Phase II trial of continuous low-dose temozolomide for patients with recurrent malignant glioma
- 格式:pdf
- 大小:180.36 KB
- 文档页数:9
Velcade® (bortezomib)Document Number: IC-0137 Last Review Date: 5/1/2018Date of Origin: 11/28/2011Dates Reviewed: 12/2011, 03/2012, 06/2012, 09/2012, 12/2012, 03/2013, 06/2013, 09/2013, 12/2013, 03/2014, 06/2014, 09/2014, 12/2014, 03/2015, 05/2015, 08/2015, 11/2015, 02/2016, 05/2016, 08/2016, 11/2016, 02/2017, 05/2017, 08/2017, 11/2017, 02/2018, 05/2018I.Length of AuthorizationCoverage will be provided for 6 months and may be renewed.II.Dosing LimitsA.Quantity Limit (max daily dose) [Pharmacy Benefit]:-Velcade 3.5 mg powder for injection: 4 vials per 21 day supplyB.Max Units (per dose and over time) [Medical Benefit]:∙140 billable units every 21 daysIII.Initial Approval CriteriaCoverage is provided in the following conditions:∙Patient aged 18 years or older; ANDMultiple myeloma †Mantle cell lymphoma †∙Used as a single agent or in combination with rituximabSystemic Light Chain Amyloidosis ‡∙Used as a single agent; OR∙Used in combination with dexamethasone with or without melphalan; OR∙Used in combination with dexamethasone and cyclophosphamideWaldenström’s macroglobulinemia/Lymphoplasmacytic Lymphoma‡∙Used as a single agent; OR∙Used in combination with dexamethasone; OR∙Used in combination with rituximab with or without dexamethasoneMulticentric Castleman’s Disease‡∙Must be used as subsequent therapy; AND∙Patient has progressed following treatment for relapsed/refractory or progressive disease;AND∙Used as a single agent or in combination with rituximabPrimary cutaneous CD30+ T-Cell Lymphoproliferative Disorders ‡∙Used as single agent for relapsed or refractory disease; ANDo Patient has primary cutaneous anaplastic large cell lymphoma (pcALCL) with multifocal lesions; ORo Patient has cutaneous ALCL with regional nodes (excludes systemic ALCL) Adult T-Cell Leukemia/Lymphoma ‡∙Must be used as a single agent for non-responders to first-line therapy for acute disease or lymphoma; ANDo Used second-line if intent is to proceed to high-dose therapy with autologous stem cell rescue (HDT/ASCR); ORo Subsequent therapy after HDT/ASCR†FDA Approved Indication(s); ‡Compendia recommended indication(s)IV.Renewal CriteriaCoverage can be renewed based upon the following criteria:∙Patient continues to meet the criteria identified in section III; AND∙Tumor response with stabilization of disease or decrease in size of tumor or tumor spread;AND∙Absence of unacceptable toxicity from the drug. Example of unacceptable toxicity include: peripheral neuropathy, hypotension, cardiac toxicity, pulmonary toxicity, posteriorreversible encephalopathy syndrome, gastrointestinal toxicity, thrombocytopenia,neutropenia, tumor lysis syndrome, hepatic toxicity, etc.V.Dosage/AdministrationVI.Billing Code/Availability InformationJcode:∙J9041– Injection, bortezomib, 0.1 mg; 1 billable unit = 0.1 mgNDC(s):∙Velcade 3.5 mg single-use vial powder for injection: 63020-0049-xxVII.References1.Velcade [package insert]. Cambridge, MA; Millennium Pharmaceuticals, Inc; June 2017.Accessed March 2018.2.Referenced with permission from the NCCN Drugs & Biologics Compendium (NCCNCompendium®) for Bortezomib. National Comprehensive Cancer Network, 2018. The NCCN Compendium® is a derivative work of the NCCN Guidelines®. NATIONALCOMPREHENSIVE CANCER NETWORK®, NCCN®, and NCCN GUIDELINES® aretra demarks owned by the National Comprehensive Cancer Network, Inc.” To view the mostrecent and complete version of the Compendium, go online to . Accessed March2018.3.Boccadoro M, Bringhen S, Gaidano G, et al, “Bortezomib, Melphalan, Prednisone, andThalidomide (VMPT) Followed by Maintenance With Bortezomib and Thalidomide (VT) forInitial Treatment of Elderly Multiple Myeloma Patients,” J Clin Oncol, 2010, 28(7s):8013[abstract 8013 from 2010 ASCO Annual Meeting].4.Palumbo A, Bringhen S, Rossi D, et al, “Bortezomib, Melphalan, Prednisone andThalidomide (VMPT) Followed by Maintenance With Bortezomib and Thalidomide for Initial Treatment of Elderly Multiple Myeloma Patients,” Blood, 2009, 114(22):128 [abstract 128from ASH 2009 Annual Meeting].5.Ghobria l IM, Hong F, Padmanabhan S, et al, “Phase II Trial of Weekly Bortezomib inCombination With Rituximab in Relapsed or Relapsed and Refractory WaldenstromMacroglobulinemia,” J Clin Oncol, 2010, 28(8):1422-8.6.First Coast Service Options, Inc. Local Coverage Determination (LCD): Bortezomib(Velcade®) (L33273) Centers for Medicare & Medicare Services, Inc. Updated on 2/2/2018 with effective date 2/8/2018. Accessed March 2018.7.National Government Services, Inc. Local Coverage Article for Bortezomib – Related to LCDL33394 (A52371). Centers for Medicare & Medicaid Services, Inc. Updated on 1/26/2018 with effective date of 2/1/2018. Accessed March 2018.8.Referenced with permission from the NCCN Drugs & Biologics Compendium (NCCNCompendium®) T-Cell Lymphomas. Version 3.2018. National Comprehensive CancerNetwork, 2018. The NCCN Compendium® is a derivative work of the NCCN Guidelines®.NATIONAL COMPREHENSIVE CANCER NETWORK®, NCCN®, and NCCNGUIDELINES® are trademarks owned by the National Comprehensive Cancer Network, Inc.” To view the most recent and complete version of the Compendium, go online to. Accessed March 2018.9.Zinzani PL, Musuraca G, Tani M, et al. Phase II trial of proteasome inhibitor bortezomib inpatients with relapsed or refractory cutaneous T-cell lymphoma. J Clin Oncol 2007;25:4293-4297.Appendix 1 – Covered Diagnosis CodesDual coding requirements:Codes Z85.72 & Z85.79 are secondary codes and must be billed in conjunction with a primary code Appendix 2 – Centers for Medicare and Medicaid Services (CMS)Medicare coverage for outpatient (Part B) drugs is outlined in the Medicare Benefit Policy Manual (Pub. 100-2), Chapter 15, §50 Drugs and Biologicals. In addition, National Coverage Determination (NCD) and Local Coverage Determinations (LCDs) may exist and compliance with these policies is required where applicable. They can be found at: /medicare-coverage-database/search/advanced-search.aspx. Additional indications may be covered at the discretion of the health plan.Medicare Part B Covered Diagnosis Codes (applicable to existing NCD/LCD):。
