Progress in the pathogenesis, prevention and treatment of renal injury caused by immune checkpoint inhibitors

Wu Huan, Yu Xiaofang

PDF(20182 KB)
Chinese Journal of Nephrology ›› 2022, Vol. 38 ›› Issue (9) : 844-849. DOI: 10.3760/cma.j.cn441217-20211126-00122
Review

Progress in the pathogenesis, prevention and treatment of renal injury caused by immune checkpoint inhibitors

Author information +
History +

Abstract

Cite this article

Download Citations
Wu Huan. , Yu Xiaofang. Progress in the pathogenesis, prevention and treatment of renal injury caused by immune checkpoint inhibitors[J]. Chinese Journal of Nephrology, 2022, 38(9): 844-849. DOI: 10.3760/cma.j.cn441217-20211126-00122.
免疫疗法是一种新型的治疗恶性肿瘤的方法,近年来以免疫检查点抑制剂(immune checkpoint inhibitors,ICIs)为代表的免疫疗法通过激活患者自身T淋巴细胞的抗肿瘤免疫功能,发挥杀灭肿瘤细胞的作用,在临床上已被广泛使用。目前,已有7种ICIs被美国食品和药物管理局(food and drugs administration,FDA)批准用于治疗肿瘤(表1),其中抗细胞毒性T淋巴细胞相关抗原4(cytotoxic T-lymphocyte-associated protein 4,CTLA-4)抗体通过阻止CTLA-4竞争共刺激分子,抗程序性死亡蛋白1(programmed death 1,PD-1)抗体与抗程序性死亡蛋白配体1(programmed death-ligand 1,PD-L1)抗体通过阻止PD-1与PD-L1结合,从而起到激活免疫系统的作用。但是,被ICIs激活的T细胞不能区分肿瘤细胞和正常组织,导致包括肾脏在内的正常组织受到免疫损伤,即免疫相关不良反应(immune-related adverse events,irAEs)。本文将简述ICIs所致肾损伤的发病机制、临床表现和防治进展。
表1 美国食品和药物管理局批准的免疫检查点抑制剂(ICIs)及其适应证[1]
分类 药物 适应证
CTLA-4抑制剂 伊匹单抗 黑色素瘤、肾细胞癌
PD-1抑制剂
尼鲁单抗
黑色素瘤、非小细胞肺癌、小细胞肺癌、肾细胞癌、经典霍奇金淋巴瘤、头颈部鳞状细胞癌、尿路上皮癌、结直肠癌、肝细胞癌
帕博利珠单抗
黑色素瘤、非小细胞肺癌、霍奇金淋巴瘤、原发性纵隔大B细胞淋巴瘤、头颈部鳞状细胞癌、尿路上皮癌、胃癌、宫颈癌、具有高度微卫星不稳定性或错配修复缺陷的实体瘤
西米普利单抗 皮肤鳞状细胞癌
PD-L1抑制剂 阿特朱单抗 非小细胞肺癌、尿路上皮癌
德鲁单抗 尿路上皮癌、非小细胞肺癌
阿维鲁单抗 默克尔细胞癌、尿路上皮癌
注:CTLA-4:细胞毒性T淋巴细胞相关抗原4;PD-1:程序性死亡蛋白1;PD-L1:程序性死亡蛋白配体1

一、 不同ICIs所致肾损伤的机制和表现

1. CTLA-4抑制剂: CTLA-4是T细胞上的一种跨膜受体,与T细胞上的共刺激受体CD28竞争B7分子,而CTLA-4与B7分子结合后可诱导T细胞失能,抑制免疫系统对肿瘤的杀伤[2]。CTLA-4抑制剂通过阻断CTLA-4与B7分子结合,从而激活T细胞,发挥抗肿瘤的作用。目前认为,CTLA-4通过调节次级淋巴器官中抗原呈递细胞(APC)和T细胞之间的相互作用来调节外周免疫耐受,故CTLA-4抑制剂作用范围更广,与PD-1和PD-L1抑制剂相比,它的irAEs也更大[3]。有研究表明,CTLA-4基因敲除小鼠会患致死性的淋巴增生性疾病,在小鼠出生5~6 d后即可检测到T细胞的活化,18~28 d后小鼠因淋巴细胞浸润到非淋巴组织而死亡[4]。此外,CTLA-4抑制剂和PD-1抑制剂都可能刺激B细胞产生抗体,而这些抗体与肾小管上皮细胞、系膜细胞或足细胞上的自身抗原结合形成免疫复合物,导致肾脏损伤[5]。Mamlouk等[6]报道3例接受CTLA-4抑制剂治疗后出现急性肾损伤(acute kidney injury,AKI)的患者,表现为镜下血尿、少量蛋白尿、白细胞尿和血肌酐升高,肾活检均提示急性间质性肾炎(acute interstitial nephritis,AIN)合并肾小球肾炎。Cortazar等[7]报道的10例接受CTLA-4抑制剂治疗后出现AKI的患者中,血肌酐峰值在2.9~11.7 mg/dl(1 mg/dl=88.4 μmol/L)之间,其中7例患者有白细胞尿,2例患者有镜下血尿,病理改变除AIN外还合并有不同程度的足突消失;同时,半数以上的患者合并至少1种肾外irAEs,如甲状腺炎、结肠炎、肝功能损伤等。值得注意的是,ICIs相关肾损伤的发生存在显著潜伏期[8],有研究提示单独使用CTLA-4抑制剂或联合使用PD-1抑制剂的患者发生AKI的潜伏期长达9周[6]
=2. PD-1抑制剂与PD-L1抑制剂: PD-1是一种表达于T细胞上的共抑制分子。据报道,肿瘤细胞过度表达PD-1配体(PD-L1或PD-L2),与T细胞上的PD-1结合后可抑制T细胞活化,从而逃避宿主免疫监视[2]。PD-1和PD-L1抑制剂导致肾损伤的机制可能是肾小管上皮细胞表达PD-L1,保护其免受T细胞介导的自身免疫损伤;当患者接受PD-L1抑制剂治疗时自身反应性T细胞被激活,攻击正常的肾脏组织,从而导致AIN。同时,其他易导致AIN的药物(如质子泵抑制剂、非甾体抗炎药等)或其代谢产物可作为一种半抗原与肾小管基底膜正常成分结合形成完全抗原,使T细胞致敏;当患者使用PD-1抑制剂后,T细胞因被再次激活而失去免疫耐受,攻击自身组织[9]。此外,有研究表明在使用ICIs前,PD-L1在一些基础肾脏病中已有表达,并且在糖尿病肾脏疾病、ANCA相关性小血管炎、狼疮肾炎中表达较为显著,这提示有基础肾脏病的患者使用ICIs后更易出现肾损伤[10]
接受PD-1或PD-L1抑制剂治疗的肾损伤患者最常见的病理表现是AIN,如间质水肿并伴有不同程度的淋巴细胞、浆细胞浸润,与其他药物如质子泵抑制剂、非甾体抗炎药等所致AIN并无显著差别;部分患者除AIN外还可伴有肾小球病变,如局灶节段性肾小球硬化、膜性肾病、微小病变性肾病等[6,11-12];少数患者也可仅表现为微小病变性肾病或IgA肾病[12-14]。就临床表现而言,AKI在ICIs所致肾损伤中最为常见,主要表现为少量蛋白尿和不同程度的血肌酐升高,少数患者可出现大量蛋白尿,部分患者有白细胞尿和镜下血尿;同时,部分患者会合并至少1种肾外irAEs[15]。本科室曾收治过1例使用帕博利珠单抗后出现蛋白尿、血尿和血肌酐升高的肺癌患者,肾活检病理提示为IgA肾病合并AIN(见图1)。近几年也有病例报告提示PD-1抑制剂相关的AIN可出现远端肾小管酸中毒[16-17],表现为先于血肌酐升高的电解质紊乱(高氯血症、低钾血症、酸中毒)。与CTLA-4抑制剂相比,PD-1抑制剂所致肾损伤发生时间更晚:单独使用尼鲁单抗的中位潜伏期为20周,单独使用帕博利珠单抗的中位潜伏期为13.5周[6],Seethapathy等[18]报道的接受PD-L1抑制剂治疗的患者发生AKI的中位潜伏期是99 d。
图1 免疫检查点抑制剂(ICIs)所致肾脏不良反应的肾脏病理学表现(×200)
注:A:HE染色;B:IgA免疫荧光

