Immune checkpoint blockade
- 格式:pdf
- 大小:1.09 MB
- 文档页数:12
免疫调节明星分子PD-1程序性死亡受体1(programmed death 1,PD-1),其最初是从凋亡的小鼠T细胞杂交瘤2B4.11克隆出来的,因此命名为“程序性死亡受体1”1。
PD-1可与其内源性配体PD-L1(B7-H1)或PD-L2(B7-DC)结合,PD-L1是PD-1的主要配体,但PD-L2与PD-1的亲和性更强,可竞争PD-L1与PD-1的结合位点2, 3。
随着后续研究的深入,研究结果显示PD-1的主要功能不是参与细胞死亡,而是作为一种重要的免疫抑制分子,其可从多个层面阻止T、B细胞的激活:1)PD-1可直接影响T细胞激活早期免疫突触(immunological synapse,IS) 的形成4;2)PD-1与配体PD-L1/2的结合可减弱激活的T细胞中TCR和共刺激信号的激活5;3)PD-L1除了与PD-1结合外,还可与B7.1结合,从而阻断CD28-B7.1共激活信号6, 7;4)PD-1在免疫抑制细胞——调节性T细胞中高度表达7。
综上可知PD-1作为免疫检测点,在免疫耐受中发挥着重要的作用。
研究表明肿瘤微环境中呈现高度免疫抑制的状态,且已在乳腺癌、前列腺癌、卵巢癌、黑色素瘤等多种癌症的肿瘤浸润性淋巴细胞(tumor-infiltrating lymphocytes ,TILs)中发现了PD-1的高表达,且PD-1的高表达与肿瘤的分级、大小、淋巴结转移、远处转移等具有相关性8, 9,说明PD-1在肿瘤发生中具有重要作用,于此同时,在多种肿瘤细胞中发现了PD-L1的高表达10, 11。
肿瘤细胞表面PD-L1的表达可能是应对肿瘤微环境的代偿机制。
PD-1蛋白由N端细胞外结合域、跨膜结构域和C端胞浆结构域三部分组成,且胞浆结构域中含有一个免疫受体酪氨酸交换模体(immunoreceptor tyrosine-based switch motif ,ITSM)和一个免疫受体酪氨酸抑制模体(immunoreceptortyrosine-based inhibitory motif,ITIM)。
检查点阻断(checkpoint blockade)是一种利用免疫反应来治疗癌症的强大新策略。
溶瘤病毒疗法则是利用病毒来消灭肿瘤的一种干预疗法。
来自Ludwig癌症研究中心的一项研究表明联合两者将有可能大大提高临床疗效。
检查点阻断(checkpoint blockade)是一种利用免疫反应来治疗癌症的强大新策略。
溶瘤病毒疗法则是利用病毒来消灭肿瘤的一种干预疗法。
来自Ludwig癌症研究中心的一项研究表明联合两者将有可能大大提高临床疗效。
发表在3月5日《科学转化医学》(Science Translational Medicine)杂志上的这项研究对联合治疗进行了评估。
研究人员首先将对人类通常无害的一种鸟病毒:新城鸡瘟病毒直接注入到小鼠体内移植的两个黑色素瘤其中一个肿瘤中,随后注入一种抗体释放免疫反应刹车。
研究人员报告称,联合疗法诱导了强力的、全身有效的抗肿瘤免疫反应,也破坏了未接受病毒感染的肿瘤。
甚至是迄今为止抵抗检查点阻断和其他免疫疗法策略的肿瘤类型也对这种联合疗法敏感。
论文的主要作者、纪念斯隆凯特琳癌症中心(MSK) Ludwig合作实验室成员DmitriyZamarin说:“许多患者从癌症免疫疗法中受益。
但是这些疗法并非对所有的患者或是所有的癌症类型有效,因为大多数的癌症能够有力地抑制免疫反应。
我们希望能够使更多的患者从免疫疗法中受益,优化应用于更多癌症类型。
”Zamarin和同事们发现,治疗效应主要是由NDV在肿瘤中诱导的一种炎症免疫反应所致。
研究中所采用的检查点阻断抗体结合了免疫细胞上的CTLA-4分子,这一分子起免疫反应刹车(或“检查点“)作用。
一个版本的这种抗体已用于癌症治疗,一项评估这种抗体联合另一种免疫疗法的临床实验证实了其效力。
研究人员注意到当将NDV注入到小鼠的一个移植肿瘤中时,杀癌免疫细胞涌入到肿瘤中。
“但让我们惊讶的是,我们发现在远端的一个肿瘤中也发生了相似的激活免疫细胞浸润,在这一肿瘤中从未检测到NDV病毒,”Zamarin说。
PD-1抑制剂单抗治疗鼻咽癌的临床试验研究进展雷小梅1,瞿家权2,谭潭3△摘要:鼻咽癌是一种EB 病毒(EBV )感染率高的头颈部恶性肿瘤,程序性死亡蛋白-1及其配体(PD-1/PD-L1)通路的激活可能是EBV 相关鼻咽癌免疫逃逸的机制之一。
与化疗相比,免疫检查点抑制剂有不良反应小、生存获益更长和耐受性更好等优势,但存在仅有少部分患者能受益的缺点。
目前,至少有4种针对该通路的PD-1抑制剂单抗已经在临床试验中取得了较好的效果,并且获批用于治疗复发或转移性鼻咽癌。
就近年来已完成和正在进行的鼻咽癌PD-1抑制剂的治疗策略、疗效评价和安全性的研究进行综述。
关键词:鼻咽肿瘤;免疫疗法;程序性细胞死亡受体1;免疫检查点抑制剂中图分类号:R739.6文献标志码:ADOI :10.11958/20211672Progress in clinical trials of PD-1inhibitor monoclonal antibody for nasopharyngeal carcinomaLEI Xiaomei 1,QU Jiaquan 2,TAN Tan 3△1Department of Laboratory,the Affiliated Chenzhou Hospital,Hengyang Medical School,University of South China,Chenzhou 421001,China;2Department of Oncology,the Eighth Affiliated Hospital,Sun Yat-sen University;3Precision Medicine Center,Chenzhou First People's Hospital△Corresponding Author and Reviser E-mail:******************Abstract:Nasopharyngeal carcinoma (NPC)is a head and neck malignant tumor with high infection rate of EBV.Theactivation of programmed death protein-1and its ligand (PD-1/PD-L1)pathway may be one of the mechanisms of immune escape from EBV-associated pared with chemotherapy,immune checkpoint inhibitors have less adverse reactions.Survival benefit is longer and the advantages of the tolerance is better,but there is only a small number of patients will benefit from faults.At present,at least four kinds of inhibitors targeting the pathway of PD-1single resistance in clinical trials have achieved good results,and it has been approved for the treatment of recurrent or metastatic nasopharyngeal carcinoma.Recent completed and ongoing studies on the PD-1inhibitor treatment strategy for NPC,the efficacy evaluationand safety studies were reviewed.Key words:nasopharyngeal neoplasms;immunotherapy;programmed cell death 1receptor;immune checkpoint inhibitor基金项目:国家自然科学基金资助项目(81703043);湖南省科技计划项目(2017SK51102、2017SK1103);湖南省卫生厅项目(20200861);陕西省精准医学中心重点实验室项目(KLTPM-SX2018-B5)作者单位:1南华大学衡阳医学院,附属郴州医院检验科(邮编421001);2中山大学附属第八医院肿瘤科;3郴州市第一人民医院精准医学中心作者简介:雷小梅(1995),女,硕士在读,主要从事肿瘤方向研究。
免疫疗法相关肿瘤模型介绍导读前两期我们介绍了常见的肿瘤动物模型,考虑到肿瘤免疫疗法不同于普通抗癌药物的作用方式和评价体系,有必要在此单独介绍一下肿瘤免疫疗法研究领域常用的肿瘤动物模型。
为此,笔者专门搜集和整理了一些相关资料,以当前较为成熟的CAR-T/TCR-T以及免疫检查点阻断技术为例,对免疫疗法肿瘤模型的特点、常用实验动物及细胞株、建立方法以及药物评价方式等关键点做个简要介绍,以期为有志于从事肿瘤免疫疗法研究的同行们提供些许参考。
背景在当今众多的癌症治疗手段中,免疫疗法无疑是近年来最为吸引人们眼球的“明星”治疗手段,普遍引起学术界及医学界的强烈关注和研究兴趣。
随着以T细胞受体T细胞技术(TCR-T)、嵌合抗原受体T细胞技术(CAR-T)以及免疫检查点阻断技术(Immune Checkpoint Blockade)为代表的新兴肿瘤免疫疗法不断取得临床上的突破和成功,奇迹频现,捷报频传,持续更新着人们对机体免疫系统潜在的强大肿瘤杀伤能力的认知,进一步提高人们战胜恶性肿瘤的信心,当然也持续激发着科研人员对“肿瘤免疫疗法”这一强大抗癌利器的研究和开发热情。
而对于肿瘤免疫疗法的研究,离不开相关动物模型的选择和建立。
事实上,目前已经取得临床成功的肿瘤免疫疗法中,无一例外都已经过大量严格的临床前动物实验进行验证、评估和预测。
可见,相关动物模型的建立对于肿瘤免疫疗法的开发和应用是至关重要且必不可少的。
免疫疗法相关肿瘤模型一、以CAR-T/TCR-T为代表的细胞过继性疗法(Adoptive Transfer)常用肿瘤动物模型细胞过继性疗法是指将供体(donor)细胞(此处主要是T淋巴细胞)经体外刺激活化或者基因修饰后再次回输入受者(Recipient)体内,从而达到相关治疗目的的治疗方式。
供体细胞可以是来源于受者自身,也可以来自于其他个体,前者称为自体移植(Autograft),后者则有两种情况,如果受者与供者属于相同种属,称为同种异体移植(Allograft),反之则为异种移植(Heterograft)。
一例肝癌患者使用卡瑞利珠单抗致反应性毛细血管增生症案例分析一、前言免疫检查点抑制剂(immune checkpoint inhibitions ,ICIs)的应用显著改善了多种肿瘤的预后,是当前肿瘤治疗中备受重视的手段。
以程序性死亡因子‐1(programmed death 1 ,PD‐1)、程序性死亡因子配体‐1(programmed death ligand 1 ,PD‐L1)和细胞毒性 T 淋巴细胞相关抗原 4(cytotoxic T lymphocyte antigen 4 ,CTLA‐4)单克隆抗体为主的免疫检查点的临床研究结果显示,单一 ICIs 临床效果有限[1]。
卡瑞利珠单抗( SHR-1210,Camrelizumab,Cam)是一种具有自主知识产权、独特结构和功能的国产ICIs,它是一种人源化免疫球蛋白G4(IgG4)型单克隆抗体(mAb),可与PD-1靶向结合,阻断其与PD-L1及程序性死亡配体2(PD-L2)之间的相互作用而恢复机体免疫功能,最终发挥抗肿瘤的作用[2]。