文章编号:1000-5404(2007)04-0311-03论著不同剂量环磷酰胺对裸鼠A549细胞移植瘤凋亡的影响盛伟利,梅同华 (重庆医科大学附属第二医院呼吸内科,重庆400010) 提 要:目的 探讨小剂量环磷酰胺对裸鼠肺腺癌移植瘤凋亡的影响。
方法 建立裸鼠人肺腺癌移植瘤模型,不同剂量环磷酰胺干预,测定抑瘤效果和副作用,免疫组化检测NF-κB P65表达,电镜、TUNEL检测肿瘤细胞凋亡情况。
结果 小剂量组经环磷酰胺处理后肿瘤体积与大剂量组和对照组相比明显减小;电镜下出现典型凋亡小体,凋亡指数明显增高(P<0.05)而P65蛋白表达比对照组显著减少(P<0.05)。
结论 小剂量环磷酰胺能抑制肺癌移植瘤增长,诱导肿瘤细胞凋亡,可能与下调N F-κB信号传导途径有关。
关键词:肺肿瘤;凋亡;环磷酰胺 中图法分类号:R73-361;R734.2;R979.1文献标识码:AE ffect of lo w-dose cycl ophospha m i d e on cell apoptosis of m i planted hu m an l ung car-ci no ma i n nude m i ceSH E NG W ei-li,ME I Tong-hua(Depart m ent of Re s p ira tory D isease s,The Second A ffilia t ed Ho s p ita l,Chongqi ng M edical U niversity, Chongqing400010,Ch i na) Abstract:Obj e c ti v e To explore the e ffec t of lo w-dose c y clopho spha m ide(CTX)on apoptosis of i m plan-ted hu m an l u ng carci n o m a i n nude m ice.M ethods B ALB/c nude m ice bea ring lung carcino m a w ere rando m-ized into severa l groups,each receiv ing diffe r ent do se of CTX respective ly.W e investigated an titum or effect and tox icity of CTX.A t t h e end o f expe ri m ent,tum orsw ere r esected fo r i m m unohist o che m ica l staining.Nuclear fac-t o rκB(NF-κB P65)w as detected and apoptosis w as m easu r ed by TUNEL.M orpho log ica l changes o f tumo r ce lls w ere observed.Results The volum e of tum ors w asm uch s m aller in m ice receiv i n g continuous lo w-do se CTX than t h at r eceiving m axi m all y tolerated dose o fCTX and no r m al sali n e(contr o l g r oup).The apop t o tic i n-dex w as i n creased and NF-κB P65decr eased in t h e m ice receiv ing lo w-do se of CTX.Concl u si o n The conti n-uous lo w-dose of CTX inhibits t h e gro w t h o f i m plan t e d t u m o r and increases tum or ce ll apop t o sis,wh ich m ay be due to t h e do w n-r egu lati o n of nuclear factorκB. K ey w ords:l u ng neop las m s;apop t o sis;cyclophospha m i d e 环磷酰胺是目前临床治疗肺癌常用的化疗药物,常规化疗产生严重副作用。
紫杉醇(PTX)在食管癌同期放化疗中的研究进展邓家营【摘要】紫杉醇(paclitaxel,PTX)是食管癌化疗中最有活性的药物之一,因其独特的作用机制和良好的耐受性,被广泛与顺铂(cisplatin,DDP)、5氟尿嘧啶(fluorouracil,5 Fu)等联合应用.PTX还有一定的放射增敏作用,作为放射增敏剂之一常被用于同期放化疗.在临床上,PTX类药物有不同的制剂类型和用药方案,本文就其在食管癌放化疗中的研究进展,在不同方案中的作用以及应用中存在的问题作一综述.【期刊名称】《复旦学报(医学版)》【年(卷),期】2014(041)005【总页数】5页(P697-700,705)【关键词】食管肿瘤;同期放化疗;紫杉醇(PTX)【作者】邓家营【作者单位】复旦大学附属肿瘤医院放疗科上海200032;复旦大学上海医学院肿瘤学系上海200032【正文语种】中文【中图分类】R735.1食管癌是我国常见的恶性肿瘤之一,也是世界范围内常见的癌症死亡原因。
因其早期症状不明显,30%~40%的食管癌确诊时已属无法手术切除的局部晚期。
食管癌单纯放疗效果不佳,5年生存率约为20%,局部未控和复发可达60%~80%。
单纯的化疗又很少获得病理完全缓解,且缓解期较短。
所以临床上更多地应用同期放化疗来治疗局部区域性食管癌非手术的患者。
顺铂(cisplatin,DDP)联合5-氟尿嘧啶(5-fluorouracil,5-Fu)是经典化疗方案,但是该方案患者耐受性差且疗效不理想。
探索更加高效、低毒并有放射增敏作用的新化疗药物是进一步提高食管癌同期放化疗疗效的重要途径之一。
紫杉醇(paclitaxel,PTX)是一种二萜类生物碱,主要从红豆杉树皮中分离提取。
作为抗肿瘤药,PTX具有独特的作用机制,一方面它可与β-微管蛋白结合并促进其聚合,诱导有丝分裂阻滞于G2/M期,从而使细胞出现分裂性死亡;另一方面,它还可以影响信号通路诱发细胞凋亡。
替莫唑胺治疗脑肿瘤的研究进展张垒;李延辉;姜晓东;王宁;霍志慧;刘亚东;王志强;郝丹丹【摘要】替莫唑胺(temozolomide,TMZ)为烷化剂-咪唑四嗪类衍生物家族中的一员,脂溶性比较好,所以容易透过血脑屏障,针对这一特点,临床已将其作为治疗脑肿瘤的主要药物.本文将从替莫唑胺在脑肿瘤治疗中的应用、与其他药物的联合使用等方面进行阐述.【期刊名称】《赤峰学院学报(自然科学版)》【年(卷),期】2017(033)015【总页数】2页(P66-67)【关键词】替莫唑胺;胶质瘤;疗效【作者】张垒;李延辉;姜晓东;王宁;霍志慧;刘亚东;王志强;郝丹丹【作者单位】赤峰学院附属医院;赤峰学院附属医院;赤峰学院附属医院;赤峰学院附属医院;赤峰学院附属医院;赤峰学院附属医院;赤峰学院附属医院;赤峰学院医学院,内蒙古赤峰 024000【正文语种】中文【中图分类】R739脑胶质瘤占颅内肿瘤的41%~44.6%,其是来源于神经外胚层的肿瘤,也被称为神经外胚层肿瘤或神经上皮肿瘤[1].脑胶质瘤具有发病率高、复发率高、病死率高、治愈率低的特点[2].胶质瘤的分类方法很多,临床常用Kernohan分类法.在不同的胶质瘤中,星形细胞瘤为最多,第二位的为胶质母细胞瘤,最后为髓母细胞瘤、室管膜瘤等.根据世界卫生组织(WHO)胶质瘤可分为Ⅰ~Ⅳ级,前两级属于低级,而Ⅲ、Ⅳ属于高级.男性明显多于女性.在临床上可依据患者的年龄、性别、脑胶质瘤生物特征及临床症状进行分析,同时结合病史及辅助检查(如超声波、放射性核素、放射学及核磁共振等)而对其进行诊断.目前,大部分脑胶质瘤治疗多以手术为主,术后辅助进行放疗和化疗[3].传统的化疗药物有卡莫司汀、洛莫司汀、顺铂、环磷酰胺等.由于很多药物不容易透过血脑屏障,从而限制了一些对癌症有较好效果的化疗药到达中枢神经系统,使得在中枢神经系统中大多数化疗药仅能达到相对低的浓度.且一些药物的异质性、耐药性以及全身副作用使以上药物化疗效果很不理性.近几年来,几种可通过血脑屏障的新抗癌药研发成功,目前用于临床的有亚硝基脲、长春新碱和替莫唑胺等.3.1 替莫唑胺的药理作用替莫唑胺是一种小分子、具有脂溶性的烷化剂,单药治疗脑胶质瘤显示了一定得作用.动物实验研究显示,替莫唑胺具有较好的透过血脑屏障的特性,其中枢浓度大约能到达血浆浓度的1/3~2/5[4,5].与传统的烷化剂类相比,替莫唑胺口服可迅速吸收,进入体内不经肝脏代谢广泛分布于全身,通透性良好,且可在脑肿瘤组织中优先聚集,在生理PH值的条件下,能自发快速的转换为活性化合物甲基三氮烯咪唑酰胺(Methyl triazene imidazole amide,MTIC),之后水解为甲基肼和5-氨基咪-4-酰胺(AIC),可作用于各个分裂时期的肿瘤细胞,将肿瘤细胞DNA 进行烷基化修饰,从而产生细胞毒作用[6].另外,TMZ还可以诱导肿瘤细胞周期停滞于G2~M期,使细胞周期同步化,从而增加肿瘤组织对放疗的敏感性[7].刘丽华等[8]研究显示,提前应用替莫唑胺预处理大鼠脑胶质瘤C6细胞后,可使该细胞的促凋亡蛋白Bax表达增加,而抑制凋亡蛋白Bcl-2表达降低,从而诱导脑胶质瘤C6细胞凋亡.常顺[9]等研究发现替莫唑胺不但通过促进活化凋亡信号通路p53/Bax可以促进脑胶质瘤C6细胞的凋亡,其还可以抑制脑胶质瘤C6细胞的增殖.替莫唑胺的以上生活学特性使其成为治疗脑胶质瘤的理想药物.3.2 临床研究情况(1)用替莫唑胺治疗术后高级别的胶质瘤患者34例[10],对术后的高级别胶质瘤患者单用TMZ化疗,平均无进展生存时间大约为10.08±6.4个月,中位无进展生存时间大约为8个月.6个月肿瘤无进展生存率大约为73.54%,9个月肿瘤无进展生存率大约为50.00%.1年肿瘤无进展生存率大约为35.29%.平均生存时间大约为12.06±6.25个月,中位生存时间大约为10个月,9个月生存率大约为76.47%,1年生存率大约为38.24%.另外,在一些临床研究报道中发现[11,12],替莫唑胺胶囊对于恶性脑胶质瘤的治疗具有较好的临床效果.TMZ治疗脑胶质瘤的有效率和生存率比较高,耐受性好,而且还可以逆转O-6-甲基鸟嘌呤-DNA甲基转移酶(O6-methylguanine-DNA-methlyltransferase,MGMT)阳性患者的耐药性,不良反应少.均显示对患者的生存时间能够显著延长,且安全性较高[13].替莫唑胺的这种作用对于年龄70岁的患者也同样有效[14].与其它化疗药物(卡莫司汀,福莫司汀等)相比,替莫唑胺对胶质母细胞瘤化疗效果更好,生存率明显高于卡莫司汀且副作用小,而耐受性好[15,16].(2)有学者对三维适形放疗联合替莫唑胺治疗与单纯适形放疗治疗恶性脑胶质瘤术后患者在疗效和安全性进行对比临床研究[17],结果显示,放疗联合替莫唑胺治疗恶性脑胶质瘤术后患者近期疗效与单纯放疗的疗效差不多,但是与单纯放疗组中位复发时间(16.3±10.4个月)相比,联合替莫唑胺组中位复发时间(22.8±8.9个月)延缓,而且治疗组1、2、3年的生存率分别比治疗组大约提高14.3%、42.6%、 42.8%.这一结果与王[18]等的研究结果一致,三维适形放疗联合替莫唑胺治疗顽固性脑胶质瘤可以防止肿瘤的进展,使患者症状缓解.联合治疗后不良反应发生较少,生活质量有所提高[17,18,19].另外,有研究报道[11]脑胶质瘤术后有残余病灶患者21例,术后采用调强放射治疗(intensity-modulated radiationtherapy,IMRT)联合同步及辅助替莫唑胺化疗治疗,按照世界卫生组织(WHO)病理分级,Ⅱ级10例,Ⅲ~Ⅳ级11例,在同期放化疗4周结束后,然后继续口服替莫唑胺化疗2周.结果显示,Ⅱ级患者1年无进展生存率100%,总的生存率也为100%;Ⅲ~Ⅳ级患者有效率约为81.8%,其中1年无进展生存率为72.7%,总生存率为81.8%.可以看出,IMRT联合同步及辅助替莫唑胺化疗治疗脑胶质近期有效率更好.上述的临床研究结果提示,替莫唑胺能够很大程度上提高患者生存率,不良反应小,与其它疗法联合治疗脑胶质瘤更是一种比较好的治疗方法.欧洲癌症研究治疗组织和加拿大癌症研究院Ⅲ临床试验结果和徐彬等研究结果也支持这一观点[19,20].综上所述,替莫唑胺单纯治疗或者联合其它化疗治疗脑胶质瘤术后患者疗效更好,不良反应少,生存率高.但是,现有的临床试验,观察时间短,对患者较为长期的生存率以及长期的治疗毒性的研究仍需要进一步更深入的研究.