Full size|PPT slide

二、 ICIs所致肾损伤的预防、诊断及治疗

1. 预防: 由于质子泵抑制剂或非甾体抗炎药具有半抗原的作用,因此需避免ICIs与这些药物联合使用。几项回顾性研究发现,同时使用质子泵抑制剂、非甾体抗炎药的患者更有可能发生AIN[15,19-20],如Manohar等[15]发现,在接受ICIs治疗后发生AIN的14例患者中,有11例使用过质子泵抑制剂,3例使用过非甾体抗炎药;Seethapathy等[18]也有类似的报道,其中5例ICIs相关AKI患者都曾使用过质子泵抑制剂、非甾体抗炎药或抗生素中的一种。但上述回顾性研究中大部分患者为白种人,目前尚不清楚种族对肾脏相关免疫不良反应有无影响。
已有研究证实联合使用CTLA-4抑制剂和PD-1抑制剂可以通过激活不同类型的T细胞来启动抗肿瘤免疫杀伤,因此联合使用ICIs可能会获得更好的抗肿瘤作用[21],但由此也会产生更严重的irAEs。从预防肾损伤而言,应避免两种ICIs的联合使用。不过,近期一项meta分析却发现,ICIs联合常规化疗药物治疗较两种ICIs联合治疗方案所产生的肾毒性更大[22]
用药前已有肾功能减退[eGFR<30 ml·min-1·(1.73 m2)-1]的患者更有可能发生肾损伤,但目前尚未明确是否与ICIs导致的免疫损伤有关。已有的研究表明,ICIs相关AKI的发生与患者年龄、性别、所患肿瘤类型无关,而与患者基础肾功能的减退程度密切相关[6,18,20,23]。在Cortazar等[20]进行的一项回顾性研究中,138例ICIs相关AKI患者被纳入病例组,276例接受ICIs治疗但未发生AKI的患者被纳入对照组,分析发现,接受ICIs治疗的Ⅳ期慢性肾脏病(chronic kidney disease,CKD)患者发生肾损伤的风险明显增加,因此,接受ICIs治疗的CKD患者应密切监测肾损伤相关指标。
近期研究表明,体重指数(BMI)≥25 kg/m2且代谢性疾病(糖尿病、血脂异常、高血压等)小于2种的患者与BMI在18.5~24.9 kg/m2之间、代谢性疾病小于2种的患者相比,前者发生2级以上irAEs的风险更高[24]。一项动物实验发现饮食诱导的肥胖小鼠T细胞上PD-1表达增加;临床研究也发现,BMI≥30 kg/m2的患者T细胞上PD-1表达增加[25]。以上研究提示,肥胖患者在接受ICIs治疗后发生irAEs的可能性更大。因此,接受ICIs治疗的肥胖患者也应密切监测肾损伤相关指标。
2. 诊断: 目前关于ICIs相关肾损伤的诊断标准尚未统一。美国临床肿瘤学会(American Society of Clinical Oncology,ASCO)建议定期监测血肌酐水平来观察患者肾功能的变化并排除其他原因导致的肾脏不良反应[26];而美国国家综合癌症网络(National Comprehensive Cancer Network,NCCN)还推荐定期监测患者尿蛋白水平[27]。但以上策略并不能起到早期诊断的作用。为了及时治疗患者,减少irAEs带来的不良反应,我们应积极寻找ICIs相关肾损伤的早期生物标志物,以达到早诊断、早治疗的目的。
近期研究提示,尿液中的新型细胞因子生物标志物白细胞介素9(IL-9)和肿瘤坏死因子α(TNF-α)可有效区分AIN、急性肾小管坏死和其他肾脏病变,有望成为早期诊断ICIs相关肾脏不良反应的无创性筛查方法[28]。一项针对PD-1抑制剂治疗实体瘤患者的研究发现,治疗前外周血淋巴细胞计数较高的患者发生irAEs的风险更大,但该项研究中仅1例发生了肾损伤[29],因此治疗前淋巴细胞计数与ICIs相关肾损伤的相关性尚待进一步证实。就影像学检查而言,Qualls等[30]首次报道ICIs相关AIN患者肾皮质18F-脱氧葡萄糖(FDG)摄取增加,而3例非AIN导致的AKI患者PET-CT扫描中FDG摄取无改变或减少。由于AIN典型的病理表现为单核细胞和淋巴细胞浸润[31],因此推测FDG摄取增强是由新陈代谢活跃的炎症细胞浸润肾小管间质所致。这一研究结果提示,PET-CT可能辅助诊断ICIs相关AIN,尤其对不能及时进行肾活检的患者。但目前该技术尚不成熟,一方面单一的病例报告证据不足,另一方面昂贵的检查费也限制了它的推广使用。
肾活检作为一种有创检查,是诊断肾脏病的金标准,但是否有必要对使用ICIs后出现肾损伤的患者都进行肾活检目前尚无定论[32]。一些学者建议在没有肾活检禁忌证的情况下进行肾活检以明确病理诊断[32-33],如一旦确认肾损伤发生的原因与ICIs无关,则不仅能避免不必要的糖皮质激素干预,而且还能继续接受原ICIs治疗。一项关于ICIs的多中心研究发现,接受肾活检的AKI患者中有93%被证实为AIN,其中87%的患者在接受糖皮质激素治疗后肾功能完全或部分恢复[20],因此有学者认为,ICIs相关肾损伤主要是AIN所致,而肾活检后也不大可能会改变既定的糖皮质激素治疗这一经验方案,故不推荐常规进行肾活检[32]
由于使用ICIs的患者常同时使用其他易导致AIN的药物,如质子泵抑制剂、非甾体抗炎药等,临床上诊断为肾损伤后需鉴别诊断是ICIs所致的肾损伤,抑或是其他药物所致的肾损伤,可进一步结合患者的用药史、发病时间、有无合并其他肾外irAEs等加以判断。一般来说,ICIs相关肾损伤的发生存在显著潜伏期,如使用CTLA-4抑制剂的潜伏时间为9周左右,使用PD-1抑制剂或PD-L1抑制剂的潜伏时间会更长;部分患者在发生肾损伤时常同时合并其他器官的irAEs,如肝功能损伤、垂体功能减退、甲状腺功能减退、结肠炎等[6-7]
3.治疗: 糖皮质激素是治疗AIN的常用药物。Manohar等[15]发现,在治疗ICIs所致肾损伤的第1个月内,糖皮质激素起始剂量较大的患者肾功能恢复得更好,提示糖皮质激素起始剂量可能在治疗中起重要作用。如患者出现激素抵抗或明显不良反应时,应考虑更换或加用免疫抑制剂,如霉酚酸酯、英夫利昔单抗或利妥昔单抗[1]。至于使用免疫抑制剂是否会影响ICIs的抗肿瘤效果,一项回顾性研究显示,加用免疫抑制剂治疗irAEs并不会影响肿瘤患者的预后,该项研究中有254例患者发生ICIs相关irAEs,其中103例接受糖皮质激素治疗,29例加用英夫利昔单抗,结果发现,与发生irAEs但未使用糖皮质激素/英夫利昔单抗治疗的患者相比,两者总生存期无明显差异[34]。Mamlouk等[6]报道6例肾活检证实为AIN伴有肾小球损伤的ICIs相关肾损伤患者接受免疫抑制剂治疗后,与未接受免疫抑制治疗的10例患者相比,其无进展生存期亦无明显差异。因此,在必要的情况下,可以考虑使用免疫抑制剂治疗ICIs所致肾损伤。
另外,在患者肾损伤恢复后能否及何时再次进行ICIs治疗也是一个亟待解决的临床问题。目前,大多数观点认为,当患者血肌酐达到基础值的3倍以上或>4.0 mg/dl时应永久停止ICIs治疗[26-27]。Taki等[35]报道1例75岁的非小细胞肺癌患者接受帕博利珠单抗治疗后发生AIN,血肌酐水平从0.75 mg/dl升高到1.5 mg/dl,糖皮质激素治疗后肾功能恢复,再次接受ICIs治疗后未见肾损伤复发。另有研究发现,对于糖皮质激素治疗后血肌酐未恢复正常,或需要较长时间(>30 d)才能恢复正常,或既往已有CKD的患者,再次接受ICIs治疗后出现永久性肾损伤的风险更高,故认为只有对于接受糖皮质激素治疗后肾功能迅速恢复的患者可以再次进行ICIs治疗[36]。因此,我们建议,是否再次进行ICIs治疗需要患者、肿瘤科医师和肾内科医师谨慎地权衡利弊,一旦再次使用,必须密切监测血肌酐等肾损伤指标以期早期识别肾脏不良反应的发生,并及时采用糖皮质激素和/或免疫抑制治疗。图2为结合ASCO指南推荐的诊治流程[26,37]
图2 ICIs所致肾脏不良反应的诊治流程[26,37]
注:ICIs:免疫检查点抑制剂;AKI:急性肾损伤; irAEs:免疫相关不良反应;AIN:急性间质性肾炎;AKI 诊断标准参考2012年KDIGO急性肾损伤临床实践指南[38],1期:血肌酐达基础值的1.5~1.9倍或上升≥0.3 mg/dl(1 mg/dl=88.4 μmol/L);2期:血肌酐达基础值的2.0~2.9倍;3期:血肌酐达基础值的3倍及以上,或升至≥4 mg/dl