卡瑞利珠单抗在2019年5月经国家药品监督管理局( NMPA) 批准上市,陆续获得了治疗多种肿瘤的适应证,正在临床上广泛使用。
新型免疫治疗较传统治疗,疗效上有进一步的提升,但是免疫治疗的同时也出现了许多免疫相关性的不良反应[3]。
二、病史简介患者某某,男,69岁,身高168cm,体重78kg,体表面积1.94m2,原发性肝癌(IIb期),患者于2021年7月无明显诱因出现腹胀,伴进食减少,无呕吐、腹痛、腹泻症状,后于2021年8月临床诊断原发性肝癌,评估无手术指征。
于8月23日、9月20日、10月17日、11月22日行卡瑞利珠单抗免疫治疗4次,并口服"索拉非尼"靶向治疗,9月10日起发生腹泻4-5次/天,严重时10余次/天,10月17日后剂量减半使用至11月17日,期间间断出现腹泻症状。
如何正确地选择肿瘤动物模型?展开全文导读在中国,每天约有7710例患者死于癌症,每分钟大概6人死于癌症,因此,癌症的研究无疑成为生物医学的重中之重,动物模型在肿瘤发生发展及药物研发上扮演着至关重要角色。
那么面对不同的肿瘤模型,究竟怎样的动物模型才可以准确反映人类肿瘤发展情况呢?这就是我们本期内容,告诉大家什么样的肿瘤动物模型才是你真正想要的。
首先我们要知道,最大的问题不是如何去建立模型,而是建立什么样的模型。
因为不同的模型会导致实验结果有较大出入,肿瘤学研究工作者应当熟悉该领域已有的研究成果,查阅全面资料后,才能为自己的课题选到合适的实验动物肿瘤模型。
第一步,需要明确你的实验目的根据设定的实验目标来选择最合适的动物模型,才能得到科学的结论和理想的结果。
肿瘤动物模型的应用一般分为:肿瘤发生发展机制研究、抗肿瘤药物筛选、免疫疗法相关研究等。
第二步,了解已有的各类常见肿瘤模型分类及特点肿瘤动物模型一般分为:①CDTX(肿瘤细胞系移植模型)②P DTX(人源肿瘤组织异种移植模型)③诱发性肿瘤动物模型④基因修饰肿瘤模型⑤自发性肿瘤动物模型;每个模型介绍详情可点击往期文章(3.4肿瘤动物模型介绍)。
第三步,进一步细化到模型的每一个不定因素主要包括以下几个不确定因素:(1)细胞系的选择根据研究的肿瘤类型以及基因型选取相应的肿瘤细胞系,用于荷瘤的细胞要保证较好的生长状态,无污染,处于对数生长期。
不同肿瘤细胞系的成瘤率不同,结合自身选取成瘤率高的细胞系进行实验。
如乳腺癌细胞有几十种,根据来源有鼠源和人源的,根据分型有三阴性与非三阴性的,如果要更贴近人类三阴性乳腺癌的研究,一般会选择易荷瘤的MDA-MB-231细胞。
(2)小鼠品系的选择肿瘤细胞系移植模型分为两种:将人的肿瘤细胞系或肿瘤组织移植到小鼠体内和将小鼠的肿瘤细胞移植到小鼠体内,前者为异种移植因此必须选择具有免疫缺陷的小鼠(如:nude,NOD/scid,NSG),后者为同种移植,因此选择与肿瘤细胞系来源一致的小鼠(C57BL/6品系来源的黑色素瘤细胞系B16F10以及路易斯肺癌细胞系LLC就要选择C57BL/6品系的小鼠)。
国际肿瘤学杂志202丨年2月第48卷第2期JInt Oncol, February 2021, Vo l. 48, No.2• 105 •.综述•免疫检查点抑制剂常见免疫相关不良反应及其管理李慧杨宇哈尔滨医科大学附属第二医院肿瘤内科150000通信作者:杨宇,Email:yangyu138****5585@163. com【摘要】目前临床上免疫检查点抑制剂(ICI)应用广泛,常见的不良反应包括皮肤、胃肠道、内分泌和肝脏不良反应,肺脏和心脏不良反应相对较少,但可能致命:、类固醇全身治疗是对抗免疫治疗相关不良反应(irAE)的主要治疗手段,如对类固醇治疗没有反应,则需要考虑使用免疫调节剂。
掌握irAE的发生率、发病机制、常见类型及其治疗策略,可为IC1在临床上的安全应用提供理论依据。
【关键词】肿瘤;免疫检查点抑制剂;免疫治疗相关不良反应;治疗策略DOI :10. 3760/cma. j. cn371439-20200828-00020Common immune-related adverse reactions of immune checkpoint inhibitors and their managementLi Hui, Yang YuDepartment of Oncology, Second Affiliated Hospital of Harbin Medical University, Harbin 150000, ChinaCorresponding author:Yang Yu, Email:yangyul3836125585@ 163. com【A bstract】At present, immune checkpoint inhibitors ( ICIs) are widely used in clinical, and thecommon adverse reactions include adverse reactions of skin, gastrointestinal tract, endocrine and liver. Adversereactions to the lungs and heart are relatively rare, but can be fatal. Systemic steroid therapy is the maintreatment for immune related adverse events ( irA Es). If there is no response to steroid therapy, animmunomodulator may be considered. Understanding the incidence, pathogenesis, common types and treatmentstrategies of irAEs can provide theoretical basis for the safe application of ICIs in clinical practice.【Key words】Neoplasms; Immune checkpoint inhibitor;Immune related adverse events;TreatmentstrategyDOI :10. 3760/cma. j. cn371439-20200828-00020免疫检查点抑制剂(immune checkpoint inhibitor, ICI)具有良好的抗肿瘤疗效,在通过阻断免疫检查 点、增强肿瘤特异性免疫反应的同时,也会非特异性 地激活免疫系统导致免疫稳态破坏,产生与治疗相关 的特殊不良反应,即免疫治疗相关不良反应(immune related adverse event,irAE)c irA E 可累及全身各个脏 器,临床表现与自身免疫性疾病有相似之处,但是患 者血清中往往检测不到自身抗体U]。
·专家论坛·益生菌调控肠道菌群对免疫检查点阻断治疗的影响高广琦,张和平(内蒙古农业大学乳品生物技术与工程教育部重点实验室,呼和浩特010018)摘要:免疫检查点阻断是一种肿瘤免疫治疗的突破性技术。
利用单克隆抗体抑制机体靶向的负调控信号,激活下游免疫反应,能够产生由细胞毒性T细胞介导的杀伤肿瘤细胞的作用。
然而这种疗法只能令少数患者受益,因此研究并开发提高免疫疗效的方法具有重要意义。
肠道菌群被认为对肿瘤免疫检查点阻断治疗的成功有重要的贡献。
研究表明,肠道菌群的存在、组成和多样性水平直接影响患者和小鼠的肿瘤治疗效果,并且免疫应答效率能通过粪菌移植或有益菌干预而改善。
肠道的特征菌群与宿主肿瘤微环境树突状细胞的抗原呈递功能和效应性T淋巴细胞的活性有显著的相关性。
这种菌群‐免疫之间的调控机制很大程度上是由肠道菌群代谢产生的短链脂肪酸或次级胆汁酸等物质通过配体‐受体的关系调控宿主基因表达,调节免疫细胞分化及活性。
另外,肠道细菌还可能通过易位定植于肿瘤组织进而刺激免疫反应。
基于这些菌群与免疫关系的研究,可确信通过避免使用抗生素、粪菌移植和益生菌干预的方法,改善或维持健康的肠道菌群结构,是优化肿瘤免疫检查点阻断治疗的有效途径。
关键词:肠道菌群;免疫检查点阻断;肿瘤免疫治疗;程序性死亡受体‐1;细胞毒性T淋巴细胞相关抗原‐4Influence of probiotic‑regulated gut microbiota on immune checkpoint blockade therapyGao Guangqi,Zhang HepingKey Laboratory of Dairy Biotechnology and Engineering,Ministry of Education,Inner Mongolia Agricultural University,Hohhot010018,Inner Mongolia,ChinaAbstract:Immune checkpoint blockade is a breakthrough technology in tumor immunotherapy.It could be summarized that monoclonal antibodies suppressed the target negative regulators to activate the downstream immune response,which produced a cytotoxic T cell mediated killing effect on tumor cells.However,this treatment can only benefit a part of patients,so it is important to find out effective ways to improve the cancer immunotherapy.Gut microbiota is considered to be an important contribution to the success of checkpoint blockade therapy.Studies showed that the presence,composition,and diversity of gut microbiota directly affected the efficacy of cancer treatment in patients and mice,and the efficiency of the immune response could be improved by fecal microbiota transplantation or beneficial bacteria intervention.There was a significant correlation between the characteristic bacteria or microbiota of intestinal tract and the antigen presentation of dendritic cells in the host tumor microenvironment as well as the activity of effector T lymphocytes.To a large extent,the regulation mechanism between gut microbiota and host immunity is that substances,such as short⁃chain fatty