Local Ablative Therapy药理药效研究 动物模型
- 格式:pdf
- 大小:341.71 KB
- 文档页数:8
功效性动物模型研究及其应用随着科学技术的发展,人们越来越注重动物模型在医学领域的应用。
在动物模型研究中,最重要的一点是其功效性。
本文将讨论功效性动物模型的研究及其在医学领域的应用。
1. 功效性动物模型的定义
功效性动物模型是一种能够模拟疾病模型的动物模型。
它可以被用来评估各种药物和治疗方法。
该模型具有以疾病病理学和生理学作为基础的生物学特征,能够准确地反映疾病的生物学特征和其反应。
2. 功效性动物模型的应用
2.1 药物研发
在药物研发中,功效性动物模型是一种重要的工具。
通过使用该模型,药物的安全性和有效性可以迅速进行评估。
功效性动物模型还可以使用在新药物的毒性实验中,以确保新药物不会产生负面影响。
2.2 疾病研究
功效性动物模型可以用来研究某些疾病的机制。
例如,糖尿病、肿瘤和心脏病等疾病可以通过实验动物模型进行研究,以便更好
地理解它们的发生机制和治疗方法。
3. 功效性动物模型的缺陷
虽然功效性动物模型可以帮助科学家研究疾病和药物,但该方
法也存在一些问题。
首先,该方法可能存在异种问题,即在某些
情况下,疾病或药物在人类上的效果与在动物模型上的效果不同。
此外,使用动物模型研究的成本也很高,而且总是存在一些伦理
问题,如动物权利和安全性方面的问题。
4. 总结
虽然存在一些缺陷,但功效性动物模型仍是一种非常有用的工具。
它可以作为研究某些疾病和药物的起点。
然而,我们在使用
该方法时需要注意其局限性,并在以后的实验中持续改进该方法,以更好地进行研究,同时也考虑到动物福利及保护。
小鼠骨折及不同治疗方式模型的研究进展引言小鼠骨折模型是研究骨折治疗新方法以及了解骨折康复过程的重要工具。
通过建立小鼠骨折模型,可以模拟人类骨折,并且方便进行不同治疗方式的研究。
随着生物医学研究的发展,越来越多的治疗方式被应用于小鼠骨折模型,包括传统治疗方法和新技术手段。
本文将介绍小鼠骨折模型及不同治疗方式的研究进展,并探讨其在骨折治疗领域的应用前景。
小鼠骨折模型的建立小鼠骨折模型的建立方法多种多样,常见的包括手术法、化学法和物理法等。
手术法是最常用的建立小鼠骨折模型的方法,通过制作骨折口、固定骨折等步骤来模拟人类骨折情况。
化学法是利用化学物质诱导小鼠骨折,常用的化学物质有胶原酶、硫酸铜等。
物理法是利用外力(如剪切力、冲击力)使小鼠骨骼受到破坏并导致骨折发生。
通过不同的建立方法,可以模拟不同程度、不同类型的骨折,从而为研究治疗方式提供多样化的样本。
传统治疗方式的研究进展传统的骨折治疗方式主要包括保守治疗和手术治疗。
保守治疗是指利用石膏、绷带等外部固定器材保护骨折部位,促进骨折愈合。
手术治疗则是通过外科手术的方式,将骨折部位复位固定,促进骨折愈合。
这些传统治疗方式在小鼠骨折模型中的应用也得到了广泛研究。
通过对不同治疗方式的比较研究,可以评估其在骨折治疗中的疗效、安全性以及对骨折愈合过程的影响。
新技术手段的研究进展随着生物医学技术的进步,越来越多的新技术手段被应用于小鼠骨折模型的研究。
其中,基因治疗和干细胞治疗是研究热点。
基因治疗通过向骨折部位输送特定基因或调控骨细胞基因表达,促进骨折愈合。
干细胞治疗则是通过将干细胞定向分化为骨细胞或骨髓基质细胞,增加骨折部位的细胞来源,从而促进骨折愈合。
其他新技术手段包括利用成骨细胞因子、3D打印技术等,通过提供骨生成材料或建立复杂的人工支架来促进骨折愈合。
应用前景小鼠骨折模型及不同治疗方式的研究进展为骨折治疗提供了重要的基础。
通过研究不同治疗方式在小鼠骨折模型中的疗效和机制,可以为人类骨折治疗提供新的思路和方法。
·3435··综述·多原发肺癌诊断与治疗的最新研究进展洪子强1,2,金大成2,白向豆1,2,崔百强1,2,苟云久2*【摘要】 随着CT 和肺癌筛查的广泛应用,多原发肺癌(MPLC )(即同个患者中发生两个或两个以上的原发恶性肺癌)的检出率逐渐增加,但如何准确鉴别MPLC 和肺内转移(IM )仍是一个难题。
组织学特征在某些情况下可以很好地鉴别MPLC 和IM ,但常需要进行分子分析,如下一代测序技术(NGS )的应用有助于鉴别MPLC 和IM 。
而对于MPLC 的治疗,手术仍然是主要的治疗方式,对于不能手术的患者,放疗和局部消融发挥着重要作用。
NGS 和新疗法(如靶向药物和免疫检查点抑制剂)的出现为MPLC 的诊疗提供了新的选择。
本研究就近年来MPLC 诊断和治疗的研究进展进行论述。
【关键词】 肺肿瘤;多原发肺癌;肿瘤转移;诊断,鉴别;肺外科手术;放射疗法;免疫检查点抑制剂;分子靶向治疗;综述【中图分类号】 R 734.2 【文献标识码】 A DOI :10.12114/j.issn.1007-9572.2022.0331洪子强,金大成,白向豆,等. 多原发肺癌诊断与治疗的最新研究进展[J ]. 中国全科医学,2022,25(27):3435-3442.[ ]HONG Z Q ,JIN D C ,BAI X D ,et al. Recent advances in the diagnosis and management of multiple primary lung cancer[J ]. Chinese General Practice ,2022,25(27):3435-3442.Recent Advances in the Diagnosis and Management of Multiple Primary Lung Cancer HONG Ziqiang 1,2,JIN Dacheng 2,BAI Xiangdou 1,2,CUI Baiqiang 1,2,GOU Yunjiu 2*1.First School of Clinical Medical ,Gansu University of Chinese Medicine ,Lanzhou 730000,China2.Department of Thoracic Surgery ,Gansu Provincial Hospital ,Lanzhou 730000,China*Corresponding author :GOU Yunjiu ,Professor ;E-mail :【Abstract 】 The wide application of lung CT scan and lung cancer screening significnatly improves the detection rateof multiple primary lung cancer (MPLC ),namely ,lung cancer patients have two or more primaries at the same time. It is a tough problem all the time that how to distinguish between MPLC and intrapulmonary metastases (IM ). Although histological feature analysis is a good means to distinguish them in some circumstances ,molecular analysis is also needed generally. Such as the application of next generation sequencing (NGS ) is useful for the distinguish between MPLC and IM. For MPLC ,surgery remains the main treatment modality. For inoperable MPLC ,radiotherapy and local ablation are important treatments. NGS and new therapies such as targeted drug therapy and immune checkpoint inhibitors have become new altenatives for the diagnosis and treatment of MPLC. This article reviews recent advances in the diagnosis and treatment of MPLC.【Key words 】 Lung neoplasms ;Multiple primary lung cancer ;Neoplasm metastasis ;Diagnosis ,differential ;Pulmonary surgical procedures ;Radiotherapy ;Immune checkpoint inhibitors ;Molecular targeted therapy ;Review管有既定的标准,但鉴别MPLC 与肺内转移(intrapulmonary metastasis,IM)仍具有挑战性,这也是胸外科医生和病理科医生长期面临的难题[4-5]。
动物模型在药物开发中的应用药物开发是一项复杂的过程,需要经历从药物候选到药物审批的多个阶段。
其中,药物开发的重要工具之一就是利用动物模型进行药物研究和开发。
本文将阐述动物模型在药物开发中的应用,依次探讨其优势、局限性以及未来发展趋势。
一、动物模型在药物开发中的优势1. 研究临床用药效果动物模型可以帮助研究人员模拟药物在人体的各种作用和反应,尤其是能够预测药物在临床上是否有效。
例如,对于治疗心脏病的药物,动物模型可以模拟出心脏疾病模型,然后引用新药并观察它是否可以达到治疗目的。
从而帮助研发人员提早评估药物的功效和安全性。
2. 测定药物物理、化学和毒理性特性动物模型可以帮助研究人员更好地了解药物的理化性质、药代动力学和药效学。
同时,以不同动物模型为基础,可以评估药物对机体产生的副作用,从而帮助调整药物的治疗范围和用量等。
在药物研究和开发的早期阶段,动物模型也可以作为筛选和排除药物的重要手段。
二、动物模型在药物开发中的局限性1. 无法完全模拟人体生理和病理状态尽管动物模型可以帮助研究人员模拟药物在人体中的各种反应和响应,但是由于人体和动物生理结构和功能的差异,动物模型无法完全模拟人体的生理和病理状态。
例如,对于某些疾病,如癌症和人类神经退行性疾病,动物模型难以准确模拟,从而导致力荐出来的药物与人类病例之间存在显著差异。
2. 可能存在动物样品的数量和质量问题药物研究和开发需要大量动物样品的支持,过程中如果出现动物伦理、质量等问题,将给研究和开发带来巨大的影响。
此外,由于涉及不同动物种类和功能影响的变化等因素,药物在不同动物模型中的测试结果会有所不同,也需要消耗额外的成本和时间来验证和确认。
三、动物模型在药物开发中的未来发展趋势1. 合理选择动物模型在未来的药物开发中,应尽可能选择与人类生理和病理状态相似的动物模型进行药物研究和开发。
例如,对于某些疾病,如心血管、神经退行性疾病等,经过慎重考虑,可能需要选择高级哺乳动物模型进行药物研究,以提高药物研究的效率和重复性。
药学研究中动物模型的选择与应用在药学研究中,动物模型是评估药物安全性和有效性的重要工具。
它们帮助科研人员理解疾病机制、评估治疗效果以及预测临床治疗的结果。
合理地选择和应用动物模型对于药物研发的成功至关重要。
本文将探讨动物模型在药学研究中的重要性,以及如何科学合理地选择和应用这些模型。
动物模型的基本概念动物模型是指在实验条件下模拟人类疾病或药物反应的动物。
这些模型通过不同的手段被诱发或基因改造,以便于研究特定的人类疾病过程和评估药物干预的效果。
常见的动物模型包括小鼠、大鼠、兔子、猪以及非人类灵长类动物等。
动物模型的分类自然发病模型:这类模型是指那些自然发生特定疾病的动物。
例如,某些腺体肿瘤在特定品系的小鼠中自然发生,适合用于研究肿瘤相关药物。
诱导性模型:通过化学、物理或生物方法诱导动物产生某种特定疾病。
例如,通过注射化合物来引起糖尿病或心脏病等疾病,可以用于新型药物的筛选和验证。
基因工程模型:借助基因编辑技术,如CRISPR/Cas9,创建具有特定遗传缺陷的动物。