Full size|PPT slide

三、 结语

ICIs在激活T细胞治疗肿瘤的同时可能会导致irAEs。肾脏相关免疫不良反应作为其中的一种,其机制尚未完全阐明,AIN是其最常见的病理类型,血肌酐水平和蛋白尿有助于临床监测和筛选ICIs相关肾损伤的发生。新型生物标志物的发现为肾损伤的早期诊断提供了可能,必要的肾活检也可以鉴别ICIs相关肾损伤发生的病因。目前,通常根据患者肾损伤的严重程度对其进行管理,停用ICIs并接受糖皮质激素治疗是首选的治疗方法。大多数出现ICIs相关肾损伤的患者在及时诊断和治疗的情况下可以实现肾脏功能完全或部分恢复,而多学科协作可以进一步提高ICIs所致肾损伤的治疗效果。

References

[1]
Herrmann SM, Perazella MA. Immune checkpoint inhibitors and immune-related adverse renal events[J]. Kidney Int Rep, 2020, 5(8): 1139-1148. DOI: 10.1016/j.ekir.2020.04.018.
Renal toxicities have been increasingly recognized as complications of the immune checkpoint inhibitors (ICIs). Recent studies have outlined the incidence and potential risk factors for nephrotoxicity. For clinicians, the key question is how to manage patients who develop these adverse renal effects. This is of paramount importance to providers as ICI use for cancer therapy becomes more widespread and nephrotoxicity increasingly develops. As clinicians encounter ICI-associated nephrotoxicity, an appropriate approach to management is required to facilitate the best outcomes in patients with cancer. Importantly, ICI rechallenge in patients who developed ICI-related acute kidney injury (AKI) is unclear and represents a conundrum for providers. Clinicians struggle with the "if, when, and how to" questions related to ICI rechallenge in this subset of patients. In addition, ICI use in the transplant population raises concerns for promoting acute rejection when treating cancer in these patients. We herein review current information on these various topics.© 2020 International Society of Nephrology. Published by Elsevier Inc.
[2]
Murakami N, Motwani S, Riella LV. Renal complications of immune checkpoint blockade[J]. Curr Probl Cancer, 2017, 41(2): 100-110. DOI: 10.1016/j.currproblcancer.2016.12.004.
[3]
Wanchoo R, Karam S, Uppal NN, et al. Adverse renal effects of immune checkpoint inhibitors: a narrative review[J]. Am J Nephrol, 2017, 45(2): 160-169. DOI: 10.1159/000455014.
Cancer immunotherapy, such as anti-cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) and anti-programmed death 1 (PD-1), has revolutionized the treatment of malignancies by engaging the patient's own immune system against the tumor rather than targeting the cancer directly. These therapies have demonstrated a significant benefit in the treatment of melanomas and other cancers.In order to provide an extensive overview of the renal toxicities induced by these agents, a Medline search was conducted of published literature related to ipilimumab-, pembrolizumab-, and nivolumab-induced kidney toxicity. In addition, primary data from the initial clinical trials of these agents and the FDA adverse reporting system database were also reviewed to determine renal adverse events. Acute interstitial nephritis (AIN), podocytopathy, and hyponatremia were toxicities caused by ipilimumab. The main adverse effect associated with both the PD-1 inhibitors was AIN. The onset of kidney injury seen with PD-1 inhibitors is usually late (3-10 months) compared to CTLA-4 antagonists related renal injury, which happens earlier (2-3 months). PD-1 as opposed to CTLA-4 inhibitors has been associated with kidney rejection in transplantation. Steroids appear to be effective in treating the immune-related adverse effects noted with these agents. Key Message: Although initially thought to be rare, the incidence rates of renal toxicities might be higher (9.9-29%) as identified by recent studies. As a result, obtaining knowledge about renal toxicities of immune checkpoint inhibitors is extremely important.© 2017 S. Karger AG, Basel.
[4]
Chambers CA, Kuhns MS, Egen JG, et al. CTLA-4-mediated inhibition in regulation of T cell responses: mechanisms and manipulation in tumor immunotherapy[J]. Annu Rev Immunol, 2001, 19: 565-594. DOI: 10.1146/annurev.immunol.19.1.565.
The T cell compartment of adaptive immunity provides vertebrates with the potential to survey for and respond specifically to an incredible diversity of antigens. The T cell repertoire must be carefully regulated to prevent unwanted responses to self. In the periphery, one important level of regulation is the action of costimulatory signals in concert with T cell antigen-receptor (TCR) signals to promote full T cell activation. The past few years have revealed that costimulation is quite complex, involving an integration of activating signals and inhibitory signals from CD28 and CTLA-4 molecules, respectively, with TCR signals to determine the outcome of a T cell's encounter with antigen. Newly emerging data suggest that inhibitory signals mediated by CTLA-4 not only can determine whether T cells become activated, but also can play a role in regulating the clonal representation in a polyclonal response. This review primarily focuses on the cellular and molecular mechanisms of regulation by CTLA-4 and its manipulation as a strategy for tumor immunotherapy.
[5]
Izzedine H, Gueutin V, Gharbi C, et al. Kidney injuries related to ipilimumab[J]. Invest New Drugs, 2014, 32(4): 769-773. DOI: 10.1007/s10637-014-0092-7.
[6]
Mamlouk O, Selamet U, Machado S, et al. Nephrotoxicity of immune checkpoint inhibitors beyond tubulointerstitial nephritis: single-center experience[J]. J Immunother Cancer, 2019, 7(1): 2. DOI: 10.1186/s40425-018-0478-8.
The approved therapeutic indication for immune checkpoint inhibitors (CPIs) are rapidly expanding including treatment in the adjuvant setting, the immune related toxicities associated with CPI can limit the efficacy of these agents. The literature on the nephrotoxicity of CPI is limited. Here, we present cases of biopsy proven acute tubulointerstitial nephritis (ATIN) and glomerulonephritis (GN) induced by CPIs and discuss potential mechanisms of these adverse effects.We retrospectively reviewed all cancer patients from 2008 to 2018 who were treated with a CPI and subsequently underwent a kidney biopsy at The University of Texas MD Anderson Cancer Center.We identified 16 cases diagnosed with advanced solid or hematologic malignancy; 12 patients were male, and the median age was 64 (range 38 to 77 years). The median time to developing acute kidney injury (AKI) from starting CPIs was 14 weeks (range 6-56 weeks). The average time from AKI diagnosis to obtaining renal biopsy was 16 days (range from 1 to 46 days). Fifteen cases occurred post anti-PD-1based therapy. ATIN was the most common pathologic finding on biopsy (14 of 16) and presented in almost all cases as either the major microscopic finding or as a mild form of interstitial inflammation in association with other glomerular pathologies (pauci-immune glomerulonephritis, membranous glomerulonephritis, C3 glomerulonephritis, immunoglobulin A (IgA) nephropathy, or amyloid A (AA) amyloidosis). CPIs were discontinued in 15 out of 16 cases. Steroids and further immunosuppression were used in most cases as indicated for treatment of ATIN and glomerulonephritis (14 of 16), with the majority achieving complete to partial renal recovery.Our data demonstrate that CPI related AKI occurs relatively late after CPI therapy. Our biopsy data demonstrate that ATIN is the most common pathological finding; however it can frequently co-occur with other glomerular pathologies, which may require immune suppressive therapy beyond corticosteroids. In the lack of predictive blood or urine biomarker, we recommend obtaining kidney biopsy for CPI related AKI.
[7]
Cortazar FB, Marrone KA, Troxell ML, et al. Clinicopathological features of acute kidney injury associated with immune checkpoint inhibitors[J]. Kidney Int, 2016, 90(3): 638-647. DOI: 10.1016/j.kint.2016.04.008.