【相关文献】〔1〕孙燕,赵平,吴孟超,等.临床肿瘤学进展[M].北京:中国协和医科大学出版社,2006.1087.〔2〕雷霆.脑肿瘤学 [M].北京:中国医药科技出版社,2005.512-515.〔3〕陈正和,陈忠平.脑胶质瘤的治疗进展[J].新医学,2015,46(7):417-422.〔4〕Stupp R,Dietrich PY,OstermannK raljevic S,etal.Promising survivalforpatientswith newly diagnosed Glioblastoma multiforme treated with concomitant radiation plus temozolomide followed by adjuvant temozolomide[J].J Clin Oncol,2002,20(5):1375-1382.〔5〕Wen P Y.Therapy for recurrent high grade gliomas: does continuous dose intense temozolomide have a role [J].J Clin Oncol,2010,28(12):1977-1979.〔6〕唐曦,胡娅,徐炎华.三维适形放疗联合替莫唑胺治疗恶性脑胶质瘤的临床研究 [J].现代生物医学进展,2009,9(15):2885-2886.〔7〕Tuettenberg J,Grobholz R, Korn T,et al.Continuous low-dose chemotherapy plus inhibition of cyclooxygenase-2 asan antiangiogenic therapy ofglioblastomamultiforme [J].J Cancer Res Clin Oncol,2005,131(1):31-40.〔8〕刘丽华,张明.替莫唑胺诱导大鼠脑救治瘤C6细胞caspase非依赖的程序性死亡[J].J south Med univ,2015,35(2):229-233.〔9〕常顺,刘永军,周虎,等.替莫唑胺抑制大鼠C6胶质瘤增殖及对P53、caspase-3蛋白表达的影响[J].中国地方病防治杂志,2016,31(11):1276-1277.〔10〕朱正权,刘亮,夏海成,等.替莫唑胺治疗34例高级别胶质瘤的临床疗效观察 [J].中国现代药物应用,2011,5(18):6-7.〔11〕印海林,吴伟莉,金风,等.调强放射治疗联合同步及辅助替莫唑胺化疗治疗脑胶质瘤术后残余病灶的临床研究[J].肿瘤,2011,31(5):428-431.〔12〕Stupp R,Mason WP,Van den Bent M J,et al.Radiotherapy plus Concomitant and Adjuvant Temozolomide for Glioblastoma[J].N Engl J Med,2005,352(10):987 -996.〔13〕李方明,田增发,聂青,等.恶性脑胶质瘤TMZ联合同步放疗临床疗效探讨[J].中国肿瘤临床,2009,36(13):721-724.〔14〕傅浩,万林林,杨灵,等.放疗联合口服替莫唑胺治疗恶性脑胶质瘤术后患者的临床疗效研究[J].Chinese General Practice,2011,14(5):1556-1558.〔15〕王计伟,陈步东,张春智,等.替莫唑胺治疗胶质母细胞瘤的长期疗效评价[J].中华神经医学杂志,2012,11(1):57-60.〔16〕武新虎,朱锡旭,沈泽天,等.替莫唑胺与福莫司汀治疗恶性脑胶质瘤的临床 [J].观察现代肿瘤医学,2012,20(6):1165-1168.〔17〕王世伟,陈福群,张春荣,等.替莫唑胺联合三维适形放疗治疗顽固性脑胶质细胞瘤的临床研究[J].中国实用医药,2011,6(3):179.〔18〕王琼,王南瑶,盛华明,等.三维适形放疗联合替莫唑胺治疗恶性脑胶质瘤的临床观察[J].中华肿瘤防治杂志,2008,15(11):843–845.〔19〕Perry JR,Belanger K,Mason WP,et al.PhaseⅡ Trial of Continuous Dose-Intense Temozo-lomide in Recurrent Malignant Glioma:RESCUE Study [J].J Clin Oncol,2010,68(2):582.〔20〕徐彬,杨超,张弩,等.三维适行放疗联合替莫唑胺治疗脑胶质瘤的观察[J].医药论坛杂志,2016,37(9):169-170.。
胃的胃肠间质瘤【概述】1.胃肠间质瘤(gastrointestinal stromal tumor,GIST)是胃肠道最常见的间叶源性肿瘤。
2.可发生在消化道的任何部位,主要发生在胃,约60%左右。
3.临床症状不典型,难以早期确诊。
4.通常瘤组织CD117 、 CD34免疫组化表达阳性。
【病因和发病机制】病因未明,几乎均为散发病例。
GIST细胞KIT或PDGFRA发生功能获得性突变,导致其表达的产物酪氨酸激酶跨膜受体蛋白持续活化,是GIST病理发生的关键,并成为最佳的治疗靶点。
【病理】组织学上GIST由梭形细胞、上皮样细胞、偶或多形性细胞组成,依据细胞形态可分为三大类:梭形细胞型(70%)、上皮样细胞型(20%)和梭形细胞/上皮样细胞混合型(10%)。
约95%的病例免疫组化CD117(和/或DOG-1)表达阳性。
结合组织形态学和免疫组化CD117表达阳性,是病理诊断的关键。
GIST的病理报告中,应至少包含肿瘤大小、部位和核分裂像,核分裂像应选择肿瘤细胞核分裂发生最集中和最多的局部,并累计其中50个高倍视野的核分裂总数。
有条件的单位,应对瘤组织作KIT和PDGFRA基因突变检测。
【临床表现】1.症状:GIST的症状均为非特异性。
(1)消化道出血是最常见的症状表现。
常见为隐蔽的慢性失血,病人可有贫血症状。
部分表现为急性上消化道大出血。
(2)可有非特异性的溃疡样疼痛。
(3)部分病人表现为上腹部包块。
(4)大约20%的GIST没有明显症状,而在体检或进行腹部手术时偶然发现。
(5)有时候一开始就表现为消化道穿孔或腹腔内出血的症状。
2.体征(1)患者表情可较为疲倦;有长期慢性出血的患者可有贫血貌;长期纳差的患者可有体重下降、消瘦、营养不良的表现。
(2)当存在较大的肿瘤时,可触及腹部包块和/或有压痛。
(3)淋巴结肿大极其罕见。
【实验室检查】(1)一般检查:慢性出血时血常规化验会发现血红蛋白、红细胞压积减少。
大便潜血可阳性。
胃肠道间质瘤基因突变及靶向治疗进展韩振宇;郑吉阳;周威;王虎;杨建军【期刊名称】《西南国防医药》【年(卷),期】2018(028)006【总页数】3页(P599-600,封3)【关键词】胃肠道间质瘤;基因突变;靶向治疗;耐药【作者】韩振宇;郑吉阳;周威;王虎;杨建军【作者单位】710032西安,第四军医大学西京医院消化病医院消化外科;710032西安,第四军医大学西京医院消化病医院消化外科;710032西安,第四军医大学西京医院消化病医院消化外科;710032西安,第四军医大学西京医院消化病医院消化外科;710032西安,第四军医大学西京医院消化病医院消化外科【正文语种】中文【中图分类】R735胃肠道间质瘤(GIST)是胃肠道最常见的间叶源性肿瘤,c-Kit和PDGFRA基因突变是GIST最常见的发病机制。
以伊马替尼为代表的酪氨酸激酶抑制剂在GIST治疗中的成功应用,使GIST成为分子靶向治疗的典范。
近年来,在GIST的发病机制、药物治疗、耐药机制等领域的研究中取得了很多进展,笔者将做一简要综述。
1 GIST基因突变1.1 c-Kit基因突变 1998年,Hirota等[1]首次在GIST中发现c-kit基因突变,突变导致酪氨酸激酶可在无配体干细胞因子结合的情况下自发持续活化,激活下游信号传导,从而提示c-kit基因突变是GIST的发病机制。
在GIST中,c-kit基因突变占75%~80%,其中11外显子突变最常见(65%~70%),外显子 9 突变次之(10%),13、17外显子突变少见[2]。
11外显子编码胞内近膜区KIT蛋白,通过阻止激酶活化环活化从而实现自抑构象,11外显子突变破坏自抑机制,使激酶发生自发活化。
在GIST中,突变方式包括点突变、缺失突变和插入突变。
11外显子基因突变GIST可以发生在胃肠道的任何部位,常见于胃,组织学上特异性的表现为梭形细胞,其中缺失突变预后较差。
9外显子编码胞外近膜配体结合结构域,突变后通过模拟与配体结合后的构象变化使激酶活化,9外显子主要突变方式为插入突变,大多发生于小肠和大肠GIST。
低剂量吉西他滨持续静脉滴注在晚期非小细胞肺癌维持治疗中的应用陈俊;陶庆松;庞林荣;李晖;徐彩虹;黄佳;郑宏瑜;杨成【摘要】目的:观察一线诱导化疗后低剂量吉西他滨持续静脉滴注维持化疗治疗晚期非小细胞肺癌(NSCLC)的疗效和安全性。
方法对收治的66例初治ⅢB/Ⅳ期NSCLC患者,采用吉西他滨1000mg/m2(第1、8天)联合顺铂75mg/m2(分3d使用)方案诱导化疗,21d为1个周期。
4个周期后有效或稳定的36例按2:1随机分为维持组(24例)和观察组(12例),维持组予低剂量吉西他滨(250mg/m2)6h持续静脉滴注,第1、8天使用,21d为1个周期,维持至疾病进展或患者对化疗毒性不能耐受。
观察组停止化疗,观察直至疾病进展。
结果维持组和观察组至疾病进展时间(TTP)分别为6.2个月和4.8个月(P=0.039);中位总生存时间分别为13.1个月和10.8个月(P=0.667)。
维持化疗不良反应较轻微,主要表现为中性粒细胞、血小板数降低、贫血及恶心、呕吐。
结论常规剂量吉西他滨联合顺铂一线化疗后序贯低剂量吉西他滨持续静脉滴注维持化疗安全、有效,应可进一步扩大样本量行研究是否有总生存时间的获益。
%Objective To evaluate the efficacy and safety of prolonged infusion of low- dose gemcitabine in maintenance treatment for advanced non- smal celllung cancer (NSCLC) after first line regimen. Methods Sixty six patients with stageⅢB/Ⅳ NSCLC admitted from March 2008 to October 2009 were enrolled. The patients received gemcitabine 1000 mg/m2 (d1 and d8)plus cisplatin 25 mg/m2 (d1, d2 and d3) every 21 d. Thirty six patients achieving objective response or stable disease fol ow-ing initial gemcitabine plus cisplatin regime were randomized to receive maintenance gemcitabine(continuous injection for 6h with a dose of 250 mg/m2 on d1 and d8, every 21d)plus best supportive care (maintenance arm), or best supportive care only (control arm). Results The time to progress (TTP) throughout the study period was 6.2 and 4.8 months for maintenance and control arms, respectively (P=0.039). Median overal survival (OS) was 13.1 months for maintenance and 10.8 months for control arms(P=0.667). The toxicity profile was mild,with neutropenia,thrombocytopenia,anemia,nausea and vomit being most common toxicities. Conclusion Maintenance treatment with low- dose gemcitabine,fol owing first line regime is feasible and it produces significantly longer TTP compare to best supportive care alone.【期刊名称】《浙江医学》【年(卷),期】2013(000)020【总页数】4页(P1823-1826)【关键词】非小细胞肺癌/维持化疗;低剂量;吉西他滨【作者】陈俊;陶庆松;庞林荣;李晖;徐彩虹;黄佳;郑宏瑜;杨成【作者单位】315040 宁波大学附属鄞州人民医院肿瘤放化疗中心;315040 宁波大学附属鄞州人民医院肿瘤放化疗中心;315040 宁波大学附属鄞州人民医院肿瘤放化疗中心;315040 宁波大学附属鄞州人民医院肿瘤放化疗中心;315040 宁波大学附属鄞州人民医院肿瘤放化疗中心;315040 宁波大学附属鄞州人民医院肿瘤放化疗中心;315040 宁波大学附属鄞州人民医院肿瘤放化疗中心;315040 宁波大学附属鄞州人民医院肿瘤放化疗中心【正文语种】中文目前,每年有超过100万的患者死于肺癌或肺癌引起的相关疾病。
