美国
本帖最后由 荷花池荒岛 于 2014-8-11 11:04 编辑

While crizotinib works very well, “at some point patients do develop resistance, typically around 9 to 12 months” after starting treatment, said Shaw.

A spectrum of secondary mutations have been identified in the ALK kinase itself. The most common is the L1196M “gatekeeper” mutation.
Amplification of the ALK fusion gene has also been identified. In about two-thirds of resistant cases, no resistance mutations in ALK are identifiable, and activation of other signaling pathways, such as the EGFR pathway, may mediate resistance to crizotinib.

Understanding these mechanisms has helped to accelerate the next wave of ALK inhibitors, said Shaw. The next generation ALK inhibitors entering the clinic have primarily been studied in ALK-positive NSCLC patients who have acquired resistance to crizotinib. Many of these drugs also target ROS1 rearrangements, Shaw noted.

The secondary ALK gene mutations have so far shown different degrees of response to the novel ALK inhibitors. The most advanced of these are LDK378 (Novartis) and alectinib (Roche).

Based on positive phase II data, LDK378 is now being tested in two phase III trials in both treatment-na&#239;ve patients and those previously treated with both chemotherapy and crizotinib. The drug has shown activity against both ALK and ROS1.

Alectinib selectively targets ALK—but not ROS1—and has shown a high response rate in crizotinib-na&#239;ve Japanese patients.

Both of these agents have been shown to be active on brain metastases.

Going forward, the next questions for researchers are whether using these new ALK inhibitors upfront could prolong the time to resistance and how to overcome resistance that develops even to these new drugs.

http://www.onclive.com/conferenc ... -Resistant-Patients


“人类只有经历过悲哀才能够知道什么是快乐,你只有经过死亡以后才知道重生意味着什么,上帝给人留下了四个字,那就是希望和等待。”
荷花池荒岛  硕士一年级 发表于 2014-8-9 23:53:06 | 显示全部楼层 来自: 美国
本帖最后由 荷花池荒岛 于 2014-8-11 11:35 编辑

The Landscape of EGFR Pathways and Personalized Management of Non-small-cell Lung Cancer
http://www.medscape.com/viewarticle/740715_1

Abstract and Introduction

Abstract

Two classes of anti-EGF receptor (EGFR) agents, monoclonal anti-EGFR antibodies and small-molecule EGFR tyrosine kinase inhibitors, have been used for the treatment of non-small-cell lung cancer (NSCLC). However, only a subset of patients will benefit from EGFR-targeted therapy. The discovery of biomarkers that select the appropriate patients for the therapy and predict the responses to the therapy is urgently needed. Molecular genetic analyses provide new insights into EGFR pathway alterations and demonstrate promise for predicting the clinical outcome of patients with NSCLC. In this article, we summarize the latest available knowledge on the clinical impact of EGFR mutations, gene copy number, EGFR overexpression, phosphorylation expression and the alteration of the EGFR pathway downstream factors in predicting the response to EGFR-targeted therapy in NSCLC patients. The role of KRAS and BRAF mutations and ALK rearrangement in lung cancer-targeted therapy, are also reviewed.

Introduction

Non-small-cell lung cancer (NSCLC) accounts for approximately 80% of lung cancers and is one of the leading causes of cancer death in North America. It is often diagnosed at an advanced stage, with only 30–40% of metastatic NSCLC patients surviving for 12 months.[1–3]

Surgery is the most effective treatment for NSCLC; however, it is usually reserved for patients whose tumors are confined to the primary site and who have no or minimal lymph node involvement – a small portion of NSCLC cases. In addition, many patients who undergo curative surgery will later develop recurrence. Platinum-based chemotherapy is the mainstay of treatment in advanced or recurrent NSCLC, but the results of the treatment are far from encouraging.[1]

Approximately 85% of all primary lung cancers are NSCLCs, which are classified into three major histologic types: adenocarcinoma, squamous cell carcinoma and large-cell carcinoma.[4] Adenocarcinoma is the most frequent histologic type of NSCLC in both genders in many parts of the world (Figure 1). It accounts for approximately 40% of lung cancers and is usually found in the periphery of the lungs.[1] The most common histologic subtypes of adenocarcinoma are papillary (37%), acinar (30%), solid (25%) and bronchioloalveolar (7%).[5] Squamous cell carcinoma accounts for approximately 25–30% of all lung cancers and is often linked to a history of smoking. This cancer tends to occur in the hilar region of the lungs near the bronchus. Large-cell carcinoma accounts for approximately 10–15% of lung cancers and may appear in any part of the lung. It tends to grow and spread quickly and carries a poor prognosis.

Cigarette smoking remains the principal cause of lung cancer. It is estimated that 85–90% of all lung cancer patients have smoked cigarettes at some time.[6] As such, 87% of lung cancer deaths are thought to result from smoking. The US Environmental Protection Agency reports that radon is the second leading cause of lung cancer after cigarette smoking and is the leading environmental cause for nonsmokers. The risk of developing NSCLC from radon is much higher in people who smoke than in those who do not. Workplace exposure to asbestos fibers is another important, but less common, risk factor for lung cancer.[7–9]

In recent years, attention has turned to the role that the EGF receptor gene (EGFR) plays in tumorigenesis and its utility as a target for therapy. Biomarkers that can reliably predict which patients may benefit from anti-EGFR therapy are urgently needed. Pathologists will play a central role in the process to determine suitable testing and interpretation of the test results. In this article, we summarize the impact of EGFR alterations in predicting response to anti-EGFR therapies and discuss currently proposed technologies and their potential clinical implications.

Overview of EGFR & NSCLC

The EGFR and members of its family play an important role in carcinogenesis through their involvement in the modulation of cell proliferation, apoptosis, cell motility and neovascularization.[10] EGFR alterations have been implicated in the pathogenesis and progression of many malignancies.[11–14] Although the exact molecular pathways by which the mutant receptors lead to carcinogenesis are not completely understood, it is clear that mutant variants of EGFR have enhanced tyrosine kinase (TK) activity.

The presence of activating mutations in EGFR was initially reported in 2004.[12,15,16] Various groups also found amplification and overexpression of EGFR.[17–21] Clinical and pathological factors such as female gender, never having smoked, East Asian ethnicity and adenocarcinoma or bronchioloalveolar histology, correlated with objective responses to single-agent TK inhibitor (TKI) therapy in NSCLC and also with the presence of somatic EGFR mutations.[10,11,15,22,23] EGFR mutations rarely occurred in squamous cell carcinoma, large-cell carcinomas or adenocarcinomas with KRAS mutations.[5,12,15,20,22–31] Of great benefit to researchers in evaluating possible contributing mutations in NSCLC and acquired resistance to targeted therapies is the Catalogue of Somatic Mutations in Cancer (COSMIC[201]). COSMIC is designed to store and display somatic mutation information and related details relating to human cancers; it is a very useful tool for both researchers and clinicians. As more advanced molecular techniques reveal further molecular mutations, centralized databases such as COSMIC will allow clinicians and researchers to stay abreast of currently available information.