Immune checkpoint inhibitors (CPIs), monoclonal antibodies that target inhibitory receptors expressed on T cells, represent an emerging class of immunotherapy used in treating solid organ and hematologic malignancies. We describe the clinical and histologic features of 13 patients with CPI-induced acute kidney injury (AKI) who underwent kidney biopsy. Median time from initiation of a CPI to AKI was 91 (range, 21 to 245) days. Pyuria was present in 8 patients, and the median urine protein to creatinine ratio was 0.48 (range, 0.12 to 0.98) g/g. An extrarenal immune-related adverse event occurred prior to the onset of AKI in 7 patients. Median peak serum creatinine was 4.5 (interquartile range, 3.6-7.3) mg/dl with 4 patients requiring hemodialysis. The prevalent pathologic lesion was acute tubulointerstitial nephritis in 12 patients, with 3 having granulomatous features, and 1 thrombotic microangiopathy. Among the 12 patients with acute tubulointerstitial nephritis, 10 received treatment with glucocorticoids, resulting in complete or partial improvement in renal function in 2 and 7 patients, respectively. However, the 2 patients with acute tubulointerstitial nephritis not given glucocorticoids had no improvement in renal function. Thus, CPI-induced AKI is a new entity that presents with clinical and histologic features similar to other causes of drug-induced acute tubulointerstitial nephritis, though with a longer latency period. Glucocorticoids appear to be a potentially effective treatment strategy. Hence, AKI due to CPIs may be caused by a unique mechanism of action linked to reprogramming of the immune system, leading to loss of tolerance.Copyright © 2016 International Society of Nephrology. Published by Elsevier Inc. All rights reserved.
[8]
Tinawi M, Bastani B. Nephrotoxicity of immune checkpoint inhibitors: acute kidney injury and beyond[J]. Cureus, 2020, 12(12): e12204. DOI: 10.7759/cureus.12204.
[9]
Koda R, Watanabe H, Tsuchida M, et al. Immune checkpoint inhibitor (nivolumab)-associated kidney injury and the importance of recognizing concomitant medications known to cause acute tubulointerstitial nephritis: a case report[J]. BMC Nephrol, 2018, 19(1): 48. DOI: 10.1186/s12882-018-0848-y.
Background: Acute tubulointerstitial nephritis (ATIN) has been increasingly recognized as an important manifestation of kidney injury associated with the use of immune checkpoint inhibitors (anti-PD-1 and anti-CTLA-4). While the exact pathophysiology remains unknown, corticosteroids are the mainstay of management.Case presentation: We describe a 67-year-old man with stage IV non-small-cell lung cancer who developed kidney injury during treatment with the anti-PD-1 antibody nivolumab. A kidney biopsy showed ATIN without granuloma formation. Considering their mechanism of action, immune checkpoint inhibitors can alter immunological tolerance to concomitant drugs that have been safely used for a long time. For more than 4 years before the initiation of nivolumab therapy, the patient had been receiving the proton pump inhibitor lansoprazole, known to cause drug-induced ATIN, without significant adverse events including kidney injury. He showed rapid improvement in kidney function in 3 days (creatinine decreased from 2.74 to 1.82 mg/dl) on discontinuation of lansoprazole. He then received 500 mg intravenous methylprednisolone for 3 days followed by 1 mg/kg/day oral prednisolone and his creatinine levels eventually stabilized around 1.7 mg/dl. Drug-induced lymphocyte stimulation test (DLST) for lansoprazole was positive.Conclusions: The rapid improvement of kidney function after discontinuation and DLST positivity indicate that lansoprazole contributed to the development of ATIN during nivolumab therapy. Considering the time course, it is plausible that nivolumab altered the long-lasting immunological tolerance against lansoprazole in this patient. To the best of our knowledge, this is the first case report of DLST positivity for a drug that had been used safely before the initiation of an immune checkpoint inhibitor. Although corticosteroid therapy is recommended, the recognition and discontinuation of concomitant drugs, especially those known to induce ATIN, is necessary for the management of kidney injury associated with anti-PD-1 therapy.
[10]
Hakroush S, Kopp SB, Tampe D, et al. Variable expression of programmed cell death protein 1-ligand 1 in kidneys independent of immune checkpoint inhibition[J]. Front Immunol, 2020, 11: 624547. DOI: 10.3389/fimmu.2020.624547.
Due to recent advantages in cancer therapy, immune checkpoint inhibitors (ICIs) are new classes of drugs targeting programmed cell death protein 1 (PD-1) or its ligand programmed cell death protein 1-ligand 1 (PD-L1) used in many cancer therapies. Acute interstitial nephritis (AIN) is a potential and deleterious immune-related adverse events (irAE) in the kidney observed in patients receiving ICIs and the most common biopsy-proven diagnosis in patients who develop acute kidney injury (AKI). Based on previous reports, AIN in patients receiving ICIs is associated with tubular positivity for PD-L1, implicating that PD-L1 positivity reflects susceptibility to develop renal complications with these agents. It remains unclear if PD-L1 positivity is acquired specifically during ICI therapy or expressed independently in the kidney.
[11]
Shirali AC, Perazella MA, Gettinger S. Association of acute interstitial nephritis with programmed cell death 1 inhibitor therapy in lung cancer patients[J]. Am J Kidney Dis, 2016, 68(2): 287-291. DOI: 10.1053/j.ajkd.2016.02.057.
Immune checkpoint inhibitors that target the programmed death 1 (PD-1) signaling pathway have recently been approved for use in advanced pretreated non-small cell lung cancer and melanoma. Clinical trial data suggest that these drugs may have adverse effects on the kidney, but these effects have not been well described. We present 6 cases of acute kidney injury in patients with lung cancer who received anti-PD-1 antibodies, with each case displaying evidence of acute interstitial nephritis (AIN) on kidney biopsy. All patients were also treated with other drugs (proton pump inhibitors and nonsteroidal anti-inflammatory drugs) linked to AIN, but in most cases, use of these drugs long preceded PD-1 inhibitor therapy. The association of AIN with these drugs in our patients raises the possibility that PD-1 inhibitor therapy may release suppression of T-cell immunity that normally permits renal tolerance of drugs known to be associated with AIN. Copyright © 2016 National Kidney Foundation, Inc. Published by Elsevier Inc. All rights reserved.
[12]
Izzedine H, Mathian A, Champiat S, et al. Renal toxicities associated with pembrolizumab[J]. Clin Kidney J, 2019, 12(1): 81-88. DOI: 10.1093/ckj/sfy100.