Phase II trial of continuous low-dose temozolomide for patients with recurrent malignant gliomaAntonio Omuro,Timothy A.Chan,Lauren E.Abrey,Mustafa Khasraw,Anne S.Reiner,Thomas J.Kaley,Lisa M.DeAngelis,Andrew ssman,Craig P.Nolan,Igor T.Gavrilovic,Adilia Hormigo,Cynthia Salvant,Adriana Heguy,Andrew Kaufman,Jason T.Huse,Katherine S.Panageas,Andreas F.Hottinger,and Ingo MellinghoffDepartments of Neurology (A.O.,L.E.A.,M.K.,T.J.K.,L.M.D.,A.B.L.,C.P.N.,I.T.G.,A.H.,C.S.,A.F.H.,I.M.),Radiation Oncology (T.A.C.),Human Oncology and Pathogenesis Program (T.A.C.,A.H.,A.K.,I.M.),Epidemiology and Biostatistics (A.S.R.,K.S.P.),Pathology (J.T.H.),Memorial Sloan-Kettering Cancer Center,New York,New YorkPresent affiliation:Royal Melbourne and Geelong Hospitals,Australia (M.K.);University of Zurich,Switzerland (L.E.A.);Columbia University Medical Center,New York,New York (A.B.L.);University of Rochester Medical Center,Rochester,New York (A.H.);Centre Hospitalier Universitaire Vaudois and University of Lausanne,Switzerland (A.F.H.);Upstate Medical University,Syracuse,New York (S.C.)Background.In this phase II trial,we investigated the efficacy of a metronomic temozolomide schedule in the treatment of recurrent malignant gliomas (MGs).Methods.Eligible patients received daily temozolomide (50mg /m 2)continuously until progression.The primary endpoint was progression-free survival rate at 6months in the glioblastoma cohort (N ¼37).In an ex-ploratory analysis,10additional recurrent grade III MG patients were enrolled.Correlative studies included eval-uation of 76frequent mutations in glioblastoma (iPLEX assay,Sequenom)aiming at establishing the frequency of potentially “drugable”mutations in patients entering re-current MG clinical trials.Results.Among glioblastoma patients,median age was 56y;median Karnofsky Performance Score (KPS)was 80;62%of patients had been treated for ≥2recurrences,including 49%of patients having failed bevacizumab.Treatment was well tolerated;clinical benefit (complete response +partial response +stable disease)was seen in 10(36%)patients.Progression-free survival rate at 6months was 19%and median overall survival was 7months.Patients with previous bevacizumab exposure survived significantly less than bevacizumab-naive pa-tients (median overall survival: 4.3mo vs 13mo;hazard ratio ¼3.2;P ¼.001),but those patients had lower KPS (P ¼.04)and higher number of recurrences (P ,.0001).Mutations were found in 13of the 38MGs tested,including mutations of EGFR (N ¼10),IDH1(N ¼5),and ERBB2(N ¼1).Conclusions.In spite of a heavily pretreated population,including nearly half of patients having failed bevacizu-mab,the primary endpoint was met,suggesting that this regimen deserves further investigation.Results in bevaci-zumab-naive patients seemed particularly favorable,while results in bevacizumab-failing patients highlight the need to develop further treatment strategies for ad-vanced MG.Clinical identifier .NCT00498927(available at /ct2/show /NCT00498927)Keywords:bevacizumab,glioblastoma,malignant glioma,metronomic chemotherapy,temozolomide.Radiotherapy with concomitant temozolomide fol-lowed by adjuvant temozolomide is the standard of care for newly diagnosed glioblastoma,1butthe optimal regimen for recurrent disease has not been defined.Clinical trials in recurrent glioblastoma haveCorresponding Author:Antonio Omuro,MD,Memorial Sloan-Kettering Cancer Center,1275York Avenue,New York,NY 10065(omuroa@).Received August 13,2012;accepted October 17,2012.Neuro-Oncology 15(2):242–250,2013.doi:10.1093/neuonc /nos295NEURO-ONCOLOGYAdvance Access publication December 14,2012#The Author(s)2012.Published by Oxford University Press on behalf of the Society for Neuro-Oncology.All rights reserved.For permissions,please e-mail:journals.permissions@.at China Pharmaceutical University on December 5, 2016/Downloaded fromreported dismal outcomes,with objective response rates (ORRs)of5%–6%,6-month progression-free survival rates(PFS6)of9%–21%,and median overall survival (OS)of6–7months.2–6Treatment with numerous targeted and nontargeted agents has been attempted without success,with the exception of bevacizumab.7–9 This agent received accelerated FDA approval for recurrent glioblastoma,based on improved ORRs of 20%–26%seen in2phase II trials.8,9However,re-sponses tend to be short-lived(median:4months),and OS has been modest(median:8–10months).Metronomic temozolomide has emerged as a well-tolerated salvage approach in recurrent glioblastoma.10–15 Preclinical studies have suggested that metronomic temozolomide may result in increased activity through depletion of the O6-methylguanine-DNA methyltransfer-ase(MGMT)protein16and anti-angiogenic properties through targeting of endothelial cells.17,18A phase II study19using metronomic,daily temozolomide(50mg/ m2/d)for glioblastoma atfirst relapse investigated3pre-established cohorts:patients with progression before com-pletion of6adjuvant cycles(“early”group),patients with progression after6cycles while still on temozolomide(“ex-tended”group),and patients who discontinued temozolo-mide after completing6cycles without progression (“rechallenge”group).The extended group did not seem to benefit(PFS6:7%),but the early and rechallenge groups achieved PFS6of27%and36%,respectively. However,these results are difficult to interpret because there are no historical controls for either of these cohorts.Moreover,results in the early group could have represented inclusion of patients with pseudoprogression, while patients in the rechallenge group could have har-bored tumors with a different biology.It is also unclear how these results apply to current treatment trends in the United States,where temozolomide tends to be continued beyond6cycles and the use of bevacizumab is frequent.We report a