Lung adenocarcinomas frequently possess EGFR mutations and frequently exhibit increased EGFR copy number.[32] A study of 334 cases of lung adenocarcinomas using PCR-based assays to detect deletions within exon 19 and the L858R mutation in exon 21 of the EGFR gene found that 23% of these tumors contained a mutation. Of those, 71% were exon 19 deletions and 29% comprised the L858R mutation in exon 21.[32] In addition, EGFR amplification, defined as greater than five EGFR signals per nucleus by FISH, was detected in 52% of EGFR-mutated tumors, but in only 6% of those lacking the mutations. EGFR mutations were present in 26% of 86 bronchioloalveolar carcinomas.[24] It appears that EGFR mutations occur much less frequently in squamous cell carcinoma than in adenocarcinoma, with a reported incidence of 0–14%.[24,33] The third type of NSCLC, large-cell carcinoma, harbors EGFR mutations very rarely, if ever.[23,34] Marchetti et al. investigated a series of 31 large-cell carcinomas and found no EGFR mutations in any case.[24]

Motoi et al. reported that EGFR mutations are particularly frequent in adenocarcinoma of the papillary subtype.[5] In a group of NSCLC patients, 13 out of 36 (35%) papillary cancers harbored EGFR mutations, in contrast to three out of 63 nonpapillary cancers (5%). Kim et al. found that papillary subtype is a significant predictor of response to gefitinib in lung adenocarcinoma, although they did not link the incidence of response to the EGFR mutation status.[35]

The clinical implications of EGFR overexpression have been studied extensively, but the results are inconclusive thus far. Recent use of phosphor-specific antibody has facilitated analysis of the correlation between phosphorylation and EGFR mutation status. In a study of 218 cases of NSCLC, McMillen et al. correlated EGFR expression status with mutation status.[36] Phosphorylation at Y1045 was noted in 52% of cases, 71% of which exhibited the presence of an EGFR mutation. Phosphorylation of Y1068 was seen in 55% of cases, but it was present in 73% of those cases with an EGFR mutation. The data demonstrate that among Chinese patients, immunohistochemical detection of p-1045 and p-1068 expression predicts EGFR mutations.

Activation mutations identified within the kinase domain of the EGFR gene led researchers to propose and develop therapeutic strategies targeting EGFR TK. Therapeutic strategies targeting the EGFR pathway offered exciting new options for the treatment of NSCLC. EGFR alterations have prompted the development of two classes of anti-EGFR agents: monoclonal anti-EGFR antibodies (e.g., cetuximab and panitumumab) and small-molecule TKIs of EGFR (e.g., gefitinib and erlotinib, among others). According to large-cohort Phase III clinical trials,[37–40] the response rates range from 15 to 37.5%. Clinical trials were initiated that employed novel agents targeting the EGFR TK. The results of these clinical trials indicated that many of the tumors harboring mutant EGFR are highly sensitive to EGFR TKIs, with 10–30% demonstrating a significant clinical response.[15,41]

Summary

Constitutive activation of EGFR TK in NSCLC is associated with carcinogenesis of NSCLC.

The incidence of EGFR mutations in NSCLC is dependent upon gender, smoking history, tumor type and ethnic background.

Female gender, zero or very light smoking history, East Asian ethnicity, adenocarcinoma or bronchioloalveolar histology correlates with objective responses to single-agent TKI therapy in NSCLC.

EGFR Pathway Alteration & Implications

EGFR, located at chromosome 7p12, spans approximately 200 kb and contains 28 exons. It is a member of the ErbB family of four closely related TK receptors: EGFR (ErbB1), HER2/c-neu (ErbB2), HER3 (ErbB3) and HER4 (ErbB4).[42,43] EGFR is activated by binding of its specific ligands. Structurally, EGFR is composed of an N-terminal extracellular ligand-binding domain, a transmembrane lipophilic segment, and a C-terminal intracellular region containing a TK domain. Multiple ligands that bind and activate EGFR have been described, including EGF and TGF-α. Upon ligand binding to EGFR, the receptors form homo- or hetero-dimers, which activate their intrinsic intracellular protein TK. The ligand binding-induced dimerization results in cross-autophosphorylation of key tyrosine residues in the cytoplasmic domain, which function as docking sites for downstream signal transducers.[44] This activation of EGFR results in initiation of signaling cascades involving several downstream pathways.[4,26,45–50] Through its influence on these pathways, EGFR induces a number of crucial cellular responses, such as proliferation, differentiation, motility and enhanced cell survival (Figure 2).[50–53]

CaptureFigure2.PNG
Figure 2.

EGFR and its signaling pathway. Structurally, an EGF receptor (EGFR) monomer is composed of an extracellular domain consisting of two ligand-binding subdomains: a transmembrane lipophilic segment and an intracellular region containing tyrosine kinase domains that occupy exons 18–24. The binding of ligands to EGFR results in autophosphorylation of key tyrosine residues in the cytoplasmic domain and activation of its intrinsic, intracellular protein tyrosine kinase activity. EGFR activation results in initiation of signaling cascades. These function to further modulate cell proliferation and survival through two downstream intermediate pathways: the PI3K–AKT–mTOR pathway and the RAS–RAF–MEK–MAPK pathway. These two intermediate pathways influence several key aspects of the cell cycle that include cell proliferation, apoptosis, migration and survival, and more complex processes such as angiogenesis.

P: Phosphorylation.

One of the major molecular alterations in the carcinogenesis of NSCLC is the activation mutation of EGFR. The mechanisms that regulate EGFR expression, such as epigenetic alteration and aberrant transcription factors, have been studied, but with inconclusive results. The significance of miRNA-128b was reported by Weiss et al., who found loss of miRNA-128b in two out of three NSCLC cell lines and in tumors from 55% of NSCLC patients.[54] miRNA-128b directly regulated EGFR expression, and loss of miRNA-128b correlated with better tumor responsiveness to gefitinib treatment and improved survival (23.4 vs 10.5 months).[54]

A strong genetic association with particular germline mutations has been shown to influence the susceptibility to EGFR TKIs (i.e., those that confer mutations in EGFR signaling). Liu et al. found that the frequencies of the -216T and CA-19 alleles are significantly higher in patients with any mutation, in particular in those with exon 19 microdeletions.[55] The -216T allele is preferentially amplified in human lung cancer specimens and cancer cell lines. These results suggest that the local haplotype structures across the EGFR gene may favor the development of carcinogenesis and thus significantly confer risk to the occurrence of EGFR mutations in NSCLC, particularly the exon 19-microdeleted cases.[55] A novel germline transmission of the EGFR mutation V843I in a family with multiple members with lung cancer has been reported.[56] The proband was a 70-year-old woman who had multiple adenocarcinomas with EGFR mutations. These observations suggest that germline EGFR V843I mutation may result in altered EGFR signaling in cases of multicentric adenocarcinoma, bronchioloalveolar carcinoma and atypical adenomatous hyperplasia, and may also play a role in the development of lung cancer in multiple family members.[56]

EGFR plays a key role in the growth and survival of many solid tumor types.[21,57] Mutations affecting EGFR activity or expression can result in cancer.[58] The EGFR TK modulates cell proliferation and survival through two downstream intermediate pathways: the PI3K–AKT–mTOR pathway and the RAS–RAF–MEK–MAPK pathway.[59] These downstream cell signaling pathways influence several critical cellular processes, including cell proliferation, apoptosis, migration and survival. They are also involved in more complex processes such as angiogenesis and tumorigenesis. Studies on EGFR oncogene activation have been focused on gene mutations, DNA copy number alterations, protein expression alterations and genetic alterations of downstream signaling molecules.