Expanded clinical experience with patients treated by pembrolizumab has accumulated. However, renal toxicities associated with this anti-programmed cell death 1 agent are poorly described because kidney histology is rarely sought. As a nephrology referral centre, we aimed to describe the clinic-biological and histopathological characteristics of pembrolizumab-related nephropathy and its response to treatment.We conducted a monocentric large case series study, including all pembrolizumab-treated cancer patients presenting a renal toxicity addressed to our centre from 2015 to 2017.A total of 12 patients (7 men) out of 676 pembrolizumab-treated patients (incidence 1.77%) were included (median age 69.75 years). Patients were referred for acute kidney injury ( = 10) and/or proteinuria ( = 2). A kidney biopsy was performed in all patients, with a median duration of use of 9 months (range 1-24 months) after the beginning of treatment. Biopsy showed that four patients had acute interstitial nephritis (AIN), whereas five had acute tubular injury (ATI) alone, one had minimal change disease (MCD) and ATI, and one had MCD alone. Pembrolizumab withdrawal coupled with corticosteroid therapy was the most effective treatment for kidney function recovery. Drug reintroduction resulted in a more severe recurrence of AIN in one patient who required maintenance of pembrolizumab. Two patients died of cancer progression with one of them developing severe renal failure requiring dialysis.In our series, ATI, AIN and MCD are the most frequent forms of kidney involvement under pembrolizumab therapy. Kidney dysfunction is usually isolated but can be severe. Use of corticosteroids in case of AIN improves the glomerular filtration rate.
[13]
Wang R, Das T, Takou A. IgA nephropathy after pembrolizumab therapy for mesothelioma[J]. BMJ Case Rep, 2020, 13(11): e237008. DOI: 10.1136/bcr-2020-237008.
[14]
Kitchlu A, Fingrut W, Avila-Casado C, et al. Nephrotic syndrome with cancer immunotherapies: a report of 2 cases[J]. Am J Kidney Dis, 2017, 70(4): 581-585. DOI: 10.1053/j.ajkd.2017.04.026.
[15]
Manohar S, Ghamrawi R, Chengappa M, et al. Acute interstitial nephritis and checkpoint inhibitor therapy: single center experience of management and drug rechallenge[J]. Kidney360, 2020, 1(1): 16-24. DOI: 10.34067/KID.0000152019.
The objective of this case cohort study was to describe our experience in the care of patients with immune checkpoint inhibitor–related acute interstitial nephritis (ICI-AIN) including rechallenge.
[16]
El Bitar S, Weerasinghe C, El-Charabaty E, et al. Renal tubular acidosis an adverse effect of PD-1 inhibitor immunotherapy[J]. Case Rep Oncol Med, 2018, 2018: 8408015. DOI: 10.1155/2018/8408015.
[17]
Charmetant X, Teuma C, Lake J, et al. A new expression of immune checkpoint inhibitors′renal toxicity: when distal tubular acidosis precedes creatinine elevation[J]. Clin Kidney J, 2020, 13(1): 42-45. DOI: 10.1093/ckj/sfz051.
The main manifestation of acute interstitial nephritis (AIN) due to immune checkpoint inhibitors is acute kidney injury. We report here a biopsy-proven AIN revealed by tubular acidosis. This case highlights that immune checkpoint inhibitor prescribers must be aware of electrolytic disorders since tubular dysfunction can precede serum creatinine increase and reveal renal toxicity.© The Author(s) 2019. Published by Oxford University Press on behalf of ERA-EDTA.
[18]
Seethapathy H, Zhao S, Strohbehn IA, et al. Incidence and clinical features of immune-related acute kidney injury in patients receiving programmed cell death ligand-1 inhibitors[J]. Kidney Int Rep, 2020, 5(10): 1700-1705. DOI: 10.1016/j.ekir.2020.07.011.
Programmed cell death receptor ligand 1 (PD-L1) inhibitors are immune checkpoint inhibitors (ICIs) with a side effect profile that may differ from other classes of ICIs such as those directed against cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) and programmed cell death 1 receptor (PD-1). Being the more recently approved class of checkpoint inhibitors, there are no studies investigating the frequency, etiology and predictors of acute kidney injury (AKI) in patients receiving PD-L1 inhibitors.This was a retrospective cohort study of patients who received PD-L1 inhibitors during 2017 to 2018 in our healthcare system. AKI was defined by a ≥1.5-fold rise in serum creatinine from baseline. The etiology of all cases of sustained AKI (lasting >48 hours) and clinical course were determined by review of electronic health records.The final analysis included 599 patients. Within 12 months of ICI initiation, 104 patients (17%) experienced AKI, and 36 (6%) experienced sustained AKI; however, only 5 (<1%) experienced suspected PD-L1-related AKI. The PD-L1-related AKI occurred a median of 99 days after starting therapy. All patients concurrently received another medication known to cause acute interstitial nephritis (proton pump inhibitors, nonsteroidal anti-inflammatory drugs, or antibiotics) at the time of the suspected PDL1-related AKI.Although AKI is common in patients receiving PD-L1 therapy, the incidence of suspected PD-L1-related AKI is low (<1%) and may be less common when compared to other classes of ICIs. This cohort provides further validation that other drugs associated with acute interstitial nephritis may be involved in the pathogenesis of ICI-related AKI.© 2020 International Society of Nephrology. Published by Elsevier Inc.
[19]
Seethapathy H, Zhao S, Chute DF, et al. The incidence, causes, and risk factors of acute kidney injury in patients receiving immune checkpoint inhibitors[J]. Clin J Am Soc Nephrol, 2019, 14(12): 1692-1700. DOI: 10.2215/CJN.00990119.
Immune checkpoint inhibitor use in oncology is increasing rapidly. We sought to determine the frequency, severity, cause, and predictors of AKI in a real-world population receiving checkpoint inhibitors.
[20]
Cortazar FB, Kibbelaar ZA, Glezerman IG, et al. Clinical features and outcomes of immune checkpoint inhibitor-associated AKI: a multicenter study[J]. J Am Soc Nephrol, 2020, 31(2): 435-446. DOI: 10.1681/ASN.2019070676.
Despite increasing recognition of the importance of immune checkpoint inhibitor-associated AKI, data on this complication of immunotherapy are sparse.We conducted a multicenter study of 138 patients with immune checkpoint inhibitor-associated AKI, defined as a ≥2-fold increase in serum creatinine or new dialysis requirement directly attributed to an immune checkpoint inhibitor. We also collected data on 276 control patients who received these drugs but did not develop AKI.Lower baseline eGFR, proton pump inhibitor use, and combination immune checkpoint inhibitor therapy were each independently associated with an increased risk of immune checkpoint inhibitor-associated AKI. Median (interquartile range) time from immune checkpoint inhibitor initiation to AKI was 14 (6-37) weeks. Most patients had subnephrotic proteinuria, and approximately half had pyuria. Extrarenal immune-related adverse events occurred in 43% of patients; 69% were concurrently receiving a potential tubulointerstitial nephritis-causing medication. Tubulointerstitial nephritis was the dominant lesion in 93% of the 60 patients biopsied. Most patients (86%) were treated with steroids. Complete, partial, or no kidney recovery occurred in 40%, 45%, and 15% of patients, respectively. Concomitant extrarenal immune-related adverse events were associated with worse renal prognosis, whereas concomitant tubulointerstitial nephritis-causing medications and treatment with steroids were each associated with improved renal prognosis. Failure to achieve kidney recovery after immune checkpoint inhibitor-associated AKI was independently associated with higher mortality. Immune checkpoint inhibitor rechallenge occurred in 22% of patients, of whom 23% developed recurrent associated AKI.This multicenter study identifies insights into the risk factors, clinical features, histopathologic findings, and renal and overall outcomes in patients with immune checkpoint inhibitor-associated AKI.Copyright © 2020 by the American Society of Nephrology.
[21]
Wei SC, Levine JH, Cogdill AP, et al. Distinct cellular mechanisms underlie anti-CTLA-4 and anti-PD-1 checkpoint blockade[J]. Cell, 2017, 170(6): 1120-1133.e17. DOI: 10.1016/j.cell.2017.07.024.
