• 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]
214064 163 荷花池荒岛 发表于 2014-3-24 12:31:48 |
荷花池荒岛  硕士一年级 发表于 2014-5-14 06:11:41 | 显示全部楼层 来自: 美国

养生篇

本帖最后由 荷花池荒岛 于 2014-9-15 09:06 编辑

1)
研究:打坐冥想25分钟 持续三天即可减压

近年来,打坐冥想在西方社会正兴起一股潮流。一项最新研究显示,打坐冥想能有效纾缓压力,带来健康益处。每天只需25分钟,连续三天就可见效。过去也有研究发现,打坐冥想可以减轻5~10%的焦虑症状,改善10~20%的忧郁情形,与抗抑郁药提供的效果类似。
据《每日邮报》7月4日报导,这项研究主导人、卡内基美隆大学迪特里希学院(Dietrich College)人文和社会科学系心理学副教授克雷斯韦尔(J. David Creswell)表示,越来越多的人认识到打坐冥想可减轻压力,但很少人知道需要多少时间,才达到效果。

在这项研究中,克雷斯韦尔和研究团队对66名18-30岁健康人士进行为期三天的实验。 一组受试者连续三天进行25分钟的打坐冥想。第二组受试者则完成为期三天的认知训练课程,被要求批判性地分析诗歌,努力提高解决问题的能力。

最后,所有受试者被要求在表情严肃的评估者面前,完成紧张的演讲和数学任务。 每个人需报告他们在完成演讲和数学任务时的压力水平,并提供唾液样品进行皮质醇测定,皮质醇又被称为“压力荷尔蒙”。

结果显示,那些进行打坐冥想的受试者在演讲和完成数学任务时,压力明显低于另一组人。表明打坐冥想可培养处理心理压力的应变能力。更有趣的是,在生理方面,打坐冥想者表现出更大的皮质醇反应。该研究报告发表在《心理神经》杂志上。
压力过大会导致人的身体出现高血压、抑郁症、不育甚至衰老等等。打坐冥想其实是一个自然轻松的过程,能使你达到舒适安静的机敏状态,并且能恢复身体的自我修复和自我平衡的机制。

打坐冥想和人体生命科学的奥秘

美国哈佛医学院的心理医生JohnDenninger正在主导一项研究,关于东方古老的修炼方式,对长期处于高压下的人们的基因和脑部活动有何影响。他发表的一项研究结果证明,一些身心技巧可以使一些与压力和免疫系统功能有关的基因打开或者关闭。

东方古老的修炼方式,对长期处于高压下的人们一些身心技巧可以使一些与压力和免疫系统功能有关的基因打开或者关闭。(Fotolia)
John Denniger是哈佛医学院的教学医院之一麻塞诸塞州总医院身心医学研究所研究主任。他说:“这确实有生理的影响,当你打坐冥想的时候,对整个身体都有效果,而不只是大脑。”

在这项研究中,Denniger把210名有长期压力困扰的研究对像分为三组,每天分别进行20分钟的活动:

第一组70个人做一种叫Kundalini的瑜伽,第二组70人打坐冥想,第三组70人听减压教育的音讯书籍。研究结果今年5月发表在医学杂志《PloS One》上,一段令人放松的活动练习,能够增强与能量代谢和胰岛素分泌有关基因的表达、抑制炎症反应和压力基因的表达,即使对于从未练过的新手也有如此效果。

现在许多西方知名企业都竞相引进冥想、瑜珈及静坐等观想(mindfulness,或译正念减压)活动,作为员工纾缓工作压力及激发创新观念的方法。热衷打坐冥想的高盛集团和艾克森美孚公司董事会成员Bill George,和喜剧演员Jerry Seinfeld,新闻集团董事长Rupert Murdoch也都在推特上也说,想尝试打坐冥想。

打坐冥想有助治疗注意力缺陷

计划不周、走神以及难以抑制冲动是认知控制能力出现了问题。现在,越来越多的研究表明,通过打坐冥想强化“心智肌肉”,可以帮助儿童和成年人应对其注意缺陷多动障碍(ADHD)和注意力缺陷障碍(ADD)。