phase II trial using a similar regimen of temozolomide50mg/m2daily but encompassing eligi-bility criteria similar to available historical controls and typical clinical trials in glioblastoma,4,20with no re-striction on timing of failure with respect to previous temozolomide use.We also performed an exploratory, comprehensive disease-specific mutational analysis to characterize the typical population of patients entering studies of recurrent malignant gliomas(MGs)from a molecular perspective.Patients and MethodsThis was a prospective,phase II trial conducted at Memorial Sloan-Kettering Cancer Center from July 2007to April2010.The main cohort comprised patients with progressive/recurrent glioblastoma;patients with recurrent World Health Organization grade III anaplas-tic astrocytoma,anaplastic oligoastrocytoma,or ana-plastic oligodendroglioma were enrolled in a separate, exploratory cohort.Inclusion criteria consisted of histologically con-firmed diagnosis of MG,previous exposure to temozolomide and radiotherapy,unequivocal tumor re-currence at enrollment,age≥18years,Karnofsky Performance Score(KPS)≥60,and adequate bonemarrow and organ function.Patients with other,concur-rent active malignancy or serious medical illness were excluded.There was no limit on number of previous re-currences or chemotherapy regimens.Treatment consisted of temozolomide given at a dose of50mg/m2daily,without interruption,until progression or development of unacceptable side effects.One cycle wasdefined as28days.Anti-emetic therapy and pneumocysto-sis prophylaxis were prescribed at the discretion of treating physicians.Treatment was held for toxicity grades≥3.Once resumed,the dose of temozolomide could bereduced by25%as clinically indicated,but if further reduc-tions were necessary,patients were removed from the study.Complete blood counts were repeated weekly,and compre-hensive chemistry panels were obtained monthly.MRIs andclinical exams were performed every2months.Responseswere evaluated using Macdonald criteria.21The studywas approved by the institutional review board;written in-formed consent was obtained from all patients.StatisticsThe primary endpoint was PFS6in glioblastoma pa-tients.A Simon2-stage design was used.In thefirststage,12patients were accrued.If at least one of thosepatients did not progress at the6-month mark,then an additional25patients(for a total of37)would beaccrued to the second stage;otherwise,the trial wouldbe terminated.At the end of the trial,if fewer than4pa-tients were progression free at6months,the studywould be declared negative.This design yields at leasta.90probability of a positive result if the true PFS6was at least20%and a.90probability of a negativeresult if the true PFS6was5%.Secondary endpoints were median PFS,OS,ORR,and toxicity as per the National Cancer Institute’sCommon Terminology Criteria of Adverse Eventsversion3.0.Survival was calculated from registrationdate(Kaplan–Meier methodology),and log-rank statis-tic was utilized for group comparisons.An additional10patients with grade III MG were enrolled in an explor-atory cohort,and a descriptive analysis was performed.MGMT Promoter Methylation Status and Evaluationof Relevant Glioblastoma MutationsAvailable formalin-fixed paraffin-embedded(FFPE)tissuesamples were evaluated for MGMT promoter methyla-tion status by real-time methylation-specific PCR as described previously,22performed by MDxHealthC.FFPE samples were also subjected to a comprehensive mu-tation screening using mass spectrometry genotyping/iPLEX assay(Sequenom)in genomic DNA,looking fora panel of76mutations described in glioblastoma (Supplementary material,Appendix S1).The iPLEXassay was performed as described by the manufacturer (Supplementary material,Appendix S2).Primers arelisted in Supplementary material,Appendix S3.Omuro et al.:Metronomic temozolomide for recurrent glioblastomaNEURO-ONCOLOGY†F E B R U A R Y2013243at China Pharmaceutical University on December 5, 2016/Downloaded fromResultsPatient CharacteristicsAccrual was completed as planned,with all37glioblas-toma patients enrolled,constituting the main study cohort.Ten additional grade III recurrent MG patients were accrued to the exploratory cohort.Patient charac-teristics are summarized in Table1.Among the glioblastoma patients,32(86%)had recurrence of a de novo glioblastoma and5(14%)had recurrent secondary glioblastoma(ie,initial diagnosis was of a grade2or3glioma,which recurred as a glio-blastoma histologically proven).Patients were heavily pretreated by the time of enrollment(Table1).All glioblastoma patients had re-ceived radiotherapy with concomitant temozolomide. Thirty(81%)patients had a history of tumor progres-sion diagnosed during adjuvant temozolomide,and7 (19%)had never progressed during temozolomide, having previously received a median of12cycles.The number of prior recurrences was1in14(38%)patients,2in11(30%),and3or more in12(32%).Other prior chemotherapies consisted of bevacizumab in18(49%) patients,other cytotoxic agents in9(24%),and experi-mental targeted therapies in8(21%).In all patients with bevacizumab exposure,bevacizumab had been given as salvage treatment,and all of those patients experienced progression while on bevacizumab.Glioblastoma Cohort—Response,PFS,and OS Patients who had measurable tumor and completed at least one cycle of treatment were considered evaluable for response(N¼28).A partial response(PR)was seen in3(11%)patients,stable disease(SD)in7 (25%),and progressive disease(PD)in18(64%);clini-cal benefit(CR+PR+SD)was seen in10(36%)pa-tients.Among the9patients considered nonevaluable for response,1had no measurable disease due to surgical resection and8did not complete thefirst cycle.All patients(N¼37)were included in the PFS and OS analysis,in an intent-to-treat fashion.A total of7pa-tients were progression free