Immune-checkpoint blockade is able to achieve durable responses in a subset of patients; however, we lack a satisfying comprehension of the underlying mechanisms of anti-CTLA-4- and anti-PD-1-induced tumor rejection. To address these issues, we utilized mass cytometry to comprehensively profile the effects of checkpoint blockade on tumor immune infiltrates in human melanoma and murine tumor models. These analyses reveal a spectrum of tumor-infiltrating T cell populations that are highly similar between tumor models and indicate that checkpoint blockade targets only specific subsets of tumor-infiltrating T cell populations. Anti-PD-1 predominantly induces the expansion of specific tumor-infiltrating exhausted-like CD8 T cell subsets. In contrast, anti-CTLA-4 induces the expansion of an ICOS Th1-like CD4 effector population in addition to engaging specific subsets of exhausted-like CD8 T cells. Thus, our findings indicate that anti-CTLA-4 and anti-PD-1 checkpoint-blockade-induced immune responses are driven by distinct cellular mechanisms.Copyright © 2017 Elsevier Inc. All rights reserved.
[22]
Liu K, Qin Z, Xu X, et al. Comparative risk of renal adverse events in patients receiving immune checkpoint inhibitors: a Bayesian network meta-analysis[J]. Front Oncol, 2021, 11: 662731. DOI: 10.3389/fonc.2021.662731.
Immune checkpoint inhibitors (ICIs) have brought a paradigm shift to cancer treatment. However, little is known about the risk of renal adverse events (RAEs) of ICI-based regimens, especially ICI combination therapy.
[23]
Isik B, Alexander MP, Manohar S, et al. Biomarkers, clinical features, and rechallenge for immune checkpoint inhibitor renal immune-related adverse events[J]. Kidney Int Rep, 2021, 6(4): 1022-1031. DOI: 10.1016/j.ekir.2021.01.013.
Immune checkpoint inhibitors (ICIs) are effective in treating several cancers; however, acute kidney injury (AKI) can occur as part as an immune-related adverse event (iRAE). Biomarkers at the time of AKI diagnosis may help determine whether they are ICI- related and guide therapeutic strategies.In this retrospective study, we reviewed patients with cancer treated with ICI therapy between 2014 and 2020 who developed AKI (defined as a ≥1.5-fold increase in serum creatinine [SCr]) that was attributed to ICI (ICI-AKI) and compared them with an adjudicated non-ICI-AKI group. Clinical and laboratory features, including SCr, serum C-reactive protein (CRP), and urine retinol binding protein/urine creatinine (uRBP/Cr) levels at AKI event were evaluated.There were 37 patients with ICI-AKI and 13 non-ICI-AKI referents in the cohort for analysis. At time of AKI, SCr, CRP, and uRBP/Cr were significantly higher in the ICI-AKI compared with the non-ICI-AKI patients (median [interquartile range (IQR)] SCr 2.0 [1.7, 2.9] vs. 1.5 [1.3, 1.6] mg/dl, serum CRP 54.0 [33.7, 90.0] vs. 3.5 [3.0, 7.9] mg/l, and uRBP/Cr 1927 [1174, 46,522] vs. 233 [127, 989] μg/g Cr, respectively,  < 0.05 for all). Compared with the referent group, time from ICI initiation to AKI was shorter in the ICI-AKI patients. Among the ICI-AKI group, complete renal recovery occurred in 39% of patients by 3 months; rechallenge occurred in 16 (43%) of patients, of whom 3 (19%) had recurrence of AKI.Our findings suggest that serum CRP and uRBP/Cr may help to differentiate AKI due to ICI from other causes.© 2021 International Society of Nephrology. Published by Elsevier Inc.
[24]
Leiter A, Carroll E, De Alwis S, et al. Metabolic disease and adverse events from immune checkpoint inhibitors[J]. Eur J Endocrinol, 2021, 184(6): 857-865. DOI: 10.1530/eje-20-1362.
Obese and overweight body mass index (BMI) categories have been associated with increased immune-related adverse events (irAEs) in patients with cancer receiving immune checkpoint inhibitors (ICIs); however, the impact of being overweight in conjunction with related metabolic syndrome-associated factors on irAEs have not been investigated. We aimed to evaluate the impact of overweight and obese BMI according to metabolic disease burden on the development of irAEs.We conducted a retrospective observational study of patients receiving ICIs at a cancer center. Our main study outcome was development of ≥grade 2 (moderate) irAEs. Our main predictor was weight/metabolic disease risk category: (1) normal weight (BMI 18.5-24.9 kg/m)/low metabolic risk (<2 metabolic diseases [diabetes, dyslipidemia, hypertension]), (2) normal weight/high metabolic risk (≥2 metabolic diseases), (3) overweight (BMI ≥25 kg/m)/low metabolic risk, and (4) overweight/high metabolic risk.Of 411 patients in our cohort, 374 were eligible for analysis. Overall, 111 (30%) patients developed ≥grade 2 irAEs. In Cox analysis, overweight/low metabolic risk was significantly associated with ≥grade 2 irAEs (hazard ratio [HR]: 2.0, 95% confidence interval [95% CI]: 1.2-3.4) when compared to normal weight/low metabolic risk, while overweight/high metabolic risk (HR: 1.3, 95% CI: 0.7-2.2) and normal weight/high metabolic risk (HR: 1.5, 95% CI: 0.7-3.0) were not.Overweight patients with fewer metabolic comorbidities were at increased risk for irAEs. This study provides an important insight that BMI should be evaluated in the context of associated metabolic comorbidities in assessing risk of irAE development and ICI immune response.
[25]
Wang Z, Aguilar EG, Luna JI, et al. Paradoxical effects of obesity on T cell function during tumor progression and PD-1 checkpoint blockade[J]. Nat Med, 2019, 25(1): 141-151. DOI: 10.1038/s41591-018-0221-5.
The recent successes of immunotherapy have shifted the paradigm in cancer treatment, but because only a percentage of patients are responsive to immunotherapy, it is imperative to identify factors impacting outcome. Obesity is reaching pandemic proportions and is a major risk factor for certain malignancies, but the impact of obesity on immune responses, in general and in cancer immunotherapy, is poorly understood. Here, we demonstrate, across multiple species and tumor models, that obesity results in increased immune aging, tumor progression and PD-1-mediated T cell dysfunction which is driven, at least in part, by leptin. However, obesity is also associated with increased efficacy of PD-1/PD-L1 blockade in both tumor-bearing mice and clinical cancer patients. These findings advance our understanding of obesity-induced immune dysfunction and its consequences in cancer and highlight obesity as a biomarker for some cancer immunotherapies. These data indicate a paradoxical impact of obesity on cancer. There is heightened immune dysfunction and tumor progression but also greater anti-tumor efficacy and survival after checkpoint blockade which directly targets some of the pathways activated in obesity.
[26]
Brahmer JR, Lacchetti C, Schneider BJ, et al. Management of immune-related adverse events in patients treated with immune checkpoint inhibitor therapy: American Society of Clinical Oncology Clinical Practice Guideline[J]. J Clin Oncol, 2018, 36(17): 1714-1768. DOI: 10.1200/JCO.2017.77.6385.