Results from a Phase III trial evaluating the EGFR TKI, gefitinib, indicated that approximately 10% of patients responded to the therapy, and no survival benefit was observed.[30] Follow-up analysis identified mutations in the TK domain of EGFR in eight out of nine responders, whereas no mutations were detectable in seven patients who did not respond to gefitinib therapy.[12]

The most widely used EGFR TKIs are gefitinib and erlotinib. These agents are reversible inhibitors that compete with ATP at the active site of the TK receptor domain.[6] They are primarily used in patients who have failed platinum-based chemotherapy.[60] In a randomized Phase III study, erlotinib significantly improved median survival from 4.7 (placebo) to 6.7 months in patients with NSCLC who had previously failed one or two chemotherapy regimens.[61] Gefitinib improved disease-related symptoms in heavily pretreated symptomatic patients with NSCLC.[30] However, in the Phase III Iressa Survival Evaluation in Advanced Lung Cancer (ISEL) trial, which included pretreated patients with recurrent disease, gefitinib failed to demonstrate a survival benefit in the overall unselected patient population compared with placebo-treated controls.[62] Recently, however, results from the randomized Phase III IRESSA NSCLC Trial Evaluating Response and Survival against Taxotere (INTEREST) indicate that gefitinib is not inferior to docetaxel in terms of overall survival, suggesting that this TKI may also be a viable option for previously treated patients with advanced NSCLC.[63]

The data from 222 publications indicate that EGFR mutations are predictive of patient response to single-agent EGFR TKI treatment in advanced NSCLC.[64]

EGFR Mutations

The EGFR mutations responsible for the constitutive activation of receptor TK are also most frequently associated with sensitivity to EGFR TKIs.[65] These mutations are associated with response rates of >70% in patients treated with either erlotinib or gefitinib.[28,66]

Receptors containing different mutations appear to have different signaling properties, but most mutations seem to affect the ATP-binding cleft, which is also where targeting TKIs bind (Figure 3).[49]

CaptureFigure3.PNG
Figure 3.

Mechanism of EGFR-activating mutations and EGFR-targeted therapy. (A) EGFR mutations render EGFR tyrosine kinase constitutively activated. Activated EGFR phosphorylates key tyrosine residues (P) in the tyrosine domain, which initiates downstream effectors. (B) At present, two categories of agents are used for inhibiting EGFR signaling: humanized antibodies and small-molecule TKIs. Antibodies inhibit ligand-dependent activation of EGFR by blocking the ligand-binding site and preventing activation. TKIs block the magnesium–ATP-binding pocket of the intracellular tyrosine kinase domain, further inhibiting autophosphorylation. This inhibition disrupts tyrosine kinase activity and abrogates intracellular downstream signaling.

TKI: Tyrosine kinase inhibitor.

In vitro studies have demonstrated that mutant EGFR has enhanced TK activity, leading to a greater sensitivity to anti-EGFR inhibition. As mentioned previously, the four most common mutations seem to be those most closely associated with TKI sensitivity. The discovery that many objective responders to TKIs harbored EGFR mutations in exons 19 and 21 was a major breakthrough in patient selection for EGFR targeting therapy.[67,68] The most frequent mutation, located in exon 19, eliminates four amino acids – leucine, arginine, glutamic acid and alanine – downstream from the lysine residue at position 745.[67,69–72] Patients with an EGFR mutation who were treated with TKI had much higher response rates and longer progression-free survival than those without a mutation (Table 1).[69]

Both retrospective and prospective studies have demonstrated that NSCLC patients carrying the described EGFR gene mutations have a significantly higher response rate to gefitinib and/or erlotinib compared with patients with wild-type EGFR.[12,15,16,28,67,68,73–76] Some patients experienced rapid complete or partial responses that were durable.[26] The discovery of somatic mutations in EGFR that correlated with sensitivity to TKIs identified a plausible and reproducible explanation for these observations.

The most commonly used methods to detect mutations are direct sequencing and real-time PCR.[73,77] Other methods include single-strand conformational polymorphism analysis[78,79] and high-resolution melting amplicon analysis.[17,80] Scorpion ARMS&#174; (QIAGEN, Germany), a multitargeted real-time PCR detection kit, allows the detection of the most prevalent somatic mutations in the EGFR that are common in human cancers. The high sensitivity and specificity of the kit permits the detection of mutations against a background of wild-type genomic DNA. The kit uses DxS Scorpions&#174; (QIAGEN) technology to detect exon 19 deletions and mutations in exons 19–21 (T790M, L858R, L861Q, G719X and S768I) and any one of three insertions into exon 19 (2307_2308ins9, 2319_2320insCAC and 2310_2311insGGT). Relative to the direct sequencing method, the other two techniques allow for the rapid detection of EGFR mutations with high sensitivity and specificity. However, confirmation of mutations via direct sequencing is necessary.[24,80,81] Standardization is essential for the clinical application of EGFR mutation tests. However, at present, there is no official guideline for these EGFR mutation tests. The number of mutation sites that are needed in the testing protocol still remains to be established. Large-scale clinical trials are also needed.

Jackman et al. studied 223 chemotherapy-naive patients with advanced NSCLC.[28] Sensitizing EGFR mutations were associated with a 67% response rate, with a time to progression (TTP) of 11.8 months and overall survival of 23.9 months. Exon 19 deletions were associated with a longer median TTP and overall survival compared with L858R (exon 21) mutations. Wild-type EGFR was associated with poor outcomes (response rate: 3%; TTP: 3.2 months), irrespective of KRAS status. EGFR genotype was more effective than clinical characteristics at selecting appropriate patients for consideration of first-line therapy with an EGFR TKI.

Studies indicate that more than 75% of patients responsive to TKI therapy have activating mutations in EGFR.[13,77] However, some rare types of EGFR mutations can confer resistance to EGFR-targeted therapies after treatment with TKIs when combined with the common activating mutations.[82–85] Clinically, patients with EGFR exon 20 mutations do not respond to gefitinib.[72] Moreover, the appearance of a secondary mutation in exon 20 (T790M) accounts for approximately 50% of acquired drug resistance.[77,86] Screening for the emergence of such mutations in circulating tumor cells from the blood of patients during the course of treatment may allow earlier identification of acquired resistance.[83,87]

Results of some preclinical studies suggest that the clinical benefit observed with EGFR TKIs is not restricted to those patients harboring EGFR mutations. This may be due to molecular factors outside of gene mutations. EGFR amplification and receptor/ligand overexpression, both of which allow for a 'gain of function' to occur, are implicated in creating a scenario of EGFR dependence that causes the sensitivity to single-agent EGFR inhibitors.[44,88] However, the data from the IRESSA Pan-Asia Study (IPASS) clearly demonstrate that patients with increased EGFR copy numbers and no EGFR mutations do not benefit from EGFR TKIs.[89]