这些模型常用于癌症、自身免疫性疾病等复杂疾病的研究。
选择动物模型的原则在选择适合的动物模型时,有几个关键因素需要考虑:1. 生物相似性选用的动物种类需在生理、生化及病理特征上与人类有高度相似性。
例如,非人灵长类动物因为与人类更接近,会被用于复杂神经系统疾病的研究。
2. 模型的可重复性一个良好的动物模型应具备可重复性,即其他研究者能够通过相同的方法和条件再现先前的发现。
因此,选择已有广泛验证和接受的标准化模型尤为重要。
3. 操作简便性实验人员对操作简单、管理便捷的动物更为青睐,以确保实验环境一致并减少操作误差。
同时也要考虑到伦理问题,保障实验动物的福利。
4. 成本效益不同动物模型所需资源成本各异,选择时需考虑预算,比如小鼠通常相对比较经济,而大型动物如猪则涉及更高的照料及管理费用。
动物模型在药学研究中的应用动物模型在药学研究中扮演多重角色,它们不仅用于基础科学研究,也广泛用于药物开发、毒性检测以及临床试验设计等领域。
消痒洗剂对急、慢性湿疹动物模型的药效研究李江萍;蒋玲;李玲【摘要】目的:观察消痒洗剂对急、慢性湿疹小鼠模型的白介素-1β(IL-1β),白介素-4(IL-4)和白介素-8 (IL-8)的表达影响,以及其抗炎止痒的效果,阐明其治疗急、慢性湿疹的作用机制.方法:分别采用2,4-二硝基氟苯(DNFB)、2,4-二硝基氯苯(DNCB)致敏建立小鼠急、慢性湿疹模型,分别给予消痒洗剂低、中、高剂量(体积浓度分别为10%、20%、40%)及阳性对照药(复方苦参洗剂)(浓度为40%)干预,比较各组炎症因子的变化及其抗炎效果;通过磷酸组胺致痒试验及二甲苯致炎试验观察消瘁洗剂的止痒、抗炎效果.结果:消痒洗剂各剂量组能显著减轻ACD小鼠模型的耳肿胀度(P<0.01)并能够减轻ACD小鼠模型血清IL-1β、IL-8(P<0.01)等炎症因子的水平;能显著减轻DTH小鼠模型的耳肿胀度(高、中剂量组P<0.01,低剂量组无差异),而所有剂量组均能够减轻DTH小鼠模型血清IL-1β、IL-8(P<0.01)的水平.消痒洗剂高、中剂量组能提高磷酸组胺对豚鼠的致瘁阀(P<0.05);消痒洗剂各剂量组均能显著抑制二甲苯所致小鼠耳廓肿胀(P<0.01).结论:消痒洗剂通过降低炎性细胞因子IL-1β、IL-8的水平来治疗急、慢性皮炎湿疹,且止痒及抗炎的效果明显.【期刊名称】《贵州科学》【年(卷),期】2016(034)006【总页数】5页(P46-50)【关键词】消痒洗剂;急慢性皮炎湿疹;IL-1β;IL-8;IL-4;止痒;抗炎【作者】李江萍;蒋玲;李玲【作者单位】贵阳中医学院第二附属医院药学部,贵州贵阳550001;贵阳中医学院第二附属医院药学部,贵州贵阳550001;贵州医科大学机能实验室,贵州贵阳550004【正文语种】中文【中图分类】R965湿疹是临床常见的皮肤病之一,是一种由多种内外因素引起过敏反应的慢性炎症性皮肤病,多由急性、亚急性湿疹反复发作迁延而来,属变态反应性疾病范畴,约占皮肤科门诊患者的20 %左右[1]。
赵刚等肿瘤立体定向消融放疗联合免疫治疗的研究进展第 11 期肿瘤立体定向消融放疗联合免疫治疗的研究进展①赵刚涂甲丁卜嘉蕊王然玉张书涵(吉林大学公共卫生学院,国家卫生健康委员会放射生物学重点实验室,长春 130021)中图分类号R734.2 文献标志码 A 文章编号1000-484X(2023)11-2461-06[摘要]近年来,随着放疗技术的迅速发展,特别是影像技术和计算机技术的发展,肿瘤立体定向消融放疗(SABR)实现了精准地将非常高剂量的射线传递到肿瘤局部,而对周围正常组织的损伤降到最低。
SABR除了对肿瘤的直接杀伤作用外,还具有免疫调节作用,SABR的作用相当于原位疫苗接种的效果,导致受照射局部肿瘤释放大量肿瘤相关抗原和损伤相关模式分子等,促进了机体抗肿瘤T细胞反应,使肿瘤的免疫原性细胞死亡增加,并可能诱发抗肿瘤的远端效应。
然而,肿瘤周围微环境通常处于免疫抑制状态,单独SABR的免疫调节作用很难奏效,因此,在SABR的同时引入免疫调节剂的联合治疗方案非常具有说服力。
免疫调节剂可解除肿瘤周围的免疫抑制状态,从而与SABR发挥协同作用,这种协同关系已在实验室模型中得到广泛证实,目前多项早期临床试验正在进行中。
[关键词]立体定向消融放疗;免疫治疗;免疫调节剂;肿瘤Progess in tumor stereotactic ablative radiotherapy combined with immuno-therapyZHAO Gang,TU Jiading,BU Jiarui,WANG Ranyu,ZHANG Shuhan. NHC Key Laboratory of Radiobiology,School of Public Health, Jilin University, Changchun 130021, China[Abstract]In recent years, with the rapid development of radiotherapy technology, especially imaging technology and computer technology, stereotactic ablative radiotherapy (SABR) has accurately delivered very high doses of radiation to the tumor region, and minimized damage to surrounding normal tissue. SABR in addition to direct effect of tumor killing,also has immunomodulatory effects, SABR effects is equivalent to in situ vaccination effect, which lead to irradiation local tumor release large tumour-associated antigens and damage associated molecular patterns,promote body anti-tumor T cell response,increase tumor immunogenic cell death, and may induce anti-tumor abscopal effect. However, the surrounding tumor microenvironment is generally in an immunosup‐pressive state, and the immunomodulatory effects of SABR alone are difficult to work, so the combination treatment regimen of immu‐nomodulators accompany with SABR is very convincing. Immunomodulators can relieve the immune suppressive state surrounding tumors and thus cooperate with SABR, which has been widely demonstrated in laboratory models, and multiple early-stage clinical trial studies are currently under way.[Key words]Stereotactic ablative radiotherapy;Immunotherapy;Immunomodulator;Tumor1 立体定向消融放疗的概念立体定向消融放疗(stereotactic ablative radio‐therapy,SABR)又称体部立体定向放射治疗(stereo‐tactic body radiation therapy,SBRT)、立体定向放射外科(stereotactic radiosurgery,SRS)等[1],本文以下统称SABR。
吡柔比星局部消融治疗兔移植性VX2肝肿瘤的疗效探讨张巍;周石;蒋天鹏;斯光晏;郭剑星;吴晓萍【摘要】目的通过探讨吡柔比星(THP)与无水酒精采用局部消融方法在治疗兔移植性VX2肝肿瘤中的疗效,了解THP采用局部消融方法在治疗肝癌中的效果.方法建立兔移植性VX2肝肿瘤模型,经超声检查后,挑选荷瘤兔24只.将其分成3组:A组空白对照组(8只)、B组THP组(8只)、C组无水乙醇组(8只).观察治疗后精神、活动、进食状态、毛发情况及两周天后兔子体重变化.治疗后两周处死兔子观察肿瘤的生长率、坏死率及肿瘤中VEGF的表达水平.结果治疗前后B组的体重分别为2.72±0.12(kg)、2.58±0.13(kg),C±组治疗前后的体重分别为2.79±0.20(kg)、2.48±0.22(kg),差异显著(P<0 05).B、C组肿瘤生长率分别为198.5±31.8%、240.3±46.0%,差异显著;肿瘤坏死率分别为60.8±7.9%、70.6±12.9%,无显著差异;VEGF表达的阳性率分别为25.0%、62.5.0%,差异显著(P<0.05)结论 THP采用局部消融疗法治疗兔移植性VX2肝肿瘤,在抑制肿瘤生长、促进肿瘤细胞凋亡、下调VEGF表达水平方面表现出一定的作用.【期刊名称】《当代医学》【年(卷),期】2010(016)023【总页数】5页(P428-432)【关键词】吡柔比星;无水乙醇;VX2;肝癌【作者】张巍;周石;蒋天鹏;斯光晏;郭剑星;吴晓萍【作者单位】浙江,325000,温州医学院附属第一医院介入科;贵州,550004,贵阳医学院附属医院介入科;贵州,550004,贵阳医学院附属医院介入科;贵州,550004,贵阳医学院附属医院介入科;贵州,550004,贵阳医学院附属医院介入科;贵州,550004,贵阳医学院附属医院介入科【正文语种】中文【中图分类】R9肝癌是常见的消化道恶性肿瘤之一,手术切除是目前治疗肝癌的最佳手段,但大多患者在就诊时已处于中晚期,已丧失手术机会,仅有9%~27%的患者适宜手术。
药理学研究中的动物模型选择概述:动物模型在药理学研究中发挥着重要的作用。
通过合适的动物模型可以更好地理解药物的机制、效果以及安全性。
然而,在选择适当的动物模型时需要考虑多个因素,包括相似性、成本和可行性等。
本文将探讨药理学研究中动物模型选择的原则和常见的应用。
一、相似性1.1 物种相似性:选择与人类生理相似度较高的动物作为研究对象,如大鼠和小鼠。
因为这些动物在生命活动、器官结构和代谢途径等方面与人类有较好的相似性,能够提供更可靠的数据。
1.2 疾病模型:根据所研究的药物治疗目标来选择与之相关的疾病模型。
例如,在心血管领域,可以使用高胆固醇饮食诱导小鼠产生高血压或者缺血再灌注损伤等模型来评估药物对这些情况下心脏功能改善效果。
二、成本和可行性2.1 成本:动物模型的建立和维护需要耗费大量资金,因此在选择时需要考虑经济实用性。
多个相关研究中常用的小鼠是较经济实惠的选择。
2.2 可行性:动物模型的选取还要根据实验室条件、技术设备和人员配备等方面进行考虑。
对于一些高度特化的模型,如果缺乏相关资源,则不宜选择。
三、常见应用3.1 药效学评价:通过动物模型可以评估药物在生物体内的药效学参数,包括药代动力学和药效学等。
这些数据对于了解药物吸收、分布、代谢和排泄过程非常重要。
3.2 治疗策略验证:在新药开发阶段,动物模型常被用来验证治疗策略的有效性。
例如,在癌症治疗研究中,使用小鼠移植瘤模型可以评估抗肿瘤药物的疗效。
3.3 安全评价:动物模型能够帮助检测潜在毒副作用,并评估药物在各种毒理学指标上的影响。
这对于药物安全性评价以及制定适当的用药指南至关重要。
四、局限性和新技术4.1 物种差异:尽管动物模型在探索药理学的研究中有很大帮助,但人类与动物之间仍然存在一定的生理、代谢差异。
因此,不能直接将动物实验结果推广到人体。
4.2 替代方法:随着科技的发展,出现了许多替代动物模型的技术,如体外细胞培养和计算机模拟等。
这些新技术可以减少对动物实验的需求,并提供更快速、精确、经济高效的研究手段。