and alive at6months,andTable1.Patient characteristicsGlioblastoma(N537)Grade III MalignantGliomas(N510) GenderMen12(32%)6(60%) Women25(68%)4(40%)Median age(range)56(31–81)58(29–73) Median KPS(range)80(60–100)80(60–100) Histology at time of enrollmentDe novo glioblastoma32(86%)–Secondary glioblastoma5(14%)–Grade III anaplastic astrocytoma–4(40%) Grade III anaplastic oligodendroglioma–5(50%) Grade III anaplastic oligoastrocytoma–1(10%) Median time between current histologic diagnosisand enrollment(range)20mo(0.3–58)18mo(0.5–177) Number of previous recurrences114(38%)4(40%) 211(30%)3(30%)≥312(32%)3(30%) Prior salvage therapiesBevacizumab18(49%)0Other cytotoxic agents9(24%)4(40%) Experimental targeted therapies8(21%)3(30%) Progression while on previous adjuvant temozolomideProgression duringfirst6adjuvant cycles21(57%)NA Progression after.6adjuvant cycles9(24%)NA Completed adjuvant cycles without progression7(19%;median of12adjuvant cycles)NAMGMT promoter methylation statusMethylated3(8%)2(20%) Unmethylated20(54%)3(30%) Invalid MGMT results10(27%)4(40%) Tissue unavailable4(11%)1(10%) Abbreviation:NA,not applicable to grade3tumors(patients received variable chemotherapy regimens at diagnosis).Omuro et al.:Metronomic temozolomide for recurrent glioblastoma244NEURO-ONCOLOGY†F E B R U A R Y2013 at China Pharmaceutical University on December 5, 2016 /Downloaded fromtherefore the primary endpoint was met.PFS6was19% (N¼37;95%confidence interval[CI]:6–32),and median PFS was2months(95%CI:1–4)(Fig.1). Median OS was7months(95%CI:5–12),and1-year OS was35%(95%CI:20–51)(Fig.1).Median follow-up for survivors was19months.Given the previously reported poor prognosis observed after bevacizumab discontinuation/failure in retrospective studies,23–27we analyzed the impact of prior bevacizumab exposure on outcomes.Patients with bevacizumab failure survived significantly less(median OS:4.3months;6-months OS:28%)than bevacizumab-naive patients(median OS:13months;6-months OS: 84%);hazard ratio(HR)¼3.2;P¼.001(Fig.2).PFS6 in the bevacizumab-failure group was11.1%versus 26.3%in the bevacizumab-naive group(P¼.07) (Fig.3).Responses were seen in3/15(20%)evaluable bevacizumab-naive patients,and no responses were observed in the bevacizumab-failure group.Among the bevacizumab-naive group,bevacizumab was given to9 (47%)patients after progression on this trial.In an exploratory fashion,we sought to investigate whether differences in pretreatment characteristics could explain the differences in outcomes according to pre-vious bevacizumab exposure(Supplementary material, Appendix S4).In a post-hoc analysis,we evaluated the dis-tribution of potential prognostic factors in bevacizumab-naive versus bevacizumab-failure patients(categorical: Fisher’s exact test;continuous:Kruskal–Wallis test) and performed a multivariate analysis using a forward stepwise Cox proportional hazards regression model with entry and exit criteria of a¼0.20(Supplementary material,Appendix S4).Bevacizumab-failure patients had a higher number of previous progressions/ chemotherapy regimens(P,.0001)and lower KPS (P¼.04)than bevacizumab-naive patients.The median time from glioblastoma diagnosis was21months in bevacizumab-failure versus13months for bevacizumab-naive patients(P¼.18).Multivariate stepwise analysis suggested that the driving variable for decreased PFS was number of progressions/salvage chemotherapies (P¼.02),favored over other candidate variables,includ-ing bevacizumab exposure.Multivariate analysis of OS showed that number of progressions/salvage chemo-therapies(P¼.05)and KPS(P¼.05),but not prior bevacizumab exposure,were the driving variables for differences in survival.There were no differences in outcome according to previous history of progression on temozolomide(PFS: P¼.58;OS:P¼.18)or according to MGMT promoter methylation status(PFS:P¼.76;OS:P¼.14),although analysis is limited by the small number of patients in whom this could be determined accurately(Table1). Grade III Recurrent Malignant Glioma Exploratory CohortAmong the10patients with grade III MG,PFS6was 30%(95%CI:1.6–58.4);median PFS was3months. Median OS was18months;1-year OS was60%(95%Fig.1.OS and PFS(glioblastoma cohort,N¼37).PFS6:19%(95%CI:6–32);median PFS:2months;1-year OS:35%;median OS:7months.Fig.2.Glioblastoma cohort(N¼37):OS according to previoushistory of bevacizumab(BEV)treatment.Patients with previousBEV failure:median OS:4.3mo,6-mo OS:28%(95%CI:7–48);patients without BEV failure:median OS:13mo;6-monthOS:84%(95%CI:68–100);HR¼3.2;P¼.001.Fig.3.Glioblastoma cohort(N¼37):PFS according to previous bevacizumab(BEV)treatment.Patients with previous BEV failure:PFS6:11.1%(95%CI:0.0–25.6),median PFS:1.5mo;patientswithout bevacizumab failure:PFS6:26.3%(95%CI:6.5–46.1),median PFS:1.8mo;HR¼1.9;P¼.07.Omuro et al.:Metronomic temozolomide for recurrent glioblastomaNEURO-ONCOLOGY†F E B R U A R Y2013245at China Pharmaceutical University on December 5, 2016/Downloaded fromCI:30–90).One patient achieved PR,4achieved SD, and5achieved PD.ToxicityTreatment was well tolerated;no patient discontinued treatment as a result of toxicity,and there were no toxic deaths.Lymphopenia was the most frequent toxic-ity(grade3¼10patients,grade4¼1),but no lymphopenia-related infection was reported.The only other grade4toxicity was thrombocytopenia(n¼1). Grade3toxicities consisted of increased alanine amino-transferase(n¼2),fatigue(2),hyponatremia(2), hypokalemia(1),pulmonary embolism(1),neuropathy (1),and infection(1).Mutational AnalysisIn an exploratory analysis,we evaluated FFPE tissue from38patients(28glioblastomas and10grade III tumors)for the presence of76mutations previously de-scribed in gliomas using mass spectrometry genotyping/ iPLEX assay(Sequenom[Supplementary material, Appendix S1]).A total of9different mutations was found