acids or secondary bile acids,produced by gut bacteria metabolism can regulate the gene expression or the differentiation and activity of immune cells through ligand⁃receptor pathways.Another possible factor can be supposed as that bacterial translocation into the tumor stimulates the systemic immune response.Based on these gut microbiota⁃host immune investigations,it can be confirmed that improving or maintaining healthy intestinal microbiota structure by avoiding the use of antibiotics,fecal bacteria transplantation or probiotic intervention is an effective way to optimize the immune checkpoint blockade treatment of tumor.Key words:Gut microbiota;Immune checkpoint blockade;Tumor immunotherapy;Programmed cell death protein⁃1;Cytotoxic T lymphocyte⁃associated antigen⁃4免疫疗法是当前用于科学研究和临床治疗多种恶性肿瘤的一类新型方法[1]。
肠道菌群和肿瘤肠道菌群一般指人体肠道内的正常微生物,如双歧杆菌,乳酸杆菌,大肠杆菌等。
人体肠道内寄生着大约10万亿个细菌,他们中的一些微生物能合成多种人体生长发育必须的维生素,有的细菌还能利用蛋白质残渣合成必需氨基酸,并参与糖类和蛋白质的代谢。
肠道菌群中的有益菌所产生的营养物质对人类的健康有着重要作用,一旦缺少会引起多种疾病,例如炎症反应和自身免疫疾病等1-3。
肠道微生物、肠道上皮细胞以及人体免疫系统三者息息相关,他们之间的相互作用以及平衡与许多疾病有着紧密的联系,癌症也不例外4。
对于肠道菌群会如何影响肿瘤发生,以前人们由于对肠道菌群认识的局限性,认为他们只能形成肠道微环境,最终通过调节肠道免疫反应影响肠癌的发生与进展5。
一个直观的例子是腹内感染或者过度使用抗生素会增加结肠直肠癌的发病几率,这是因为肠道内部(肠道微生物、肠道上皮细胞以及人体免疫系统)的平衡被打破了,肠道微生物影响增强结肠致癌作用6。
同时一些肠道微生物的代谢产物能够直接减缓致癌作用或者抑制肿瘤发生。
临床研究确认肠道菌群可以用来筛查直肠癌7。
研究人员还发现肠道在发生炎症时,肠道微生物的拓扑结构发生变化,最终会导致宿主免疫系统的变化8, 9。
2013年两篇发表在science上的文章报道引发了关于研究肠道菌群对肿瘤影响的新浪潮。
他们发现肠道微生物可以显著影响包括环磷酰胺(cyclophosphamide)等几个抗癌药物所引起的宿主免疫反应,微生物可以通过影响药物活性影响肠外器官的肿瘤治疗,这使得关于肠道菌群的研究成为肿瘤1研究的热点。
相比于具有丰富肠道微生物的小鼠,无菌小鼠对于肿瘤靶向性治疗的反应较差。
环磷酰胺药物可以改变动物的肠道菌群组成,并使一些菌种到达淋巴器官促进免疫细胞反应能力,最终提高环磷酰胺效力,而无菌小鼠则对这种药物耐药。
因此,肠道微生物不仅影响肠道局部炎症,而且影响了全身炎症的形成,进而影响肠道外器官癌症的进展10, 11。
Immune Checkpoint Blockade:A Common Denominator Approach to Cancer TherapySuzanne L.Topalian,1,*Charles G.Drake,2,3and Drew M.Pardoll31Department of Surgery2The Brady Urological Institute3Department of OncologySidney Kimmel Comprehensive Cancer Center and Johns Hopkins University School of Medicine,Baltimore,MD21287,USA*Correspondence:stopali1@/10.1016/ell.2015.03.001The immune system recognizes and is poised to eliminate cancer but is held in check by inhibitory receptors and ligands.These immune checkpoint pathways,which normally maintain self-tolerance and limit collateral tissue damage during anti-microbial immune responses,can be co-opted by cancer to evade immune destruction.Drugs interrupting immune checkpoints,such as anti-CTLA-4,anti-PD-1,anti-PD-L1,and others in early development,can unleash anti-tumor immunity and mediate durable cancer regressions.The com-plex biology of immune checkpoint pathways still contains many mysteries,and the full activity spectrum of checkpoint-blocking drugs,used alone or in combination,is currently the subject of intense study.In the current era in oncology emphasizing personalized therapy, immune checkpoint blockade is distinguished by its‘‘common denominator’’approach.Although the vast somatic mutational diversity found in most human cancers creates challenges for therapies targeting individual mutations,it exposes a panoply of new antigens for potential immune recognition.However,cells of the adaptive and innate immune systems that recognize and are poised to attack cancer are held in check by molecular pathways that suppress their activation and effector functions. The seminal observation that blocking the prototypical immune checkpoint receptor cytotoxic T lymphocyte antigen4(CTLA-4) could mediate tumor regression in murine models(Leach et al., 1996)led to the clinical development and approval of anti-CTLA-4as a treatment for patients with advanced melanoma (Hodi et al.,2010).Subsequently,drugs blocking the distinct checkpoints Programmed Death1(PD-1)and its major ligand PD-L1have shown great promise in treating many diverse cancer types,fueling the intensive examination of a growing cohort of unique checkpoint molecules as potential therapeutic targets. This has revealed new treatment options for patients and has revolutionized our approach to cancer therapy.Biology of Immune Checkpoints:The BasicsThe rapid-fire clinical successes from blocking CTLA-4and PD-1, thefirst checkpoint receptors to be discovered,have opened prospects for extending the potential of cancer immunotherapy by inhibiting more recently discovered checkpoint ligands and re-ceptors.It is clear that,despite some commonalities,CTLA-4and PD-1have distinct patterns of expression,signaling pathways, and mechanisms of action.Although discovered over20years ago,there are still many unanswered questions about their biology,particularly in the context of cancer.The CD28/CTLA-4System of Immune ModulationThe conventional wisdom underlying our vision of how CTLA-4 blockade mediates tumor regression is that it systemically acti-vates T cells that are encountering antigens.CTLA-4represents the paradigm for regulatory feedback inhibition.Its engagement down-modulates the amplitude of T cell responses,largely by in-hibiting co-stimulation by CD28,with which it shares the ligands CD80(B7.1)and CD86(B7.2)(Figure1;Lenschow et al.,1996). As