Purpose To increase awareness, outline strategies, and offer guidance on the recommended management of immune-related adverse events in patients treated with immune checkpoint inhibitor (ICPi) therapy. Methods A multidisciplinary, multi-organizational panel of experts in medical oncology, dermatology, gastroenterology, rheumatology, pulmonology, endocrinology, urology, neurology, hematology, emergency medicine, nursing, trialist, and advocacy was convened to develop the clinical practice guideline. Guideline development involved a systematic review of the literature and an informal consensus process. The systematic review focused on guidelines, systematic reviews and meta-analyses, randomized controlled trials, and case series published from 2000 through 2017. Results The systematic review identified 204 eligible publications. Much of the evidence consisted of systematic reviews of observational data, consensus guidelines, case series, and case reports. Due to the paucity of high-quality evidence on management of immune-related adverse events, recommendations are based on expert consensus. Recommendations Recommendations for specific organ system-based toxicity diagnosis and management are presented. While management varies according to organ system affected, in general, ICPi therapy should be continued with close monitoring for grade 1 toxicities, with the exception of some neurologic, hematologic, and cardiac toxicities. ICPi therapy may be suspended for most grade 2 toxicities, with consideration of resuming when symptoms revert to grade 1 or less. Corticosteroids may be administered. Grade 3 toxicities generally warrant suspension of ICPis and the initiation of high-dose corticosteroids (prednisone 1 to 2 mg/kg/d or methylprednisolone 1 to 2 mg/kg/d). Corticosteroids should be tapered over the course of at least 4 to 6 weeks. Some refractory cases may require infliximab or other immunosuppressive therapy. In general, permanent discontinuation of ICPis is recommended with grade 4 toxicities, with the exception of endocrinopathies that have been controlled by hormone replacement. Additional information is available at www.asco.org/supportive-care-guidelines and www.asco.org/guidelineswiki.
[27]
Perazella MA, Sprangers B. Checkpoint inhibitor therapy-associated acute kidney injury: time to move on to evidence-based recommendations[J]. Clin Kidney J, 2021, 14(5): 1301-1306. DOI: 10.1093/ckj/sfab052.
Immune checkpoint inhibitors (ICIs) have revolutionized cancer treatment since their introduction ∼15 years ago. However, these monoclonal antibodies are associated with immune-related adverse events that can also affect the kidney, resulting in acute kidney injury (AKI), which is most commonly due to acute tubulointerstitial nephritis (ATIN). Limited data are available on the true occurrence of ICI-associated AKI. Furthermore, evidence to guide the optimal management of ICI-associated AKI in clinical practice is lacking. In this issue, Oleas report a single-center study of patients with nonhematologic malignancies who received ICI treatment during a 14-month period, experienced AKI and underwent a kidney biopsy at the Vall d'Hebron University Hospital. Importantly, they demonstrate that only a minority of ICI-associated AKI patients was referred to the nephrology service and kidney biopsy was only performed in 6.4% of patients. Although the authors add to our knowledge about ICI-associated AKI, their article also highlights the need for the development of noninvasive diagnostic markers for ICI-associated ATIN, the establishment of treatment protocols for ICI-associated ATIN and recommendations for optimal ICI rechallenge in patients with previous ICI-associated AKI.© The Author(s) 2021. Published by Oxford University Press on behalf of ERA-EDTA.
[28]
Moledina DG, Wilson FP, Pober JS, et al. Urine TNF-α and IL-9 for clinical diagnosis of acute interstitial nephritis[J]. JCI Insight, 2019, 4(10): DOI: 10.1172/jci.insight.127456.
[29]
Diehl A, Yarchoan M, Hopkins A, et al. Relationships between lymphocyte counts and treatment-related toxicities and clinical responses in patients with solid tumors treated with PD-1 checkpoint inhibitors[J]. Oncotarget, 2017, 8(69): 114268-114280. DOI: 10.18632/oncotarget.23217.
[30]
Qualls D, Seethapathy H, Bates H, et al. Positron emission tomography as an adjuvant diagnostic test in the evaluation of checkpoint inhibitor-associated acute interstitial nephritis[J]. J Immunother Cancer, 2019, 7(1): 356. DOI: 10.1186/s40425-019-0820-9.
Acute interstitial nephritis is an immune-related adverse event that can occur in patients receiving immune checkpoint inhibitor therapy. Differentiating checkpoint inhibitor-associated acute interstitial nephritis from other causes of acute kidney injury in patients with cancer is challenging and can lead to diagnostic delays and/or unwarranted immunosuppression. In this case report, we assess the use of F-flourodeoxyglucose positron-emission tomography imaging as an alternative diagnostic modality in the evaluation of potential acute interstitial nephritis.A 55-year-old woman with metastatic vulvar melanoma underwent treatment with two cycles of ipilimumab plus nivolumab, followed by seven cycles of nivolumab combined with radiation therapy. During her treatment, she developed non-oliguric acute kidney injury to a creatinine of 4.5 mg/dL from a baseline of 0.5 mg/dL. A clinical diagnosis of acute interstitial nephritis was made, and steroids were initiated, with rapid improvement of her acute kidney injury. Retrospectively, four positron-emission tomography scans obtained for cancer staging purposes were reviewed. We found a markedly increased F-flourodeoxyglucose uptake in the renal cortex at the time acute interstitial nephritis was diagnosed compared to baseline. In three cases of acute kidney injury due to alternative causes there was no increase in F-flourodeoxyglucose uptake from baseline.To our knowledge, this is the first report describing increased F-flourodeoxyglucose uptake in the renal cortex in a patient with checkpoint inhibitor-associated acute interstitial nephritis. Our findings suggest that F-flourodeoxyglucose positron-emission tomography may be a valuable test for diagnosing immune-mediated nephritis, particularly in patients where timely kidney biopsy is not feasible.
[31]
Katagiri D, Masumoto S, Katsuma A, et al. Positron emission tomography combined with computed tomography (PET-CT) as a new diagnostic tool for acute tubulointerstitial nephritis (AIN) in oliguric or haemodialysed patients[J]. NDT PLUS, 2010, 3(2): 155-159. DOI: 10.1093/ndtplus/sfp188.
[32]
Perazella MA. Kidney biopsy should be performed to document the cause of immune checkpoint inhibitor-associated acute kidney injury: commentary[J]. Kidney360, 2020, 1(3): 166-168. DOI: 10.34067/KID.0001072019.
[33]
Zheng K, Qiu W, Wang H, et al. Clinical recommendations on diagnosis and treatment of immune checkpoint inhibitor-induced renal immune-related adverse events[J]. Thorac Cancer, 2020, 11(6): 1746-1751. DOI: 10.1111/1759-7714.13405.
Immune checkpoint inhibitors (ICIs) are nowadays widely used in clinical oncology treatment, and significantly improve the prognosis of cancer patients. However, overactivation of T cells and related signaling pathways caused by ICIs can also induce immune-related adverse effects (irAEs). Renal immune side-effects are relatively rare, but some are serious and fatal. Acute kidney injury (AKI), diagnosed mainly by percentage increases in serum creatinine (sCr), is the most common clinical manifestation, while acute tubulointerstitial nephritis (ATIN) is the main cause of ICI-related AKI. Urinalysis analysis and sediment evaluation, 24 hour urine protein and sCr are helpful in screening and monitoring renal irAEs. Multiple potential causes for AKI are involved during cancer therapy, and should be differentiated from the immune mechanisms of ICIs. Under these circumstances, a renal biopsy should be considered which is essential for clinical decision-making. Steroids are an effective treatment option for renal irAEs. Most patients who experience ICI-related ATIN achieve a partial or complete renal recovery with prompt diagnosis and treatment. Multidisciplinary collaborations of different specialists will improve the effectiveness and outcome in the management of ICI irAEs.© 2020 The Authors. Thoracic Cancer published by China Lung Oncology Group and John Wiley & Sons Australia, Ltd.