in13patients(Table2),including8(29%)glio-blastomas and5(50%)grade III MGs.Mutations were found in EGFR(N¼10),IDH1(N¼5),and ERBB2(N¼1).All IDH1mutations were found in grade III(N¼3)or secondary glioblastomas(N¼2) and none in recurrent primary glioblastomas.EGFR extracellular-domain mutations were found in both glio-blastomas(N¼7)and grade III gliomas(N¼3).None of the analyzed mutations involving phosphatidylinosi-tol3-kinase(PI3K),BRAF,or KRAS was found. Table2summarizes the mutations found,along with patient characteristics and treatment outcomes.DiscussionThis study was designed to test the efficacy of metro-nomic temozolomide in the typical population enrolled in clinical trials for recurrent glioblastoma,irrespective of prior treatments.The primary efficacy endpoint (based on pre–bevacizumab-era historical controls) was met,with19%PFS6,despite the inclusion of a heavily pretreated population,including nearly half of the patients with a history of bevacizumab failure.This regimen was therefore deemed active,warranting further investigation.Enrollment to this trial coincided with the rise of bev-acizumab for glioblastoma salvage therapy in the United States.Although this confounded the interpretation of our results in comparison with historical controls,it also provided the unique opportunity to evaluate the same cytotoxic regimen in patients who were bevacizu-mab naive or had failed bevacizumab.Results in bevacizumab-naive glioblastoma seemed particularly fa-vorable(PFS6:26%,median OS:13mo)and compara-ble to other active salvage treatments;activity was also observed in grade III tumors.Given the excellent toxicity profile and ease of drug administration,this regimencould potentially constitute a viable alternative to beva-cizumab and nitrosoureas as second-line treatment forglioblastoma and a suitable platform for combinationwith targeted agents.Conversely,bevacizumab-failurepatients achieved a much shorter PFS6of11%and median OS of4.3months,highlighting the needfor alternative therapies in this population.Several phase II trials testing intensified temozolo-mide schedules have been conducted(Table3),but theoptimal regimen remains to be defined.A direct compar-ison across trials is difficult,given the varying inclusioncriteria and study settings,but overall it appears thatsigns of activity were observed irrespective of theregimen used.A regimen of comparable efficacy usedtemozolomide150mg/m2given7/14days;toxicitieswere more frequent,likely reflecting the150%greatercumulative dose of this agent in comparison with ourregimen.13It must also be noted that an intensified adju-vant temozolomide schedule(75–100mg/m2×21d q 4wk)failed to improve survival in a recent phase IIItrial in newly diagnosed glioblastoma,28althoughthose results may not be applicable to recurrentdisease,given potential differences in chemosensitivityand patient selection.Dedicated randomized trials arethus needed to define the role and optimal metronomictemozolomide schedule in recurrent glioblastoma.Interestingly,results of trials combining daily temozolo-mide and bevacizumab29,30(Tables3and4)were disap-pointing and comparable or inferior to our single-agentresults.This raises the intriguing question of whetherbevacizumab could decrease the activity of temozolo-mide when given at low doses,perhaps by decreasingdrug penetration.31–34To our knowledge,this is thefirst prospective study re-porting outcomes in bevacizumab-failure patients inwhom bevacizumab was discontinued.Because of fearsof a rebound effect,bevacizumab is often continueddespite progression on the drug.23–27,35Three smallprospective studies in bevacizumab-failure patients usedcontinued bevacizumab with different cytotoxic agentsand found a median OS of3–6months(Table4).9,36,37The most favorable results were observed with combinedbevacizumab,carboplatin,and irinotecan(PFS6:16%;median OS:6mo),37but48%of patients in that studyhad failed bevacizumab as part offirst-line treatment,whereas the other studies included patients failingsalvage bevacizumab;moreover,that same drug combi-nation was not active in bevacizumab-naive patients.38Therefore,outcomes of continued bevacizumab regi-mens were poor and not superior to our results,suggest-ing that it is justifiable to discontinue bevacizumab afterbevacizumab failure39,40and that conducting clinicaltrials in that setting is feasible.Interestingly,our mul-tivariate analysis suggested that the poor outcomes inbevacizumab failures seemed to be at least partially ex-plained by poor KPS and greater number of progressions(Supplementary material,Appendix S4),although thepossibility that bevacizumab exposure may induce amore aggressive,invasive phenotype41,42cannot be en-tirely ruled out.Omuro et al.:Metronomic temozolomide for recurrent glioblastoma246NEURO-ONCOLOGY†F E B R U A R Y2013 at China Pharmaceutical University on December 5, 2016 /Downloaded fromFinally,we performed a Sequenom-based tissue anal-ysis in order to characterize the frequency of glioma-associated mutations43within patients who enroll in clinical trials for recurrent disease.A total of13(38%) patients displayed one or more mutations involving the EGFR,IDH1,and ERBB2genes,but overall theTable2.Mutations found in the patients enrolled in the trial with corresponding patient characteristics and clinical outcomes(N¼38)Pt Path MGMT Time from Diagnosis(mo)Prior BEV Response PFS(mo)OS(mo) EGFR_P596L1AA Unmethylated0.6No PD 1.637.332GBM Invalid30Yes NE0.370.373GBM Invalid22No SD7.417.24a AO Invalid 1.6No PD 1.838.95GBM Invalid55No PD 1.919.8 EGFR_C620Y6GBM Unmethylated17Yes SD 5.938.2 EGFR_G598V7GBM Unmethylated31Yes PD 1.5 2.4 EGFR_I91L8GBM Unmethylated17Yes NE 1.2 2.6 EGFR_T263P9AA Methylated26No SD8.0315.57 EGFR_V651M10a GBM(secondary)Methylated22No PR 5.4712.63 IDH1_R132L_H11AO Methylated163No SD 1.77 3.134a AO Invalid 1.6No PD 