EGFR Copy Number Alterations

EGFR is frequently over-represented or amplified in NSCLC, which is commonly associated with EGFR overexpression.[90] Increased EGFR copy numbers may result from gene amplification or polysomy of chromosome 7. The incidence of EGFR amplification ranges from 12 to 59%, depending on the patients selected and the technology used.[64,88,91,92] Some, but not all, studies have revealed that positive EGFR amplification is associated with significantly better survival after treatment with a TKI.[88,93] Gain of EGFR copy number has been consistently associated with a favorable outcome after EGFR TKI therapy; it has also been proposed to be a potential biomarker of TKI responsiveness.[91,94]

Somatic EGFR mutations consistently correlate with improved response rates; by contrast, the results of studies investigating EGFR copy number as a predictor of response to TKIs have been inconsistent.[25,95] Overall, EGFR mutations seem to have higher sensitivity and specificity for predicting response to TKIs than EGFR copy number gain status.[64] High copy numbers of EGFR have been detected in approximately 30% of NSCLC patients using FISH, and are reportedly associated with better responses to TKI therapy,[73,96] although the EGFR mutation status of those cases was unclear. Approximately 70% of patients with EGFR copy number gain also had EGFR somatic mutations, a fact that clouds the true significance of EGFR copy number gain. IPASS demonstrated that EGFR mutation was the strongest predictor of improved progression-free survival. There was a high degree of overlap between EGFR mutation positivity and high EGFR gene copy numbers: of 245 patients with high EGFR copy numbers whose EGFR mutation status was also known, 190 (78%) were also EGFR mutation-positive. This suggests that the improved outcome in high EGFR copy number patients is being driven by the EGFR mutation-positive overlap.[89] Some researchers have suggested that high EGFR copy numbers can be used as a predictive biomarker for response and survival benefit in patients with NSCLC who receive EGFR TKI therapy.[88] However, the data from the IPASS trial clearly demonstrate that patients with increased EGFR copy numbers and no EGFR mutations do not benefit from EGFR TKIs.[89] Hirsch et al. suggested that although EGFR mutations and high copy numbers are both predictive of response to erlotinib in NSCLC, EGFR copy number was a more powerful predictor of differential survival benefit from erlotinib.[88]

There are several methods for detecting and determining EGFR copy number and dosage, including FISH,[73,88,97] chromogenic in situ hybridization[92,98] and real-time quantitative PCR.[20,99,100] When EGFR copy number was measured by PCR, it was found that increased EGFR copy number was significantly associated with prolonged survival, indicating a potential prognostic value of EGFR copy number.[64,101] The patients with EGFR gain demonstrated a higher disease control rate (67 vs 26%), longer TTP (9.0 vs 2.5 months) and prolonged survival time (18.7 vs 7.0 months).[64] It is noteworthy that EGFR copy number is used as a predictor for response to TKI therapy largely because it is correlated with EGFR mutations – EGFR mutations are the best predictors. Hirsch et al. investigated 229 chemotherapy-naive patients with advanced-stage NSCLC in a Phase II clinical trial.[88] Among the 76 patients analyzed by FISH, 59.2% had increased EGFR copy numbers, as indicated by four or more gene copies per cell in >40% of the cells. Response was higher in EGFR-amplified patients (45%) versus the EGFR-unamplified patients (26%). Those patients with EGFR amplification had a median progression-free survival time of 6 months compared with 3 months for patients without amplification. Median overall survival was 15 months for the EGFR-amplified group, while it was only 7 months for patients without amplification.[102]

Despite a majority of studies demonstrating that high EGFR copy number correlates with better response and increased survival in NSCLC patients treated with EGFR TKIs, debate remains about its true predictive value. Some studies suggest that when compared with EGFR mutations, EGFR gene copy number is a less sensitive and less specific marker and may not be considered clinically suitable for patient selection.[64] Douillard et al. also reported that EGFR mutation demonstrates greater predictive power than EGFR copy number in therapy response and progression-free survival.[103] Further studies are necessary to resolve these discrepant findings.

EGFR Overexpression

The clinical implications of EGFR overexpression have been studied extensively. However, the results have been inconclusive thus far. Immunohistochemistry-based assays measuring EGFR expression could not reliably predict the response to EGFR TKI therapy. Overexpression of EGFR has been demonstrated in 40–80% of cases of NSCLC and has been associated with a poor prognosis.[104–106] The initial assumption was that EGFR antibodies would be more effective in tumors with robust overexpression of EGFR. However, early clinical studies were unable to demonstrate a distinct correlation between EGFR expression and the likelihood of response to EGFR inhibition with targeted antibodies.[107] In addition, studies suggest that immunohistochemistry-based assays measuring EGFR expression do not serve as reliable predictors of response to cetuximab therapy.[108]

Increased response rates after treatment with a TKI have been demonstrated in patients with positive EGFR immunostaining in some studies, but not in others.[104,105,109,110] In multivariate analyses, EGFR expression level was associated with an objective response or adverse prognosis in NSCLC.[93,110] Several investigations into the prognostic significance of EGFR expression revealed no association with survival benefit.[102,105,111] Therefore, EGFR overexpression by itself is not prognostic of survival in NSCLC. It has been suggested that the nonoptimized cut-off value for EGFR-positive immunostaining and/or lack of standardization in staining procedures and guidelines may explain the discordance among these studies.[111]

EGFR Mutation-specific Antibodies

Since the use of EGFR overexpression as a prognostic marker has largely been unproductive, considerable efforts have been made to develop antibodies that react specifically with the mutant form of EGFR. Cell Signaling Technology, Inc. (MA, USA) has developed two mutant-specific antibodies against the most common mutant forms of EGFR: the 15-base pair/5-amino acid deletion (E746-A750del) in exon 19 and the L858R point mutation in exon 21.[112] Yu et al. investigated EGFR genotypes of 40 NSCLC tumor samples by immunohistochemistry with these antibodies and confirmed the immunohistochemistry results by DNA sequencing.[112] Detection of mutant EGFR by these two antibodies was performed by western blotting, immunofluorescence and immunohistochemistry. The sensitivity and specificity of these antibodies in a 340-sample panel of paraffin-embedded NSCLC tumors was 92 and 99%, respectively, compared with direct sequencing and mass spectrometry-based DNA sequencing. These results demonstrate that mutation-specific antibodies provide a rapid, sensitive, specific and cost–effective method to identify lung cancer patients who may respond to EGFR-targeted therapies. Brevet et al. evaluated the two mutation-specific monoclonal antibodies for the detection of EGFR mutations by immunohistochemistry on 218 paraffin-embedded lung adenocarcinomas.[112] The EFGR L858R mutant antibody showed a sensitivity of 95%, a positive predictive value of 99% and a specificity of 76%, with a positive cut-off of (2+).[113] A positive threshold of (2+) will effectively reduce the false-positive rate and enhance the predictive power of immunohistochemistry assays to 100%, with a minimal reduction in sensitivity. The immunostaining scoring was based on cytoplasmic and/or membrane staining intensity as follows: (0+) = no staining or faint staining intensity in <10% of tumor cells; (1+) = faint staining in >10% of tumor cells; (2+) = moderate staining; and (3+) = strong staining. Therefore, immunohistochemistry using mutation-specific antibodies can be used to screen for patients who may be candidates for EGFR inhibitors.[113]

Phosphorylated Form of EGFR

Aberrant activation of EGFR is a recognized component of cancer development and progression.[59] In addition, recent data indicate that both EGFR mutations and the activation status of EGFR, defined by phosphorylation, might have a strong impact on the clinical course of NSCLC.[114] The two major EGFR signaling pathways, PI3K–AKT–mTOR and RAS–RAF–MAPK, mediate EGFR effects on cell proliferation and survival. The activation of these pathways is dependent on the phosphorylation status of the components. Investigations to date indicate that the major molecular alteration involved in the carcinogenesis of NSCLC is an activation mutation. The mechanisms that regulate EGFR expression, such as epigenetic alteration and aberrant transcription factors have been studied but are not yet conclusive.