a‘‘master T cell co-stimulator,’’CD28engagement amplifies TCR signaling when the T cell receptor(TCR)is also engaged by cognate peptide-major histocompatibility complex(MHC) (Schwartz,1992).However,CTLA-4has a much higher affinity for both CD80and CD86compared with CD28(Linsley et al., 1994),so its expression on activated T cells dampens CD28 co-stimulation by out-competing CD28binding and,possibly, also via depletion of CD80and CD86via‘‘trans-endocytosis’’(Qureshi et al.,2011).Because CD80and CD86are expressed on antigen-presenting cells(APCs; e.g.,dendritic cells and monocytes)but not on non-hematologic tumor cells,CTLA-4’s suppression of anti-tumor immunity has been viewed to reside primarily in secondary lymphoid organs where T cell activation occurs rather than within the tumor microenvironment(TME). Furthermore,CTLA-4is predominantly expressed on CD4+‘‘helper’’and not CD8+‘‘killer’’T cells.Therefore,heightened CD8responses in anti-CTLA-4-treated patients likely occur indi-rectly through increased activation of CD4+cells.Of note,a few studies suggest that CTLA-4can act as a direct inhibitory recep-tor of CD8T cells(Fallarino et al.,1998;Chambers et al.,1998), although this role in down-modulating anti-tumor CD8T cell re-sponses remains to be directly demonstrated.The specific signaling pathways by which CTLA-4inhibits T cell activation are still under investigation,although activation of the phosphatases SHP2and PP2A appears to be important in counteracting both tyrosine and serine/threonine kinase sig-nals induced by TCR and CD28(Rudd et al.,2009).CTLA-4 engagement also interferes with the‘‘TCR stop signal,’’which maintains the immunological synapse long enough for extended or serial interactions between TCR and its peptide-MHC ligand (Schneider et al.,2006).Naive and resting memory T cells ex-press CD28,but not CTLA-4,on the cell surface,allowing co-stimulation to dominate upon antigen recognition.However, CTLA-4is rapidly mobilized to the cell surface fromintracellular450Cancer Cell27,April13,2015ª2015Elsevier Inc.protein stores,allowing feedback inhibition to occur within an hour after antigen engagement.The central role of CTLA-4in maintaining immune tolerance is dramatically demonstrated by the rapidly lethal systemic immune hyperactivation pheno-type of Ctla-4knockout mice (Tivol et al.,1995;Waterhouse et al.,1995).In humans,anti-CTLA-4treatment induces ex-pression of activation markers on circulating T cells (Maker et al.,2005)and a high rate of inflammatory side effects (Phan et al.,2003).However,because melanoma patients appear to possess an unusually high proportion of tumor-reactive T cells,anti-tumor responses balance autoimmune toxicity and provide a clinical benefit to roughly 20%of patients with this disease (see below).PD-1:Similarities to and Differences from CTLA-4The PD-1system of immune modulation bears similarities to CTLA-4as well as key distinctions (Parry et al.,2005).Similar to CTLA-4,PD-1is absent on resting naive and memory T cells and is expressed upon TCR engagement.However,in contrast to CTLA-4,PD-1expression on the surface of activated T cells requires transcriptional activation and is therefore delayed (6–12hr).Also in contrast to CTLA-4,PD-1contains a conven-tional immunoreceptor tyrosine inhibitory motif (ITIM)as well as an immunoreceptor tyrosine switch motif (ITSM).PD-1’s ITIM and ITSM bind the inhibitory phosphatase SHP-2.PD-1engage-ment can also activate the inhibitory phosphatase PP2A.PD-1engagement directly inhibits TCR-mediated effector functions and increases T cell migration within tissues,thereby limiting the time that a T cell has to survey the surface of interacting cells for the presence of cognate peptide-MHC complexes.Therefore,T cells may ‘‘pass over’’target cells expressing lower levels of peptide-MHC complexes (Honda et al.,2014).In contrast to CTLA-4,PD-1blockade is viewed to work predominantly within the TME,where its ligands are commonly overexpressed by tumor cells as well as infiltrating leukocytes (Keir et al.,2008).This mechanism is thought to reflect its im-portant physiologic role in restraining collateral tissue damage during T cell responses to infection.In addition,tumor-infiltrating lymphocytes (TILs)commonly express heightened levels of PD-1and are thought to be ‘‘exhausted’’because of chronic stimulation by tumor antigens,analogous to the exhausted phenotype seen in murine models of chronic viral infection,which is partially reversible by PD-1pathway blockade (Barber et al.,2006).Importantly,the phenotypes of murine knockouts of PD-1and its two known ligands are very mild,consisting of late-onset organ-specific inflammation,particularly when crossed to auto-immune-prone mouse strains (Nishimura et al.,1999,2001).This contrasts sharply with the Ctla-4knockout phenotype and highlights the importance of the PD-1pathway in restricting pe-ripheral tissue inflammation.Furthermore,it is consistent with clinical observations that autoimmune side effects of anti-PD-1drugs