0061117栏目中药药物评价>>非临床安全性和有效性评价标题银屑病药效学动物模型及其在中药新药研究中应用的思考作者韩玲部门正文内容审评一部韩玲摘要:本文综述了银屑病药效学动物模型的进展,对治疗银屑病中药新药药效学研究方面存在的问题进行了分析,提出了应结合银屑病发病机制的研究进展完善药效学方法学研究的建议。
银屑病是一种慢性复发性皮肤病,其发病机制复杂,至今尚未明了。
可能是一种具有基因遗传特性的疾病,也与感染、内分泌机能障碍、免疫功能紊乱等有关。
中医学认为以风湿毒邪内侵为标,血虚、血燥为根源,血热、血虚、血瘀是本病的基本病机,故清热解毒、凉血、活血化瘀、祛风除湿成为治疗本病的基本法。
银屑病的组织病理学主要表现为角质形成细胞过度增生、炎症细胞聚集和真皮乳头部血管增生扩张三大特征,从而出现角化过度、角化不全、颗粒层消失、棘细胞层增厚等。
尽管许多不同的动物模型能复制银屑病的某个方面的病理特征,并能解释部分病因,但长期以来仍缺乏一种能够研究所有有关的潜在因素的动物模型。
即-缺乏理想的动物模型。
一、银屑病药效学动物模型银屑病动物模型是根据银屑病的特征,人为建立的一种与之相似的模型。
有人总结了银屑病动物模型的建立过程,包括以下4个不断完善的阶段:1.诱发性动物模型该模型是通过工方法在动物,如豚鼠、大鼠等皮肤上诱发银屑病样增生。
其方法包括紫外线照射、化学刺激剂外涂或药物外涂、缺乏必须脂肪酸的饲料喂养等。
这些方法均为过度增殖模型,可引起表皮增生加快。
但这些方法仅是针对表皮损伤的一种迟发反应,持续时间较短。
如缺乏必须脂肪酸饲料喂养的大鼠模型,因缺乏必须脂肪酸,故可使大鼠皮肤逐渐出现角化过度、棘层肥厚、基底细胞有丝分裂加快,DNA合成增加等变化,表皮细胞呈慢性增生状态,与药物诱发的急性表皮增生相比,其与银屑病更为相似。
如心得安诱导银屑病动物模型,是利用心得安特有的药理作用,即心得安可阻断角阮细胞的β-肾上腺能受体而降低了细胞内cAMP水平所致。
Introduction: Many patients with oncogene-driven non–small-cell lung cancer (NSCLC) treated with tyrosine kinase inhibitors expe-rience limited sites of disease progression. This study investigated retrospectively the benefits of local ablative therapy (LAT) to central nervous system (CNS) and/or limited systemic disease progression and continuation of crizotinib or erlotinib in patients with metastatic ALK gene rearrangement (ALK+) or EGFR -mutant (EGFR -MT) NSCLC, respectively.Methods: Patients with metastatic ALK + NSCLC treated with crizo-tinib (n = 38) and EGFR -MT NSCLC treated with erlotinib (n = 27) were identified at a single institution. Initial response to the respective kinase inhibitors, median progression-free survival (PFS1), and site of first progression were recorded. A subset of patients with either nonlep-tomeningeal CNS and/or four sites or fewer of extra-CNS progression (oligoprogressive disease) suitable for LA T received either radiation or surgery to these sites and continued on the same tyrosine kinase inhibi-tors. The subsequent median progression-free survival from the time of first progression (PFS2) and pattern of progression were recorded.Results: Median progression-free survival in ALK + patients on crizotinib was 9.0 months, and 13.8 months for EGFR -MT patients on erlotinib. T wenty-five of 51 patients (49%) who progressed were deemed suitable for local therapy (15 ALK +, 10 EGFR -MT; 24 with radiotherapy, one with surgery) and continuation of the same targeted therapy. Post-LAT, 19 of 25 patients progressed again, with median PFS2 of 6.2 months.Discussion: Oncogene-addicted NSCLC with CNS and/or limited systemic disease progression (oligoprogressive disease) on relevant targeted therapies is often suitable for LAT and continuation of thetargeted agent, and is associated with more than 6 months of addi-tional disease control.Key Words: EGFR -mutant non–small-cell lung cancer, anaplastic lymphoma kinase gene arrangement non–small-cell lung cancer, Radiation therapy, Oligoprogressive disease.(J Thorac Oncol. 2012;7: 1807–1814)Patients with metastatic non–small-cell lung cancer (NSCLC) with anaplastic lymphoma kinase gene rear-rangements (ALK+) or epidermal growth factor receptor-mutations (EGFR -MTs) have high response rates and long progression-free survival times when treated with crizotinib or EGFR tyrosine kinase inhibitors (TKIs), such as erlo-tinib, respectively.1–6 However, progression inevitably occurs because of either inadequate central nervous system (CNS) penetration of the drug in some cases of CNS progression, or biological change in the tumor such as the development of new kinase domain mutations in the drug target or the devel-opment of alternate oncogenic drivers.7–16Although studies of many novel agents are ongoing, there are no currently approved targeted therapies specific for treatment of such patients upon progression. Although continuation of the TKI therapy by itself with no local therapy to slow the progression or continuation of the TKI in combination with chemotherapy have been advocated as options for these patients,17–20 the current standard therapeutic option at the time of progression is to treat the patient with cytotoxic chemotherapy alone. Local therapies, such as radiotherapy or surgery, have had little role outside symptom palliation in this setting. However, radiation therapy of isolated CNS progression in patients with EGFR -mutant NSCLC being treated with EGFR-TKIs and continued systemic administration of the TKI if there is no evidence of systemic progression has recently been described.21 Such an approach relies on the logic that CNS progression could reflect inadequate drug penetration rather than a change in the biology of the cancer. Therefore, the patient is unlikely to have developed systemic resistance to the drug and may be deriving significant ongoing benefit from its use.Copyright © 2012 by the International Association for the Study of Lung CancerISSN: 1556-0864/12/0712-1807Local Ablative Therapy of Oligoprogressive Disease Prolongs Disease Control by Tyrosine Kinase Inhibitors in Oncogene-Addicted Non–Small-Cell Lung CancerAndrew J. Weickhardt, MBBS, DmedSc,* Benjamin Scheier, MD,* Joseph Malachy Burke, MD,* Gregory Gan, MD,‡ Xian Lu, MSc,‡ Paul A. Bunn, Jr., MD,* Dara L. Aisner, MD, PhD,§ Laurie E. Gaspar, MD, MBA,‡ Brian D. Kavanagh, MD, MPH,‡ Robert C. Doebele, MD, PhD,*and D. Ross Camidge, MD, PhD**Department of Medicine, Division of Medical Oncology, †Department of Radiation Oncology, University of Colorado Cancer Center, Colorado; ‡Department of Biostatistics and Informatics, Colorado School of Public Health and University of Colorado, Colorado; and §D epartment of Pathology, University of Colorado Cancer Center, Colorado.Disclosure: Drs. Weickhardt, Doeble, and Camidge have received speaking honoraria from Pfizer. The other authors declare no conflicts of interest.Address for Correspondence: Andrew J. Weickhardt, MBBS, D medSc, University of Colorado Cancer Center, Aurora, CO 80045. E-mail: andrew.weickhardt@O RIGINAL ARTICLEWeickhardt et al. Journal of Thoracic Oncology • Volume 7, Number 12, December 2012Building on this logic, the approach we describe here uses local therapies to ablate sites of oligoprogressive disease that occurs systemically, as well as in the CNS, and continu-ing the same targeted therapy and is based on two underly-ing hypotheses. First, given our increasing knowledge about the different mechanisms of acquired resistance to TKIs in EGFR-MT and ALK+ disease, we hypothesized that any biological change mediating acquired resistance occurs as a stochastic clonal event that favors survival in accordance with Darwinian evolutionary principles.14–16 Consequently, if treated with ablative therapy before widespread dissemination of the resistant clone, disease control may be prolonged until either a new event occurs or resistant clones that have dis-seminated expand enough to become detectable. Second, we hypothesized that there is ongoing benefit from the targeted therapy in other sites of (nonprogressing) disease because of continuing suppression of sensitive clones that have not yet developed acquired resistance. Consistent with this, patients with EGFR-MT disease who progress often experience a dis-ease flare when the EGFR-TKI is discontinued,22 and rechal-lenge of these patients with the same EGFR-TKI after only a short time off therapy can lead to re-responses.23,24 In addi-tion, treatment beyond progression of EGFR-MT NSCLC with an EGFR-TKI has been associated with improved overall survival, compared with those in whom the TKI was perma-nently discontinued.25 Analogous benefits of the continuation of trastuzumab beyond progression have been well described in metastatic breast cancer.26–29This study describes a single-institution experience of using local ablative therapy (LAT) and continuation of the same targeted therapy to treat ALK+ and EGFR-MT meta-static NSCLC patients who progress either within the CNS and/or at limited systemic sites (oligoprogressive disease) while on crizotinib or erlotinib, respectively. In most cases we used stereotactic body radiation therapy (SBRT) as our LAT of choice. SBRT has previously been shown to be highly effec-tive in achieving local control in a variety of organs, without significant toxicity.30–37MATERIALS AND METHODSPatientsPatients eligible for inclusion in this retrospective analy-sis included all patients with histologically confirmed ALK+ or EGFR-MT metastatic NSCLC at the University of Colorado Cancer Center treated with crizotinib, or erlotinib between May 2005 and December 2011 with adequate follow-up data. Patients were identified through a query of the Colorado Molecular Correlates database for ALK+ patients determined by break-apart fluorescent in situ hybridization assay or EGFR mutation positive patients (exon 19 deletions or exon 21 L858R mutations) determined either through direct sequenc-ing or allele-specific polymerase chain reaction assays. An institutional review board approved protocol permits clinical correlates to be made on all patients seen at the University of Colorado in whom molecular analyses have been conducted within the Colorado Molecular Correlates laboratory.Baseline clinical characteristics were determined by retrospective collection from electronic records, including age at diagnosis (taken at date of diagnostic biopsy), sex, tumor histology, prior therapy, method of CNS imaging before initiation with erlotinib or crizotinib, date of diagnosis of any known CNS involvement, treatment of any known CNS involvement before the initiation of erlotinib or crizotinib, smoking status, and sites of metastatic disease. If patients did not have imaging of the CNS within 3 months before com-mencing TKI therapy and had no previous history of CNS metastases, they were assessed as having unknown CNS status. Smoking status was categorized as current (smoked within less than a year before start of therapy), former (quit more than a year before start of therapy), or never (less than a 100 lifetime cigarettes).All ALK+ patients received crizotinib (Xalkori, Pfizer, La Jolla, CA) starting at 250 mg twice a day on either the phase I expansion cohort of PROFILE 100138 or the nonran-domized phase II PROFILE 1005 clinical trial,39 and received staging every 8 weeks (PROFILE 1001), or every 6 weeks (PROFILE 1005) with computer tomography (CT) or positron emission tomography (PET)/CT. Imaging of the brain at either baseline or on treatment in these trials was not mandatory for any patient but was performed at