Phosphorylation at tyrosine 845 in the kinase domain of EGFR may stabilize the activation loop, which maintains the receptor in an active state and provides a binding surface for substrate proteins.[115] Phosphorylation of two additional tyrosines, 1068 and 1173, mediates the direct binding of growth factor receptor-bound protein 2. Furthermore, tyrosine 1068 is involved in the activation of the MAPK signaling pathway.[116]

Detection of activated EGFR is conducted by using anti-phospho-EGFR antibodies directed at EGFR in its phosphorylated state. Phosphorylations in the carboxyl-terminus of EGFR play a key role in the recruitment of signaling molecules and activation of downstream signaling pathways.[115,117] In a study by Endoh et al., involving 97 NSCLC cases, patients with phospho-EGFR-positive tumors demonstrated a prolonged survival, although the follow-up period was relatively short.[117] Hijiya et al. investigated 21 cases of NSCLC for correlations between the presence of EGFR mutations and the EGFR phosphorylation status by immunohistochemistry with antibodies recognizing EGFR that was phosphorylated at tyrosine 992 and tyrosine 1173, respectively.[118] The mutation status of EGFR was strongly correlated with immunoreactivity for phosphorylated tyrosine 992, indicating a clear potential for using anti-phospho-EGFR antibodies as a surrogate marker of EGFR mutations and thus predicting the clinical response to tyrosine antagonist therapy.

The immunohistochemical evaluation of NSCLC with anti-phospho-EGFR antibodies is potentially useful in the clinical prediction of responsiveness to EGFR-targeted therapy. However, further testing and evaluation are needed to determine its true clinical implication.

EGFRvIII

The newly characterized EGFR mutant, EGFRvIII, results from an in-frame deletion of exons 2–7 of the coding sequence, which has been found to be generated by gene rearrangement or aberrant mRNA splicing.[119,120] The variant form has a deletion of 267 amino acids in the extracellular domain of normal EGFR, creating a unique epitope at the fusion junction. A number of functional differences between EGFRvIII and EGFR have been characterized.[120,121] Although EGFRvIII fails to bind EGF, its intracellular TK is constitutively activated, allowing the receptor to undergo tyrosine autophosphorylation.[122,123] These studies provide further evidence that EGFRvIII expressed in NSCLC is phosphorylated and, hence, activated. The data suggest that the sustained activation of EGFRvIII may play a role in the pathogenesis of NSCLC and, therefore, EGFRvIII is a potential therapeutic target for NSCLC.[114] Antibodies directed to this tumor-specific variant of EGFR provide an alternative targeting strategy. It has been demonstrated that systemic treatment of mice bearing tumors expressing EGFRvIII with monoclonal antibodies specific for EGFRvIII inhibited tumor growth and extended animal survival.[124,125] Antibodies that have an affinity for EGFRvIII, but without an affinity to wild-type EGFR, provide an alternative tool for detecting this mutation variant.[126]

The role of EGFRvIII mutations in the pathogenesis of NSCLC is unclear. Reported incidences of EGFRvIII mutation in NSCLC vary from 0 to 42%.[127,128] These differences may be due to differences in the tumor composition (histological type) or to technical considerations, such as the threshold for the result interpretations. Studies using immunohistochemical assays with EGFRvIII mutant-specific antibodies suggest that this mutation is present in multiple other tumor types and is not exclusive to NSCLC.[114,129] However, owing to the large size and complex genomic structure (28 exons spanning ~190 kb) of EGFR and its large intron 1 (123 kb), where genomic deletions frequently occur, it has been difficult to assess and verify the existence of the EGFRvIII mutations at the genomic level.[127] To evaluate the clinical impact of EGFRvIII in NSCLC, Okamoto et al. investigated EGFRvIII expression in 76 cases of NSCLC by immunohistochemistry, using a monoclonal antibody specific for this EGFR variant. EGFRvIII expression was found in 39% (30/76) of NSCLC; however, genetic analysis of EGFRvIII mutations only generated a 3% positive rate compared with the 39% immunopositivity rate.[114] Okamoto et al. found that EGFRvIII was also observed in several normal tissue components of the lung, which raised the question of the clinical implications for this detection methodology.[114]

Studies of small-molecule TKIs have demonstrated clinical responses in NSCLC patients whose tumors bear EGFR kinase domain mutations. However, the efficacy of these inhibitors against tumors with the EGFRvIII mutation remains unclear. Ji et al. determined that EGFRvIII mutations were present in 5% (3/56) of human squamous cell lung carcinomas, but found no EGFRvIII mutations in a large cohort of human lung adenocarcinomas (0/123).[127] In their study, EGFRvIII-bearing tumors seemed relatively resistant to some TKIs, but responsive to others.[130] In an in vivo system, treatment with an irreversible EGFR inhibitor, HKI-272, dramatically reduced the size of EGFRvIII-driven murine tumors within 1 week.[126] A total of 7 days of erlotinib treatment led to an average reduction of 45% in tumor volume in the three treated mice. By contrast, those treated with HKI-272 demonstrated a reduction of 88%. The Ba/F3 cells, transformed with the EGFRvIII mutant, were relatively resistant to gefitinib and erlotinib in vitro, but sensitive to HKI-272, suggesting that TKI treatment is potentially efficacious for cancers harboring the EGFRvIII mutation.

Summary

Current studies of alterations of the EGFR pathway have been focused on gene mutations, gene copy-number alterations, protein expression alterations and downstream genetic alterations.

Four activating mutations – exon 18 (G719A/C), exon 21 (L858R and L861Q), and in-frame deletions in exon 19 – are the dominant mutations present in NSCLCs.

Patients with an EGFR mutation, who were treated with TKIs, had much higher response rates and longer progression-free survival than patients without EGFR mutations who had the same treatment.

Acquisition of a new mutation in exon 20 can confer resistance to TKI treatment.

Overexpression of EGFR has been found in 40–80% of cases, but its usefulness as a predictive marker remains controversial.

Sustained activation of EGFRvIII is implicated in the pathogenesis of squamous cell carcinoma and, thus, EGFRvIII is a potential therapeutic target in this subset of NSCLCs.