研究表明,通过所谓的正念冥想强化“心智肌肉”,可以帮助儿童和成年人应对其注意缺陷多动障碍(ADHD)和注意力缺陷障碍(ADD)。

虽然大部分青少年ADHD患者可从第一年的治疗中受益,但这些效果通常在第三年或更早的时候就开始逐渐减弱。加州大学欧文分校(University of California, Irvine)心理学家詹姆斯•M•斯旺森(James M•Swanson)说,“然而,打坐冥想似乎能对可减少ADHD活动的那部分脑区加以训练。”

一般认知控制能力从4至12岁左右开始逐渐增强,然后就进入平台期,冲动水准在16岁左右会达到顶峰,大多数人在20多岁的时候会达到成年人水准。健康成年人在七、八十岁时会开始明显减弱,体现出无法记住名字或单词。

现在,专家们提出,强化认知控制这种精神能力可能对治疗ADHD和ADD特别有帮助。加州大学三藩市分校(University of California, San Francisco)的神经科学家亚当•加扎利(Adam Gazzaley)博士说,冥想是一种认知控制锻炼,它可以增强“人对内部干扰进行自我调节的能力”。

《临床神经生理学》(Clinical Neurophysiology)上最近一份报告显示,成年ADD患者可从正念训练加认知治疗中受益,他们在精神功能方面的改善与服用药物的受试者相当。加州大学伯克利分校(University of California, Berkeley)发展精神病理学专家史蒂芬•欣肖(Stephen Hinshaw)表示,探索正念冥想等非药物干预效用的时机已经成熟。

CaptureDZ01.PNG CaptureDZ02.PNG


2)
美营养师推荐:“7对食物黄金搭档”抗癌

各种食物皆有其多元的营养成分和特有功效,有时单独食用并无显著效果,但如果与其它食物相互搭配食用,不但味道更佳,还能促进有益成分的吸收,更好地发挥抗癌保健的效果。

绿茶●柠檬
绿茶中抗氧化剂儿茶酚可迫使癌细胞自我毁灭,降低心脏病和脑中风危险,还能快速燃烧脂肪。美国《食品科学》杂志刊登一项近期研究发现,绿茶中加入柠檬的维生素C能帮助人体吸收3倍的儿茶素,且避免过早被消化系统分解。

柠檬绿茶可渗透到细胞,将细胞内因新陈代谢产生之毒素及废料予以清除,使血液纯净,并将体内蕴含之杂质毒素如:铅、汞、重金属、辐射物、铜酸、农药、尿酸、酒精等对人体有害之物质排出体外。

鸡蛋●牛奶
牛奶富含高密度蛋白质,氨基酸最全面,维生素与矿物质含量最高,搭配含有维生素D的鸡蛋(特别是蛋黄),可促进人体对钙质的吸收,进而改善骨骼和心脏健康。

圣路易斯大学(Saint Louis University)吉儿‧范德华(Jillon Vander Wa)博士研究称,短时间内,鸡蛋配牛奶的早餐组合,除了能提供蛋白外,也能增加饱足感,更有助控制食欲。

番茄●橄榄油
番茄中的番茄红素是预防癌症、心脏病、衰老的主要功臣。番茄红素比其他种类的胡萝卜素和维生素E来得高,已被证实有预防多种癌症的效果。

《自由基生物学与医学》杂志刊登的一项新研究显示,番茄红素具有脂溶性,与橄榄油或鳄梨一起食用,可使人体吸收更多番茄红素。而且是脂溶性,所以食用时加入油脂,会使肠道更容易吸收,比纯喝番茄汁来的有用。而橄榄油含较多单元不饱和脂肪酸,比较不容易受热变质,另外具有降血压、防癌等功能。

国外有研究称,将番茄和橄榄油一起烹煮,食用一定的量一周后,就会使血液中番茄红素的抗氧化效力升高,防癌效果更好。

大蒜●鱼肉
大蒜可以防治动脉硬化、降低血脂、预防心肌梗塞,具有降脂抗凝作用,还可防止脑血栓的形成。日本在研究功能食品中,发现多种鱼肉含有血压升高抑制肽,有显著降血压的作用,且能预防动脉硬化,有多种有益心脏健康的微量元素,并具有降脂抗炎功效。

《美国临床营养学杂志》刊登一项研究发现,鱼肉与大蒜搭配,可全面降低总胆固醇和坏胆固醇(LDL)水准。因此,烹调鱼时加入大蒜更美味健康。

巧克力●苹果
将苹果沾上融化的黑巧克力食用,美味健康又防癌。(fotolia)