are generally milder and less frequent than with anti-CTLA-4.Despite the conventional wisdom that CTLA-4acts early in T cell activation in secondary lymphoid tissues whereas PD-1in-hibits execution of effector T cell responses in tissue and tumors,this distinction is not absolute.Beyond its role in dampening acti-vation of effector T cells,CTLA-4plays a major role in driving the suppressive function of T regulatory (Treg)cells (Wing et al.,2008;Peggs et al.,2009).Tregs,which broadly inhibit effector T cell responses,are typically concentrated in tumor tissues and are thought to locally inhibit anti-tumor immunity.Therefore,CTLA-4blockade may affect intratumoral immune responses by inactivating tumor-infiltrating Treg cells.Recent evidence has demonstrated anti-tumor effects from CTLA-4blockade even when S1P inhibitors block lymphocyte egress from lymph nodes (Spranger et al.,2014),indicating that this checkpoint exerts at least some effects directly in the TME as opposed to secondary lymphoid tissues.Conversely,PD-1has been shown to play a role in early fate decisions of T cells recognizing antigens pre-sented in the lymph node.In particular,PD-1engagement limits the initial ‘‘burst size’’of T cells upon antigen exposure and can partially convert T cell tolerance induction to effector differentia-tion (Goldberg et al.,2007).Complex Receptor-Ligand Interactions in the PD-1Pathway:Links and Analogies to the CD28/CTLA-4SystemThe receptor-ligand interactions of the PD-1system appear to be even more complex than the CD28/CTLA-4system (Figure 1).The two ligands for PD-1are PD-L1(B7-H1,CD274)and PD-L2(B7-DC,CD273),which share 37%sequence homology and arose via gene duplication (Dong et al.,1999,Latchman et al.,2001;Tseng et al.,2001).However,their regulation is highly divergent.PD-L1is induced on activated hematopoietic cells and on epithelial cells by the inflammatory cytokine interferon (IFN)-g ,which is produced by some activated T and natural killer (NK)cells.PD-L2has much more selective expression on acti-vated dendritic cells and some macrophages.It is induced toaFigure plex Interactions between the CTLA-4/CD28and PD-1Families of Receptors and LigandsShown are the defined interactions between the co-inhibitory (checkpoint)receptors CTLA-4and PD-1and their ligands and related receptors.The two known ligands for CTLA-4are CD80(B7.1)and CD86(B7.2).CD86can ‘‘backward-signal’’into APCs when engaged by CTLA-4,inducing the immune inhibitory enzyme IDO.CD80and CD86also bind the co-stimulatory receptor CD28on T cells.Recently,another B7family member,inducible costimulator ligand (ICOS-L),which was discovered as the ligand for the co-stimulatory receptor ICOS (not shown),has been reported to bind to CD28,leading to co-stimulation independent of CD80or CD86.The two defined ligands for PD-1,PD-L1(B7-H1)and PD-L2(B7-DC),bind to additional molecules.PD-L1binds CD80molecules expressed on activated T cells,mediating inhibition.Addi-tionally,PD-L1on APCs appears to provide inhibitory signals (backward signaling)when it is engaged by PD-1.PD-L2binds another molecule,RGMb,which is expressed on macrophages and some epithelial cell types and ap-pears to deliver an inhibitory immune signal through an as yet undefined mechanism.Although not identified,genetic evidence from PD-1knockout T cells and knockout mice suggests the existence of another receptor for PD-L2that is co-stimulatory.Cancer Cell 27,April 13,2015ª2015Elsevier Inc.451much greater extent by interleukin 4(IL-4)than by IFN-g ,further emphasizing differences in regulation of expression of PD-L1and PD-L2.Beyond their role as ligands for PD-1,PD-L1and PD–L2appear to have additional partners,indicating additional layers of immune modulation.An unexpected molecular interaction be-tween PD-L1and CD80has been discovered (Butte et al.,2007;Park et al.,2010)whereby CD80expressed on activated T cells (and possibly APCs)can function as a receptor rather than a ligand,delivering inhibitory signals when engaged by PD-L1.The relevance of this interaction in tumor immune resistance has not yet been determined.Recently,PD-L2has been shown to bind to repulsive guidance molecule b (RGMb),which itself binds at least three other molecules in cis (neogenin and BMP receptors type I and II)(Xiao et al.,2014).This interaction ap-pears to be inhibitory independent of PD-1,as demonstrated in a pulmonary tolerance model.Finally,evidence from murine models suggests that PD-L2and,possibly,PD-L1may bind to a co-stimulatory T cell receptor (Shin et al.,2003,2005),an arrangement reminiscent of the CD80/CD86ligand pair for the co-stimulatory CD28and co-inhibitory CTLA-4receptors.Understanding the roles of these various interactions in cancer is highly relevant for the development of immunomodulatory drugs and the discovery of biomarkers predictive of therapeutic response.Mechanisms of PD-1Ligand Induction:Implications for Cancer ImmunotherapyA key finding that encouraged the development of drugs block-ing the PD-1pathway for cancer