EGFR-targeted Therapy & Mechanisms of Resistance

There are two major approaches for inhibiting EGFR signaling: to prevent ligand binding to the extracellular domain with a monoclonal antibody and to inhibit the intracellular TK activity with a small molecule. Use of the latter approach was the first method to be attempted clinically.[131] The EGFR TKIs are reversible competitive inhibitors of the TK domain of EGFR that bind to its ATP-binding site. Somatic activating mutations of the EGFR gene, increased gene copy number and certain clinical and pathological features have been associated with dramatic tumor responses and favorable clinical outcomes with these agents in patients with NSCLC. The majority of these patients inevitably acquire resistance to EGFR TKIs. Recent data indicate that a secondary mutation, such as T790M, expression of HGF, PTEN and/or early growth response-1 and changes in the epithelial-to-mesenchymal transition, were associated with EGFR TKI resistance. Uramoto et al. found that strong expression of HGF was detected in six out of eight specimens with the T790M mutation.[132] Three out of eight cases (38%) demonstrated a loss of PTEN in samples with the T790M mutation. A loss of early growth response-1 was detected in two out of seven cases (29%), including one tumor without PTEN. Four out of seven cases (57%) demonstrated positive expression of phosphorylated Akt. A change in the epithelial-to-mesenchymal transition status between pre- and post-treatment was observed in four out of nine cases (44%). These results suggest that alterations in gene or protein expression can account for all mechanisms by which tumors acquire resistance to EGFR TKIs.[132] This phenomenon suggests the existence of complicated relationships between acquired resistance-related genes.

Somatic activating mutations in EGFR are identified in a subset of NSCLC that responds to TKIs. As noted previously, acquisition of drug resistance has been linked to a specific secondary somatic mutation, EGFR T790M. Bell et al. described a family in which multiple members developed NSCLC associated with germline EGFR mutations of T790M.[25] These observations implicate altered EGFR signaling as a culprit in the genetic susceptibility to lung cancer in families with an increased incidence of NSCLC.

We propose an algorithm for molecular testing for patients with NSCLC (Figure 4). A stepwise approach, based on the frequency of specific mutations, is used to assess lung cancer patients for specific findings, which will allow proper therapeutic stratification for targeted therapy.


Figure 4.

Suggested algorithm for molecular testing for patients with non-small-cell lung cancer. A stepwise approach is used to test lung cancer patients according to the known frequencies of various mutations. Small-cell lung cancers are excluded from testing. NSCLC, which accounts for approximately 85% of all lung cancers, is tested for the presence of EGFR mutations. A positive test, found in approximately 20% of Caucasians and 40% of East Asians, predicts an 80% probability of response to EGFR TKI therapy. Nonmutated EGFR is found in approximately 80% of Caucasians and 60% of East Asians. These patients are further tested for EML4–ALK mutations. EML4–ALK mutations are found in only 3% of patients with NSCLC, but the mutation predicts a 53% probability of response to targeted therapy. Cases lacking EML4–ALK mutations may undergo additional testing.

EGFR: EGF receptor; Mu: Mutation; NSCLC: Non-small-cell lung cancer; SCC: Small-cell carcinoma; TKI: Tyrosine kinase inhibitor.

EGFR-targeted Therapy Approaches

Monoclonal Antibodies Monoclonal antibodies, such as cetuximab and panitumumab, are either chimeric mouse–human or fully humanized antibodies targeting the extracellular domain of EGFR and thereby inhibiting the binding of activating ligands to the receptor. This class of treatment inhibits ligand-dependent activation of EGFR and inhibits the downstream pathways, which cause cell cycle progression, cell growth and angiogenesis (Figure 3A). In addition, binding of the antibody initiates EGFR internalization and degradation, which leads to signal termination.[108,133] Fully humanized antibodies such as panitumumab, have a high affinity for EGFR and a longer half-life.[134] Although EGFR is frequently expressed in patients with NSCLC, the clinical efficacy of treatment with anti-EGFR antibodies is limited to only a subset of patients.

Tyrosine kinase inhibitors Tyrosine kinase inhibitors are synthetic small molecules that block the magnesium–ATP-binding pocket of the intracellular TK domain.[108] Several TKIs, such as gefitinib and erlotinib, are specific for EGFR, whereas others inhibit other receptors in addition to EGFR, such as HER2 and VEGF receptor 2. TKIs block ligand-induced receptor autophosphorylation by binding to the TK domain and disrupting TK activity, thereby abrogating intracellular downstream signaling (Figure 3B & Table 1).

Mechanisms of Resistance to EGFR-targeted Therapy

Acquired Resistance Caused by a Secondary Mutation Although EGFR mutations are associated with enhanced sensitivity to gefitinib and erlotinib, not all tumors that have activating mutations are associated with an enhanced response. The efficacy of EGFR TKIs is limited owing to either primary or acquired resistance after therapy. Most patients who initially respond to gefitinib and erlotinib eventually become resistant and experience progressive disease.

It is known that four mutations result in TKI drug sensitivity: point mutations in exon 18 (G719A/C) and exon 21 (L858R and L861Q), as well as in-frame deletions in exon 19, which eliminate four amino acids – leucine, arginine, glutamic acid and alanine – downstream of the lysine residue at position 745.[67,69–72] However, insertion mutations of exon 20 at D770–N771 were associated with EGFR TKI resistance.[60,135] This observation was confirmed in an in vitro model in which insertion mutations in exon 20 rendered transformed cells less responsive to EGFR TKIs compared with the sensitizing mutations of exons 19 and 21.[135]

Two established mechanisms of acquired resistance consist of additional mutations in the EGFR gene acquired during the course of treatment that change the protein coding sequence and amplification of other oncogene signaling pathways.[85,136–138]

Kobayashi et al. reported a gefitinib-resistant advanced NSCLC patient who had a relapse after 2 years of complete remission due to treatment with gefitinib.[86] The DNA sequence of EGFR at relapse revealed the presence of a second point mutation, resulting in a T790M mutation of EGFR. Structural modeling and biochemical studies showed that this second mutation led to gefitinib resistance.[86] The same mutation was confirmed by Pao et al. through molecular analysis of EGFR in patients with acquired resistance to gefitinib or erlotinib.[139] The gefitinib-resistant cases contain the same secondary mutation (T790M) in the kinase domain.[22] Codon 790 of EGFR is considered to be the 'gatekeeper' residue, which is an important determinant of inhibitor specificity in the ATP-binding pocket of EGFR.[108] Substitution of this residue in EGFR with a bulky methionine may cause resistance by steric interference with the binding of TKIs, including gefitinib and erlotinib.[86,139,140] This mutation may confer a survival advantage to the tumor and is probably selected for while the patient is receiving anti-EGFR TKI treatment.[25,84] These findings have led to the development of irreversible EGFR TKIs in an effort to effectively target this mechanism of resistance.[140]

The role of oncogenic activation of EGFR downstream effectors, such as KRAS, BRAF, PIK3CA and PTEN, in response to therapy is discussed extensively in a series of studies.[47,53] The RAS–MAPK and PI3K–AKT pathways are major signaling networks linking EGFR activation to cell proliferation and survival.[141,142] Mutations in these downstream effectors of EGFR signaling could lead to resistance to EGFR inhibitors.[136–138] The discovery of molecular aberrations, such as MET kinase amplification or mutations of EML4–ALK fusion, which causes constitutive activation of RAS–RAF–MEK, has provided further insight and validation into factors limiting the therapeutic efficacy of EGFR inhibitors.[46,143,144]

KRAS Mutations KRAS plays a key role in the EGFR signaling network. The KRAS proto-oncogene encodes KRAS G-protein, which plays a critical role in the RAS–MAPK signaling pathway downstream of many growth factor receptors, including EGFR.