1.838.912AO Invalid25No SD 6.921.110a GBM(secondary)Methylated22No PR 5.4712.63 IDH1_R132C_G_S13GBM(secondary)Unmethylated10No NE0.4 1.7 ERBB2_L49H10a GBM(secondary)Methylated22No PR 5.4712.63Abbreviations:Pt,patient;Path,most recent pathology prior to enrollment;BEV,bevacizumab.a Patients with.1mutation found.Table3.Phase II trials of metronomic temozolomide schedules for glioblastomaReference N TMZ Daily Dose,Schedule ORR(%)PFS6(%)Median OS(mo) Khan112775mg/m2,42/70d14278Brandes123375mg/m2,21/28d93010Wick1364150mg/m2;7/14d15449Perry1050mg/m2,28/28d33Early progression on adjuvant TMZ327NA27Progression on extended TMZ07NA28Rechallenge1136NAKong153840–50m2,28/28d53213Abacioglu1416100mg/m2,21/28d7257Verhoeff301550mg/m2,28/28d+bevacizumab10mg/kg q21d NA74Desjardins293250mg/m2,28/28d+bevacizumab10mg/kg q14d28196This study3750mg/m2,28/28dAll patients11197Bevacizumab failures0114Bevacizumab naive202613 Abbreviations:TMZ,temozolomide;ORR,objective response rate(complete+partial responses);NA,not available.Omuro et al.:Metronomic temozolomide for recurrent glioblastomaNEURO-ONCOLOGY†F E B R U A R Y2013247 at China Pharmaceutical University on December 5, 2016 /Downloaded fromindividual frequency of tested mutations was low.The on-cogenic role of some of the identified extracellular-domain EGFR mutations has been validated in functional assays,44,45and testing EGFR inhibitors in those patients could be of interest.However,none of the33mutations involving the PI3K complex was found,suggesting that a very large number of recurrent patients would have to be screened in order to study a PI3K inhibitor in a trial re-stricted to PI3K mutated tumors.In addition,the wide variability in outcome observed in patients with IDH1 mutations was unexpected.These mutations have been reported to confer a better prognosis,46but the outcome of these patients in this trial was variable(OS:3–21 mo),likely influenced by pretreatment characteristics such as time from initial diagnosis and previous therapies (Table2).This suggests that even in trials for molecularly selected tumors,further patient selection and stratifica-tion based on pretreatment clinical characteristics may be necessary,which will add to the feasibility challenges that such trials will face.Of note,Sequenom-based meth-odology requires that point mutations be chosen and pre-specified;other panels have been proposed,47,48including mutations that are different from the ones we tested.The adoption of next-generation sequencing may overcome this limitation in the future.A few other limitations apply to this study.Our mul-tivariate analysis was performed post hoc and was based on a relatively small number of patients.We used Macdonald criteria for assessment of response,which allows for a comparison with historical controls;criteria of the Revised Assessment of Neuro-Oncology49have since been proposed and may define progression earlier.The median time between diagnosis and enroll-ment was unusually long(21mo),although the longest times were observed among bevacizumab-failure pa-tients,who fared poorly;therefore,selection bias does not seem to explain the activity observed.In summary,we provide further evidence that daily-dose temozolomide is a safe and active regimen in recur-rent glioblastoma.We also demonstrate the differences in outcomes between bevacizumab-naive and bevacizumab-failure patients in the prospective setting and provide historical controls that can be used for sample-size calculation and clinical trial interpretation in bevacizumab-failure patients.Ourfindings support the development of separate trials or stratification ac-cording to bevacizumab exposure but also highlight the prognostic importance of other variables,such as number of previous recurrences and KPS,for that purpose.Importantly,our results in bevacizumab fail-ures were comparable to trials of continuation of bevaci-zumab after progression,suggesting that one should not fear discontinuing bevacizumab for the purposes of pur-suing alternative treatments or clinical trials.Supplementary Material Supplementary material is available at Neuro-Oncology Journal online(http://neuro-oncology.oxfordjournals. org/).AcknowledgmentsPresented in part at the American Society of Clinical Oncology(ASCO)annual meeting2010and Society for Neuro-Oncology(SNO)annual meeting2010.Conflict of interest statement.L.E.A.is currently an employee of Hoffmann–La Roche.M.K.has received honoraria from Roche.A.O.has received compensation for consulting from Roche.A.B.L.has received com-pensation for consulting from Merck/Schering-Plough, BMS,Campus Bio,Cephalon,Eisai,Genentech,GSK, Imclone,Sigma Tau,and Abbott and reports research funding from Merck/Schering-Plough,Genentech, Keryx,Sigma Tau,Astra Zeneca,Medimmune,and Boehringer Ingelheim.FundingThis study was an investigator-initiated clinical trial funded by the Memorial Sloan-Kettering Cancer Center Department of Neurology Research and De-velopment Fund.Schering-Plough/Merck provided drug,partialfinancial support,and analysis of MGMT promoter methylation status.Table4.Outcomes of clinical trials conducted in recurrent glioblastoma patients with history of bevacizumab(BEV)failureReference Regimen N Patients who had Failed BEVas Part of First-LineTreatment PFS6(%)MedianOS(mo)ORRReardon36BEV+daily TMZ50mg/m2100030 BEV+VP-1650mg/m2(3wk on/1wkoff)1315%8 4.70 All pts2315%4 4.10 Reardon37BEV+carboplatin+CPT-112548%16 5.80 Kreisl9BEV+CPT-11190≤5a NA0 This study Daily TMZ50mg/m2,no BEV18011 4.30 Abbreviations:ORR,objective response rate(complete+partial responses);TMZ,temozolomide;VP-16,etoposide;NA,not available.a Only1patient received.3cycles.Omuro et al.:Metronomic temozolomide for recurrent glioblastoma248NEURO-ONCOLOGY†F E B R U A R Y2013 at China Pharmaceutical University on December 5, 2016 /Downloaded from。