immunotherapy was that PD-1ligands are upregulated in many human cancers (Dong et al.,2002),whereas PD-1is highly expressed on tumor-infiltrating lymphocytes (Ahmadzadeh et al.,2009;Sfanos et al.,2009).Indeed,PD-L1appears to be the major ligand expressed in solid tumors,whereas PD-L2(together with PD-L1)is highly ex-pressed in certain subsets of B cell lymphomas (Ansell et al.,2015).Exploration of this phenomenon as a central process by which cancers resist elimination by endogenous tumor-specific T cells revealed two mechanisms for PD-1ligand upregulation in cancer,known as intrinsic and adaptive immune resistance (Figure 2).These mechanisms are not mutually exclusive and may co-exist in the same TME.Intrinsic resistance refers to the constitutive expression of PD-L1by tumor cells because of ge-netic alterations or activation of certain signaling pathways,such as the AKT pathway and STAT3,which are commonly acti-vated in many cancers (Parsa et al.,2007;Marzec et al.,2008).Although PD-L1induction by AKT and STAT3signaling has been demonstrated in some tumor cell lines,the importance of this intrinsic pathway in PD-L1expression by tumors in vivo re-mains to be determined.Genetic alterations in B cell lymphoma subtypes can drive expression of either or both PD-L1and PD-L2.Primary mediastinal lymphomas commonly display gene fu-sions between MHC class II transactivator (CIITA)and PD-L1or PD-L2,placing PD-1ligands under the transcriptional control of the CIITA promoter,which is highly active in B cell lymphomas (Steidl et al.,2011).A significant subset of Hodgkin’s lymphoma has amplification of chromosome 9p23-24,where PD-L1and PD-L2reside,resulting in overexpression of both ligands.Other cancers,such as a subset of Epstein-Barr virus-induced gastric cancers,also display gene amplification with consequent induc-tion of PD-L1and PD-L2.The second mechanism,adaptive resistance,refers to the induction of PD-L1expression on tumor cells in response to spe-cific cytokines,in particular IFN-g .Because IFN-g is only pro-duced by activated Th1-type helper CD4cells,activatedCD8Figure 2.Two General Mechanisms for Expression of Checkpoint Ligands in the TMEThe examples in this figure use the PD-1ligand PD-L1for illustrative purposes,although the concept likely applies to multiple checkpoint ligands.Top:innate immune resistance.In some tumors,consti-tutive oncogenic signaling,such as through activa-tion of the AKT pathway or gene amplification,can upregulate PD-L1expression on tumor cells inde-pendently of inflammatory signals in the TME.Bot-tom:adaptive immune resistance refers to PD-L1induction in tumors as an adaptation to the sensing of an immune attack.In adaptive resistance,PD-L1is not constitutively expressed but,rather,is induced by inflammatory signals,such as IFN-g produced by T cells attempting to execute an active anti-tumor response.Expression of PD-L1in a non-uniform distribution associated with lymphocyte infiltrates suggests adaptive induction in response to immune reactivity within the TME.Adaptive resistance can be generated by cytokine-induced PD-L1expression on either tumor cells themselves or on leukocytes (macrophages,myeloid suppres-sor cells,dendritic cells,or even lymphocytes)in the TME.Inhibition of tumor-specific T cells by PD-L1-or PD-L2-expressing leukocytes may in-volve cross-presentation of tumor antigens so that PD-1-dependent inhibition is in cis .Adaptive resis-tance may be a common mechanism for the intra-tumoral expression of multiple immune checkpoint molecules.452Cancer Cell 27,April 13,2015ª2015Elsevier Inc.cells,and NK cells,this mechanism represents an adaptation of tumor cells upon‘‘sensing’’an inflammatory immune microenvi-ronment that‘‘threatens’’the tumor.Indeed,human tumors show significant correlations between PD-L1expression,levels of T cell infiltration,and IFN-g in the TME(Taube et al.,2012; Spranger et al.,2013).Other inhibitory molecules in the TME, such as indoleamine2030dioxygenase(IDO),which inhibits immu-nity locally via conversion of tryptophan to kynurenines,are also induced by IFN-g and coordinately upregulated with PD-L1.The concept of adaptive resistance does not solely apply to induction of PD-L1on tumor cells.Early studies demonstrated that PD-L1 expression on myeloid cells,including dendritic cells,can signifi-cantly impair activation of tumor-specific T cells.Inhibition of T cell responses can be mediated by PD-L1+suppressive myeloid cells or dendritic cells(DCs)in the TME as well as in tu-mor-draining lymph nodes(Curiel et al.,2003).In some tumors, such as microsatellite instability(MSI)colon cancer,myeloid rather than tumor cells are the major cell type expressing PD-L1 (Llosa et al.,2015).A recent report suggests that PD-L1expres-sion by infiltrating myeloid cells rather than tumor cells is more predictive of response to PD-1pathway blockade(Herbst et al., 2014).The relative importance of PD-L1expression on leukocytes in the TME,which would provide‘‘third-party’’inhibition,versus direct expression