investigator discretion. All EGFR-MT patients received erlotinib (Tarceva, Astellas, Farmingdale, NY) starting at 150 mg once a day, with two of 27 receiving erlotinib in combination with the insulin-like growth factor-1 receptor monoclonal antibody cixutumumab (Imclone, New Y ork, NY) as part of a clinical trial.40,41 Baseline and ongoing CNS and body imaging with mag-netic resonance imaging (MRI), CT and/or PET/CT for the 25 of 27 EGFR-MT patients treated off study was performed according to investigator discretion. The two EGFR-mutated NSCLC patients treated with erlotinib and cixutumumab on trial had interval body CT scans performed every 6 weeks while on study, although both withdrew from study after 7 and 8 months before progression to continue erlotinib alone, and had staging performed from this time according to investiga-tor discretion. Median progression-free survival (PFS1) was calculated from time of initiation of targeted therapy to first progression of disease (as per Response Evaluation Criteria in Solid Tumors version 1.1 ) or clinical progression (as assessed by clinician), or death from any cause (using Kaplan–Meier methods). Sites of first progression (CNS or external to the CNS [eCNS]) were documented.On the basis of institutional practice, patients who progressed on their oral targeted therapy who had either lep-tomeningeal disease, more than four sites of eCNS progres-sion, poor performance status (Eastern Cooperative Oncology Group >2) or poor tolerance of their targeted therapy were not considered suitable for LAT (n=26). A subset of patients with progression after initial treatment with either crizotinib or erlo-tinib with either nonleptomeningeal CNS progression and/or less than 4 sites of eCNS progression, adequate performance status (Eastern Cooperative Oncology Group <2), and good tolerance of their targeted therapy (n = 25) were considered for LAT to the site(s) of progression and continuation of the same oral targeted therapy. Before LAT, patients underwentJournal of Thoracic Oncology • Volume 7, Number 12, December 2012Local Ablative Therapy of Oligoprogression in NSCLCa biopsy of the site of their progressive disease to determine the molecular mechanism of resistance to targeted therapy if this was determined to be safe by their treating oncologist and interventional radiologist which, in part, have been reported separately.16 Patients were instructed to withhold their oral targeted therapy on the days of local therapy with radiation and restart on the day after radiation was completed, with no change in dosage. Those patients who received surgical LAT were instructed to withhold the TKI until the surgical team considered it appropriate to recommence oral dosing.The characteristics and timing of local ablative therapy (SBRT, standard radiation therapy [XRT], stereotactic radio-surgery [SRS], whole brain radiation therapy [WBRT] or surgery), and number of disease sites treated was recorded. Electronic records of patients who received radiation or sur-gery were reviewed for evidence of relevant systemic or local toxicity related to the volume irradiated for 6 months from the end of the LAT, including but not limited to fatigue and headaches after CNS irradiation; pneumonitis after lung irra-diation; radiation-induced liver disease after liver irradiation; and skin toxicity after any SBRT or XRT. PFS2 was measured from the time of first progression until second progression on the same targeted therapy, using Response Evaluation Criteria in Solid Tumors 1.1 or death from any cause. Data analysis was performed up to January 1, 2012.Statistical AnalysisStatistical analysis for creation of Kaplan–Meier curves was performed using Prism V software (Graphpad, San Diego, CA). Median survival time, confidence intervals, and a mul-tivariate analysis with a Cox proportional hazards model was performed using version 9.3 of SAS/STAT software (SAS institute Inc., Cary, NC).ResultsThirty-eight ALK+ patients received crizotinib, 28 (74%) of whom had progressed at the time of analysis. Twenty-seven EGFR-MT patients received erlotinib, 23 (85%) of whom had progressed at the time of analysis. Patient characteristics are summarized in Table 1. The majority of patients (63 of 65, 97%) had adenocarcinoma histology, and the median age was 58 years. Collectively, 19 of 65 patients (29%) had known CNS metastases before commencement with targeted therapy, but 29 of 65 patients (45%) with no history of CNS metasta-ses did not have MRI or CT imaging of their brain performed in the 3 months before commencement on either drug. The median duration of follow-up was 20 months. The PFS1 of the 65 NSCLC patients treated with either crizotinib or erlotinib was 10.3 months (9.0 months for ALK+ patients, 13.8 months for EGFR-MT patients [Table 1]).Among 28 ALK+ patients who had progressed at the time of the analysis, 13 (46%) first progressed in the CNS at PFS1 (two of whom progressed simultaneously in the CNS and eCNS) and the CNS failure rate was similar in the 18 ALK+ patients who had a documented CNS status (14 with CNS metastases, four with no CNS metastases) before com-mencing crizotinib (seven of 18, 39%). Among EGFR-MT patients, five of 23 patients (22%) first progressed in the CNS at PFS1 (two simultaneously in CNS and eCNS), and again the CNS failure rate was similar in the subgroup of 10 EGFR-MT patients who had a documented CNS status (five with CNS metastases, five with no CNS metastases) before commencing erlotinib (two of 10, 20%).Of the 28 ALK+ patients who had progressed, 15 of 28 (54%) received LAT after first progression and were treated beyond progression with crizotinib. Of the 23 EGFR-MT patients who had progressed, 10 (43.5%) received LAT afterTABLE 1. Demographics of PatientsALL PTS ALK+EGFR-MT ALL PTSTBP ALK +TBPEGFR-MTTBPNo.65382725/51 (49%)15/28 (54%)10/23 (43%) Age (median, yr)585560585059 Range23–8023–8046–7523–7523–7254–75 Female sex37 (57%)18 (47%)19 (70%)16 (64%)8 (53%)8 (80%) Never smokers43 (66%)26 (68%)17 (63%)15 (60%)11 (73%) 4 (40%) Previous lines RxMean 2.53 1.85 2.5 2.9 2.0 Range0–61–61–40–41–40–2 CNS status pre C1None17 (26%)10 (26%)7 (26%) 5 (20%) 4 (27%) 1 (10%) Present19 (29%)14 (37%) 5 (18%)8 (32%) 5 (33%) 3 (30%) Unknown29 (45%)14 (37%)15 (56%)12 (48%) 6 (40%) 6 (60%) PFS1 (mo)Median10.39.013.89.89.012.0 95% CI8.9–13.8 6.5–12.88.9–16.48.8–13.8 6.5–12.0 6.5–19.0 No events512823251510PTS, patients; ALK +, ALK positive as defined by fluorescence in situ hybridization; TBP, treated beyond progression with local ablative therapy; CNS, central nervous system; CI, confidence interval; PFSI, median progression-free survival; EGFR-MT, EGFR-mutant.Weickhardt et al. Journal of Thoracic Oncology • Volume 7, Number 12, December 2012first progression and were treated beyond progression with erlotinib. Overall, 25 of 51 patients (49%) received LAT at first progression. All patients who received LAT recom-menced their TKI after therapy. The median time from PFS1 to the start of LAT was 3.7 weeks. The PFS1 was 9.8 months for all 25 patients with oligoprogressive disease, who received LAT and continuation of targeted therapy (9.0 months for ALK+ patients, 12.0 months for EGFR-MT patients). The PFS1 was 12.8 months for all patients with progression, who did not receive LAT and continuation of targeted therapy (7.2 months for ALK+ patients, 13.9 months for EGFR-MT patients Table 1).The pattern of progression at PFS1 for those 25 patients treated with LAT for oligoprogressive disease is shown in Table 2. Seventeen of the 25 patients (68%) had restaging of their CNS with an MRI of the brain at the time of PFS1. Seventeen of the 25 patients (68%) had systemic restaging with PET/CT at the time of PFS1, with all others using CT scanning. Thirteen patients (nine ALK+, four EGFR-MT) first progressed in the CNS, with 10 of 13 patients (77%) only having progression in their CNS while still having control of systemic disease outside the CNS. All six patients with fewer than four CNS metastases were treated with SRS. A single patient with eight sites of cerebral metastases was treated with SRS to each site at an outside institution. Otherwise, all other patients with four or more CNS metastases received WBRT. The majority of the 15 patients who progressed out-side the CNS and were treated with local therapy were treated with SBRT (15–54 Gy, median 40 Gy), with eight of 15 (53%) having a single site of progression treated. Up to four eCNS sites were treated (median 2), with the most common sites being bone and lung. One patient underwent an adre-nalectomy, and two patients were treated with XRT to bone metastases (either with 20 Gy in five fractions or 30 Gy in 10 fractions).For the 17 patients who received CNS restaging at PFS1, the median interval of CNS restaging between PFS1 and PFS2 was 3.1 months. The median interval of systemic restaging between PFS1 and PFS2 was 2.1 months. The median follow-up post-LAT at the time of analysis was 9.4 months. For the 25 patients who received LAT and continued on targeted therapy, the median PFS2 from the time of PFS1 was 6.2 months (Table 3, Fig. 1). The median PFS2 in patients with initial CNS only progression was 7.1 months. Median PFS2 in patients with initial eCNS progression, including three patients who had CNS progression detected within a month of systemic progression, was 4.0 months. The pattern of progression at PFS2 is shown in Table 3. Of patients who progressed initially in the CNS, 50% next progressed outside the CNS. Similarly, of patients who progressed initially outside the CNS, 53% next progressed outside the CNS again. At the time of analysis, six of 25 patients (24%) had not progressed again after local therapy post-PFS1 after a median follow-up of 7 months. There was a trend for patients whose time to first progression was less than or equal to 12 months to have a shorter time to second progression, (hazard ratio=3.45, 95% confidence intervals 0.92–12.99, p = 0.067) but this was not statistically significant.The majority of adverse events relating to ablative ther-apy occurred in patients having WBRT. Radiation-induced liver damage was not observed in the patient who received liver SBRT. Grade 3 fatigue was reported in two patients within the 6 months after WBRT, but there were no other doc-umented grade 3/4 adverse events attributable to radiotherapy (Table 4).DISCUSSIONOncologists have traditionally discontinued or changed