[34]
Horvat TZ, Adel NG, Dang TO, et al. Immune-related adverse events, need for systemic immunosuppression, and effects on survival and time to treatment failure in patients with melanoma treated with ipilimumab at Memorial Sloan Kettering Cancer Center[J]. J Clin Oncol, 2015, 33(28): 3193-3198. DOI: 10.1200/JCO.2015.60.8448.
Ipilimumab is a standard treatment for metastatic melanoma, but immune-related adverse events (irAEs) are common and can be severe. We reviewed our large, contemporary experience with ipilimumab treatment outside of clinical trials to determine the frequency of use of systemic corticosteroid or anti-tumor necrosis factor α (anti-TNFα) therapy and the effect of these therapies on overall survival (OS) and time to treatment failure (TTF).We reviewed retrospectively the medical records of patients with melanoma who had received treatment between April 2011 and July 2013 with ipilimumab at the standard dose of 3 mg/kg. We collected data on patient demographics, previous and subsequent treatments, number of ipilimumab doses, irAEs and how they were treated, and overall survival.Of the 298 patients, 254 (85%) experienced an irAE of any grade. Fifty-six patients (19%) discontinued therapy because of an irAE, most commonly diarrhea. Overall, 103 patients (35%) required systemic corticosteroid treatment for an irAE; 29 (10%) also required anti-TNFα therapy. Defining TTF as either starting a new treatment or death, estimated median TTF was 5.7 months. Twelve percent of patients experienced long-term disease control without receiving additional antimelanoma therapy. OS and TTF were not affected by the occurrence of irAEs or the need for systemic corticosteroids.IrAEs are common in patients treated with ipilimumab. In our experience, approximately one-third of ipilimumab-treated patients required systemic corticosteroids, and almost one-third of those required further immune suppression with anti-TNFα therapy. Practitioners and patients should be prepared to treat irAEs and should understand that such treatment does not affect OS or TTF.© 2015 by American Society of Clinical Oncology.
[35]
Taki T, Oda N, Fujioka Y, et al. Successful treatment of non-small-cell lung cancer with atezolizumab following tubulointerstitial nephritis due to pembrolizumab[J]. Intern Med, 2020, 59(13): 1639-1642. DOI: 10.2169/internalmedicine.4260-19.
[36]
Sise ME, Seethapathy H, Reynolds KL. Diagnosis and management of immune checkpoint inhibitor-associated renal toxicity: illustrative case and review[J]. Oncologist, 2019, 24(6): 735-742. DOI: 10.1634/theoncologist.2018-0764.
Immune checkpoint inhibitors (ICIs) are monoclonal antibodies directed at negative regulatory components on T cells, such as cytotoxic T lymphocyte-associated antigen 4, programmed cell death-1 (PD-1), and its ligand, programmed cell death ligand-1. ICIs initate antitumor immunity; however, these agents are associated with immune-related adverse events (irAEs) that may affect a variety of organs. Renal irAEs most commonly present with asymptomatic acute kidney injury (AKI), which is often detected by routine laboratory testing. The severity of AKI associated with irAEs ranges from mild (grade 1-2) to severe (grade 3-4). It is often challenging to diagnose because this group of patients often have multiple reasons to have AKI (dehydration, sepsis, or nephrotoxic medication exposure). We present an illustrative case of a 60-year-old man with metastatic melanoma who presented with AKI during treatment with nivolumab and review the literature to address frequently asked questions concerning the diagnosis and management of renal irAEs in patients with advanced cancer. Importantly, most patients will recover completely, and some may tolerate a rechallenge of ICI therapy, with prompt and effective treatment. KEY POINTS: Renal immune-related adverse events (irAEs) are less frequently reported than other irAEs; however, it is possible that available data underestimate their true incidence because of missed diagnoses and under-reporting. Although severe renal irAEs are more easily detected, smaller rises in creatinine may not be appreciated or may be attributed to other causes, because the differential diagnosis of acute kidney injury (AKI) in patients with cancer is broad.Baseline creatinine should be established prior to beginning immune checkpoint inhibitorss (ICIs), and it should be monitored with every cycle. If a patient develops AKI, the ICI should be held while the evaluation is pursued. A thorough workup of suspected renal irAEs must exclude other potential causes of AKI such as infection, dehydration, urinary tract obstruction, and nephrotoxin exposure.Acute kidney injury after ICI therapy does not appear to be more common in patients with baseline estimated glomerular filtration rate <60 mL per min per 1.73 m. One particular concern, however, is that those with baseline renal disease have less "renal reserve," and repeated AKI events may push a patient closer to end-stage renal disease. Thus, clinicians must exert caution when rechallenging patients with pre-existing renal disease with ICI therapy in the event of a prior AKI from ICI-related allergic interstitial nephritis.© AlphaMed Press 2019.
[37]
Perazella MA, Shirali AC. Immune checkpoint inhibitor nephrotoxicity: what do we know and what should we do?[J]. Kidney Int, 2020, 97(1): 62-74. DOI: 10.1016/j.kint.2019.07.022.
Immune checkpoint inhibitors have dramatically improved cancer therapy for many patients. These humanized monoclonal antibodies against various immune checkpoints (receptors and ligands) effectively treat a number of malignancies by unleashing the immune system to destroy cancer cells. These drugs are not excreted by the kidneys or liver, have a long half-life, and undergo receptor-mediated clearance. Although these agents have greatly improved the prognosis of many cancers, immune-related end organ injury is a complication that has come to light in clinical practice. Although less common than other organ involvement, kidney lesions resulting in acute kidney injury and/or proteinuria are being described. Acute tubulointerstitial nephritis is the most common lesion seen on kidney biopsy, while acute tubular injury and glomerular lesions occur less commonly. Clinical findings and laboratory tests are suboptimal in predicting the underlying renal lesion, making kidney biopsy necessary in the majority of cases to definitely diagnose the lesion and potentially guide therapy. Immune checkpoint inhibitor discontinuation and corticosteroid therapy are recommended for acute tubulointerstitial nephritis. Based on a handful of cases, re-exposure to these drugs in patients who previously developed acute tubulointerstitial nephritis has been mixed. Although it is unclear whether re-exposure is appropriate, it should perhaps be considered in patients with limited options. When this approach is taken, patients should be closely monitored for recurrence of acute kidney injury. Treatment of cancer in patients with a kidney transplant with immune checkpoint inhibitors risks the development of acute rejection in some patients and requires close surveillance.Copyright © 2019 International Society of Nephrology. Published by Elsevier Inc. All rights reserved.
[38]
Khwaja A. KDIGO clinical practice guidelines for acute kidney injury[J]. Nephron Clin Pract, 2012, 120(4): c179-c184. DOI: 10.1159/000339789.
PDF(20182 KB)

2228

Accesses

0

Citation

Detail

Sections
Recommended

/