by the tumor cells,remains to be determined. Implications of Adaptive Immune ResistanceThe adaptive resistance mechanism of intratumoral PD-L1in-duction,together with the broad therapeutic activity of PD-1 pathway blockade in human cancer,validates one of the most important tenets underlying cancer immunology and immuno-therapy,namely,that many cancer patients contain a significant repertoire of tumor-specific T cells capable of killing their tumor save for the adaptive induction of immune checkpoints in the TME.It also implies that PD-L1expression in the tumor repre-sents a measure of the potential for a patient’s immune system to recognize the tumor.One of the major unanswered questions is this:what are the dominant antigenic targets that T cells recog-nize when checkpoints are blocked?Circumstantial evidence supports the notion that neoantigens created by the multiple somatic mutations in cancers provide such targets.Indeed,a recent report has demonstrated that melanomas with higher mutational loads were more responsive to anti-CTLA-4therapy (Snyder et al.,2014).Also,the tumor types that have been shown to respond to anti-PD-1/PD-L1therapy tend to be those with higher median mutational loads(i.e.,carcinogen-induced can-cers such as melanoma,lung,bladder,and head and neck can-cers).However,there has been much evidence over the past20 years that shared self-antigens upregulated in tumors by epige-netic mechanisms(e.g.,cancer-testis antigens)are also able to provide selective tumor targeting.The relative importance of mutation-dependent,tumor-specific neoantigens versus tumor-associated self-antigens as T cell targets remains to be determined.Finally,the adaptive resistance mechanism has profound im-plications for developing synergistic combinatorial cancer im-munotherapies.One of the most promising general approaches to immunotherapy utilizes positive drivers of anti-tumor immune responses,such as vaccines,intratumoral injection of immune activators,and co-stimulatory receptor agonists.These modal-ities with the potential to enhance anti-tumor responses would also be expected to enhance the adaptive induction of check-points like PD-1ligands.This has,in fact,been demonstrated in animal models of vaccination(Fu et al.,2014).Therefore,pos-itive drivers of anti-tumor immunity may be synergistic with PD-1 pathway inhibitors.Such approaches are just beginning to enter the clinic.Clinical Impact of Drugs Blocking CTLA-4and PD-1Anti-CTLA-4The anti-CTLA-4monoclonal antibodies(mAbs)ipilimumab,a fully human IgG1(Bristol-Myers Squibb),and tremelimumab,a fully human IgG2(Pfizer,MedImmune),were thefirst immune checkpoint-blocking drugs to enter clinical testing in oncology. Although designed as CTLA-4-blocking mAbs,these drugs have recently been postulated to have unique functions en-dowed by their specific isotypes,with evidence suggesting that ipilimumab may deplete Tregs overexpressing CTLA-4 (Selby et al.,2013).In2011,ipilimumab was approved in the United States and Europe as therapy for advanced unresectable melanoma based on results from two phase III trials showing significant extensions in overall survival(OS)(Hodi et al.,2010; Robert et al.,2011).Long-term follow-up in a pooled meta-anal-ysis of1,861melanoma patients receiving ipilimumab in phase II or III trials revealed durable survival in approximately20%,in some cases extending to10years(Schadendorf et al.,2015). Interestingly,this survival rate is approximately double the observed rate of tumor regressions measured by standard oncologic criteria( 10%complete responses[CRs]and partial responses[PRs]).Factors contributing to this phenomenon may include prolonged disease stabilization,unconventional ‘‘immune-related’’response patterns,or a heightened respon-siveness of ipilimumab-refractory patients to subsequent thera-pies.Although tremelimumab,a distinct CTLA-4-blocking mAb, showed promise in early-phase melanoma trials,it did not meet its designated endpoint when randomized against standard chemotherapy in afirst-line phase III melanoma trial(Ribas et al.,2013).Ipilimumab has so far shown only modest anti-tumor effects in non-melanoma cancers,and tremelimumab is still in early testing for these indications(reviewed in Weber,2014).Kidney,lung, and prostate cancer have been the most intensively studied. In a phase II study of metastatic renal cell carcinoma(RCC, n=61),a partial response rate of10%was observed with ipilimu-mab monotherapy(Yang et al.,2007).In lung cancer,treatment-naive patients with non-small-cell lung cancer(NSCLC)(n=204) or extensive disease small-cell lung cancer(ED-SCLC)(n=130) received standard chemotherapy alone or combined with ipili-mumab during initial(‘‘concurrent’’)or later(‘‘phased’’)chemo-therapy cycles in a phase III trial(Lynch et al.,2012;Reck et al.,2013).For both diseases,a brief but statistically significant 1-month extension of progression-free survival measured by im-mune-related criteria(irPFS)was observed in patients receiving phased ipilimumab plus