美国心脏病专家霍利•格莱恩杰博士表示,黑巧克力含有丰富的抗氧化剂黄酮醇,其中儿茶酸与茶中的含量一样多,儿茶酸能增强免疫力,预防癌症。苹果有维生素C、β-胡萝卜素、胡萝卜素、茄红素、维生素E、有机酸,使细胞不易癌化,免于受到活性氧化伤害,进而预防癌症的发生,而苹果皮与皮下存有多数抗癌物质,若削皮则降低功效。将苹果沾上融化的黑巧克力食用,美味健康。

咖哩●胡椒
咖哩成分之一的姜黄粉具有抗氧化成分,有助于抗击癌症、糖尿病、预防心血管疾病和认知障碍症等多种疾病。美国约翰霍普金斯大学医学院杰迪洛博士的研究也发现,其中的姜黄和槲黄素,可以有效地减少肠息肉。而黑胡椒中的辛辣物质胡椒碱,可使人体对咖哩中的活性物质姜黄素的吸收率提高1000倍,在烹煮食物同时使用两种调料,抗炎、抗癌效果更佳。

燕麦●奇异果
人体的铁如果不足,将会造成疲倦、降低免疫力和血压降低,甚至还会影响记忆力和学习能力。研究显示,来自肉品的血红素铁容易被人体吸收,而谷物和各种蔬菜的非血红素铁则较难被吸收利用,若能与维生素C并用,会有助于人体对铁的吸收。

奇异果含有丰富的维他命C,且被人体吸收利用率高达94%,可以提高免疫力,阻断致癌因子“亚硝酸胺”的形成。而燕麦含有丰富的维生素和矿物质,对于预防骨质疏松症、帮助伤口愈合皆有成效,同时有预防贫血的功用,也可补充奇异果所缺乏的维生素B6。


3)
你吃过吗?苹果熟吃养生有奇效

苹果不仅模样讨人喜欢,营养也多样化,既不贵又香脆可口,可谓自然界的好水果。但苹果一般都生吃,对于保护其水溶性维生素来说是最好的途径。而煮苹果吃,一般人家很少这样做,但吃煮熟的苹果确实有独特的养生功效。

苹果的营养有哪些?据台湾行政院卫生署食品卫生处公布,苹果的植物性化学物质有胡萝卜素、鞣酸、苹果酸、柠檬酸、硒石酸、枸橼酸、丹宁酸、苹果香醇(香气)。在红色及紫红色的苹果中,含有山茶酚及懈皮素(两者存在于果皮内)、植物性凝血素、β-胡萝卜素、茄红素。

另外,美国康乃尔大学的研究发现,100克新鲜的苹果所含的抗氧化元素相当于1500mg的维生素C。科学家将苹果萃取物用于抑制实验鼠的癌细胞,其效果可达57%。

苹果煮熟吃 抗菌效果好
研究发现,苹果加热后,苹果内所含的多酚类天然抗氧化物质含量会大幅增加。这类物质不仅能降血糖血脂、抑制自由基而抗氧化、抗炎杀菌,还能抑制血浆胆固醇升高。

中国民间利用熟苹果治疗腹泻非常普遍。而苹果中富含的果胶,又是一种能够溶于水的膳食纤维,不能被人体消化。果胶能在肠内吸附水分,使粪便变得柔软而容易排出。果胶还具有降低血浆胆固醇水平、刺激肠内益生菌群的生长、消炎和刺激免疫的机能。

另外,熟苹果所含的碘是香蕉的8倍,是橘子的13倍,因此熟苹果也是防治大脖子病的最佳水果之一。

熟苹果的养生功效
1、苹果金橘饮:苹果300克、金橘15个、胡萝卜100克,洗净切碎榨汁入锅煮沸,调入蜂蜜即可。具有健脾和胃、理气化痰的功效。

2、苹果山药汤:苹果500克,山药30克,麦芽30克,芡实10克。苹果洗净切块,山药、麦芽、芡实洗净与苹果一同入锅,加适量水,大火煮沸后改小火熬煮1个半小时。早晚温服。有益脾胃、助消化、止腹泻的功效。

3、苹果煲猪肉:苹果若干个切成块,带皮猪肉一块,花生若干,桂圆肉若干,除猪肉外,其余材料一起放,若体质怕寒,可放陈皮一块,至水开后放猪肉,煲至一个小时左右即可。有清心润肺温胃的功效,特别是在干燥的天气喝,对皮肤有滋润的作用。