One of the most important discoveries for the clinical management of colorectal carcinoma has been the association of mutations in KRAS and the efficacy of monoclonal antibodies targeting EGFR, such as panitumumab and cetuximab. Some tumors harbor somatic mutations in exon 2 of KRAS that compromise the hydrolysis of RAS-bound GTP to GDP, resulting in constitutive activation of the RAS pathway.[145] In the presence of a KRAS mutation, EGFR pathway activation cannot be significantly inhibited by cetuximab or panitumumab, which acts upstream of the KRAS protein, diminishing the efficacy of the agents.

An activating mutation of KRAS is present in 15–30% of NSCLC cases[26,146,147] and accounts for approximately 35–45% of TKI-nonresponsive cases.[148] Approximately 30% of lung adenocarcinomas contain activating KRAS mutations, which are associated with resistance to EGFR TKIs.[22] It is noteworthy that the presence of a KRAS mutation is common in NSCLC, but the occurrences of KRAS and EGFR mutations seem to be mutually exclusive.[4,27,149–152] EGFR and KRAS mutations are rarely if ever detected in the same tumor, suggesting that they may perform functionally equivalent roles in lung tumorigenesis.[58,153] However, there is growing evidence that coexistence of EGFR and KRAS mutations is possible,[27,151,154] although the frequency is low. Due to the limited number of cases, it is difficult to obtain conclusive results; however, the available data suggests a negative association between EGFR/KRAS mutation and EGFR TKI responsiveness.[27,151,154]

It remains unclear whether assessment of KRAS mutation status will prove to be clinically useful with regard to anti-EGFR therapy.[50] Although an association between the presence of a KRAS mutation and lack of response to EGFR TKIs has been observed, it remains indeterminate whether this association is clinically relevant with respect to progression-free and overall survival. Investigations of KRAS mutation status as a negative predictor of outcome after anti-EGFR therapy have been undertaken, but small sample sizes due to low prevalence of KRAS mutations have limited the power of such studies. Some investigators have reported that KRAS mutation is a negative predictor of response to anti-EGFR monoclonal antibodies and also an important mechanism of resistance to TKIs in NSCLC.[26] Unlike colorectal cancer, KRAS mutations do not seem to identify patients who do not benefit from anti-EGFR monoclonal antibodies in NSCLC.

KRAS mutations are almost exclusively detected in codons 12 and 13 of exon 2, which may result in EGFR-independent intracellular signal transduction activation. In a study by Eberhard et al., EGFR exons 18–21 and KRAS exon 2 mutations were investigated via sequencing in tumors of 274 patients.[27] KRAS mutations were present in 21% of tumors, which were associated with significantly decreased TTP and survival in patients treated with erlotinib plus chemotherapy. Others have reported that KRAS mutation status did not impact EGFR TKI therapy.[28] In a study that included 223 chemotherapy-naive patients with advanced NSCLC treated with erlotinib or gefitinib monotherapy, EGFR mutations were associated with a 67% response rate. Wild-type EGFR was associated with poorer outcomes, regardless of KRAS mutation status.[28]

A study by Wang et al. utilizing PCR-restriction fragment length polymorphism analysis investigated the KRAS mutations in codons 12 and 13 in 273 NSCLC cases.[155] Of the 120 patients who received EGFR TKI therapy, only 5.3% (one out of 19) of the patients with a KRAS mutation demonstrated a response compared with a 29.7% response rate for patients lacking a KRAS mutation.[152] Furthermore, the median progression-free survival time of patients with a KRAS mutation was 2.5 months compared with 8.8 months for patients with wild-type KRAS.

A meta-analysis by Linardou et al. provided empirical evidence that somatic mutations of the KRAS oncogene are highly specific negative predictors of response to single-agent EGFR TKIs in advanced NSCLC.[148] Among 17 publications, 165 out of 1008 (16%) NSCLC patients presented with KRAS mutations. The presence of KRAS mutations was significantly associated with an absence of response to TKIs in these patients.

Having an intact KRAS is necessary, but not sufficient, to derive benefit from EGFR inhibition, and additional mechanisms of resistance to EGFR inhibitors exist.

KRAS testing scenarios in the management of NSCLC are summarized in Figure 5. The incidence of KRAS mutations in NSCLC is reportedly up to 20%.[156] In the subset of tumors with KRAS mutations, less than 3% also contain an EGFR mutation, and the remaining 97% of tumors with KRAS mutations have wild-type EGFR. Both KRAS/EGFR double mutations and wild-type EGFR are associated with nonresponsiveness to EGFR-targeted therapy.

Figure 5.


KRAS testing scenarios in the management of non-small-cell lung cancer. NSCLC accounts for approximately 85% of all lung cancers. The incidence of KRAS mutations in NSCLC is reportedly up to 20%. In the subset of patients with KRAS mutation, less than 3% of the tumors also contain an EGFR mutation; the remaining 97% have wild-type EGFR. Both KRAS/EGFR double mutations and wild-type EGFR are associated with nonresponsiveness to EGFR-targeted therapy. Of the 80% of NSCLCs that do not have KRAS mutations, approximately 20% harbor EGFR mutations, which are associated with an 80% likelihood of response to EGFR TKI therapy.

EGFR: EGF receptor; Mu: Mutation; NSCLC: Non-small-cell lung cancer; TKI: Tyrosine kinase inhibitor.

lung without KRAS and EGFR mutations.[160] Of these, ten adenocarcinomas with the BRAF-V600E mutation were identified. BRAF mutations were reported more frequently in micropapillary lung adenocarcinoma.[161] De Oliveira Duarte Achcar and colleagues analyzed the clinical and molecular profile of 15 primary micropapillary adenocarcinomas and found BRAF mutations in three cases (20%). The BRAF-V600E mutation-bearing tumors had a slight female predilection (6:4 female:male). The elderly patients were found to have a greater than expected incidence of intralobar satellite nodules and N2 node involvement.[161] The adenocarcinomas were largely of mixed type, with a high incidence of papillary (80%) and lepidic growth (50%). However, due to the relatively small sample size, it is yet to be determined whether BRAF mutant tumors represent a distinct subset of lung adenocarcinoma.[162]

Mutations in BRAF have been shown to impair responsiveness to panitumumab or cetuximab in patients with colorectal carcinomas. This initial retrospective work was performed on a cohort of 132 patients.[163] The results showed that none of the patients who experienced a response displayed BRAF mutations, whereas 11 of 79 nonresponders (14.0%) carried a BRAF-V600E allele.

Nevertheless, BRAF mutations are rarely detected in NSCLC when compared with KRAS mutations.[162,164,165] A total of 80% of the reported mutations are located within the kinase domain of BRAF.[159] Brose et al. identified activating BRAF mutations in five out of 292 cases (1.7%) of NSCLC. Among these mutations, three were found in exon 11 and two in exon 15.[164] It has been proposed that mutations in BRAF, a downstream signaling molecule of EGFR, predict clinical response to EGFR inhibitors, but this has yet to be validated in a larger number of cases.[157] Notably, a single substitution of glutamic acid for valine at codon 600 (V600E) accounts for approximately 90% of the BRAF missense mutations found in human tumors.[159] Considering BRAF is a serine/threonine kinase that is commonly activated by a somatic point mutation in human cancer, it may provide new therapeutic opportunities in a subset of NSCLC.