systemic therapy when there is objective evidence of radio-logical or clinical progression, intolerable toxicity, or com-pletion of a fixed number of treatment cycles. However, in cases of progression on a previously beneficial targeted agent for molecularly subtyped cancer other options may exist. Specifically, our experience suggests that when patients with EGFR-MT or ALK+ NSCLC progress on erlotinib or crizotinib, respectively, and the progression occurs in only a limited number of sites (oligoprogressive disease) it may be reasonable to consider LAT to the sites of progression andTABLE 2. Sites of Oligoprogression and LAT Treatment ModalityNo. of PTS SRS WBRT SBRT XRT Surgery CNS as site of first progressionLesions < 466———Lesions > 4716———eCNS as site of first progressionBone7——52—Lung7——7——Lymph node2——2——Adrenal2——1—1 Liver1——1——LAT, local ablative therapy; SRS, stereotactic radiosurgery; WBRT, whole brain radiation therapy; SBRT, stereotactic body radiation therapy; XRT, external beam radiotherapy; CNS, central nervous system of disease; eCNS, extra-CNS site of disease; PTS, patients.TABLE 3. Outcomes from LAT and Continuation of Targeted TherapySite of FirstProgressionNo. ofPatientsPFS1 (mo)(CI)PFS2 (mo)(CI)Site of SecondProgression CNS1010.97.1 2 (20%)No prog7.3–18.3 1.7–11.3 3 (30%)CNS5 (50%)eCNS eCNS a159.0 4.0 4 (27%)No prog6.5–13.8 2.7–7.4 3 (20%)CNS8 (53%)eCNS All patients259.8 6.2 6 (24%)No prog8.8–13.8 3.7–8.07 (28%)CNS12 (48%)eCNSa Includes three patients who progressed eCNS and CNS at PFS1.CI, confidence interval; CNS, central nervous system as site of disease; eCNS, extra-CNS sites of disease; PFS1, median progression-free survival.Journal of Thoracic Oncology • Volume 7, Number 12, December 2012 Local Ablative Therapy of Oligoprogression in NSCLCcontinuation of the TKI (Table 5, Fig. 2). Forty-nine percent of patients treated with either erlotinib or crizotinib who pro-gressed at our institution were deemed appropriate for this treatment strategy.Patients treated with crizotinib or erlotinib in this series had a median PFS1 of 10.3 months, consistent with literature precedent.1–6 Although retrospective series of radiotherapy used in oligometastatic disease at diagnosis in metastatic NSCLC report good local control rates and better overall survival than in historical controls,42–48 there is little published data about the use of local therapy for oligoprogressive disease on therapy. This study suggests that in patients with EGFR -MT or ALK + NSCLC on erlotinib or crizotinib therapy who develop either less than four systemic progressive lesions (themaximum treated in this series) and/or CNS progression, LAT (either radiation or surgery) and continuation of the TKI may extend disease control by over 6 months. Our results expand on recently published work on the role of LAT in patients with EGFR-MT. A Japanese group reported a median eCNS PFS2 of 5.6 months after LAT of isolated CNS progression in 17 NSCLC patients who had achieved at least stable disease for more than 6 months on an EGFR-TKI,21 and an American group reported a median PFS2 of 10 months in 18 NSCLC patients with EGFR-MT after LAT of isolated sites of eCNS progression.49In our series, no patients receiving LAT had radiologi-cal evidence of leptomeningeal disease, which is associated with poor outcomes, lack of clear effective therapy, and there-fore unlikely to be suited to a local treatment approach.50 Strikingly, nearly half (13 of 28, 46%) of all ALK+patientsFIGURE 1. A , PFS1 and PFS1+PFS2 survival curves of all 25 patients treated with LAT. B , Ten patients treated with LAT who first pro-gressed only in the CNS. C , Fifteen patients treated with LAT who first progressed in extra-CNS locations, including three patients with simul-taneous CNS and eCNS progression. PFS1, median progression-free sur-vival; PFS2, progression-free survival from the time of first p rogression; LAT, local ablative therapy; CNS, central nervous system; eCNS, extra-CNS.TABLE 4. Potential Treatment-Related Toxicity within 6 Months of Completing LATWBRT (n = 6)Other Ablative Therapy (n = 19)Grade 1/2Grade 3/4Grade 1/2Grade 3/4Alopecia 6000Fatigue1230Memory impairment 3000Nausea 1010Anorexia1010Emotional lability 2000Headaches1000Chest-wall tenderness1LAT, local ablative therapy; WBRT, whole brain radiotherapy.TABLE 5. Suggested Criteria for Considering Local Ablative Therapy of Oligoprogressive Disease and Treatment with a TKI beyond Progression Includea1. A LK positive or EGFR -mutant metastatic non–small-cell lung cancer2. R elevant TKI (e.g., crizotinib or erlotinib) is well tolerated3. O ligoprogressive disease on TKI therapy, defined as: C NS progression without leptomeningeal disease amenable to WBRT, SRS, or surgical resection. P rogression in ≤ 4 extra-CNS sites amenable to SBRT, XRT, or surgical resection.aBased on the practices within this study.WBRT, whole brain radiation therapy; SRS, stereotactic radiosurgery; SBRT,stereotactic body radiation therapy; XRT, conventionally fractionated radiation therapy; TKI, tyrosine kinase inhibitor; EGFR, epidermal growth factor recepetor.Weickhardt et al. Journal of Thoracic Oncology • Volume 7, Number 12, December 2012progressed first in the