4、煮苹果:将苹果连皮切成六至八瓣,放入冷水锅内煮,待水开后,将苹果取出,连皮吃下。每天一次,每次一个,连吃7~10个可治愈。可防治嘴唇生热疮、牙龈发炎、舌裂等内热现象。

5、煮鸡肉咖哩或炖牛肉加入苹果,能增添食材香气又能养生可谓一举双得。

苹果醋 让你内外皆美:
用醋泡苹果是很特殊的吃法,醋可以平衡体质酸碱度,醋把苹果的营养与香气释放出来。

苹果醋做法:把苹果数个洗净(视容器大小决定苹果数量),去芯,连皮切成薄片,一层苹果、一层冰糖(或蜂蜜)放入容器内(容器最好用玻璃瓶)。把酿造米醋倒入容器中淹过苹果,封盖置于阴凉处一个月后即可加水稀释饮用。

苹果醋有以下几种功效:
1、泡脚:用苹果醋泡脚可舒缓肿胀的脚,减轻疼痛感。对于孕妇,跑步者,高跟鞋爱好者而言,效果立竿见影。

2、防脱皮:出外爬山或到海边晒太阳,回家后皮肤被晒得发红,可以用干净的棉布蘸稀释的苹果醋在发红皮肤处轻擦,可减少两天后的脱皮现象。

3、防粉刺:苹果醋有消炎作用。棉花球沾几滴稀释苹果醋,擦在额头和鼻子的T区或者其他容易干燥的地方,有利于防止粉刺。如皮肤较敏感无法适应醋的酸性,擦后脸部如有红肿现象需即刻用清水洗净。

4、沐浴润肤:泡澡时加入两茶匙的苹果醋有助身体排毒,对皮肤有滋润和亮泽的效果。

5、洗发可护发:用苹果醋加一点小苏打粉自制天然洗发剂,洗后头发会变得更有光泽,可防头发毛躁,刺激头发生长。

6、防口臭:苹果醋稀释漱口不仅能去除牙菌斑,还能预防口臭。



“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 2014-5-27 03:02:34 | 显示全部楼层 来自: 美国
SUNRONG302 发表于 2014-5-26 19:48
我刚才不知在哪里看见你说不忌糖可以防脑转,这个论点太新奇。以前听说美国做过试验,癌细胞会抢着吃糖,所 ...

阿姨,您好,

是我在Belinda家的治疗贴里说的。

我母亲生病以后,我就四处打听经验。不能吃糖还是大多数人的意见,现实中好像还没有听到谁反对过。

比起正常细胞的新陈代谢,癌细胞的新陈代谢据说需要更多的能量。血液中的葡萄糖就是提供这个新陈代谢所需要的能量的主要来源之一。从我看的资料,总结出来的是血液中的葡萄糖高是不好的,不仅促进癌细胞的繁殖,还会有一些其它的对人体较为负面的影响。我们吃糖是会造成血液中的葡萄糖升高的。

我看到的一个观点是, 如果癌细胞得不到更多的能量,它会游离到脑部,因为脑部的葡萄糖含量高。怎么说呢,我只能说我不排斥这样的解释,因为我认为癌细胞的适应能力很强,这种游离是有可能的。但是癌细胞想游离也不见得就能够游离,得有载体才行。所以还要取决于其它的条件和所处的环境。而且有人肯定也会问,如果脑部的葡萄糖含量高,为什么癌细胞会向骨、肝、肾转移,对不对? 所以,我想这个只能是一个说法,不能一刀切。

那么要不要吃糖呢?我个人感觉是,在没有发现脑转移前,少量食糖或含糖量高的食物可能还是可以考虑的,不一定一点糖不沾。

我在说上面的那个看到的观点时,底气是不足的,原因是有些人在发现肺有问题的同时就已经脑转了。从肺有问题到脑转的时间段里,这些人的饮食习惯不会有主动性的改变,假设他们当中有人有吃糖的习惯。那么肺原病灶出现、吃糖、脑转,就是上面那个观点的反证。

不管怎么样,加上您家,已经有4家了。我以后仍然会留意这类的例子,会记着有过这样一个观点。

希望阿姨的治疗顺顺利利! 谢谢。




“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 23:52:02 | 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。”

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