chemotherapy compared with chemo-therapy alone.In NSCLC,there was also a significant1-month extension of PFS measured by standard criteria in the phased ipilimumab arm.Although ipilimumab did not have a significant impact on OS in either NSCLC or ED-SCLC,a subset analysis appeared to show improved activity in patients with squamous NSCLC,providing the basis for an ongoing phase III trial of Cancer Cell27,April13,2015ª2015Elsevier Inc.453ipilimumab plus chemotherapy in this histology.Similarly,trials of ipilimumab in metastatic castration-resistant prostate cancer (mCRPC)have yielded weak but positive signals of activity.In phase I/II trials in which patients received ipilimumab alone or combined with systemic granulocyte-macrophage colony-stim-ulating factor or focal radiotherapy,prostate-specific antigen re-ductions of R 50%were observed in some patients,and isolated examples of measurable tumor regression were reported (Fong et al.,2009;Slovin et al.,2013),supporting further study.In a phase III trial of ipilimumab versus placebo after bone-directed radiotherapy in 799patients with docetaxel-refractory mCRPC,median OS was 11.2versus 10.0months,respectively (p =0.053),failing to meet the trial’s primary endpoint (Kwon et al.,2014).However,there was a statistically significant 1-month improvement in PFS and a suggestion that OS was prolonged in a subgroup of patients with favorable prognostic features.A separate phase III trial of ipilimumab in chemotherapy-naive pa-tients with asymptomatic or minimally symptomatic mCRPC without visceral metastases has recently completed accrual.Valuable clinical experience gained from studies of anti-CTLA-4mAbs paved a path for accelerated development of other drugs in class by providing a framework for treatment strat-egy,toxicity management,and efficacy evaluation.New princi-ples emerged that distinguished immune checkpoint blockade from traditional cancer therapies.First,a new category of side effects,so-called ‘‘immune-related adverse events’’(irAEs),was recognized and characterized,leading to algorithm devel-opment for early detection and management.Drug-related irAEs were severe in 15%–30%of patients receiving anti-CTLA-4,sometimes resulting in fatalities.These irAEs were associated with inflammation in normal tissues such as the gut,skin,and endocrine glands and resembled phenotypes observed in hu-man CTLA-4heterozygotes with reduced CTLA-4expression (Topalian and Sharpe,2014).Their occurrence in individuals with no prior history of autoimmunity validates the mechanism of action of anti-CTLA-4in ‘‘releasing the brakes’’on immune re-sponses and underscores the precarious balance that normallyexists between self-tolerance and autoimmunity.Second,a new category of clinical response termed ‘‘immune-related response’’was recognized in which major and durable tumor regressions could occur after apparent initial disease progres-sion on treatment (Wolchok et al.,2009).Tumor enlargement measured by conventional radiologic scans may result from drug-induced inflammation at tumor sites or could reflect actual tumor growth followed by delayed regression.Such phenomena pose challenges for the appropriate management of individual patients and the selection of informative endpoints for trials of immune checkpoint-blocking drugs.Drugs Blocking the PD-1PathwayInformation garnered from trials of anti-CTLA-4agents fast-for-warded the development of drugs blocking PD-1or its major ligand,PD-L1.As predicted by murine models,these drugs have heightened tumor selectivity and reduced toxicity com-pared with anti-CTLA-4,supporting their administration in an outpatient setting.Furthermore,although they are effective against advanced treatment-refractory melanoma,with recent regulatory approvals for two anti-PD-1drugs in this setting,they also appear to have a much broader spectrum of anti-tumor activity than anti-CTLA-4.Reproducible and durable regressions of epithelial cancers (lung,head and neck,and bladder cancers,among others)have catapulted the launching of hundreds of ongoing clinical trials in diverse disease indications.Although several different anti-PD-1/PD-L1-blocking mAbs are currently in clinical testing (Table 1),the fact that anti-tumor activity has been observed with all of them highlights the PD-1pathway as a dominant intratumoral immunosuppressive pathway and a key target in cancer therapy.The first-in-human trial of nivolumab anti-PD-1provided sem-inal evidence that this treatment approach could potentially impact diverse cancer types,including common epithelial can-cers,with objective responses reported in patients with mela-noma,kidney,and colorectal cancer (Brahmer et al.,2010).A transient tumor regression in one patient with NSCLC provided the impetus for investigating a larger NSCLCcohortTable 1.Drugs in Clinical Development that Block PD-1or PD-L1Isotype and bFc-modified mAbs were engineered to abrogate ADCC and complement-dependent cytotoxicity (CDC).454Cancer Cell 27,April 13,2015ª2015Elsevier Inc.。