CNS, with the majority (11 of 13) 85% still responding or with stable disease systemically, making a LAT approach combined with ongoing use of crizotinib particularly attractive within this group. In patients without baseline CNS imaging with documented CNS progression, it is not possible to categorically state whether new CNS lesions reflect true CNS progression or simply the new discovery of lesions that preexisted. However, the fact that the rates of CNS progression were very similar among those with known CNS status at baseline (39%), suggests that the predominant effect is one of true CNS progression. Failure in the CNS may be because of inadequate crizotinib exposures rather than a change in the dominant biology of the tumor.10,51–53 Similar data relating to the potential for the CNS to represent a rela-tive sanctuary site with respect to EGFR-TKI therapy for EGFR -MT disease also exist.10 In contrast, systemic mech-anisms of resistance to these drugs relate to several differ-ent biological changes in the tumor, such as kinase domain mutations in the target enzyme or the development of addi-tional oncogenic drivers.13,15,54–56 It is uncertain whether the potential for there to be different explanations for failure in CNS and systemic sites accounts for a trend toward improved PFS2 in patients receiving LAT for isolated CNS progres-sion at PFS1 relative to those patients receiving LAT for sys-temic progression. In our series, patients with isolated CNS progression had a median time to next progression of over 7 months, as compared with a PFS2 of 4.0 months in patients who experienced first progression outside the CNS; however, this difference was not statistically significant (hazard ratio for progression 0.85, 95% confidence interval 0.29–2.47, p = 0.76).There are several limitations of this study. Safety data on radiation-related side effects within this study were col-lected and graded retrospectively. However, the safety of com-bining aggressive, ablative-intent SRS or SBRT regiments with TKI- or monoclonal antibody-based EGFR inhibition has been reported for both CNS and extracranial sites, so the apparent good tolerability of our approach would not be unexpected.57–60There was a lack of standardized timing interval in systemic restaging patients taking erlotinib although not on clinical trial, and no standardized timing of CNS staging in either the crizotinib or erlotinib group. The median interval of restaging between PFS1 and PFS2 was 3.1 months for CNS in those with restaging MRI at PFS1, and 2.1 months for eCNS sites of disease, which is less than half the time interval of the additional apparent disease control from LAT. On the basis of institutional precedent, we limited the number of eCNS lesions considered for LAT to four or fewer sites, and in most cases the number of CNS lesions considered for SRS as opposed to WBRT to less than four. Emerging data suggest that SRS alone might be appropriate for a higher number of brain metastases as long as the total burden of tumor is limited,61 and there would be an opportunity to avoid the neurocognitive toxicity associated with WBRT.62 Similarly, whether the treatment of symptomatic and asymptomatic CNS metastases is equally beneficial to the patient remains unknown. All patients who received LAT continued to receive their TKI post-LAT, therefore, although we can comment on the outcomes associated with the combined approach, we cannot distinguish the specific contribution of each element.Perhaps most importantly, we do not have a comparator group to judge the true benefit of our LAT/TKI continuation approach. Historical controls of other chemotherapies in NSCLC cannot accurately be used given that this was a retrospective review of a molecularly defined population, treated across several different lines of therapy. Of note, as the PFS1 in the LAT -treated group and the non-LAT–treated group were comparable (10.3 versus 12.8 months), we do not seem to have preselected a more indolent population for LAT within this study. Although we have estimated the time to the next progression event in the LAT -treated group, additional LAT at the time of second progression was considered in several cases when only further oligoprogressive disease was manifested (data not shown). Consequently, in any prospective evaluation, comparing this approach, for example, to some standard chemotherapy in a defined line of treatment, both the clear delineation of the criteria for considering the initial and any repeat LAT acceptable and an assessment of the benefit of the approach on overall survival and quality of life and not just the PFS before the next intervention may be most informative. Within this study, at the time of analysis only four of 25 of the LAT group and 10 of 26 of the non-LAT–treated progressive group had died and therefore overall survival data are notmature.FIGURE 2. Proposed schema for incorporating local ablative therapy into therapy at time of first progression with ALK + or EGFR -MT NSCLC patients treated with TKI therapy. ALK +, anaplastic lymphoma kinase gene rearrangement; EGFR -MT NSCLC, epidermal growth factor receptor-mutant non–small-cell lung cancer; TKI, tyrosine kinase inhibitors.。