ALK Rearrangement ALK encodes a TK receptor found in a number of fusion proteins consisting of the intracellular kinase domain of ALK and the amino terminal portions of different genes.[166,167] A subset of NSCLC cases harbor a transforming fusion gene, EML4–ALK, within the genome. To date, seven gene fusion variants have been reported in NSCLCs. All involve the intracellular TK domain of ALK starting at a portion encoded by exon 20.[168] This fusion is formed as the result of a small inversion within the short arm of chromosome 2 that joins EML4 to ALK, inv(2)(p21;p23), which encodes an activated TK protein.[169,170] Several other transforming EML4–ALK fusion gene variants have also been identified, involving various EML4 exons and ALK.[171,172] ALK can also fuse with some other rare fusion partners, such as KIF5B and TFG.[173]

Activated ALK is involved in the inhibition of apoptosis and the promotion of cellular proliferation through activation of downstream PI3K–AKT and MAPK signaling pathways.[174] The EML4–ALK fusion is a rare abnormality detected in approximately 6% of patients with NSCLC.[144,169,175] Fusion of the EML4–ALK gene and its associated EML4–ALK product may lead to constitutive activation of the RAS–RAF–MEK–MAPK pathway.[47] In addition, two other less frequent ALK fusions in lung cancer have been reported.[176]

Non-small-cell lung cancer cases harboring EML4–ALK are characteristically found to have wild-type EGFR, as well as wild-type KRAS.[177,178] In addition, patients with these tumors tend to be younger, have an advanced clinical stage at presentation, have never smoked and their tumors exhibit solid histology, often with a component of signet ring cells.[178,179]

Patients with this alteration demonstrated an extraordinary response to the MET–ALK inhibitor, PF-02341066, in a Phase I/II trial.[144] Ten out of 19 patients (53%) experienced objective responses. A total of 15 out of 19 patients (79%) demonstrated disease control at 8 weeks, lasting as long as 40 weeks in some. Only four patients demonstrated disease progression. Kwak et al. screened 1500 NSCLC patients and identified 82 patients with advanced ALK-positive disease.[180] The vast majority (96%) of these cancers were adenocarcinomas. A total of 94% of these patients had received at least one prior therapy. All of these patients were treated with crizotinib, an oral small-molecule ALK TKI. The disease control rate was 90%, which included a 57% response rate plus 33% with stable disease. Furthermore, Rodig et al. evaluated the incidence and the characteristics of ALK-rearranged lung adenocarcinomas within the western population to elucidate the optimal diagnostic modality to detect ALK rearrangements in routine clinical practice.[179] In their study, 358 lung adenocarcinomas were tested for ALK rearrangement by FISH and immunohistochemistry. ALK rearrangement again demonstrated an association with younger age, never having smoked, advanced clinical stage, and a solid histology with signet ring cells in some cases. Of note, none of the ALK-rearranged tumors harbored coexisting EGFR mutations. The results of this study demonstrate that ALK rearrangements are uncommon in the western population, but represent a distinct clinical entity with unique attributes and the possibility of distinct treatment options. For suspected cases, dual diagnostic testing with FISH and immunohistochemistry should be considered in order to accurately identify lung adenocarcinomas with ALK rearrangement.[179] A study by Zhang et al. involving 266 Chinese patients with NSCLC revealed approximately similar results.[178]

Shaw and colleagues investigated 141 NSCLC cases and found that 19 (13%) contained EML4–ALK fusion, 31 (22%) demonstrated EGFR mutantations and 91 (65%) were wild-type for both.[144] Several studies provide consistent evidence that EML4–ALK and EGFR mutations are mutually exclusive.[144,169,177,181] However, Tiseo et al. reported a 48-year-old Caucasian man who had never smoked and who was diagnosed with NSCLC with a concomitant EGFR mutation and ALK translocation that was resistant to erlotinib therapy.[182] This may be representative of the nature of this subgroup of NSCLC.[154]

No EGFR mutations in the EML4–ALK cohort and no instances of ALK rearrangement in the EGFR cohort have been found.[144] Among patients with metastatic disease, EML4–ALK rearrangements were associated with resistance to EGFR TKI treatment.

Compared with the EGFR-mutated and wild-type EGFR/ALK cohorts, patients with EML4–ALK fusion gene-positive tumors were significantly younger and more likely to be male.[144] However, another study demonstrated that males and females are equally affected.[139] Patients with EML4–ALK-positive tumors are more likely to be never/light smokers. A total of 18 of the 19 EML4–ALK-positive tumors were adenocarcinomas, predominantly of the signet ring cell subtype.[144] EML4–ALK defines a molecular subset of NSCLC with distinct clinical characteristics. Specifically, patients who harbor this mutation do not benefit from EGFR TKIs and should be directed to trials investigating ALK-targeted agents.

Summary

Two classes of anti-EGFR agents – monoclonal anti-EGFR antibodies and small-molecule EGFR TKIs – are currently used in EGFR-targeted therapy.

Additional mutations in EGFR are an important mechanism of acquired resistance to EGFR-targeted therapy.

Some mutations, such as exon 20 D770–N771 and T790M, are associated with EGFR TKI resistance.

Oncogenic activation of EGFR downstream effectors, such as KRAS, BRAF, PIK3CA and deletion of PTEN, is associated with resistance to TKI therapy.

KRAS mutation accounts for approximately 35–45% of TKI-nonresponsive NSCLC patients.

KRAS and BRAF mutations, as well as ALK rearrangements, are mutually exclusive with EGFR mutations.

ALK rearrangements are more common in younger patients who have never smoked, and their tumors often exhibit solid architecture, signet ring cell histology and wild-type EGFR and KRAS.

Conclusion

The EGFR is an effective therapeutic target in treating NSCLC. EGFR mutations have been used as a selection criterion for EGFR TKIs and are also used as a predictive marker for responsiveness to EGFR-targeted therapy. However, evidence is beginning to demonstrate that NSCLC may be composed of multiple subsets of tumors, each with its own molecular abnormalities.

Identification of the relevant molecular subtypes of this heterogeneous disease and selecting patients for the appropriate targeting agents is critical in the personalized therapy of NSCLC. KRAS/BRAF mutations, which are mutually exclusive with EGFR mutations, are rare in NSCLC but may be important mechanisms in the etiology and prediction of resistance to EGFR TKI therapy.

In conclusion, one unifying predictive model does not apply to all tumor types, and the larger goal of discovering a predictive marker to guide patient selection for EGFR-targeted therapy remains elusive. EGFR alteration markers need to be further evaluated in combination with clinical data to provide clear rationales for future therapeutic strategies in the treatment of NSCLC.


“人类只有经历过悲哀才能够知道什么是快乐,你只有经过死亡以后才知道重生意味着什么,上帝给人留下了四个字,那就是希望和等待。”

发表回复

您需要登录后才可以回帖 登录 | 立即注册

本版积分规则

  • 回复
  • 转播
  • 评分
  • 分享
帮助中心
网友中心
购买须知
支付方式
服务支持
资源下载
售后服务
定制流程
关于我们
关于我们
友情链接
联系我们
关注我们
官方微博
官方空间
微信公号
快速回复 返回顶部 返回列表